1
|
Fujii Y, Asadi Z, Mehla K. Cathepsins: Emerging targets in the tumor ecosystem to overcome cancers. Semin Cancer Biol 2025; 112:150-166. [PMID: 40228591 DOI: 10.1016/j.semcancer.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Cathepsins, a group of lysosomal peptidases, have traditionally been recognized as tumor facilitators. Recent research, however, highlights their critical role in orchestrating cancer and the tumor microenvironment (TME). Primality, cathepsins degrade extracellular matrix, enabling cancer cells to invade and metastasize, while also promoting vascular endothelial infiltration and subsequent angiogenesis. Additionally, cathepsins boost fibroblast growth, thereby supporting tumor progression. More importantly, cathepsins are pivotal in modulating immune cells within the TME by regulating their recruitment, antigen processing and presentation, differentiation, and cell death, primarily contributing to immune suppression. Given their overexpression in tumors and elevated levels in the circulation of cancer patients, it is crucial to consider the systemic effects of cathepsins. Although the comprehensive role of cathepsins in cancer patients' bodies remains underexplored, they likely influence systemic immunity and inflammation, cellular metabolism, muscle wasting, and distant metastasis through their unique proteolytic functions. Notably, cathepsins also confer resistance to chemoradiotherapy by rewriting the cellular profile within the TME. In this context, promising results are emerging from studies combining cathepsin inhibitors with conventional therapies to suppress tumor development effectively. This review aims to decipher the cathepsin-driven networks within cancer cells and the TME, detailing their contribution to chemoradioresistance by reshaping both micro- and macroenvironments. Furthermore, we explore current and future perspectives on therapies targeting cathepsins' interactions, offering insights into innovative treatment strategies.
Collapse
Affiliation(s)
- Yuki Fujii
- Department of Oncology Science, University of Oklahoma Health Sciences, Oklahoma City, OK 73014, USA
| | - Zahra Asadi
- Department of Oncology Science, University of Oklahoma Health Sciences, Oklahoma City, OK 73014, USA; Department of Pathology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Kamiya Mehla
- Department of Oncology Science, University of Oklahoma Health Sciences, Oklahoma City, OK 73014, USA; Department of Microbiology and Immunology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA; Department of Pathology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA.
| |
Collapse
|
2
|
Su K, Tang M, Wu J, Ye N, Jiang X, Zhao M, Zhang R, Cai X, Zhang X, Li N, Peng J, Lin L, Wu W, Ye H. Mechanisms and therapeutic strategies for NLRP3 degradation via post-translational modifications in ubiquitin-proteasome and autophagy lysosomal pathway. Eur J Med Chem 2025; 289:117476. [PMID: 40056798 DOI: 10.1016/j.ejmech.2025.117476] [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: 12/13/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
The NLRP3 inflammasome is crucial for immune responses. However, its overactivation can lead to severe inflammatory diseases, underscoring its importance as a target for therapeutic intervention. Although numerous inhibitors targeting NLRP3 exist, regulating its degradation offers an alternative and promising strategy to suppress its activation. The degradation of NLRP3 is primarily mediated by the proteasomal and autophagic pathways. The review not only elaborates on the traditional concepts of ubiquitination and NLRP3 degradation but also investigates the important roles of indirect regulatory modifications, such as phosphorylation, acetylation, ubiquitin-like modifications, and palmitoylation-key post-translational modifications (PTMs) that influence NLRP3 degradation. Additionally, we also discuss the potential targets that may affect NLRP3 degradation during the proteasomal and autophagic pathways. By unraveling these complex regulatory mechanisms, the review aims to enhance the understanding of NLRP3 regulation and its implications for developing therapeutic strategies to combat inflammatory diseases.
Collapse
Affiliation(s)
- Kaiyue Su
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Wu
- Key Laboratory of Hydrodynamics (Ministry of Education), School of Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Neng Ye
- Scaled Manufacturing Center of Biological Products, Management Office of National Facility for Translational Medicine, West China Hospital, Sichuan University Chengdu 610041, China
| | - Xueqin Jiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Min Zhao
- Laboratory of Metabolomics and Drug-induced Liver Injury, Department of Gastroenterology & Hepatology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruijia Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoying Cai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinlu Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Peng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Lin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenshuang Wu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Haoyu Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Ferrari CR, Hannig M, Buzalaf MAR. Acquired pellicle engineering: a fascinating approach to prevent demineralization. J Appl Oral Sci 2025; 33:e20240359. [PMID: 40332163 PMCID: PMC12061453 DOI: 10.1590/1678-7757-2024-0359] [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] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 05/08/2025] Open
Abstract
The acquired enamel pellicle (AEP) consists of an organic, acellular, and bacteria-free film, formed in vivo as a result of biomolecules adsorption onto the tooth surface. It is composed of proteins, glycoproteins, lipids, phospholipids, and other macromolecules, such as carbohydrates. The AEP formation process is complex and can be divided into three stages: initiation, development, and maturation. The pellicle has two main layers: the globular and basal layers. The basal layer offers the most protection against demineralization, as the subsequent globular layer is weaker and less tenacious. The formation of the AEP can be influenced by various factors, such as the physicochemical properties of the teeth, location in the oral cavity, pathologies, and even the oral microbiota. With the advancement of "omics" techniques, it has been possible to observe the presence of acid-resistant proteins in the AEP, which allowed the development of the "acquired pellicle engineering" strategy. This strategy involves enriching and modifying the basal layer with acid-resistant proteins. Among these proteins, hemoglobin, statherin-derived peptide, and a protein derived from sugarcane stand out. The objective of this literature review is to provide a comprehensive overview of the AEP, detailing its composition, formation process, and protective functions. Additionally, the review aims to explore recent advances in the field of "acquired pellicle engineering," highlighting the acid-resistant proteins of the AEP and their potential applications in dentistry. Finally, the review intends to highlight the clinical implications of these findings and how they may contribute to the development of new strategies for the prevention and treatment of dental pathologies according to published studies.
Collapse
Affiliation(s)
- Carolina Ruis Ferrari
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Bauru, Brasil
| | - Matthias Hannig
- Saarland University, Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, Homburg, Germany
| | | |
Collapse
|
4
|
Sun Z, Chen H, Li C, Yang H, Ling J, Chang A, Zhao H, Zhuo X. Are cathepsins a risk factor for papillary thyroid carcinoma? A bidirectional two-sample mendelian randomization analysis. Eur Arch Otorhinolaryngol 2025; 282:2607-2615. [PMID: 39757267 DOI: 10.1007/s00405-024-09176-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is the most common type of endocrine tumor, and its incidence is on the rise. Observational studies have linked cathepsins, an endolysosomal cysteine protein hydrolase, to the malignant progression of several tumors, including PTC. However, the causal relationship between cathepsins and PTC remains unclear. The purpose of this study was to investigate the causal relationship between cathepsins and PTC using a bidirectional two-sample Mendelian randomization (MR) analysis. METHODS Publicly available databases were used to obtain data on cathepsins and PTCs. Single nucleotide polymorphisms were screened for instrumental variables. Causality was evaluated using five methods. Heterogeneity and sensitivity analyses were performed to evaluate the stability of the results. RESULTS The analysis revealed a significant association between cathepsin Z (CTSZ) and the risk of PTC (IVW, OR = 1.170, 95% CI: 1.035-1.102, P = 0.011). However, no association was found in the inverse analysis (IVW, OR = 1.006, 95% CI: 0.982-1.031, P = 0.612). The stability and reliability of the results of this study were indicated by both heterogeneity and sensitivity. CONCLUSIONS This study confirmed the association between CTSZ and an increased risk of PTC. This finding has important implications for clinical practice, as it may help to predict and screen for PTC at an early stage, as well as provide some guidance for therapeutic strategies against CTSZ.
Collapse
Affiliation(s)
- Zhen Sun
- Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Huarong Chen
- Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Changya Li
- Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Hao Yang
- People's Hospital of Qianxinan Prefecture, Guizhou Province, Xingyi, Guizhou, 562400, China
| | - Junjun Ling
- Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Aoshuang Chang
- Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Houyu Zhao
- Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China.
| | - Xianlu Zhuo
- Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China.
| |
Collapse
|
5
|
Podvin S, Florio J, Spencer B, Mante M, Guzman E, Arias C, Mosier C, Phan VV, Yoon MC, Almaliti J, O’Donoghue AJ, Gerwick WH, Rissman RA, Hook V. Activation of Cytosolic Cathepsin B Activity in the Brain by Traumatic Brain Injury and Inhibition by the Neutral pH Selective Inhibitor Probe Z-Arg-Lys-AOMK. ACS Chem Neurosci 2025; 16:1297-1308. [PMID: 40130579 PMCID: PMC11969537 DOI: 10.1021/acschemneuro.4c00577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/30/2025] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
Cathepsin B has been shown to contribute to deficits in traumatic brain injury (TBI), an important risk factor for Alzheimer's disease (AD). Cathepsin B is elevated in TBI and AD patients, as well as in animal models of these conditions. Knockout of the cathepsin B gene results in amelioration of TBI-induced motor dysfunction and improvement of AD memory deficit in mice. The mechanism of cathepsin B pathogenesis in these brain disorders has been hypothesized to involve its translocation to the cytosol from its normal lysosomal location. This study, therefore, evaluated brain cytosolic cathepsin B activity in the controlled cortical impact (CCI) mouse model of TBI. CCI-TBI resulted in motor deficits demonstrated by the rotarod assay, brain tissue lesions, and disorganization of the hippocampus. Significantly, CCI-TBI increased cytosolic cathepsin B activity in the brain cortex in the ipsilateral brain hemisphere that received the CCI-TBI injury, with a concomitant decrease in the lysosomal fraction. Cathepsin B activity was monitored using the substrate Z-Nle-Lys-Arg-AMC which specifically detects cathepsin B activity but not other cysteine proteases. The normal lysosomal distribution of cathepsin B was observed by its discrete localization in brain cortical cells. CCI-TBI resulted in a more diffuse cellular distribution of cathepsin B consistent with translocation to the cytosol. Further studies utilized the novel neutral pH-selective inhibitor, Z-Arg-Lys-AOMK, that specifically inhibits cathepsin B at neutral pH 7.2 of the cytosol but not at acidic pH 4.6 of lysosomes. Daily administration of Z-Arg-Lys-AOMK (ip), beginning 1 day before CCI-TBI, resulted in the reduction of the increased cytosolic cathepsin B activity induced by CCI-TBI. The inhibitor also reduced cathepsin B activities in homogenates of the brain cortex and hippocampus which were increased by CCI-TBI. Furthermore, the Z-Arg-Lys-AOMK inhibitor resulted in the reduction of motor function deficit resulting from CCI-TBI. These findings demonstrate the activation of cytosolic cathepsin B activity in CCI-TBI mouse brain injury.
Collapse
Affiliation(s)
- Sonia Podvin
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Jazmin Florio
- Department
of Physiology and Neuroscience, USC Alzheimer’s
Therapeutic Research Institute, 9880 Mesa Rim Road, San Diego, California 92121, United States
| | - Brian Spencer
- Department
of Physiology and Neuroscience, USC Alzheimer’s
Therapeutic Research Institute, 9880 Mesa Rim Road, San Diego, California 92121, United States
| | - Michael Mante
- Department
of Physiology and Neuroscience, USC Alzheimer’s
Therapeutic Research Institute, 9880 Mesa Rim Road, San Diego, California 92121, United States
| | - Estefani Guzman
- Department
of Physiology and Neuroscience, USC Alzheimer’s
Therapeutic Research Institute, 9880 Mesa Rim Road, San Diego, California 92121, United States
| | - Carlos Arias
- Department
of Physiology and Neuroscience, USC Alzheimer’s
Therapeutic Research Institute, 9880 Mesa Rim Road, San Diego, California 92121, United States
| | - Charles Mosier
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Von V. Phan
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, California 92093, United States
| | - Michael C. Yoon
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Jehad Almaliti
- Center
for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, La Jolla, California 92093, United States
- Department
Pharmaceutical Sciences, College of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Anthony J. O’Donoghue
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - William H. Gerwick
- Center
for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, La Jolla, California 92093, United States
| | - Robert A. Rissman
- Department
of Physiology and Neuroscience, USC Alzheimer’s
Therapeutic Research Institute, 9880 Mesa Rim Road, San Diego, California 92121, United States
| | - Vivian Hook
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
- Department
of Neurosciences and Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
6
|
Saadh MJ, Muhammad FA, Albadr RJ, Sanghvi G, Jyothi SR, Kundlas M, Joshi KK, Rakhmatullaev A, Taher WM, Alwan M, Jawad MJ, Ali Al-Nuaimi AM. Inflammasomes and Cardiovascular Disease: Linking Inflammation to Cardiovascular Pathophysiology. Scand J Immunol 2025; 101:e70020. [PMID: 40170223 DOI: 10.1111/sji.70020] [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: 03/15/2025] [Revised: 03/15/2025] [Accepted: 03/22/2025] [Indexed: 04/03/2025]
Abstract
Cardiovascular diseases (CVDs) remain a leading cause of global mortality, driven by risk factors such as dyslipidemia, hypertension and diabetes. Recent research has highlighted the critical role of inflammasomes, particularly the NLRP3 inflammasome, in the pathogenesis of various CVDs, including hypertension, atherosclerosis, myocardial infarction and heart failure. Inflammasomes are intracellular protein complexes that activate inflammatory responses through the production of pro-inflammatory cytokines such as IL-1β and IL-18, contributing to endothelial dysfunction, plaque formation and myocardial injury. This review provides a comprehensive overview of the structure, activation mechanisms and pathways of inflammasomes, with a focus on their involvement in cardiovascular pathology. Key activation pathways include ion fluxes (K+ efflux and Ca2+ signalling), endoplasmic reticulum (ER) stress, mitochondrial dysfunction and lysosomal destabilisation. The review also explores the therapeutic potential of targeting inflammasomes to mitigate inflammation and improve outcomes in CVDs. Emerging strategies include small-molecule inhibitors, biologics and RNA-based therapeutics, with a particular emphasis on NLRP3 inhibition. Additionally, the integration of artificial intelligence (AI) in cardiovascular research offers promising avenues for identifying novel biomarkers, predicting disease risk and developing personalised treatment strategies. Future research directions should focus on understanding the interactions between inflammasomes and other immune components, as well as genetic regulators, to uncover new therapeutic targets. By elucidating the complex role of inflammasomes in CVDs, this review underscores the potential for innovative therapies to address inflammation-driven cardiovascular pathology, ultimately improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology Faculty of Science, Marwadi University, Rajkot, Gujarat, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, Uttarakhand, India
- Graphic Era Deemed to Be University, Dehradun, Uttarakhand, India
| | - Akmal Rakhmatullaev
- Department of Faculty Pediatric Surgery, Tashkent Pediatric Medical Institute, Tashkent, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
7
|
Zhao Y, Zhuang Y, Shi J, Fan H, Lv Q, Guo X. Cathepsin B induces kidney diseases through different types of programmed cell death. Front Immunol 2025; 16:1535313. [PMID: 40129990 PMCID: PMC11930809 DOI: 10.3389/fimmu.2025.1535313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
Cathepsin B (CTSB), a key cysteine protease, plays essential roles in physiological and pathological processes. As research progresses, interest in how CTSB triggers different types of programmed cell death (PCD) to induce the onset and development of diseases is increasing. Several recent studies suggest that different types of PCD mediated by CTSB play key roles in kidney diseases. In this review, we outline the fundamental mechanisms by which CTSB triggers different types of PCD in several kidney diseases and discuss the function of CTSB in various segments of the kidney. Moreover, we explore the possibilities and prospects of using CTSB as a therapeutic target for kidney diseases.
