1
|
Wang H, Wang T, Yan S, Tang J, Zhang Y, Wang L, Xu H, Tu C. Crosstalk of pyroptosis and cytokine in the tumor microenvironment: from mechanisms to clinical implication. Mol Cancer 2024; 23:268. [PMID: 39614288 PMCID: PMC11607834 DOI: 10.1186/s12943-024-02183-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
In the realm of cancer research, the tumor microenvironment (TME) plays a crucial role in tumor initiation and progression, shaped by complex interactions between cancer cells and surrounding non-cancerous cells. Cytokines, as essential immunomodulatory agents, are secreted by various cellular constituents within the TME, including immune cells, cancer-associated fibroblasts, and cancer cells themselves. These cytokines facilitate intricate communication networks that significantly influence tumor initiation, progression, metastasis, and immune suppression. Pyroptosis contributes to TME remodeling by promoting the release of pro-inflammatory cytokines and sustaining chronic inflammation, impacting processes such as immune escape and angiogenesis. However, challenges remain due to the complex interplay among cytokines, pyroptosis, and the TME, along with the dual effects of pyroptosis on cancer progression and therapy-related complications like cytokine release syndrome. Unraveling these complexities could facilitate strategies that balance inflammatory responses while minimizing tissue damage during therapy. This review delves into the complex crosstalk between cytokines, pyroptosis, and the TME, elucidating their contribution to tumor progression and metastasis. By synthesizing emerging therapeutic targets and innovative technologies concerning TME, this review aims to provide novel insights that could enhance treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Tao Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Shuxiang Yan
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jinxin Tang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yibo Zhang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Liming Wang
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410011, China.
| | - Haodong Xu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Shenzhen Research Institute of Central South University, Guangdong, 518063, China.
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
2
|
Banerjee S, Halder P, Das S, Maiti S, Withey JH, Mitobe J, Chowdhury G, Kitahara K, Miyoshi SI, Mukhopadhyay AK, Dutta S, Koley H. Trivalent outer membrane vesicles-based combination vaccine candidate induces protective immunity against Campylobacter and invasive non-typhoidal Salmonella in adult mice. Med Microbiol Immunol 2024; 213:21. [PMID: 39407046 DOI: 10.1007/s00430-024-00805-z] [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: 10/07/2024] [Indexed: 12/28/2024]
Abstract
Campylobacter and invasive non-typhoidal Salmonella (iNTS) are among the most common causative agents of gastroenteritis worldwide. As of now, no single combination licensed vaccine is available for public health use against both iNTS and Campylobacter species. Outer-membrane vesicles (OMVs) are nanoscale proteoliposomes released from the surface of gram-negative bacteria during log phase and harbor a variety of immunogenic proteins. Based on epidemiology of infections, we formulated a novel trivalent outer membrane vesicles (TOMVs)-based vaccine candidate against Campylobacter jejuni (CJ), Salmonella Typhimurium (ST) and Salmonella Enteritidis (SE). Isolated OMVs from CJ, ST and SE were combined in equal ratios for formulation of TOMVs and 5 µg of the developed vaccine candidate was used for intraperitoneal immunization of adult BALB/c mice. Immunization with TOMVs significantly activated both the humoral and cellular arm of adaptive immune response. Robust bactericidal effect was elicited by TOMVs immunized adult mice sera. TOMVs immunization induced long-term protective efficacy against CJ, ST and SE infections in mice. The study illustrates the ability of TOMVs-based combination immunogen in eliciting broad-spectrum protective immunity against prevalent Campylobacter and iNTS pathogens. According to the findings, TOMVs can work as a potent combination-based acellular vaccine candidate for amelioration of Campylobacter and iNTS-mediated gastroenteritis.
Collapse
Affiliation(s)
- Soumalya Banerjee
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Sanjib Das
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Suhrid Maiti
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Jeffrey H Withey
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jiro Mitobe
- Department of Infectious Diseases, Kyorin University School of Medicine, Tokyo, Japan
| | - Goutam Chowdhury
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
- Collaborative Research Centre of Okayama University for Infectious Diseases at ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Kei Kitahara
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Collaborative Research Centre of Okayama University for Infectious Diseases at ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Shin-Ichi Miyoshi
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Collaborative Research Centre of Okayama University for Infectious Diseases at ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Asish Kumar Mukhopadhyay
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Shanta Dutta
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata, 700010, India.