Collapse
Affiliation(s)
- Yunlong Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Yong Zhuang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Jie Shi
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Qi Lv
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Xiaoqin Guo
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| |
Collapse
|
8
|
Hazem SH, Saad KM, Samaha MM. Protective effects of BTK inhibition by acalabrutinib on cisplatin-induced renal and testicular injury in mice: Modulation of mTOR/AMPK, NLRP3/GSDMD-N, and apoptotic pathways. Int Immunopharmacol 2025; 149:114256. [PMID: 39938312 DOI: 10.1016/j.intimp.2025.114256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/07/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Cisplatin-induced nephrotoxicity and testicular injury pose significant challenges during chemotherapy. AIM The current study evaluates the efficacy of acalabrutinib (ACB), a Bruton's tyrosine kinase inhibitor, in mitigating cisplatin-induced damage in renal and testicular tissues in mice. METHODS Testicular and renal toxicity was induced by a single I.P. injection of cisplatin (25 mg/kg). Mice were randomized into four groups: Normal (treated with vehicle), Cis (cisplatin + vehicle), Cis + ACB (6 mg/kg), and Cis + ACB (12 mg/kg). ACB was administered orally for three consecutive days, starting at Day 0 (1 h before single I.P. injection of cisplatin) and continued for Day 1 and Day 2. RESULTS ACB treatment (6 mg/kg and 12 mg/kg) significantly improved renal function by reducing serum creatinine, BUN, and KIM-1 levels, while also attenuating inflammation and apoptosis, as evidenced by decreased NLRP3, CD68, and caspase-3 expression. Additionally, it mitigated molecular damage by downregulating mTOR, AMPK, and GSDMD-N. In testicular tissues, ACB preserved structure, restored spermatogenesis, and improved sperm viability and testosterone levels. The protective effects were associated with reduced inflammation, apoptosis, and pyroptosis, indicated by lower levels of cathepsin L, NLRP3, and GSDMD-N. CONCLUSIONS These findings suggest that ACB offers a promising therapeutic approach to reduce the adverse effects of cisplatin, potentially enhancing the overall efficacy and safety of chemotherapy regimens.
Collapse
Affiliation(s)
- Sara H Hazem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516 Egypt.
| | - Karim M Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516 Egypt.
| | - Mahmoud M Samaha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516 Egypt.
| |
Collapse
|
9
|
Li S, Hu Z, Pan W, Wu H, Peng W, Wu Y, Jiang F, Peng X. Discovery of Highly Potent and Orally Bioavailable Histone Deacetylase 3 Inhibitors as Immunomodulators and Enhancers of DNA-Damage Response in Cancer Therapy. J Med Chem 2025; 68:3212-3237. [PMID: 39873221 DOI: 10.1021/acs.jmedchem.4c02445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Histone deacetylase 3 (HDAC3) is a well-established target for cancer therapy. Herein, we developed LSQ-28 as a novel HDAC3 inhibitor, which exhibited high HDAC3 inhibitory activity (IC50 = 42 nM, SI > 161) and displayed potent antiproliferative activity against four cancer cells and further demonstrated excellent antimigratory, anti-invasive, and antiwound healing activities. Further studies revealed that LSQ-28 induced a dose-dependent increase in Ac-H3 expression and promoted the degradation of PD-L1. Additionally, LSQ-28 enhanced the DNA damage response induced by PARP inhibitor, as evidenced by regulated expression of PARP1 and γ-H2AX. Notably, LSQ-28 also possessed favorable pharmacokinetic properties with significant oral bioavailability (F = 95.34%). Importantly, the combination of LSQ-28 with the PD-L1 inhibitor NP-19 could enhance antitumor immune response (TGI = 80%). When combined with olaparib, LSQ-28 significantly enhanced the in vivo tumor-suppression activity (TGI = 91%). Collectively, LSQ-28 represents a promising HDAC3 inhibitor for further exploration in cancer therapeutic strategies.
Collapse
Affiliation(s)
- Shuqing Li
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Zhihao Hu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Wanyi Pan
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Haiyan Wu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Weijie Peng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Yi Wu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Feng Jiang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Xiaopeng Peng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| |
Collapse
|
10
|
Wu X, Tian Y, Wang H, Chen H, Hou H, Hu Q. Dual Regulation of Nicotine on NLRP3 Inflammasome in Macrophages with the Involvement of Lysosomal Destabilization, ROS and α7nAChR. Inflammation 2025; 48:61-74. [PMID: 38717634 DOI: 10.1007/s10753-024-02036-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/23/2024] [Accepted: 04/23/2024] [Indexed: 02/09/2025]
Abstract
Nicotine, the primary alkaloid in tobacco products, has been shown to have immunoregulatory function in at least 20 diseases. The biological mechanism of action of nicotine immunoregulation is complex, resulting in an improvement of some disease states and exacerbation of others. Given the central role of the NLRP3 inflammasome in macrophages among multiple inflammatory diseases, this study examined how nicotine alters NLRP3 inflammasome activation in macrophages. NLRP3 inflammasome activation was examined mechanistically in the context of different nicotine dosages. We show NLRP3 inflammasome activation, apoptosis-associated speck-like protein (ASC) expression, caspase-1 activity and subsequent IL-1β secretion were positively correlated with nicotine in a dose-dependent relationship, and destabilization of lysosomes and ROS production were also involved. At high concentrations of nicotine surpassing 0.25 mM, NLRP3 inflammasome activity declined, along with increased expression of the anti-inflammatory Alpha7 nicotinic acetylcholine receptor (α7nAChR) and the inhibition of TLR4/NF-κB signaling. Consequently, high doses of nicotine also reduced ASC expression, caspase-1 activity and IL-1β secretion in macrophages. Collectively, these results suggest a dual regulatory function of nicotine on NLRP3 inflammasome activation in macrophages, that is involved with the pro-inflammatory effects of lysosomal destabilization and ROS production. We also show nicotine mediates anti-inflammatory effects by activating α7nAChR at high doses.
Collapse
Affiliation(s)
- Xiaqing Wu
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China
| | - Yushan Tian
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China
| | - Hongjuan Wang
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China
| | - Huan Chen
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China
| | - Hongwei Hou
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China.
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China.
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China.
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China.
| | - Qingyuan Hu
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China.
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China.
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China.
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China.
| |
Collapse
|
11
|
Durmus S, Gelisgen R, Hajiyeva R, Adrovic A, Yildiz M, Yucesan E, Barut K, Kasapcopur O, Uzun H. miR-21 and cathepsin B in familial Mediterranean fever: novel findings regarding their impact on disease severity. BMJ Paediatr Open 2025; 9:e003064. [PMID: 39779193 PMCID: PMC11749570 DOI: 10.1136/bmjpo-2024-003064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/22/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE The limited predictive effect of genotype on familial Mediterranean fever (FMF) phenotype suggests that epigenetic factors and alternative mechanisms that may cause IL-1β release could contribute to phenotypic heterogeneity. The objective of this study was to examine the role of IL-1β levels and miR-21-5p, cathepsin B and pyrin levels, which were identified as potential factors causing IL-1β release through the use of bioinformatics tools, in the pathogenesis of FMF and their relationship with disease severity. MATERIALS AND METHODS 50 paediatric patients with FMF and 40 healthy children were enrolled in this study. Patients were divided into subgroups according to Pras disease severity score. Serum miR-21-5p expression levels were assessed by qRT-PCR, while serum pyrin, IL-1β and cathepsin B levels were determined by ELISA. RESULTS Serum miR-21-5p was significantly downregulated in FMF patients compared with the control group (p<0.001), while serum pyrin, IL-1β and cathepsin B levels were markedly elevated (p<0.001 for each). Only miR-21-5p was negatively correlated with IL-1β (r=-0.855; p<0.001). In moderately severe FMF patients, miR-21-5p exhibited a statistically significant downregulation (p<0.001), whereas IL-1β and cathepsin B showed a statistically significant increase (p<0.001 and p<0.05, respectively). Furthermore, the Pras score showed a strong negative correlation (r=-0.738; p<0.001) with miR-21-5p levels. Multivariate logistic regression showed that in FMF, a one-unit decrease in miR-21 increased disease severity risk 6.76-fold, while a one-unit increase in cathepsin B raised it 1.71-fold. CONCLUSION This might be considered one of the mechanisms for subclinical inflammation in paediatric FMF patients through increased activation of cytokines via the downregulation of miR-21-5p. Our findings suggest that miR-21-5p and IL-1β play key roles in subclinical inflammation, and these molecules might be a potential therapeutic target.
Collapse
Affiliation(s)
- Sinem Durmus
- Medical Biochemistry, İzmir Katip Çelebi University Faculty of Medicine, Izmir, Türkiye
| | - Remise Gelisgen
- Medical Biochemistry, Istanbul University-Cerrahpasa Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | - Ramila Hajiyeva
- Medical Biochemistry, Istanbul University-Cerrahpasa Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | - Amra Adrovic
- Pediatric Rheumatology, Istanbul University-Cerrahpasa Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | - Mehmet Yildiz
- Pediatric Rheumatology, Istanbul University-Cerrahpasa Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | - Emrah Yucesan
- Neurogenetics, Istanbul University-Cerrahpasa Institute of Neurological Sciences, Istanbul, Türkiye
| | - Kenan Barut
- Pediatric Rheumatology, Istanbul University-Cerrahpasa Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | - Ozgur Kasapcopur
- Pediatric Rheumatology, Istanbul University-Cerrahpasa Cerrahpasa Faculty of Medicine, Istanbul, Türkiye
| | - Hafize Uzun
- Medical Biochemistry, Istanbul Atlas University Faculty of Medicine, Istanbul, Türkiye
| |
Collapse
|
12
|
Conesa-Bakkali R, Morillo-Huesca M, Martínez-Fábregas J. Non-Canonical, Extralysosomal Activities of Lysosomal Peptidases in Physiological and Pathological Conditions: New Clinical Opportunities for Cancer Therapy. Cells 2025; 14:68. [PMID: 39851495 PMCID: PMC11763575 DOI: 10.3390/cells14020068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Lysosomes are subcellular compartments characterised by an acidic pH, containing an ample variety of acid hydrolases involved in the recycling of biopolymers. Among these hydrolases, lysosomal proteases have merely been considered as end-destination proteases responsible for the digestion of waste proteins, trafficked to the lysosomal compartment through autophagy and endocytosis. However, recent reports have started to unravel specific roles for these proteases in the regulation of initially unexpected biological processes, both under physiological and pathological conditions. Furthermore, some lysosomal proteases are no longer restricted to the lysosomal compartment, as more novel non-canonical, extralysosomal targets are being identified. Currently, lysosomal proteases are accepted to play key functions in the extracellular milieu, attached to the plasma membrane and even in the cytosolic and nuclear compartments of the cell. Under physiological conditions, lysosomal proteases, through non-canonical, extralysosomal activities, have been linked to cell differentiation, regulation of gene expression, and cell division. Under pathological conditions, these proteases have been linked to cancer, mostly through their extralysosomal activities in the cytosol and nuclei of cells. In this review, we aim to provide a comprehensive summary of our current knowledge about the extralysosomal, non-canonical functions of lysosomal proteases, both under physiological and pathological conditions, with a particular interest in cancer, that could potentially offer new opportunities for clinical intervention.
Collapse
Affiliation(s)
- Ryan Conesa-Bakkali
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
| | - Macarena Morillo-Huesca
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
| | - Jonathan Martínez-Fábregas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Biología, Universidad de Sevilla, Avenida Reina Mercedes, 41012 Sevilla, Spain
| |
Collapse
|
13
|
Spector L, Subramanian N. Revealing the dance of NLRP3: spatiotemporal patterns in inflammasome activation. IMMUNOMETABOLISM (COBHAM, SURREY) 2025; 7:e00053. [PMID: 39816134 PMCID: PMC11731036 DOI: 10.1097/in9.0000000000000053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/01/2024] [Indexed: 01/18/2025]
Abstract
The nucleotide-binding domain, leucine-rich repeat, and pyrin domain containing-protein 3 (NLRP3) inflammasome is a multiprotein complex that plays a critical role in the innate immune response to both infections and sterile stressors. Dysregulated NLRP3 activation has been implicated in a variety of autoimmune and inflammatory diseases, including cryopyrin-associated periodic fever syndromes, diabetes, atherosclerosis, Alzheimer's disease, inflammatory bowel disease, and cancer. Consequently, fine-tuning NLRP3 activity holds significant therapeutic potential. Studies have implicated several organelles, including mitochondria, lysosomes, the endoplasmic reticulum (ER), the Golgi apparatus, endosomes, and the centrosome, in NLRP3 localization and inflammasome assembly. However, reports of conflict and many factors regulating interactions between NLRP3 and subcellular organelles remain unknown. This review synthesizes the current understanding of NLRP3 spatiotemporal dynamics, focusing on recent literature that elucidates the roles of subcellular localization and organelle stress in NLRP3 signaling and its crosstalk with other innate immune pathways converging at these organelles.
Collapse
Affiliation(s)
- Lauren Spector
- Institute for Systems Biology, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Naeha Subramanian
- Institute for Systems Biology, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
Zhao K, Sun Y, Zhong S, Luo JL. The multifaceted roles of cathepsins in immune and inflammatory responses: implications for cancer therapy, autoimmune diseases, and infectious diseases. Biomark Res 2024; 12:165. [PMID: 39736788 DOI: 10.1186/s40364-024-00711-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/19/2024] [Indexed: 01/01/2025] Open
Abstract
The cathepsin family comprises lysosomal proteases that play essential roles in various physiological processes, including protein degradation, antigen presentation, apoptosis, and tissue remodeling. Dysregulation of cathepsin activity has been linked to a variety of pathological conditions, such as cancer, autoimmune diseases, and neurodegenerative disorders. Understanding the functions of cathepsins is crucial for gaining insights into their roles in both health and disease, as well as for developing targeted therapeutic approaches. Emerging research underscores the significant involvement of cathepsins in immune cells, particularly T cells, macrophages, dendritic cells, and neutrophils, as well as their contribution to immune-related diseases. In this review, we systematically examine the impact of cathepsins on the immune system and their mechanistic roles in cancer, infectious diseases, autoimmune and neurodegenerative disorders, with the goal of identifying novel therapeutic strategies for these conditions.
Collapse
Affiliation(s)
- Kexin Zhao
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hengyang, Hunan, 421001, China
- MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hengyang, Hunan, 421001, China
| | - Yangqing Sun
- Department of Oncology, Hunan Provincial People's Hospital, Changsha, Hunan, 410005, China
| | - Shangwei Zhong
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hengyang, Hunan, 421001, China
- MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hengyang, Hunan, 421001, China
| | - Jun-Li Luo
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hengyang, Hunan, 421001, China.
- MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hengyang, Hunan, 421001, China.
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, USC, Hengyang, Hunan, 410008, China.
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hengyang, Hunan, 421001, China.
| |
Collapse
|
15
|
Rizzo GP, Sanches RC, Chavero C, Bianchi DS, Apuzzo E, Herrera SE, Agazzi ML, Cortez ML, Marmisollé WA, Keitelman IA, Trevani AS, Oliveira SC, Azzaroni O, Smaldini PL, Docena GH. Poly(allylamine)/tripolyphosphate nanocomplex coacervate as a NLRP3-dependent systemic adjuvant for vaccine development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601578. [PMID: 39005275 PMCID: PMC11244956 DOI: 10.1101/2024.07.01.601578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Nanotechnology plays a crucial role in vaccine development. It allows the design of functional nanoparticles (NPs) that can act both as antigen carriers and as adjuvants to enhance the immune response. The present study aims to evaluate complex coacervate-like NPs composed of poly(allylamine hydrochloride) (PAH) and tripolyphosphate (TPP) as a safe vehicle and adjuvant for systemic vaccines. We investigated the activation of different antigen-presenting cells (APCs) with NPs and their adjuvanticity in Balbc/c and different KO mice that were intraperitoneally immunized with NP-OVA. We found that NPs increased the expression of CD86 and MHCII and promoted the production and secretion of interleukin-1β (IL-1β) and IL-18 through the inflammasome NLRP3 when macrophages and dendritic cells were co-incubated with LPS and NPs. We evidenced an unconventional IL-1β release through the autophagosome pathway. The inhibition of autophagy with 3-methyladenine reduced the LPS/NPs-induced IL-1β secretion. Additionally, our findings showed that the systemic administration of mice with NP-OVA triggered a significant induction of serum OVA-specific IgG and IgG2a, an increased secretion of IFN-γ by spleen cells, and high frequencies of LT CD4 + IFN-γ + and LT CD8 + IFN-γ + . Our findings show that NPs promoted the inflammasome activation of innate cells with Th1-dependent adjuvant properties, making them valuable for formulating novel preventive or therapeutic vaccines for infectious and non-infectious diseases.
Collapse
|
16
|
Jangra J, Bajad NG, Singh R, Kumar A, Singh SK. Identification of novel potential cathepsin-B inhibitors through pharmacophore-based virtual screening, molecular docking, and dynamics simulation studies for the treatment of Alzheimer's disease. Mol Divers 2024; 28:4381-4401. [PMID: 38517648 DOI: 10.1007/s11030-024-10821-z] [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: 11/28/2023] [Accepted: 02/03/2024] [Indexed: 03/24/2024]
Abstract
Cathepsin B is a cysteine protease lysosomal enzyme involved in several physiological functions. Overexpression of the enzyme enhances its proteolytic activity and causes the breakdown of amyloid precursor protein (APP) into neurotoxic amyloid β (Aβ), a characteristic hallmark of Alzheimer's disease (AD). Therefore, inhibition of the enzyme is a crucial therapeutic aspect for treating the disease. Combined structure and ligand-based drug design strategies were employed in the current study to identify the novel potential cathepsin B inhibitors. Five different pharmacophore models were developed and used for the screening of the ZINC-15 database. The obtained hits were analyzed for the presence of duplicates, interfering PAINS moieties, and structural similarities based on Tanimoto's coefficient. The molecular docking study was performed to screen hits with better target binding affinity. The top seven hits were selected and were further evaluated based on their predicted ADME properties. The resulting best hits, ZINC827855702, ZINC123282431, and ZINC95386847, were finally subjected to molecular dynamics simulation studies to determine the stability of the protein-ligand complex during the run. ZINC123282431 was obtained as the virtual lead compound for cathepsin B inhibition and may be a promising novel anti-Alzheimer agent.
Collapse
Affiliation(s)
- Jatin Jangra
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Nilesh Gajanan Bajad
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ravi Singh
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India.
| |
Collapse
|
17
|
Zhou Y, Dong W, Wang L, Ren S, Wei W, Wu G. Cystatin C Attenuates Perihematomal Secondary Brain Injury by Inhibiting the Cathepsin B/NLRP3 Signaling Pathway in a Rat Model of Intracerebral Hemorrhage. Mol Neurobiol 2024; 61:9646-9662. [PMID: 38676809 DOI: 10.1007/s12035-024-04195-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
Secondary brain injury (SBI) is a noticeable contributor to the high mortality and morbidity rates associated with intracerebral hemorrhage (ICH), and effective treatment options remain limited. Cystatin C (CysC) emerges as a novel candidate for SBI intervention. The therapeutic effects and underlying mechanisms of CysC in mitigating SBI following ICH were explored in the current research. An in vivo ICH rat model was established by injecting autologous blood into the right caudate nucleus. Western blotting (WB) was utilized to assess the levels of CysC, cathepsin B (CTSB), and the NLRP3 inflammasome. Subsequently, the ICH rat model was treated with exogenous CysC supplementation or CysC knockdown plasmids. Various parameters, including Evans blue (EB) extravasation, brain water content, and neurological function in rats, were examined. RT-qPCR and WB were employed to determine the expression levels of CTSB and the NLRP3 inflammasome. The co-expression of CTSB, CysC, and NLRP3 inflammasome with GFAP, NeuN, and Iba1 was assessed through double-labeled immunofluorescence. The interaction between CysC and CTSB was investigated using double-labeled immunofluorescence and co-immunoprecipitation. The findings revealed an elevation of CysC expression level, particularly at 24 h after ICH. Exogenous CysC supplementation alleviated severe brain edema, neurological deficit scores, and EB extravasation induced by ICH. Conversely, CysC knockdown produced opposite effects. The expression levels of CTSB and the NLRP3 inflammasome were significantly risen following ICH, and exogenous CysC supplement attenuated their expression levels. Double-labeled immunofluorescence illustrated that CysC, CTSB, and the NLRP3 inflammasome were predominantly expressed in microglial cells, and the interaction between CysC and CTSB was evidenced. CysC exhibited potential in ameliorating SBI following ICH via effectively suppressing the activation of the NLRP3 inflammasome mediated by CTSB specifically in microglial cells. These findings underscore the prospective therapeutic efficacy of CysC in the treatment of ICH-induced complications.
Collapse
Affiliation(s)
- Yongfang Zhou
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Wentao Dong
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Likun Wang
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Siying Ren
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Weiqing Wei
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Guofeng Wu
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
18
|
Surve D, Fish A, Debnath M, Pinjari A, Lorenzana A, Piya S, Peyton S, Kulkarni A. Sprayable inflammasome-inhibiting lipid nanorods in a polymeric scaffold for psoriasis therapy. Nat Commun 2024; 15:9035. [PMID: 39426974 PMCID: PMC11490495 DOI: 10.1038/s41467-024-53396-x] [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: 03/03/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024] Open
Abstract
Localized delivery of inflammasome inhibitors in phagocytic macrophages could be promising for psoriasis treatment. The present work demonstrates the development of non-spherical lipid nanoparticles, mimicking pathogen-like shapes, consisting of an anti-inflammatory inflammasome inhibiting lipid (pyridoxine dipalmitate) as a trojan horse. The nanorods inhibit inflammasome by 3.8- and 4.5-fold compared with nanoellipses and nanospheres, respectively. Nanorods reduce apoptosis-associated speck-like protein and lysosomal rupture, restrain calcium influx, and mitochondrial reactive oxygen species. Dual inflammasome inhibitor (NLRP3/AIM-2-IN-3) loaded nanorods cause synergistic inhibition by 21.5- and 59-folds compared with nanorods and free drug, respectively alongside caspase-1 inhibition. The NLRP3/AIM-2-IN-3 nanorod when transformed into a polymeric scaffold, simultaneously and effectively inhibits RNA levels of NLRP3, AIM2, caspase-1, chemokine ligand-2, gasdermin-D, interleukin-1β, toll-like receptor 7/ 8, and IL-17A by 6.4-, 1.6-, 2.0-, 13.0-, 4.2-, 24.4-, 4.3-, and 1.82-fold, respectively in psoriatic skin in comparison to Imiquimod positive control group in an in-vivo psoriasis-like mice model.
Collapse
Affiliation(s)
- Dhanashree Surve
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Adam Fish
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Maharshi Debnath
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Aniruddha Pinjari
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Adrian Lorenzana
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Sumi Piya
- Pathology Department, University of Massachusetts-Chan Medical School, Baystate Medical Center, Springfield, MA, 01199, USA
| | - Shelly Peyton
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA.