| |
Collapse
|
3
|
Alipour S, Mardi A, Shajari N, Kazemi T, Sadeghi MR, Ahmadian Heris J, Masoumi J, Baradaran B. Unmasking the NLRP3 inflammasome in dendritic cells as a potential therapeutic target for autoimmunity, cancer, and infectious conditions. Life Sci 2024; 348:122686. [PMID: 38710282 DOI: 10.1016/j.lfs.2024.122686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/13/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Proper and functional immune response requires a complex interaction between innate and adaptive immune cells, which dendritic cells (DCs) are the primary actors in this coordination as professional antigen-presenting cells. DCs are armed with numerous pattern recognition receptors (PRRs) such as nucleotide-binding and oligomerization domain-like receptors (NLRs) like NLRP3, which influence the development of their activation state upon sensation of ligands. NLRP3 is a crucial component of the immune system for protection against tumors and infectious agents, because its activation leads to the assembly of inflammasomes that cause the formation of active caspase-1 and stimulate the maturation and release of proinflammatory cytokines. But, when NLRP3 becomes overactivated, it plays a pathogenic role in the progression of several autoimmune disorders. So, NLRP3 activation is strictly regulated by diverse signaling pathways that are mentioned in detail in this review. Furthermore, the role of NLRP3 in all of the diverse immune cells' subsets is briefly mentioned in this study because NLRP3 plays a pivotal role in modulating other immune cells which are accompanied by DCs' responses and subsequently influence differentiation of T cells to diverse T helper subsets and even impact on cytotoxic CD8+ T cells' responses. This review sheds light on the functional and therapeutic role of NLRP3 in DCs and its contribution to the occurrence and progression of autoimmune disorders, prevention of diverse tumors' development, and recognition and annihilation of various infectious agents. Furthermore, we highlight NLRP3 targeting potential for improving DC-based immunotherapeutic approaches, to be used for the benefit of patients suffering from these disorders.
Collapse
Affiliation(s)
- Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Shajari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Sadeghi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Yang C, Qu L, Wang R, Wang F, Yang Z, Xiao F. Multi-layered effects of Panax notoginseng on immune system. Pharmacol Res 2024; 204:107203. [PMID: 38719196 DOI: 10.1016/j.phrs.2024.107203] [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: 02/21/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
Recent research has demonstrated the immunomodulatory potential of Panax notoginseng in the treatment of chronic inflammatory diseases and cerebral hemorrhage, suggesting its significance in clinical practice. Nevertheless, the complex immune activity of various components has hindered a comprehensive understanding of the immune-regulating properties of Panax notoginseng, impeding its broader utilization. This review evaluates the effect of Panax notoginseng to various types of white blood cells, elucidates the underlying mechanisms, and compares the immunomodulatory effects of different Panax notoginseng active fractions, aiming to provide the theory basis for future immunomodulatory investigation.
Collapse
Affiliation(s)
- Chunhao Yang
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China
| | - Liping Qu
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China; Innovation Materials Research and Development Center, Botanee Research Institute, Shanghai Jiyan Biomedical Development Co., Ltd., Shanghai 201702, China
| | - Rui Wang
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China
| | - Feifei Wang
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China; Innovation Materials Research and Development Center, Botanee Research Institute, Shanghai Jiyan Biomedical Development Co., Ltd., Shanghai 201702, China
| | - Zhaoxiang Yang
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China
| | - Fengkun Xiao
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China.
| |
Collapse
|
5
|
Tölken LA, Paulikat AD, Jachmann LH, Reder A, Salazar MG, Medina LMP, Michalik S, Völker U, Svensson M, Norrby-Teglund A, Hoff KJ, Lammers M, Siemens N. Reduced interleukin-18 secretion by human monocytic cells in response to infections with hyper-virulent Streptococcus pyogenes. J Biomed Sci 2024; 31:26. [PMID: 38408992 PMCID: PMC10898077 DOI: 10.1186/s12929-024-01014-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/20/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Streptococcus pyogenes (group A streptococcus, GAS) causes a variety of diseases ranging from mild superficial infections of the throat and skin to severe invasive infections, such as necrotizing soft tissue infections (NSTIs). Tissue passage of GAS often results in mutations within the genes encoding for control of virulence (Cov)R/S two component system leading to a hyper-virulent phenotype. Dendritic cells (DCs) are innate immune sentinels specialized in antigen uptake and subsequent T cell priming. This study aimed to analyze cytokine release by DCs and other cells of monocytic origin in response to wild-type and natural covR/S mutant infections. METHODS Human primary monocyte-derived (mo)DCs were used. DC maturation and release of pro-inflammatory cytokines in response to infections with wild-type and covR/S mutants were assessed via flow cytometry. Global proteome changes were assessed via mass spectrometry. As a proof-of-principle, cytokine release by human primary monocytes and macrophages was determined. RESULTS In vitro infections of moDCs and other monocytic cells with natural GAS covR/S mutants resulted in reduced secretion of IL-8 and IL-18 as compared to wild-type infections. In contrast, moDC maturation remained unaffected. Inhibition of caspase-8 restored secretion of both molecules. Knock-out of streptolysin O in GAS strain with unaffected CovR/S even further elevated the IL-18 secretion by moDCs. Of 67 fully sequenced NSTI GAS isolates, 28 harbored mutations resulting in dysfunctional CovR/S. However, analyses of plasma IL-8 and IL-18 levels did not correlate with presence or absence of such mutations. CONCLUSIONS Our data demonstrate that strains, which harbor covR/S mutations, interfere with IL-18 and IL-8 responses in monocytic cells by utilizing the caspase-8 axis. Future experiments aim to identify the underlying mechanism and consequences for NSTI patients.