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
19
|
Henedak NT, El-Abhar HS, Soubh AA, Abdallah DM. NLRP3 Inflammasome: A central player in renal pathologies and nephropathy. Life Sci 2024; 351:122813. [PMID: 38857655 DOI: 10.1016/j.lfs.2024.122813] [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: 02/12/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
The cytoplasmic oligomer NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome has been implicated in most inflammatory and autoimmune diseases. Here, we highlight the significance of NLRP3 in diverse renal disorders, demonstrating its activation in macrophages and non-immune tubular epithelial and mesangial cells in response to various stimuli. This activation leads to the release of pro-inflammatory cytokines, contributing to the development of acute kidney injury (AKI), chronic renal injury, or fibrosis. In AKI, NLRP3 inflammasome activation and pyroptotic renal tubular cell death is driven by contrast and chemotherapeutic agents, sepsis, and rhabdomyolysis. Nevertheless, inflammasome is provoked in disorders such as crystal and diabetic nephropathy, obesity-related renal fibrosis, lupus nephritis, and hypertension-induced renal damage that induce chronic kidney injury and/or fibrosis. The mechanisms by which the inflammatory NLRP3/ Apoptosis-associated Speck-like protein containing a Caspase recruitment domain (ASC)/caspase-1/interleukin (IL)-1β & IL-18 pathway can turn on renal fibrosis is also comprehended. This review further outlines the involvement of dopamine and its associated G protein-coupled receptors (GPCRs), including D1-like (D1, D5) and D2-like (D2-D4) subtypes, in regulating this inflammation-linked renal dysfunction pathway. Hence, we identify D-related receptors as promising targets for renal disease management by inhibiting the functionality of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Nada T Henedak
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6(th) of October City, Giza, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt, Cairo 11835, Egypt
| | - Ayman A Soubh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6(th) of October City, Giza, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| |
Collapse
|
20
|
Bai S, Martin-Sanchez F, Brough D, Lopez-Castejon G. Pyroptosis leads to loss of centrosomal integrity in macrophages. Cell Death Discov 2024; 10:354. [PMID: 39117604 PMCID: PMC11310477 DOI: 10.1038/s41420-024-02093-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 08/10/2024] Open
Abstract
NLRP3 forms a multiprotein inflammasome complex to initiate the inflammatory response when macrophages sense infection or tissue damage, which leads to caspase-1 activation, maturation and release of the inflammatory cytokines interleukin-1β (IL-1β) and IL-18 and Gasdermin-D (GSDMD) mediated pyroptosis. NLRP3 inflammasome activity must be controlled as unregulated and chronic inflammation underlies inflammatory and autoimmune diseases. Several findings uncovered that NLRP3 inflammasome activity is under the regulation of centrosome localized proteins such as NEK7 and HDAC6, however, whether the centrosome composition or structure is altered during the inflammasome activation is not known. Our data show that levels of the centrosomal scaffold protein pericentrin (PCNT) are reduced upon NLRP3 inflammasome activation via different activators in human and murine macrophages. PCNT loss occurs in the presence of membrane stabilizer punicalagin, suggesting this is not a consequence of membrane rupture. We found that PCNT loss is dependent on NLRP3 and active caspases as MCC950 and pan caspase inhibitor ZVAD prevent its degradation. Moreover, caspase-1 and GSDMD are both required for this NLRP3-mediated PCNT loss because absence of caspase-1 or GSDMD triggers an alternative regulation of PCNT via its cleavage by caspase-3 in response to nigericin stimulation. PCNT degradation occurs in response to nigericin, but also other NLRP3 activators including lysomotropic agent L-Leucyl-L-Leucine methyl ester (LLOMe) and hypotonicity but not AIM2 activation. Our work reveals that the NLRP3 inflammasome activation alters centrosome composition highlighting the need to further understand the role of this organelle during inflammatory responses.
Collapse
Affiliation(s)
- Siyi Bai
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, M13 9PT, UK
| | - Fatima Martin-Sanchez
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, M13 9PT, UK
- Department of Pharmacology, Faculty of Medicine, University of Murcia, Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Faculty of Medicine, University of Murcia, 30120, Murcia, Spain
| | - David Brough
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, M13 9PT, UK
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK
| | - Gloria Lopez-Castejon
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK.
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
21
|
Lockwood TD. Coordination chemistry suggests that independently observed benefits of metformin and Zn 2+ against COVID-19 are not independent. Biometals 2024; 37:983-1022. [PMID: 38578560 PMCID: PMC11255062 DOI: 10.1007/s10534-024-00590-5] [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: 11/24/2023] [Accepted: 02/12/2024] [Indexed: 04/06/2024]
Abstract
Independent trials indicate that either oral Zn2+ or metformin can separately improve COVID-19 outcomes by approximately 40%. Coordination chemistry predicts a mechanistic relationship and therapeutic synergy. Zn2+ deficit is a known risk factor for both COVID-19 and non-infectious inflammation. Most dietary Zn2+ is not absorbed. Metformin is a naked ligand that presumably increases intestinal Zn2+ bioavailability and active absorption by cation transporters known to transport metformin. Intracellular Zn2+ provides a natural buffer of many protease reactions; the variable "set point" is determined by Zn2+ regulation or availability. A Zn2+-interactive protease network is suggested here. The two viral cysteine proteases are therapeutic targets against COVID-19. Viral and many host proteases are submaximally inhibited by exchangeable cell Zn2+. Inhibition of cysteine proteases can improve COVID-19 outcomes and non-infectious inflammation. Metformin reportedly enhances the natural moderating effect of Zn2+ on bioassayed proteome degradation. Firstly, the dissociable metformin-Zn2+ complex could be actively transported by intestinal cation transporters; thereby creating artificial pathways of absorption and increased body Zn2+ content. Secondly, metformin Zn2+ coordination can create a non-natural protease inhibitor independent of cell Zn2+ content. Moderation of peptidolytic reactions by either or both mechanisms could slow (a) viral multiplication (b) viral invasion and (c) the pathogenic host inflammatory response. These combined actions could allow development of acquired immunity to clear the infection before life-threatening inflammation. Nirmatrelvir (Paxlovid®) opposes COVID-19 by selective inhibition the viral main protease by a Zn2+-independent mechanism. Pending safety evaluation, predictable synergistic benefits of metformin and Zn2+, and perhaps metformin/Zn2+/Paxlovid® co-administration should be investigated.
Collapse
Affiliation(s)
- Thomas D Lockwood
- Department Pharmacology and Toxicology, School of Medicine, Wright State University, Dayton, OH, 45435, USA.
| |
Collapse
|
22
|
Hang W, Bu C, Cui Y, Chen K, Zhang D, Li H, Wang S. Research progress on the pathogenesis and prediction of pneumoconiosis among coal miners. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2024; 46:319. [PMID: 39012521 DOI: 10.1007/s10653-024-02114-z] [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: 04/13/2024] [Accepted: 07/02/2024] [Indexed: 07/17/2024]
Abstract
Pneumoconiosis is the most common occupational disease among coal miners, which is a lung disease caused by long-term inhalation of coal dust and retention in the lungs. The early stage of this disease is highly insidious, and pulmonary fibrosis may occur in the middle and late stages, leading to an increase in patient pain index and mortality rate. Currently, there is a lack of effective treatment methods. The pathogenesis of pneumoconiosis is complex and has many influencing factors. Although the characteristics of coal dust have been considered the main cause of different mechanisms of pneumoconiosis, the effects of coal dust composition, particle size and shape, and coal dust concentration on the pathogenesis of pneumoconiosis have not been systematically elucidated. Meanwhile, considering the irreversibility of pneumoconiosis progression, early prediction for pneumoconiosis patients is particularly important. However, there is no early prediction standard for pneumoconiosis among coal miners. This review summarizes the relevant research on the pathogenesis and prediction of pneumoconiosis in coal miners in recent years. Firstly, the pathogenesis of coal worker pneumoconiosis and silicosis was discussed, and the impact of coal dust characteristics on pneumoconiosis was analyzed. Then, the early diagnostic methods for pneumoconiosis have been systematically introduced, with a focus on image collaborative computer-aided diagnosis analysis and biomarker detection. Finally, the challenge of early screening technology for miners with pneumoconiosis was proposed.
Collapse
Affiliation(s)
- Wenlu Hang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu Province, People's Republic of China
| | - Chunlu Bu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu Province, People's Republic of China
| | - Yuming Cui
- School of Mechatronic Engineering, Jiangsu Normal University, Xuzhou, 221000, Jiangsu Province, People's Republic of China
| | - Kai Chen
- School of Materials Science and Physics, China University of Mining and Technology, Xuzhou, 221000, Jiangsu Province, People's Republic of China
| | - Dekun Zhang
- School of Materials Science and Physics, China University of Mining and Technology, Xuzhou, 221000, Jiangsu Province, People's Republic of China
| | - Haiquan Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu Province, People's Republic of China.
- School of Chemical Engineering & Technology, China University of Mining and Technology, Xuzhou, 221000, Jiangsu Province, People's Republic of China.
| | - Songquan Wang
- School of Mechatronic Engineering, Jiangsu Normal University, Xuzhou, 221000, Jiangsu Province, People's Republic of China.
| |
Collapse
|
23
|
Kang H, Choi SW, Kim JY, Oh SJ, Kim SJ, Lee MS. ER-to-lysosome Ca 2+ refilling followed by K + efflux-coupled store-operated Ca 2+ entry in inflammasome activation and metabolic inflammation. eLife 2024; 12:RP87561. [PMID: 38953285 PMCID: PMC11219040 DOI: 10.7554/elife.87561] [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] [Indexed: 07/03/2024] Open
Abstract
We studied lysosomal Ca2+ in inflammasome. Lipopolysaccharide (LPS) + palmitic acid (PA) decreased lysosomal Ca2+ ([Ca2+]Lys) and increased [Ca2+]i through mitochondrial ROS, which was suppressed in Trpm2-KO macrophages. Inflammasome activation and metabolic inflammation in adipose tissue of high-fat diet (HFD)-fed mice were ameliorated by Trpm2 KO. ER→lysosome Ca2+ refilling occurred after lysosomal Ca2+ release whose blockade attenuated LPS + PA-induced inflammasome. Subsequently, store-operated Ca2+entry (SOCE) was activated whose inhibition suppressed inflammasome. SOCE was coupled with K+ efflux whose inhibition reduced ER Ca2+ content ([Ca2+]ER) and impaired [Ca2+]Lys recovery. LPS + PA activated KCa3.1 channel, a Ca2+-activated K+ channel. Inhibitors of KCa3.1 channel or Kcnn4 KO reduced [Ca2+]ER, attenuated increase of [Ca2+]i or inflammasome activation by LPS + PA, and ameliorated HFD-induced inflammasome or metabolic inflammation. Lysosomal Ca2+ release induced delayed JNK and ASC phosphorylation through CAMKII-ASK1. These results suggest a novel role of lysosomal Ca2+ release sustained by ER→lysosome Ca2+ refilling and K+ efflux through KCa3.1 channel in inflammasome activation and metabolic inflammation.
Collapse
Affiliation(s)
- Hyereen Kang
- Severance Biomedical Science Institute, Yonsei University College of MedicineSeoulRepublic of Korea
| | - Seong Woo Choi
- Department of Physiology and Ion Channel Disease Research Center, Dongguk University College of MedicineGyeongjuRepublic of Korea
| | - Joo Young Kim
- Department of Pharmacology and Brain Korea 21 Project for Medical Sciences, Yonsei University College of MedicineSeoulRepublic of Korea
| | - Soo-Jin Oh
- Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang University College of MedicineCheonanRepublic of Korea
| | - Sung Joon Kim
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Myung-Shik Lee
- Severance Biomedical Science Institute, Yonsei University College of MedicineSeoulRepublic of Korea
- Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang University College of MedicineCheonanRepublic of Korea
| |
Collapse
|
24
|
Nishioka ST, Snipper J, Lee J, Schapiro J, Zhang RZ, Abe H, Till A, Okumura CYM. Group A Streptococcus induces lysosomal dysfunction in THP-1 macrophages. Infect Immun 2024; 92:e0014124. [PMID: 38722166 PMCID: PMC11237432 DOI: 10.1128/iai.00141-24] [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: 04/01/2024] [Accepted: 04/11/2024] [Indexed: 06/12/2024] Open
Abstract
The human-specific bacterial pathogen group A Streptococcus (GAS) is a significant cause of morbidity and mortality. Macrophages are important to control GAS infection, but previous data indicate that GAS can persist in macrophages. In this study, we detail the molecular mechanisms by which GAS survives in THP-1 macrophages. Our fluorescence microscopy studies demonstrate that GAS is readily phagocytosed by macrophages, but persists within phagolysosomes. These phagolysosomes are not acidified, which is in agreement with our findings that GAS cannot survive in low pH environments. We find that the secreted pore-forming toxin Streptolysin O (SLO) perforates the phagolysosomal membrane, allowing leakage of not only protons but also large proteins including the lysosomal protease cathepsin B. Additionally, GAS recruits CD63/LAMP-3, which may contribute to lysosomal permeabilization, especially in the absence of SLO. Thus, although GAS does not inhibit fusion of the lysosome with the phagosome, it has multiple mechanisms to prevent proper phagolysosome function, allowing for persistence of the bacteria within the macrophage. This has important implications for not only the initial response but also the overall functionality of the macrophages, which may lead to the resulting pathologies in GAS infection. Our data suggest that therapies aimed at improving macrophage function may positively impact patient outcomes in GAS infection.
Collapse
Affiliation(s)
- Scott T. Nishioka
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Joshua Snipper
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Jimin Lee
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Joshua Schapiro
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Robert Z. Zhang
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Hyewon Abe
- Biology Department, Occidental College, Los Angeles, California, USA
| | - Andreas Till
- Division of Biological Sciences, University of California San Diego, La Jolla, California, USA
- The San Diego Center for Systems Biology, University of California San Diego, La Jolla, California, USA
- University Hospital of Bonn, Bonn, Germany
| | | |
Collapse
|
25
|
Ri-Wen, Yang YH, Zhang TN, Liu CF, Yang N. Targeting epigenetic and post-translational modifications regulating pyroptosis for the treatment of inflammatory diseases. Pharmacol Res 2024; 203:107182. [PMID: 38614373 DOI: 10.1016/j.phrs.2024.107182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Inflammatory diseases, including infectious diseases, diabetes-related diseases, arthritis-related diseases, neurological diseases, digestive diseases, and tumor, continue to threaten human health and impose a significant financial burden despite advancements in clinical treatment. Pyroptosis, a pro-inflammatory programmed cell death pathway, plays an important role in the regulation of inflammation. Moderate pyroptosis contributes to the activation of native immunity, whereas excessive pyroptosis is associated with the occurrence and progression of inflammation. Pyroptosis is complicated and tightly controlled by various factors. Accumulating evidence has confirmed that epigenetic modifications and post-translational modifications (PTMs) play vital roles in the regulation of pyroptosis. Epigenetic modifications, which include DNA methylation and histone modifications (such as methylation and acetylation), and post-translational modifications (such as ubiquitination, phosphorylation, and acetylation) precisely manipulate gene expression and protein functions at the transcriptional and post-translational levels, respectively. In this review, we summarize the major pathways of pyroptosis and focus on the regulatory roles and mechanisms of epigenetic and post-translational modifications of pyroptotic components. We also illustrate these within pyroptosis-associated inflammatory diseases. In addition, we discuss the effects of novel therapeutic strategies targeting epigenetic and post-translational modifications on pyroptosis, and provide prospective insight into the regulation of pyroptosis for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Ri-Wen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yu-Hang Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Tie-Ning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Chun-Feng Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Ni Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
26
|
Zhu L, Guo L, Xu J, Xiang Q, Tan Y, Tian F, Du X, Zhang S, Wen T, Liu L. Postprandial Triglyceride-Rich Lipoproteins-Induced Lysosomal Dysfunction and Impaired Autophagic Flux Contribute to Inflammation in White Adipocytes. J Nutr 2024; 154:1619-1630. [PMID: 38008361 DOI: 10.1016/j.tjnut.2023.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Obesity and postprandial hypertriglyceridemia, characterized by an increase in triglyceride-rich lipoproteins (TRLs), cause chronic low-grade inflammation. It is unclear how postprandial TRLs affect inflammation in white adipocytes. OBJECTIVES The objectives of the study were to explore the inflammatory response of postprandial TRLs in white adipocytes and investigate the possible mechanism. METHODS We measured postprandial triglyceride (TG) and high-sensitivity C-reactive protein (hsCRP) concentrations in 204 recruited subjects and treated white adipocytes from mice with postprandial TRLs from above patients with hypertriglyceridemia. RESULTS Serum hsCRP concentrations and BMI were positively related to TG concentrations in the postprandial state. Postprandial TRLs increased mRNA and protein expression of inflammatory factors, including interleukin-1β, via the NOD-like receptor protein 3 (NLRP3)/Caspase-1 pathway, and impaired autophagy flux in white adipocytes of mice. TRLs also induced lysosomal damage as evidenced by the reduced protein expression of lysosome-associated membrane proteins-1 and Cathepsin L. Inhibition of Cathepsin B, NLRP3, and mTOR signaling improved autophagy/lysosome dysfunction and inhibited the activation of the NLRP3/Caspase-1 pathway and inflammatory factors induced by TRLs in white adipocytes. CONCLUSIONS Our results suggest that postprandial hypertriglyceridemia causes chronic inflammation in adipocytes through TRL-induced lysosomal dysfunction and impaired autophagic flux in an mTOR-dependent manner.
Collapse
Affiliation(s)
- Liyuan Zhu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Central South University, Changsha, Hunan, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan, PR China
| | - Liling Guo
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Central South University, Changsha, Hunan, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan, PR China
| | - Jin Xu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Central South University, Changsha, Hunan, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan, PR China
| | - Qunyan Xiang
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Central South University, Changsha, Hunan, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan, PR China
| | - Yangrong Tan
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Central South University, Changsha, Hunan, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan, PR China
| | - Feng Tian
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Central South University, Changsha, Hunan, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan, PR China
| | - Xiao Du
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Central South University, Changsha, Hunan, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan, PR China
| | - Shilan Zhang
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Central South University, Changsha, Hunan, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan, PR China; Department of Cardiovascular Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai, PR China
| | - Tie Wen
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Emergency Medicine and Difficult Diseases Institute, Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Ling Liu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, PR China; Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Central South University, Changsha, Hunan, PR China; Cardiovascular Disease Research Center of Hunan Province, Changsha, Hunan, PR China.
| |
Collapse
|
27
|
An Z, Ding W. Syntaxin17 Restores Lysosomal Function and Inhibits Pyroptosis Caused by Acinetobacter baumannii. J Microbiol 2024; 62:315-325. [PMID: 38451450 DOI: 10.1007/s12275-024-00109-0] [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: 09/08/2023] [Revised: 12/12/2023] [Accepted: 01/04/2024] [Indexed: 03/08/2024]
Abstract
Acinetobacter baumannii (A. baumannii) causes autophagy flux disorder by degrading STX17, resulting in a serious inflammatory response. It remains unclear whether STX17 can alter the inflammatory response process by controlling autolysosome function. This study aimed to explore the role of STX17 in the regulation of pyroptosis induced by A. baumannii. Our findings indicate that overexpression of STX17 enhances autophagosome degradation, increases LAMP1 expression, reduces Cathepsin B release, and improves lysosomal function. Conversely, knockdown of STX17 suppresses autophagosome degradation, reduces LAMP1 expression, augments Cathepsin B release, and accelerates lysosomal dysfunction. In instances of A. baumannii infection, overexpression of STX17 was found to improve lysosomal function and reduce the expression of mature of GSDMD and IL-1β, along with the release of LDH, thus inhibiting pyroptosis caused by A. baumannii. Conversely, knockdown of STX17 led to increased lysosomal dysfunction and further enhanced the expression of mature of GSDMD and IL-1β, and increased the release of LDH, exacerbating pyroptosis induced by A. baumannii. These findings suggest that STX17 regulates pyroptosis induced by A. baumannii by modulating lysosomal function.
Collapse
Affiliation(s)
- Zhiyuan An
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China.
| | - Wenyi Ding
- Department of Clinical Laboratory, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, 100730, People's Republic of China
| |
Collapse
|
28
|
Yang K, Jeltema D, Yan N. Innate immune sensing of macromolecule homeostasis. Adv Immunol 2024; 161:17-51. [PMID: 38763701 DOI: 10.1016/bs.ai.2024.03.004] [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] [Indexed: 05/21/2024]
Abstract
The innate immune system uses a distinct set of germline-encoded pattern recognition receptors to recognize molecular patterns initially thought to be unique to microbial invaders, named pathogen-associated molecular patterns. The concept was later further developed to include similar molecular patterns originating from host cells during tissue damage, known as damage-associated molecular patterns. However, recent advances in the mechanism of monogenic inflammatory diseases have highlighted a much more expansive repertoire of cellular functions that are monitored by innate immunity. Here, we summarize several examples in which an innate immune response is triggered when homeostasis of macromolecule in the cell is disrupted in non-infectious or sterile settings. These ever-growing sensing mechanisms expand the repertoire of innate immune recognition, positioning it not only as a key player in host defense but also as a gatekeeper of cellular homeostasis. Therapeutics inspired by these advances to restore cellular homeostasis and correct the immune system could have far-reaching implications.
Collapse
Affiliation(s)
- Kun Yang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Devon Jeltema
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Nan Yan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
29
|
Eriksson I, Öllinger K. Lysosomes in Cancer-At the Crossroad of Good and Evil. Cells 2024; 13:459. [PMID: 38474423 PMCID: PMC10930463 DOI: 10.3390/cells13050459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Although it has been known for decades that lysosomes are central for degradation and recycling in the cell, their pivotal role as nutrient sensing signaling hubs has recently become of central interest. Since lysosomes are highly dynamic and in constant change regarding content and intracellular position, fusion/fission events allow communication between organelles in the cell, as well as cell-to-cell communication via exocytosis of lysosomal content and release of extracellular vesicles. Lysosomes also mediate different forms of regulated cell death by permeabilization of the lysosomal membrane and release of their content to the cytosol. In cancer cells, lysosomal biogenesis and autophagy are increased to support the increased metabolism and allow growth even under nutrient- and oxygen-poor conditions. Tumor cells also induce exocytosis of lysosomal content to the extracellular space to promote invasion and metastasis. However, due to the enhanced lysosomal function, cancer cells are often more susceptible to lysosomal membrane permeabilization, providing an alternative strategy to induce cell death. This review summarizes the current knowledge of cancer-associated alterations in lysosomal structure and function and illustrates how lysosomal exocytosis and release of extracellular vesicles affect disease progression. We focus on functional differences depending on lysosomal localization and the regulation of intracellular transport, and lastly provide insight how new therapeutic strategies can exploit the power of the lysosome and improve cancer treatment.
Collapse
Affiliation(s)
- Ida Eriksson
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden;
| | | |
Collapse
|
30
|
Inoue E, Minatozaki S, Shimizu S, Miyamoto S, Jo M, Ni J, Tozaki-Saitoh H, Oda K, Nonaka S, Nakanishi H. Human β-Defensin 3 Inhibition of P. gingivalis LPS-Induced IL-1β Production by BV-2 Microglia through Suppression of Cathepsins B and L. Cells 2024; 13:283. [PMID: 38334675 PMCID: PMC10854704 DOI: 10.3390/cells13030283] [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: 12/25/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024] Open
Abstract
Cathepsin B (CatB) is thought to be essential for the induction of Porphyromonas gingivalis lipopolysaccharide (Pg LPS)-induced Alzheimer's disease-like pathologies in mice, including interleukin-1β (IL-1β) production and cognitive decline. However, little is known about the role of CatB in Pg virulence factor-induced IL-1β production by microglia. We first subjected IL-1β-luciferase reporter BV-2 microglia to inhibitors of Toll-like receptors (TLRs), IκB kinase, and the NLRP3 inflammasome following stimulation with Pg LPS and outer membrane vesicles (OMVs). To clarify the involvement of CatB, we used several known CatB inhibitors, including CA-074Me, ZRLR, and human β-defensin 3 (hBD3). IL-1β production in BV-2 microglia induced by Pg LPS and OMVs was significantly inhibited by the TLR2 inhibitor C29 and the IκB kinase inhibitor wedelolactonne, but not by the NLRPs inhibitor MCC950. Both hBD3 and CA-074Me significantly inhibited Pg LPS-induced IL-1β production in BV-2 microglia. Although CA-074Me also suppressed OMV-induced IL-1β production, hBD3 did not inhibit it. Furthermore, both hBD3 and CA-074Me significantly blocked Pg LPS-induced nuclear NF-κB p65 translocation and IκBα degradation. In contrast, hBD3 and CA-074Me did not block OMV-induced nuclear NF-κB p65 translocation or IκBα degradation. Furthermore, neither ZRLR, a specific CatB inhibitor, nor shRNA-mediated knockdown of CatB expression had any effect on Pg virulence factor-induced IL-1β production. Interestingly, phagocytosis of OMVs by BV-2 microglia induced IL-1β production. Finally, the structural models generated by AlphaFold indicated that hBD3 can bind to the substrate-binding pocket of CatB, and possibly CatL as well. These results suggest that Pg LPS induces CatB/CatL-dependent synthesis and processing of pro-IL-1β without activation of the NLRP3 inflammasome. In contrast, OMVs promote the synthesis and processing of pro-IL-1β through CatB/CatL-independent phagocytic mechanisms. Thus, hBD3 can improve the IL-1β-associated vicious inflammatory cycle induced by microglia through inhibition of CatB/CatL.
Collapse
Affiliation(s)
- Erika Inoue
- Faculty of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (E.I.); (S.M.); (S.S.); (S.M.); (M.J.)
| | - Shiyo Minatozaki
- Faculty of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (E.I.); (S.M.); (S.S.); (S.M.); (M.J.)
| | - Sachi Shimizu
- Faculty of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (E.I.); (S.M.); (S.S.); (S.M.); (M.J.)
| | - Sayaka Miyamoto
- Faculty of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (E.I.); (S.M.); (S.S.); (S.M.); (M.J.)
| | - Misato Jo
- Faculty of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (E.I.); (S.M.); (S.S.); (S.M.); (M.J.)
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China;
| | - Hidetoshi Tozaki-Saitoh
- Department of Pharmaceutical Sciences, School of Pharmacy at Fukuoka, International University of Health and Welfare, Okawa 831-8501, Japan;
| | - Kosuke Oda
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women’s University, Yasuhigashi, Hiroshima 731-0153, Japan; (K.O.); (S.N.)
| | - Saori Nonaka
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women’s University, Yasuhigashi, Hiroshima 731-0153, Japan; (K.O.); (S.N.)
| | - Hiroshi Nakanishi
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women’s University, Yasuhigashi, Hiroshima 731-0153, Japan; (K.O.); (S.N.)
| |
Collapse
|
31
|
Ziegler AR, Dufour A, Scott NE, Edgington-Mitchell LE. Ion Mobility-Based Enrichment-Free N-Terminomics Analysis Reveals Novel Legumain Substrates in Murine Spleen. Mol Cell Proteomics 2024; 23:100714. [PMID: 38199506 PMCID: PMC10862022 DOI: 10.1016/j.mcpro.2024.100714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Aberrant levels of the asparaginyl endopeptidase legumain have been linked to inflammation, neurodegeneration, and cancer, yet our understanding of this protease is incomplete. Systematic attempts to identify legumain substrates have been previously confined to in vitro studies, which fail to mirror physiological conditions and obscure biologically relevant cleavage events. Using high-field asymmetric waveform ion mobility spectrometry (FAIMS), we developed a streamlined approach for proteome and N-terminome analyses without the need for N-termini enrichment. Compared to unfractionated proteomic analysis, we demonstrate FAIMS fractionation improves N-termini identification by >2.5 fold, resulting in the identification of >2882 unique N-termini from limited sample amounts. In murine spleens, this approach identifies 6366 proteins and 2528 unique N-termini, with 235 cleavage events enriched in WT compared to legumain-deficient spleens. Among these, 119 neo-N-termini arose from asparaginyl endopeptidase activities, representing novel putative physiological legumain substrates. The direct cleavage of selected substrates by legumain was confirmed using in vitro assays, providing support for the existence of physiologically relevant extra-lysosomal legumain activity. Combined, these data shed critical light on the functions of legumain and demonstrate the utility of FAIMS as an accessible method to improve depth and quality of N-terminomics studies.