Collapse
Affiliation(s)
- Lea A Tölken
- Department of Molecular Genetics and Infection Biology, University of Greifswald, Greifswald, Germany
| | - Antje D Paulikat
- Department of Molecular Genetics and Infection Biology, University of Greifswald, Greifswald, Germany
| | - Lana H Jachmann
- Department of Molecular Genetics and Infection Biology, University of Greifswald, Greifswald, Germany
| | - Alexander Reder
- Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | | | - Laura M Palma Medina
- Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Stephan Michalik
- Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Mattias Svensson
- Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Anna Norrby-Teglund
- Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Katharina J Hoff
- Institute of Mathematics and Computer Science, University of Greifswald, Greifswald, Germany
| | - Michael Lammers
- Department of Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Nikolai Siemens
- Department of Molecular Genetics and Infection Biology, University of Greifswald, Greifswald, Germany.
| |
Collapse
|
6
|
Clausen BE, Amon L, Backer RA, Berod L, Bopp T, Brand A, Burgdorf S, Chen L, Da M, Distler U, Dress RJ, Dudziak D, Dutertre CA, Eich C, Gabele A, Geiger M, Ginhoux F, Giusiano L, Godoy GJ, Hamouda AEI, Hatscher L, Heger L, Heidkamp GF, Hernandez LC, Jacobi L, Kaszubowski T, Kong WT, Lehmann CHK, López-López T, Mahnke K, Nitsche D, Renkawitz J, Reza RA, Sáez PJ, Schlautmann L, Schmitt MT, Seichter A, Sielaff M, Sparwasser T, Stoitzner P, Tchitashvili G, Tenzer S, Tochoedo NR, Vurnek D, Zink F, Hieronymus T. Guidelines for mouse and human DC functional assays. Eur J Immunol 2023; 53:e2249925. [PMID: 36563126 DOI: 10.1002/eji.202249925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022]
Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. Recent studies have provided evidence for an increasing number of phenotypically distinct conventional DC (cDC) subsets that on one hand exhibit a certain functional plasticity, but on the other hand are characterized by their tissue- and context-dependent functional specialization. Here, we describe a selection of assays for the functional characterization of mouse and human cDC. The first two protocols illustrate analysis of cDC endocytosis and metabolism, followed by guidelines for transcriptomic and proteomic characterization of cDC populations. Then, a larger group of assays describes the characterization of cDC migration in vitro, ex vivo, and in vivo. The final guidelines measure cDC inflammasome and antigen (cross)-presentation activity. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.
Collapse
Affiliation(s)
- Björn E Clausen
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Ronald A Backer
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Luciana Berod
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Tobias Bopp
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Anna Brand
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sven Burgdorf
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Luxia Chen
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Meihong Da
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ute Distler
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Regine J Dress
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Germany
| | - Charles-Antoine Dutertre
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Christina Eich
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Anna Gabele
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Melanie Geiger
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Lucila Giusiano
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Gloria J Godoy
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Ahmed E I Hamouda
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Lukas Hatscher
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Gordon F Heidkamp
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Lola C Hernandez
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas Jacobi
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Tomasz Kaszubowski
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Wan Ting Kong
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Germany
| | - Tamara López-López
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karsten Mahnke
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Dominik Nitsche
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Jörg Renkawitz
- Biomedical Center (BMC), Walter Brendel Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, LMU Munich, Munich, Germany
| | - Rifat A Reza
- Biomedical Center (BMC), Walter Brendel Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, LMU Munich, Munich, Germany
| | - Pablo J Sáez
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Schlautmann
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Madeleine T Schmitt
- Biomedical Center (BMC), Walter Brendel Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, LMU Munich, Munich, Germany
| | - Anna Seichter
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Malte Sielaff
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Tim Sparwasser
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Patrizia Stoitzner
- Department of Dermatology, Venerology & Allergology, Medical University Innsbruck, Innsbruck, Austria
| | - Giorgi Tchitashvili
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Stefan Tenzer
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Helmholtz Institute for Translational Oncology Mainz (HI-TRON Mainz), Mainz, Germany
| | - Nounagnon R Tochoedo
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Damir Vurnek
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Fabian Zink
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Thomas Hieronymus
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
- Institute of Cell and Tumor Biology, RWTH Aachen University, Medical Faculty, Germany
| |
Collapse
|
7
|
Li Y, Sun Y, Zhang Y, Li Q, Wang S, Curtiss R, Shi H. A Bacterial mRNA-Lysis-Mediated Cargo Release Vaccine System for Regulated Cytosolic Surveillance and Optimized Antigen Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303568. [PMID: 37867213 PMCID: PMC10667801 DOI: 10.1002/advs.202303568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/14/2023] [Indexed: 10/24/2023]
Abstract
Engineered vector-based in vivo protein delivery platforms have made significant progress for both prophylactic and therapeutic applications. However, the lack of effective release strategies results in foreign cargo being trapped within the vector, restricting the provision of significant performance benefits and enhanced therapeutic results compared to traditional vaccines. Herein, the development of a Salmonella mRNA interferase regulation vector (SIRV) system is reported to overcome this challenge. The genetic circuits are engineered that (1) induce self-lysis to release foreign antigens into target cells and (2) activate the cytosolic surveillance cGAS-STING axis by releasing DNA into the cytoplasm. Delayed synthesis of the MazF interferase regulates differential mRNA cleavage, resulting in a 36-fold increase in the delivery of foreign antigens and modest activation of the inflammasome, which collectively contribute to the marked maturation of antigen-presenting cells (APCs). Bacteria delivering the protective antigen SaoA exhibits excellent immunogenicity and safety in mouse and pig models, significantly improving the survival rate of animals challenged with multiple serotypes of Streptococcus suis. Thus, the SIRV system enables the effective integration of various modular components and antigen cargos, allowing for the generation of an extensive range of intracellular protein delivery systems using multiple bacterial species in a highly efficient manner.