Collapse
Affiliation(s)
- Alexander R Ziegler
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Antoine Dufour
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - Nichollas E Scott
- Department of Microbiology and Immunology, Peter Doherty Institute, The University of Melbourne, Parkville, Victoria, Australia.
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
32
|
Gairola S, Sinha A, Kaundal RK. Linking NLRP3 inflammasome and pulmonary fibrosis: mechanistic insights and promising therapeutic avenues. Inflammopharmacology 2024; 32:287-305. [PMID: 37991660 DOI: 10.1007/s10787-023-01389-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/25/2023] [Indexed: 11/23/2023]
Abstract
Pulmonary fibrosis is a devastating disorder distinguished by redundant inflammation and matrix accumulation in the lung interstitium. The early inflammatory cascade coupled with recurring tissue injury orchestrates a set of events marked by perturbed matrix hemostasis, deposition of matrix proteins, and remodeling in lung tissue. Numerous investigations have corroborated a direct correlation between the NLR family pyrin domain-containing 3 (NLRP3) activation and the development of pulmonary fibrosis. Dysregulated activation of NLRP3 within the pulmonary microenvironment exacerbates inflammation and may incite fibrogenic responses. Nevertheless, the precise mechanisms through which the NLRP3 inflammasome elicits pro-fibrogenic responses remain inadequately defined. Contemporary findings suggest that the pro-fibrotic consequences stemming from NLRP3 signaling primarily hinge on the action of interleukin-1β (IL-1β). IL-1β instigates IL-1 receptor signaling, potentiating the activity of transforming growth factor-beta (TGF-β). This signaling cascade, in turn, exerts influence over various transcription factors, including SNAIL, TWIST, and zinc finger E-box-binding homeobox 1 (ZEB 1/2), which collectively foster myofibroblast activation and consequent lung fibrosis. Here, we have connected the dots to illustrate how the NLRP3 inflammasome orchestrates a multitude of signaling events, including the activation of transcription factors that facilitate myofibroblast activation and subsequent lung remodeling. In addition, we have highlighted the prominent role played by various cells in the formation of myofibroblasts, the primary culprit in lung fibrosis. We also provided a concise overview of various compounds that hold the potential to impede NLRP3 inflammasome signaling, thus offering a promising avenue for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Shobhit Gairola
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP, 226002, India
| | - Antarip Sinha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP, 226002, India
| | - Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP, 226002, India.
| |
Collapse
|
33
|
Caceres L, Abogunloko T, Malchow S, Ehret F, Merz J, Li X, Sol Mitre L, Magnani N, Tasat D, Mwinyella T, Spiga L, Suchanek D, Fischer L, Gorka O, Colin Gissler M, Hilgendorf I, Stachon P, Rog-Zielinska E, Groß O, Westermann D, Evelson P, Wolf D, Marchini T. Molecular mechanisms underlying NLRP3 inflammasome activation and IL-1β production in air pollution fine particulate matter (PM 2.5)-primed macrophages. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 341:122997. [PMID: 38000727 PMCID: PMC10804998 DOI: 10.1016/j.envpol.2023.122997] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023]
Abstract
Exposure to air pollution fine particulate matter (PM2.5) aggravates respiratory and cardiovascular diseases. It has been proposed that PM2.5 uptake by alveolar macrophages promotes local inflammation that ignites a systemic response, but precise underlying mechanisms remain unclear. Here, we demonstrate that PM2.5 phagocytosis leads to NLRP3 inflammasome activation and subsequent release of the pro-inflammatory master cytokine IL-1β. Inflammasome priming and assembly was time- and dose-dependent in inflammasome-reporter THP-1-ASC-GFP cells, and consistent across PM2.5 samples of variable chemical composition. While inflammasome activation was promoted by different PM2.5 surrogates, significant IL-1β release could only be observed after stimulation with transition-metal rich Residual Oil Fly Ash (ROFA) particles. This effect was confirmed in primary human monocyte-derived macrophages and murine bone marrow-derived macrophages (BMDMs), and by confocal imaging of inflammasome-reporter ASC-Citrine BMDMs. IL-1β release by ROFA was dependent on the NLRP3 inflammasome, as indicated by lack of IL-1β production in ROFA-exposed NLRP3-deficient (Nlrp3-/-) BMDMs, and by specific NLRP3 inhibition with the pharmacological compound MCC950. In addition, while ROFA promoted the upregulation of pro-inflammatory gene expression and cytokines release, MCC950 reduced TNF-α, IL-6, and CCL2 production. Furthermore, inhibition of TNF-α with a neutralizing antibody decreased IL-1β release in ROFA-exposed BMDMs. Using electron tomography, ROFA particles were observed inside intracellular vesicles and mitochondria, which showed signs of ultrastructural damage. Mechanistically, we identified lysosomal rupture, K+ efflux, and impaired mitochondrial function as important prerequisites for ROFA-mediated IL-1β release. Interestingly, specific inhibition of superoxide anion production (O2•-) from mitochondrial respiratory Complex I, but not III, blunted IL-1β release in ROFA-exposed BMDMs. Our findings unravel the mechanism by which PM2.5 promotes IL-1β release in macrophages and provide a novel link between innate immune response and exposure to air pollution PM2.5.
Collapse
Affiliation(s)
- Lourdes Caceres
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| | - Tijani Abogunloko
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| | - Sara Malchow
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Fabienne Ehret
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Faculty of Biology, University of Freiburg, 79104, Freiburg im Breisgau, Germany
| | - Julian Merz
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Xiaowei Li
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Lucia Sol Mitre
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| | - Natalia Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| | - Deborah Tasat
- Universidad Nacional de General San Martín, Escuela de Ciencia y Tecnología, B1650, General San Martín, Argentina
| | - Timothy Mwinyella
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Lisa Spiga
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Dymphie Suchanek
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Larissa Fischer
- Faculty of Biology, University of Freiburg, 79104, Freiburg im Breisgau, Germany; Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Mark Colin Gissler
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Peter Stachon
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Eva Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| | - Dennis Wolf
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany.
| | - Timoteo Marchini
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| |
Collapse
|
34
|
Ren Y, Wu H, Tan M, Chen J, Duan Z, Zhu B, Ruan X, Yu Q, Li S, Liu X, Liu Y, Si Y. Acetylation of MOB1 mediates polyphyllin II-reduced lysosome biogenesis in breast cancer by promoting the cytoplasmic retention of the YAP/TFEB coactivator complex. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155152. [PMID: 37922793 DOI: 10.1016/j.phymed.2023.155152] [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: 06/20/2023] [Revised: 09/27/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Autophagy‒lysosome abnormalities are associated with the malignant progression of cancer. Transcription factor EB (TFEB) is the master transcriptional regulator of the autophagy‒lysosome machinery, and its abnormal activity is associated with autophagy-lysosome dysfunction. Polyphyllin II (PPII), an active steroidal saponin isolated from the rhizomes of Paris polyphylla, has been demonstrated to have antitumor activity. PURPOSE Here, we explored the antitumor activity of PPII in breast cancer (BC) and further clarified its mechanism. METHODS Autophagosome was detected by transmission electron microscopy, an autophagy indicator system, and western blot. The effect of PPII on lysosomal activity was evaluated by flow cytometry, a lysosomal cathepsin activity assay, and acridine orange staining. The effect of PPII on the signaling pathway was evaluated by Western blot, gene expression measurement, gene alterations. The binding of PPII and MOB1 was examined through a drug affinity responsive target stability assay. The pharmacokinetic parameters of PPII were evaluated in Sprague-Dawley rats. RESULTS PPII exhibits therapeutic potential in BC by inducing the accumulation of autophagosome. PPII promotes the cytoplasmic retention of YAP/TFEB, which is responsible for the accumulation of autophagosome in BC. PPII activates Hippo signaling to promote cytoplasmic retention of YAP. PPII activates Hippo signaling by accelerating acetylation of MOB1 through a direct binding interaction. CONCLUSION Taken together, these results confirm that acetylation of MOB1 mediates PPII-induced autophagosome accumulation in BC by promoting cytoplasmic retention of the YAP/TFEB coactivator complex. PPII is expected to be a drug candidate for the treatment of BC based on lysosomal biosynthesis.
Collapse
Affiliation(s)
- Yuliang Ren
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Hui Wu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Miao Tan
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Junjie Chen
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Zhongqi Duan
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Bingxin Zhu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuzhi Ruan
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qingqing Yu
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shuzhen Li
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuewen Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Yuan Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
35
|
Zhang Z, Zhang Y, Zhang M, Yu C, Yang P, Xu M, Ling J, Wu Y, Zhu Z, Chen Y, Shi A, Liu X, Zhang J, Yu P, Zhang D. Food-derived peptides as novel therapeutic strategies for NLRP3 inflammasome-related diseases: a systematic review. Crit Rev Food Sci Nutr 2023; 65:1433-1464. [PMID: 38153262 DOI: 10.1080/10408398.2023.2294164] [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] [Indexed: 12/29/2023]
Abstract
NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3), a member of the nucleotide-binding domain (NOD) and leucine-rich repeat sequence (LRR) protein (NLR) family, plays an essential role in the inflammation initiation and inflammatory mediator secretion, and thus is also associated with many disease progressions. Food-derived bioactive peptides (FDBP) exhibit excellent anti-inflammatory activity in both in vivo and in vitro models. They are encrypted in plant, meat, and milk proteins and can be released under enzymatic hydrolysis or fermentation conditions, thereby hindering the progression of hyperuricemia, inflammatory bowel disease, chronic liver disease, neurological disorders, lung injury and periodontitis by inactivating the NLRP3. However, there is a lack of systematic review around FDBP, NLRP3, and NLRP3-related diseases. Therefore, this review summarized FDBP that exert inhibiting effects on NLRP3 inflammasome from different protein sources and detailed their preparation and purification methods. Additionally, this paper also compiled the possible inhibitory mechanisms of FDBP on NLRP3 inflammasomes and its regulatory role in NLRP3 inflammasome-related diseases. Finally, the progress of cutting-edge technologies, including nanoparticle, computer-aided screening strategy and recombinant DNA technology, in the acquisition or encapsulation of NLRP3 inhibitory FDBP was discussed. This review provides a scientific basis for understanding the anti-inflammatory mechanism of FDBP through the regulation of the NLRP3 inflammasome and also provides guidance for the development of therapeutic adjuvants or functional foods enriched with these FDBP.
Collapse
Affiliation(s)
- Ziqi Zhang
- The Second Clinical Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi, China
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuan Zhang
- School of Public Health, Nanchang University, Jiangxi, China
| | - Meiying Zhang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Chenfeng Yu
- Huankui College, Nanchang University, Jiangxi, China
| | - Pingping Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Zicheng Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ao Shi
- School of Medicine, St. George University of London, London, UK
| | - Xiao Liu
- Cardiology Department, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Deju Zhang
- The Second Clinical Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi, China
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong
| |
Collapse
|
36
|
Amaral MP, Cardoso FD, de Farias IS, de Souza RQ, Matteucci KC, Torrecilhas AC, Bortoluci KR. NAIP/NLRC4 inflammasome participates in macrophage responses to Trypanosoma cruzi by a mechanism that relies on cathepsin-dependent caspase-1 cleavage. Front Immunol 2023; 14:1282856. [PMID: 38124741 PMCID: PMC10731265 DOI: 10.3389/fimmu.2023.1282856] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Inflammasomes are large protein complexes that, once activated, initiate inflammatory responses by activating the caspase-1 protease. They play pivotal roles in host defense against pathogens. The well-established role of NAIP/NLRC4 inflammasome in bacterial infections involves NAIP proteins functioning as sensors for their ligands. However, recent reports have indicated the involvement of NLRC4 in non-bacterial infections and sterile inflammation, even though the role of NAIP proteins and the exact molecular mechanisms underlying inflammasome activation in these contexts remain to be elucidated. In this study, we investigated the activation of the NAIP/NLRC4 inflammasome in response to Trypanosoma cruzi, the protozoan parasite responsible for causing Chagas disease. This parasite has been previously demonstrated to activate NLRP3 inflammasomes. Here we found that NAIP and NLRC4 proteins are also required for IL-1β and Nitric Oxide (NO) release in response to T. cruzi infection, with their absence rendering macrophages permissive to parasite replication. Moreover, Nlrc4 -/- and Nlrp3 -/- macrophages presented similar impaired responses to T. cruzi, underscoring the non-redundant roles played by these inflammasomes during infection. Notably, it was the live trypomastigotes rather than soluble antigens or extracellular vesicles (EVs) secreted by them, that activated inflammasomes in a cathepsins-dependent manner. The inhibition of cathepsins effectively abrogated caspase-1 cleavage, IL-1β and NO release, mirroring the phenotype observed in Nlrc4 -/-/Nlrp3 -/- double knockout macrophages. Collectively, our findings shed light on the pivotal role of the NAIP/NLRC4 inflammasome in macrophage responses to T. cruzi infection, providing new insights into its broader functions that extend beyond bacterial infections.
Collapse
Affiliation(s)
- Marcelo Pires Amaral
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Felipe Daniel Cardoso
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Ingrid Sancho de Farias
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Rafael Queiroz de Souza
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Kely Catarine Matteucci
- Plataforma de Medicina Translacional, Fundação Oswaldo Cruz (FIOCRUZ), Faculdade de Medicina de Ribeirão Preto (FMRP), Ribeirão Preto, SP, Brazil
| | - Ana Claudia Torrecilhas
- Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), Diadema, SP, Brazil
| | - Karina Ramalho Bortoluci
- Departamento de Farmacologia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| |
Collapse
|
37
|
Zhao S, Hu Q, Jiang H, Zhao Y, Wang Y, Feng C, Li X. Multi-omics analysis of oxidative stress and apoptosis in hepatopancreas cells induced by Polyascus gregaria parasitizing the Eriocheir sinensis. FISH & SHELLFISH IMMUNOLOGY 2023; 143:109180. [PMID: 37863124 DOI: 10.1016/j.fsi.2023.109180] [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: 08/11/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/22/2023]
Abstract
Polyascus gregaria, a parasitic barnacle, poses a significant threat to Eriocheir sinensis farms by inhibiting crab growth. However, the molecular and pathological mechanisms behind P. gregaria infection in the hepatopancreas of E. sinensis remain unclear. In this study, we investigated the impact and underlying mechanisms of P. gregaria infection on E. sinensis through analyzing the infected hepatopancreatic tissues by tandem mass tag technology and RNA-Seq high-throughput sequencing. Among the identified 10,693 differentially expressed genes, 294 genes were significantly altered following P. gregaria infection, including 92 upregulated and 202 downregulated genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses further revealed the involvement of these genes in oxidative decomposition, lipid metabolism, inflammation, and hepatopancreas metabolism. Meanwhile, the identified 253 differentially expressed proteins, including 143 upregulated and 110 downregulated proteins, are mainly related to cellular and metabolic processes, catalytic activity, and cell components. The pathway analysis indicated their enrichment in glycolysis/gluconeogenesis, oxidative phosphorylation, endoplasmic reticulum protein processing, and actin cytoskeleton regulation. The involvement of these differentially expressed genes and proteins in the peroxisome proliferator-activated receptors pathway during host immune responses against P. gregaria infection has been highlighted. Furthermore, pathological examinations and biochemical indicators jointly demonstrated the hepatopancreatic damage and increased oxidative stress and apoptosis in the infected E. sinensis. Collectively, our study provides crucial insights into the mechanisms underlying the E. sinensis-P. gregaria interactions, and may contribute to the development of novel strategies for parasite control and reducing economic losses in aquaculture.
Collapse
Affiliation(s)
- Shiwei Zhao
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Qingbiao Hu
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Hongbo Jiang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Yingying Zhao
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Yanping Wang
- Linong Testing (Binzhou) Co., Ltd., Binzhou Bohai Advanced Technology Research Institute, Binzhou, 256600, China
| | - Chengcheng Feng
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Xiaodong Li
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| |
Collapse
|
38
|
Quan JH, Gao FF, Ma TZ, Ye W, Gao X, Deng MZ, Yin LL, Choi IW, Yuk JM, Cha GH, Lee YH, Chu JQ. Toxoplasma gondii Induces Pyroptosis in Human Placental Trophoblast and Amniotic Cells by Inducing ROS Production and Activation of Cathepsin B and NLRP1/NLRP3/NLRC4/AIM2 Inflammasome. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:2047-2065. [PMID: 37741453 DOI: 10.1016/j.ajpath.2023.08.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/25/2023]
Abstract
Toxoplasma gondii infection in pregnant women may cause fetal anomalies; however, the underlying mechanisms remain unclear. The current study investigated whether T. gondii induces pyroptosis in human placental cells and the underlying mechanisms. Human placental trophoblast (BeWo and HTR-8/SVneo) and amniotic (WISH) cells were infected with T. gondii, and then reactive oxygen species (ROS) production, cathepsin B (CatB) release, inflammasome activation, and pyroptosis induction were evaluated. The molecular mechanisms of these effects were investigated by treating the cells with ROS scavengers, a CatB inhibitor, or inflammasome-specific siRNA. T. gondii infection induced ROS generation and CatB release into the cytosol in placental cells but decreased mitochondrial membrane potential. T. gondii-infected human placental cells and villi exhibited NLRP1, NLRP3, NLRC4, and AIM2 inflammasome activation and subsequent pyroptosis induction, as evidenced by increased expression of ASC, cleaved caspase-1, and mature IL-1β and gasdermin D cleavage. In addition to inflammasome activation and pyroptosis induction, adverse pregnancy outcome was shown in a T. gondii-infected pregnant mouse model. Administration of ROS scavengers, CatB inhibitor, or inflammasome-specific siRNA into T. gondii-infected cells reversed these effects. Collectively, these findings show that T. gondii induces NLRP1/NLRP3/NLRC4/AIM2 inflammasome-dependent caspase-1-mediated pyroptosis via induction of ROS production and CatB activation in placental cells. This mechanism may play an important role in inducing cell injury in congenital toxoplasmosis.
Collapse
Affiliation(s)
- Juan-Hua Quan
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Fei Fei Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Tian-Zhong Ma
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Wei Ye
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Xiang Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Ming-Zhu Deng
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Lan-Lan Yin
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - In-Wook Choi
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Jae-Min Yuk
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Guang-Ho Cha
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Young-Ha Lee
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
| | - Jia-Qi Chu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China.
| |
Collapse
|
39
|
Krantz M, Eklund D, Särndahl E, Hedbrant A. A detailed molecular network map and model of the NLRP3 inflammasome. Front Immunol 2023; 14:1233680. [PMID: 38077364 PMCID: PMC10699087 DOI: 10.3389/fimmu.2023.1233680] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 10/16/2023] [Indexed: 12/18/2023] Open
Abstract
The NLRP3 inflammasome is a key regulator of inflammation that responds to a broad range of stimuli. The exact mechanism of activation has not been determined, but there is a consensus on cellular potassium efflux as a major common denominator. Once NLRP3 is activated, it forms high-order complexes together with NEK7 that trigger aggregation of ASC into specks. Typically, there is only one speck per cell, consistent with the proposal that specks form - or end up at - the centrosome. ASC polymerisation in turn triggers caspase-1 activation, leading to maturation and release of IL-1β and pyroptosis, i.e., highly inflammatory cell death. Several gain-of-function mutations in the NLRP3 inflammasome have been suggested to induce spontaneous activation of NLRP3 and hence contribute to development and disease severity in numerous autoinflammatory and autoimmune diseases. Consequently, the NLRP3 inflammasome is of significant clinical interest, and recent attention has drastically improved our insight in the range of involved triggers and mechanisms of signal transduction. However, despite recent progress in knowledge, a clear and comprehensive overview of how these mechanisms interplay to shape the system level function is missing from the literature. Here, we provide such an overview as a resource to researchers working in or entering the field, as well as a computational model that allows for evaluating and explaining the function of the NLRP3 inflammasome system from the current molecular knowledge. We present a detailed reconstruction of the molecular network surrounding the NLRP3 inflammasome, which account for each specific reaction and the known regulatory constraints on each event as well as the mechanisms of drug action and impact of genetics when known. Furthermore, an executable model from this network reconstruction is generated with the aim to be used to explain NLRP3 activation from priming and activation to the maturation and release of IL-1β and IL-18. Finally, we test this detailed mechanistic model against data on the effect of different modes of inhibition of NLRP3 assembly. While the exact mechanisms of NLRP3 activation remains elusive, the literature indicates that the different stimuli converge on a single activation mechanism that is additionally controlled by distinct (positive or negative) priming and licensing events through covalent modifications of the NLRP3 molecule. Taken together, we present a compilation of the literature knowledge on the molecular mechanisms on NLRP3 activation, a detailed mechanistic model of NLRP3 activation, and explore the convergence of diverse NLRP3 activation stimuli into a single input mechanism.
Collapse
Affiliation(s)
- Marcus Krantz
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Daniel Eklund
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Eva Särndahl
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Alexander Hedbrant
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| |
Collapse
|
40
|
Stoka V, Vasiljeva O, Nakanishi H, Turk V. The Role of Cysteine Protease Cathepsins B, H, C, and X/Z in Neurodegenerative Diseases and Cancer. Int J Mol Sci 2023; 24:15613. [PMID: 37958596 PMCID: PMC10650516 DOI: 10.3390/ijms242115613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Papain-like cysteine proteases are composed of 11 human cysteine cathepsins, originally located in the lysosomes. They exhibit broad specificity and act as endopeptidases and/or exopeptidases. Among them, only cathepsins B, H, C, and X/Z exhibit exopeptidase activity. Recently, cysteine cathepsins have been found to be present outside the lysosomes and often participate in various pathological processes. Hence, they have been considered key signalling molecules. Their potentially hazardous proteolytic activities are tightly regulated. This review aims to discuss recent advances in understanding the structural aspects of these four cathepsins, mechanisms of their zymogen activation, regulation of their activities, and functional aspects of these enzymes in neurodegeneration and cancer. Neurodegenerative effects have been evaluated, particularly in Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and neuropsychiatric disorders. Cysteine cathepsins also participate in tumour progression and metastasis through the overexpression and secretion of proteases, which trigger extracellular matrix degradation. To our knowledge, this is the first review to provide an in-depth analysis regarding the roles of cysteine cathepsins B, H, C, and X in neurodegenerative diseases and cancer. Further advances in understanding the functions of cysteine cathepsins in these conditions will result in the development of novel, targeted therapeutic strategies.
Collapse
Affiliation(s)
- Veronika Stoka
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia;
- Jožef Stefan International Postgraduate School, SI-1000 Ljubljana, Slovenia
| | - Olga Vasiljeva
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia;
- CytomX Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Hiroshi Nakanishi
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan;
| | - Vito Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia;
- Jožef Stefan International Postgraduate School, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
41
|
Theofilis P, Oikonomou E, Chasikidis C, Tsioufis K, Tousoulis D. Inflammasomes in Atherosclerosis-From Pathophysiology to Treatment. Pharmaceuticals (Basel) 2023; 16:1211. [PMID: 37765019 PMCID: PMC10537692 DOI: 10.3390/ph16091211] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Atherosclerosis, a chronic inflammatory disease characterized by arterial plaque accumulation, remains a significant global health challenge. In recent years, inflammasomes, the intracellular multiprotein complexes crucial for initiating innate immune responses, have emerged as key players in atherosclerosis pathophysiology. This review article aims to provide a comprehensive overview of the current understanding of inflammasome activation and its impact on atherosclerosis development and progression. We explore the intricate interplay between traditional cardiovascular risk factors and inflammasome activation, leading to the perpetuation of inflammatory cascades that drive plaque formation and instability. The review focuses on the molecular mechanisms underlying inflammasome activation, including the role of pattern recognition receptors and cytokines in this process. Moreover, we discuss the contribution of inflammasomes to endothelial dysfunction, foam cell formation, and vascular inflammation. Additionally, recent advances in therapeutic strategies targeting inflammasomes are examined, including pharmacological agents and potential immunomodulatory approaches. By collating and analyzing the current evidence, this review provides valuable insights into the potential of inflammasome-targeted therapies for atherosclerosis management and treatment. Understanding the pivotal role of inflammasomes in atherosclerosis pathophysiology offers promising prospects for developing effective and personalized therapeutic interventions that can mitigate the burden of this prevalent cardiovascular disorder and improve patient outcomes.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Department of Cardiology, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.)
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Chasikidis
- Department of Cardiology, General Hospital of Corinth, 20100 Corinth, Greece
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.)
| | - Dimitris Tousoulis
- 1st Department of Cardiology, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.)
| |
Collapse
|
42
|
Yoon M, Phan V, Podvin S, Mosier C, O’Donoghue AJ, Hook V. Distinct Cleavage Properties of Cathepsin B Compared to Cysteine Cathepsins Enable the Design and Validation of a Specific Substrate for Cathepsin B over a Broad pH Range. Biochemistry 2023; 62:2289-2300. [PMID: 37459182 PMCID: PMC10399199 DOI: 10.1021/acs.biochem.3c00139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/11/2023] [Indexed: 08/02/2023]
Abstract
The biological and pathological functions of cathepsin B occur in acidic lysosomes and at the neutral pH of cytosol, nuclei, and extracellular locations. Importantly, cathepsin B displays different substrate cleavage properties at acidic pH compared to neutral pH conditions. It is, therefore, desirable to develop specific substrates for cathepsin B that measure its activity over broad pH ranges. Current substrates used to monitor cathepsin B activity consist of Z-Phe-Arg-AMC and Z-Arg-Arg-AMC, but they lack specificity since they are cleaved by other cysteine cathepsins. Furthermore, Z-Arg-Arg-AMC monitors cathepsin B activity at neutral pH and displays minimal activity at acidic pH. Therefore, the purpose of this study was to design and validate specific fluorogenic peptide substrates that can monitor cathepsin B activity over a broad pH range from acidic to neutral pH conditions. In-depth cleavage properties of cathepsin B were compared to those of the cysteine cathepsins K, L, S, V, and X via multiplex substrate profiling by mass spectrometry at pH 4.6 and pH 7.2. Analysis of the cleavage preferences predicted the tripeptide Z-Nle-Lys-Arg-AMC as a preferred substrate for cathepsin B. Significantly, Z-Nle-Lys-Arg-AMC displayed the advantageous properties of measuring high cathepsin B specific activity over acidic to neutral pHs and was specifically cleaved by cathepsin B over the other cysteine cathepsins. Z-Nle-Lys-Arg-AMC specifically monitored cathepsin B activity in neuronal and glial cells which were consistent with relative abundances of cathepsin B protein. These findings validate Z-Nle-Lys-Arg-AMC as a novel substrate that specifically monitors cathepsin B activity over a broad pH range.