Collapse
Affiliation(s)
- Yu‐an Li
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225000China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou225000China
| | - Yanni Sun
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225000China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou225000China
| | - Yuqin Zhang
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225000China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou225000China
| | - Quan Li
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225000China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou225000China
| | - Shifeng Wang
- Department of Infectious Diseases and ImmunologyCollege of Veterinary MedicineUniversity of FloridaGainesvilleFL32611‐0880USA
| | - Roy Curtiss
- Department of Infectious Diseases and ImmunologyCollege of Veterinary MedicineUniversity of FloridaGainesvilleFL32611‐0880USA
| | - Huoying Shi
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225000China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou225000China
- Joint International Research Laboratory of Agriculture and Agri‐Product SafetyYangzhou University (JIRLAAPS)Yangzhou225000China
| |
Collapse
|
8
|
Dai Y, Zhou J, Shi C. Inflammasome: structure, biological functions, and therapeutic targets. MedComm (Beijing) 2023; 4:e391. [PMID: 37817895 PMCID: PMC10560975 DOI: 10.1002/mco2.391] [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: 02/24/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 10/12/2023] Open
Abstract
Inflammasomes are a group of protein complex located in cytoplasm and assemble in response to a wide variety of pathogen-associated molecule patterns, damage-associated molecule patterns, and cellular stress. Generally, the activation of inflammasomes will lead to maturation of proinflammatory cytokines and pyroptotic cell death, both associated with inflammatory cascade amplification. A sensor protein, an adaptor, and a procaspase protein interact through their functional domains and compose one subunit of inflammasome complex. Under physiological conditions, inflammasome functions against pathogen infection and endogenous dangers including mtROS, mtDNA, and so on, while dysregulation of its activation can lead to unwanted results. In recent years, advances have been made to clarify the mechanisms of inflammasome activation, the structural details of them and their functions (negative/positive) in multiple disease models in both animal models and human. The wide range of the stimuli makes the function of inflammasome diverse and complex. Here, we review the structure, biological functions, and therapeutic targets of inflammasomes, while highlight NLRP3, NLRC4, and AIM2 inflammasomes, which are the most well studied. In conclusion, this review focuses on the activation process, biological functions, and structure of the most well-studied inflammasomes, summarizing and predicting approaches for disease treatment and prevention with inflammasome as a target. We aim to provide fresh insight into new solutions to the challenges in this field.
Collapse
Affiliation(s)
- Yali Dai
- Institute of Rocket Force MedicineState Key Laboratory of Trauma and Chemical PoisoningArmy Medical UniversityChongqingChina
| | - Jing Zhou
- Institute of Rocket Force MedicineState Key Laboratory of Trauma and Chemical PoisoningArmy Medical UniversityChongqingChina
- Institute of ImmunologyArmy Medical UniversityChongqingChina
| | - Chunmeng Shi
- Institute of Rocket Force MedicineState Key Laboratory of Trauma and Chemical PoisoningArmy Medical UniversityChongqingChina
| |
Collapse
|
9
|
Hatscher L, Kaszubowski T, Amon L, Dudziak D, Heger L. Circumventing pyroptosis via hyperactivation shapes superior immune responses of human type 2 dendritic cells compared to type 3 dendritic cells. Eur J Immunol 2023; 53:e2250123. [PMID: 36724513 DOI: 10.1002/eji.202250123] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 01/22/2023] [Accepted: 01/30/2023] [Indexed: 02/03/2023]
Abstract
Exploiting inflammasome activation in dendritic cells (DCs) is a promising approach to fight cancer and to augment adjuvant-induced immune responses. As inflammasome formation is typically accompanied by pyroptosis, hyperactivation-defined as inflammasome activation in the absence of pyroptosis-represents a mechanism of circumventing cell death of DCs while simultaneously benefitting from inflammasome signaling. We previously demonstrated a unique specialization for inflammasome responses and hyperactivation of human cDC2 among all human DC subsets. As recent investigations revealed heterogeneity among the human cDC2 population, we aimed to analyze whether the two recently identified cDC2 subpopulations DC2 and DC3 harbor similar or different inflammasome characteristics. Here, we report that both DC2 and DC3 are inflammasome competent. We show that DC3 generally induce stronger inflammasome responses, which are associated with higher levels of cell death. Although DC2 release lower levels of inflammasome-dependent IL-1β, they induce stronger CD4+ T cell responses than DC3, which are predominantly skewed toward a TH 1/TH 17 phenotype. Thus, mainly DC2 seem to be able to enter a state of hyperactivation, resulting in enhanced T cell stimulatory capacity.
Collapse
Affiliation(s)
- Lukas Hatscher
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Tomasz Kaszubowski
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, 91052, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg, 91054, Erlangen, Germany
- Medical Immunology Campus Erlangen, 91054, Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, 91052, Erlangen, Germany
| |
Collapse
|
10
|
Vigneron C, Py BF, Monneret G, Venet F. The double sides of NLRP3 inflammasome activation in sepsis. Clin Sci (Lond) 2023; 137:333-351. [PMID: 36856019 DOI: 10.1042/cs20220556] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 03/02/2023]
Abstract
Sepsis is defined as a life-threatening organ dysfunction induced by a dysregulated host immune response to infection. Immune response induced by sepsis is complex and dynamic. It is schematically described as an early dysregulated systemic inflammatory response leading to organ failures and early deaths, followed by the development of persistent immune alterations affecting both the innate and adaptive immune responses associated with increased risk of secondary infections, viral reactivations, and late mortality. In this review, we will focus on the role of NACHT, leucin-rich repeat and pyrin-containing protein 3 (NLRP3) inflammasome in the pathophysiology of sepsis. NLRP3 inflammasome is a multiproteic intracellular complex activated by infectious pathogens through a two-step process resulting in the release of the pro-inflammatory cytokines IL-1β and IL-18 and the formation of membrane pores by gasdermin D, inducing a pro-inflammatory form of cell death called pyroptosis. The role of NLRP3 inflammasome in the pathophysiology of sepsis can be ambivalent. Indeed, although it might protect against sepsis when moderately activated after initial infection, excessive NLRP3 inflammasome activation can induce dysregulated inflammation leading to multiple organ failure and death during the acute phase of the disease. Moreover, this activation might become exhausted and contribute to post-septic immunosuppression, driving impaired functions of innate and adaptive immune cells. Targeting the NLRP3 inflammasome could thus be an attractive option in sepsis either through IL-1β and IL-18 antagonists or through inhibition of NLRP3 inflammasome pathway downstream components. Available treatments and results of first clinical trials will be discussed.
Collapse
Affiliation(s)
- Clara Vigneron
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Bénédicte F Py
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Guillaume Monneret
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Edouard Herriot Hospital, Lyon, France
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Fabienne Venet
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
11
|
Hesemans E, Saffarzadeh N, Maksoudian C, Izci M, Chu T, Rios Luci C, Wang Y, Naatz H, Thieme S, Richter C, Manshian BB, Pokhrel S, Mädler L, Soenen SJ. Cu-doped TiO 2 nanoparticles improve local antitumor immune activation and optimize dendritic cell vaccine strategies. J Nanobiotechnology 2023; 21:87. [PMID: 36915084 PMCID: PMC10009859 DOI: 10.1186/s12951-023-01844-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/06/2023] [Indexed: 03/16/2023] Open
Abstract
Nanoparticle-mediated cancer immunotherapy holds great promise, but more efforts are needed to obtain nanoformulations that result in a full scale activation of innate and adaptive immune components that specifically target the tumors. We generated a series of copper-doped TiO2 nanoparticles in order to tune the kinetics and full extent of Cu2+ ion release from the remnant TiO2 nanocrystals. Fine-tuning nanoparticle properties resulted in a formulation of 33% Cu-doped TiO2 which enabled short-lived hyperactivation of dendritic cells and hereby promoted immunotherapy. The nanoparticles result in highly efficient activation of dendritic cells ex vivo, which upon transplantation in tumor bearing mice, exceeded the therapeutic outcomes obtained with classically stimulated dendritic cells. Efficacious but simple nanomaterials that can promote dendritic cancer cell vaccination strategies open up new avenues for improved immunotherapy and human health.
Collapse
Affiliation(s)
- Evelien Hesemans
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Neshat Saffarzadeh
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Christy Maksoudian
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Mukaddes Izci
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Tianjiao Chu
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Carla Rios Luci
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Yuqing Wang
- Leibniz Institute for Materials Engineering IWT, Badgasteiner Straße 3, 28359, Bremen, Germany.,Faculty of Production Engineering, University of Bremen, Badgasteiner Straße 1, 28359, Bremen, Germany
| | - Hendrik Naatz
- Leibniz Institute for Materials Engineering IWT, Badgasteiner Straße 3, 28359, Bremen, Germany.,Faculty of Production Engineering, University of Bremen, Badgasteiner Straße 1, 28359, Bremen, Germany
| | | | | | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Suman Pokhrel
- Leibniz Institute for Materials Engineering IWT, Badgasteiner Straße 3, 28359, Bremen, Germany.,Faculty of Production Engineering, University of Bremen, Badgasteiner Straße 1, 28359, Bremen, Germany
| | - Lutz Mädler
- Leibniz Institute for Materials Engineering IWT, Badgasteiner Straße 3, 28359, Bremen, Germany.,Faculty of Production Engineering, University of Bremen, Badgasteiner Straße 1, 28359, Bremen, Germany
| | - Stefaan J Soenen
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium. .,Leuven Cancer Institute, KU Leuven, Leuven, Belgium. .,KU Leuven Institute of Physics-Based Modeling for In Silico Health, KU Leuven, Leuven, Belgium.