Collapse
Affiliation(s)
- Michael
C. Yoon
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, San Diego, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, La Jolla, San Diego, California 92093, United States
| | - Von Phan
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, San Diego, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, La Jolla, San Diego, California 92093, United States
| | - Sonia Podvin
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, San Diego, California 92093, United States
| | - Charles Mosier
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, San Diego, California 92093, United States
| | - Anthony J. O’Donoghue
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, San Diego, California 92093, United States
| | - Vivian Hook
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, San Diego, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, La Jolla, San Diego, California 92093, United States
- Department
of Neurosciences and Department of Pharmacology, School of Medicine, University of California, La Jolla, San Diego, California 92093, United States
| |
Collapse
|
43
|
Kim JK, Jung HJ, Hyun M, Lee JY, Park JH, Suh SI, Baek WK, Kim HA. Resistance of hypervirulent Klebsiella pneumoniae to cathepsin B-mediated pyroptosis in murine macrophages. Front Immunol 2023; 14:1207121. [PMID: 37457695 PMCID: PMC10342201 DOI: 10.3389/fimmu.2023.1207121] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/31/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Hypervirulent Klebsiella pneumoniae (hvKp) has emerged as a clinically significant global pathogen in the last decade. However, the host immune responses of the macrophages during hvKp infection are largely unknown. In the present study, we aimed to compare the cytotoxic effects of hvKp and classical K. pneumoniae (cKp) in murine macrophages. Results We found that the activation of caspase-1 -dependent pyroptosis was higher in cKp-infected macrophages compared with that in hvKp-infected macrophages. In Caspase-1 deficiency macrophages, pyroptosis diminished during infection. Both hvKp and cKp strains led to nucleotide-binding and oligomerization domain-like receptor protein 3 (NLRP3) inflammasome formation and lysosomal cathepsin B activation, thus resulting in pyroptosis. Compared with the cKp strain, the hvKp strain inhibited these phenomena in murine macrophages. Conclusion HvKp infection resulted in different levels of pyroptosis via the activation of cathepsin B-NLRP3-caspase-1 in murine macrophages. Therefore, the manipulation of pyroptotic cell death is a potential target for host response during hvKp infection in macrophages.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Hui-Jung Jung
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Miri Hyun
- Department of Infectious Diseases, Keimyung University Dongsan Hospital, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Ji Yeon Lee
- Department of Infectious Diseases, Keimyung University Dongsan Hospital, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine and Brain Korea 21 Plus Project Team, Chonnam National University, Gwangju, Republic of Korea
| | - Seong-Il Suh
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Won-Ki Baek
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Hyun ah Kim
- Department of Infectious Diseases, Keimyung University Dongsan Hospital, Keimyung University School of Medicine, Daegu, Republic of Korea
| |
Collapse
|
44
|
Morandini AC, Savio LEB. Editorial: Inflammasome, purinergic signaling, and immunometabolism in oral health and disease. FRONTIERS IN ORAL HEALTH 2023; 4:1236507. [PMID: 37397347 PMCID: PMC10313182 DOI: 10.3389/froh.2023.1236507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 07/04/2023] Open
Affiliation(s)
- Ana Carolina Morandini
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Augusta, GA, United States
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States
| | - Luiz Eduardo Baggio Savio
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
45
|
Wang H, Li J, Zhang H, Wang M, Xiao L, Wang Y, Cheng Q. Regulation of microglia polarization after cerebral ischemia. Front Cell Neurosci 2023; 17:1182621. [PMID: 37361996 PMCID: PMC10285223 DOI: 10.3389/fncel.2023.1182621] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Stroke ranks second as a leading cause of death and permanent disability globally. Microglia, innate immune cells in the brain, respond rapidly to ischemic injury, triggering a robust and persistent neuroinflammatory reaction throughout the disease's progression. Neuroinflammation plays a critical role in the mechanism of secondary injury in ischemic stroke and is a significant controllable factor. Microglia activation takes on two general phenotypes: the pro-inflammatory M1 type and the anti-inflammatory M2 type, although the reality is more complex. The regulation of microglia phenotype is crucial to controlling the neuroinflammatory response. This review summarized the key molecules and mechanisms of microglia polarization, function, and phenotypic transformation following cerebral ischemia, with a focus on the influence of autophagy on microglia polarization. The goal is to provide a reference for the development of new targets for the treatment for ischemic stroke treatment based on the regulation of microglia polarization.
Collapse
Affiliation(s)
- Hao Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jingjing Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Han Zhang
- School of Medicine, Nantong University, Nantong, China
| | - Mengyao Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Lifang Xiao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Yitong Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Qiong Cheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| |
Collapse
|
46
|
Jiang H, Dong Z, Xia X, Li X. Cathepsins in oral diseases: mechanisms and therapeutic implications. Front Immunol 2023; 14:1203071. [PMID: 37334378 PMCID: PMC10272612 DOI: 10.3389/fimmu.2023.1203071] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023] Open
Abstract
Cathepsins are a type of lysosomal globulin hydrolase and are crucial for many physiological processes, including the resorption of bone matrix, innate immunity, apoptosis, proliferation, metastasis, autophagy, and angiogenesis. Findings regarding their functions in human physiological processes and disorders have drawn extensive attention. In this review, we will focus on the relationship between cathepsins and oral diseases. We highlight the structural and functional properties of cathepsins related to oral diseases, as well as the regulatory mechanisms in tissue and cells and their therapeutic uses. Elucidating the associated mechanism between cathepsins and oral diseases is thought to be a promising strategy for the treatment of oral diseases and may be a starting point for further studies at the molecular level.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Zuoxiang Dong
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xiaomin Xia
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Xue Li
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| |
Collapse
|
47
|
Menkhorst E, Santos LL, Zhou W, Yang G, Winship AL, Rainczuk KE, Nguyen P, Zhang JG, Moore P, Williams M, Lê Cao KA, Mansell A, Dimitriadis E. IL11 activates the placental inflammasome to drive preeclampsia. Front Immunol 2023; 14:1175926. [PMID: 37292200 PMCID: PMC10244672 DOI: 10.3389/fimmu.2023.1175926] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction Preeclampsia is a life-threatening disorder of pregnancy unique to humans. Interleukin (IL)11 is elevated in serum from pregnancies that subsequently develop early-onset preeclampsia and pharmacological elevation of IL11 in pregnant mice causes the development of early-onset preeclampsia-like features (hypertension, proteinuria, and fetal growth restriction). However, the mechanism by which IL11 drives preeclampsia is unknown. Method Pregnant mice were administered PEGylated (PEG)IL11 or control (PEG) from embryonic day (E)10-16 and the effect on inflammasome activation, systolic blood pressure (during gestation and at 50/90 days post-natal), placental development, and fetal/post-natal pup growth measured. RNAseq analysis was performed on E13 placenta. Human 1st trimester placental villi were treated with IL11 and the effect on inflammasome activation and pyroptosis identified by immunohistochemistry and ELISA. Result PEGIL11 activated the placental inflammasome causing inflammation, fibrosis, and acute and chronic hypertension in wild-type mice. Global and placental-specific loss of the inflammasome adaptor protein Asc and global loss of the Nlrp3 sensor protein prevented PEGIL11-induced fibrosis and hypertension in mice but did not prevent PEGIL11-induced fetal growth restriction or stillbirths. RNA-sequencing and histology identified that PEGIL11 inhibited trophoblast differentiation towards spongiotrophoblast and syncytiotrophoblast lineages in mice and extravillous trophoblast lineages in human placental villi. Discussion Inhibition of ASC/NLRP3 inflammasome activity could prevent IL11-induced inflammation and fibrosis in various disease states including preeclampsia.
Collapse
Affiliation(s)
- Ellen Menkhorst
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Leilani L. Santos
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Wei Zhou
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Guannan Yang
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
- Department of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, Australia
| | - Amy L. Winship
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Katarzyna E. Rainczuk
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Philana Nguyen
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Jian-Guo Zhang
- Walter and Eliza Hall Institute, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Paddy Moore
- Abortion and Contraception, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Michelle Williams
- Biomedical Animal Facility, The University of Melbourne, Parkville, VIC, Australia
| | - Kim-Anh Lê Cao
- Department of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, Australia
| | - Ashley Mansell
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Evdokia Dimitriadis
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
48
|
Sbai O, Bazzani V, Tapaswi S, McHale J, Vascotto C, Perrone L. Is Drp1 a link between mitochondrial dysfunction and inflammation in Alzheimer's disease? Front Mol Neurosci 2023; 16:1166879. [PMID: 37251647 PMCID: PMC10213291 DOI: 10.3389/fnmol.2023.1166879] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Recent advances highlight that inflammation is critical to Alzheimer Disease (AD) pathogenesis. Indeed, several diseases characterized by inflammation are considered risk factors for AD, such as type 2 diabetes, obesity, hypertension, and traumatic brain injury. Moreover, allelic variations in genes involved in the inflammatory cascade are risk factors for AD. AD is also characterized by mitochondrial dysfunction, which affects the energy homeostasis of the brain. The role of mitochondrial dysfunction has been characterized mostly in neuronal cells. However, recent data are demonstrating that mitochondrial dysfunction occurs also in inflammatory cells, promoting inflammation and the secretion of pro-inflammatory cytokines, which in turn induce neurodegeneration. In this review, we summarize the recent finding supporting the hypothesis of the inflammatory-amyloid cascade in AD. Moreover, we describe the recent data that demonstrate the link between altered mitochondrial dysfunction and the inflammatory cascade. We focus in summarizing the role of Drp1, which is involved in mitochondrial fission, showing that altered Drp1 activation affects the mitochondrial homeostasis and leads to the activation of the NLRP3 inflammasome, promoting the inflammatory cascade, which in turn aggravates Amyloid beta (Ab) deposition and tau-induced neurodegeneration, showing the relevance of this pro-inflammatory pathway as an early event in AD.
Collapse
Affiliation(s)
- Oualid Sbai
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis, Tunisia
| | | | | | - Joshua McHale
- Department of Medicine, University of Udine, Udine, Italy
| | - Carlo Vascotto
- Department of Medicine, University of Udine, Udine, Italy
- IMol Polish Academy of Sciences, Warsaw, Poland
| | - Lorena Perrone
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
49
|
Abstract
As an important sensor in the innate immune system, NLRP3 detects exogenous pathogenic invasions and endogenous cellular damage and responds by forming the NLRP3 inflammasome, a supramolecular complex that activates caspase-1. The three major components of the NLRP3 inflammasome are NLRP3, which captures the danger signals and recruits downstream molecules; caspase-1, which elicits maturation of the cytokines IL-1β and IL-18 and processing of gasdermin D to mediate cytokine release and pyroptosis; and ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain), which functions as a bridge connecting NLRP3 and caspase-1. In this article, we review the structural information that has been obtained on the NLRP3 inflammasome and its components or subcomplexes, with special focus on the inactive NLRP3 cage, the active NLRP3-NEK7 (NIMA-related kinase 7)-ASC inflammasome disk, and the PYD-PYD and CARD-CARD homotypic filamentous scaffolds of the inflammasome. We further implicate structure-derived mechanisms for the assembly and activation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Jianing Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA;
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA;
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
50
|
Alijagic A, Hedbrant A, Persson A, Larsson M, Engwall M, Särndahl E. NLRP3 inflammasome as a sensor of micro- and nanoplastics immunotoxicity. Front Immunol 2023; 14:1178434. [PMID: 37143682 PMCID: PMC10151538 DOI: 10.3389/fimmu.2023.1178434] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Micro- and nanoplastics (MNPs) are emerging pollutants with scarcely investigated effects on human innate immunity. If they follow a similar course of action as other, more thoroughly investigated particulates, MNPs may penetrate epithelial barriers, potentially triggering a cascade of signaling events leading to cell damage and inflammation. Inflammasomes are intracellular multiprotein complexes and stimulus-induced sensors critical for mounting inflammatory responses upon recognition of pathogen- or damage-associated molecular patterns. Among these, the NLRP3 inflammasome is the most studied in terms of activation via particulates. However, studies delineating the ability of MNPs to affect NLRP3 inflammasome activation are still rare. In this review, we address the issue of MNPs source and fate, highlight the main concepts of inflammasome activation via particulates, and explore recent advances in using inflammasome activation for assessment of MNP immunotoxicity. We also discuss the impact of co-exposure and MNP complex chemistry in potential inflammasome activation. Development of robust biological sensors is crucial in order to maximize global efforts to effectively address and mitigate risks that MNPs pose for human health.
Collapse
Affiliation(s)
- Andi Alijagic
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | - Alexander Hedbrant
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Alexander Persson
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Maria Larsson
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | - Magnus Engwall
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | - Eva Särndahl
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|