| |
Collapse
|
12
|
Wang S, Chen Y, Ling Z, Li J, Hu J, He F, Chen Q. The role of dendritic cells in the immunomodulation to implanted biomaterials. Int J Oral Sci 2022; 14:52. [PMCID: PMC9636170 DOI: 10.1038/s41368-022-00203-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
Considering the substantial role played by dendritic cells (DCs) in the immune system to bridge innate and adaptive immunity, studies on DC-mediated immunity toward biomaterials principally center on their adjuvant effects in facilitating the adaptive immunity of codelivered antigens. However, the effect of the intrinsic properties of biomaterials on dendritic cells has not been clarified. Recently, researchers have begun to investigate and found that biomaterials that are nonadjuvant could also regulate the immune function of DCs and thus affect subsequent tissue regeneration. In the case of proteins adsorbed onto biomaterial surfaces, their intrinsic properties can direct their orientation and conformation, forming “biomaterial-associated molecular patterns (BAMPs)”. Thus, in this review, we focused on the intrinsic physiochemical properties of biomaterials in the absence of antigens that affect DC immune function and summarized the underlying signaling pathways. Moreover, we preliminarily clarified the specific composition of BAMPs and the interplay between some key molecules and DCs, such as heat shock proteins (HSPs) and high mobility group box 1 (HMGB1). This review provides a new direction for future biomaterial design, through which modulation of host immune responses is applicable to tissue engineering and immunotherapy.
Collapse
Affiliation(s)
- Siyuan Wang
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Yanqi Chen
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Zhaoting Ling
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Jia Li
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Jun Hu
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Fuming He
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Qianming Chen
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| |
Collapse
|
13
|
Bencze D, Fekete T, Pfliegler W, Szöőr Á, Csoma E, Szántó A, Tarr T, Bácsi A, Kemény L, Veréb Z, Pázmándi K. Interactions between the NLRP3-Dependent IL-1β and the Type I Interferon Pathways in Human Plasmacytoid Dendritic Cells. Int J Mol Sci 2022; 23:12154. [PMID: 36293012 PMCID: PMC9602791 DOI: 10.3390/ijms232012154] [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: 09/06/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Generally, a reciprocal antagonistic interaction exists between the antiviral type I interferon (IFN) and the antibacterial nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3)-dependent IL-1β pathways that can significantly shape immune responses. Plasmacytoid dendritic cells (pDCs), as professional type I IFN-producing cells, are the major coordinators of antiviral immunity; however, their NLRP3-dependent IL-1β secretory pathway is poorly studied. Our aim was to determine the functional activity of the IL-1β pathway and its possible interaction with the type I IFN pathway in pDCs. We found that potent nuclear factor-kappa B (NF-κB) inducers promote higher levels of pro-IL-1β during priming compared to those activation signals, which mainly trigger interferon regulatory factor (IRF)-mediated type I IFN production. The generation of cleaved IL-1β requires certain secondary signals in pDCs and IFN-α or type I IFN-inducing viruses inhibit IL-1β production of pDCs, presumably by promoting the expression of various NLRP3 pathway inhibitors. In line with that, we detected significantly lower IL-1β production in pDCs of psoriasis patients with elevated IFN-α levels. Collectively, our results show that the NLRP3-dependent IL-1β secretory pathway is inducible in pDCs; however, it may only prevail under inflammatory conditions, in which the type I IFN pathway is not dominant.
Collapse
Affiliation(s)
- Dóra Bencze
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Tünde Fekete
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Walter Pfliegler
- Department of Molecular Biotechnology and Microbiology, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary
| | - Árpád Szöőr
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Eszter Csoma
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Antónia Szántó
- Division of Clinical Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tünde Tarr
- Division of Clinical Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Lajos Kemény
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Kitti Pázmándi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
14
|
D’haese S, Laeremans T, den Roover S, Allard SD, Vanham G, Aerts JL. Efficient Induction of Antigen-Specific CD8+ T-Cell Responses by Cationic Peptide-Based mRNA Nanoparticles. Pharmaceutics 2022; 14:pharmaceutics14071387. [PMID: 35890284 PMCID: PMC9321026 DOI: 10.3390/pharmaceutics14071387] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
A major determinant for the success of mRNA-based vaccines is the composition of the nanoparticles (NPs) used for formulation and delivery. Cationic peptides represent interesting candidate carriers for mRNA, since they have been shown to efficiently deliver nucleic acids to eukaryotic cells. mRNA NPs based on arginine-rich peptides have previously been demonstrated to induce potent antigen-specific CD8+ T-cell responses. We therefore compared the histidine-rich amphipathic peptide LAH4-L1 (KKALLAHALHLLALLALHLAHALKKA) to the fully substituted arginine variant (LAH4-L1R) for their capacity to formulate mRNA and transfect dendritic cells (DCs). Although both peptides encapsulated mRNA to the same extent, and showed excellent uptake in DCs, the gene expression level was significantly higher for LAH4-L1. The LAH4-L1–mRNA NPs also resulted in enhanced antigen presentation in the context of MHC I compared to LAH4-L1R in primary murine CD103+ DCs. Both peptides induced DC maturation and inflammasome activation. Subsequent ex vivo stimulation of OT-I splenocytes with transfected CD103+ DCs resulted in a high proportion of polyfunctional CD8+ T cells for both peptides. In addition, in vivo immunization with LAH4-L1 or LAH4-L1R–mRNA NPs resulted in proliferation of antigen-specific T cells. In conclusion, although LAH4-L1 outperformed LAH4-L1R in terms of transfection efficiency, the immune stimulation ex vivo and in vivo was equally efficient.
Collapse
Affiliation(s)
- Sigrid D’haese
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (S.D.); (T.L.); (S.d.R.)
| | - Thessa Laeremans
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (S.D.); (T.L.); (S.d.R.)
| | - Sabine den Roover
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (S.D.); (T.L.); (S.d.R.)
| | - Sabine D. Allard
- Department of Internal Medicine (IRG), Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium;
| | - Guido Vanham
- Department of Virology, Institute of Tropical Medicine, University of Antwerp, 2000 Antwerp, Belgium;
| | - Joeri L. Aerts
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (S.D.); (T.L.); (S.d.R.)
- Correspondence:
| |
Collapse
|
15
|
Challagundla N, Saha B, Agrawal-Rajput R. Insights into inflammasome regulation: cellular, molecular, and pathogenic control of inflammasome activation. Immunol Res 2022; 70:578-606. [PMID: 35610534 DOI: 10.1007/s12026-022-09286-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
Abstract
Maintenance of immune homeostasis is an intricate process wherein inflammasomes play a pivotal role by contributing to innate and adaptive immune responses. Inflammasomes are ensembles of adaptor proteins that can trigger a signal following innate sensing of pathogens or non-pathogens eventuating in the inductions of IL-1β and IL-18. These inflammatory cytokines substantially influence the antigen-presenting cell's costimulatory functions and T helper cell differentiation, contributing to adaptive immunity. As acute and chronic disease conditions may accompany parallel tissue damage, we analyze the critical role of extracellular factors such as cytokines, amyloids, cholesterol crystals, etc., intracellular metabolites, and signaling molecules regulating inflammasome activation/inhibition. We develop an operative framework for inflammasome function and regulation by host cell factors and pathogens. While inflammasomes influence the innate and adaptive immune components' interplay modulating the anti-pathogen adaptive immune response, pathogens may target inflammasome inhibition as a survival strategy. As trapped between health and diseases, inflammasomes serve as promising therapeutic targets and their modus operandi serves as a scientific rationale for devising better therapeutic strategies.
Collapse
Affiliation(s)
- Naveen Challagundla
- Immunology lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382007, India
| | - Bhaskar Saha
- National Centre for Cell Science, Lab-5, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Reena Agrawal-Rajput
- Immunology lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382007, India.
| |
Collapse
|
16
|
Braunlin E, Abrahante JE, McElmurry R, Evans M, Smith M, Seelig D, O'Sullivan MG, Tolar J, Whitley CB, McIvor RS. Contribution of the innate and adaptive immune systems to aortic dilation in murine mucopolysaccharidosis type I. Mol Genet Metab 2022; 135:193-205. [PMID: 35165009 PMCID: PMC9109621 DOI: 10.1016/j.ymgme.2022.01.104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/23/2021] [Accepted: 01/31/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Adult immunocompetent male C57Bl/6 mucopolysaccharidosis, type I (MPSI) mice develop aortic insufficiency (AI), dilated ascending aortas and decreased cardiac function, findings not observed in immune incompetent adult male NSG MPSI mice. We sought to determine why. METHODS Cardiac ultrasound measurements of ascending aorta and left ventricular dimensions and Doppler interrogation for AI were performed in 6-month-old male B6 MPSI (N = 12), WT (N = 6), NSG MPSI (N = 8), NSG (N = 6) mice. Urinary glycosaminoglycans, RNA sequencing with quantitative PCR were performed and aortic pathology assessed by routine and immunohistochemical staining on subsets of murine aortas. RESULTS Ascending aortic diameters were significantly greater, left ventricular function significantly decreased, and AI significantly more frequent in B6 MPSI mice compared to NSG MPSI mice (p < 0.0001, p = 0.008 and p = 0.02, respectively); NSG and B6 WT mice showed no changes. Urinary glycosaminoglycans were significantly greater in B6 and NSG MPSI mice and both were significantly elevated compared to WT controls (p = 0.003 and p < 0.0001, respectively). By RNA sequencing, all 11 components of the inflammasome pathway were upregulated in B6 MUT, but only Aim2 and Ctsb in NSG MUT mice and none in WT controls. Both B6 and NSG MUT mice demonstrated variably-severe intramural inflammation, vacuolated cells, elastin fragmentation and disarray, and intense glycosaminoglycans on histological staining. B6 MPSI mice demonstrated numerous medial MAC2+ macrophages and adventitial CD3+ T-cells while MAC2+ macrophages were sparse and CD3+ T-cells absent in NSG MPSI mice. CONCLUSIONS Aortic dilation, AI and decreased cardiac function occur in immunocompetent B6 MPSI male mice but not in immune incompetent NSG MPSI mice, unrelated to GAG excretion, upregulation of Ctsb, or routine histologic appearance. Upregulation of all components of the inflammasome pathway in B6 MUT, but not NSG MUT mice, and abundant medial MAC2 and adventitial CD3 infiltrates in B6, but not NSG, MPSI aortas differentiated the two strains. These results suggest that the innate and adaptive immune systems play a role in these cardiac findings which may be relevant to human MPSI.
Collapse
Affiliation(s)
- Elizabeth Braunlin
- Department of Pediatrics University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Juan E Abrahante
- University of Minnesota Informatics Institute University of Minnesota, Minneapolis, MN, USA.
| | - Ron McElmurry
- Department of Pediatrics University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Michael Evans
- Biostatistical Design and Analysis Center Clinical and Translational Science Institute University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Miles Smith
- Department of Genetics, Cell Biology and Development University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Davis Seelig
- Comparative Pathology Shared Resource, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, St. Paul, MN, USA.
| | - M Gerard O'Sullivan
- Comparative Pathology Shared Resource, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, St. Paul, MN, USA.
| | - Jakub Tolar
- Department of Blood and Marrow Transplant University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Chester B Whitley
- Gene Therapy Center Department of Pediatrics University of Minnesota Medical School Minneapolis, MN, USA.
| | - R Scott McIvor
- Department of Genetics, Cell Biology and Development University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
17
|
Zhang S, Wang H, Liu Y, Tao T, Zeng Z, Zhou Y, Wang M. Nocardia rubra cell-wall skeleton influences the development of cervical carcinoma by promoting the antitumor effect of macrophages and dendritic cells. Cancer Med 2022; 11:1249-1268. [PMID: 34994088 PMCID: PMC8894708 DOI: 10.1002/cam4.4526] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023] Open
Abstract
Background As an immune enhancer, Nocardia rubra cell‐wall skeleton (Nr‐CWS) has been used to treat persistent human papillomavirus infection and cervical precancerous lesions. However, it is still unclear whether it can be used to treat cervical carcinoma. Methods In our study, the aim was to determine whether Nr‐CWS affects the apoptosis of cervical carcinoma cells by enhancing the antitumor effect of dendritic cells and macrophages in vivo and in vitro. Results The experimental results showed that Nr‐CWS can promote the activity of dendritic cells and macrophages and reduce their apoptosis. It also increased the cytokines IL‐6, IL‐12, TNF‐ɑ, and IL‐1β secreted by dendritic cells and macrophages and reduced their PD‐L1 expression. In vitro, Nr‐CWS inhibited the proliferation, colony forming ability of HeLa and SiHa cervical carcinoma cell lines cultured with macrophages, and more cells were blocked in G2/M phase. Nr‐CWS promoted TNF‐ɑ/TNFR1/caspase‐8‐mediated apoptosis by increasing macrophages secretion of TNF‐ɑ and inhibited cell migration and invasion regulated by the WNT/β‐catenin‐EMT pathway. Nr‐CWS also reduced the expression of the cervical carcinoma genes E6 and E7 thereby increasing expression of p53 gene and decreasing expression of PD‐L1 gene. In vivo, Nr‐CWS inhibited tumor growth and decreased the expression of E6, E7, PD‐L1, P16, Ki67, and PCNA in tumors. Conclusions Therefore, our results suggest that Nr‐CWS can promote apoptosis of cervical carcinoma cells by enhancing the antitumor effect of dendritic cells and macrophages.
Collapse
Affiliation(s)
- Siyang Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Han Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yisi Liu
- Department of Obstetrics and Gynecology, Cancer Hospital of China Medical University, Shenyang, China
| | - Tao Tao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhi Zeng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yingying Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Min Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
The Impact of NLRP3 Activation on Hematopoietic Stem Cell Transplantation. Int J Mol Sci 2021; 22:ijms222111845. [PMID: 34769275 PMCID: PMC8584591 DOI: 10.3390/ijms222111845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 01/12/2023] Open
Abstract
NLR family pyrin domain-containing 3 (NLRP3) is an intracellular protein that after recognizing a broad spectrum of stressors, such as microbial motifs and endogenous danger signals, promotes the activation and release of the pro-inflammatory cytokines IL-1β and IL-18, thus playing an essential role in the innate immune response. Several blood cell types, including macrophages, dendritic cells, and hematopoietic stem and progenitor cells (HSPCs), express NLRP3, where it has been implicated in various physiological and pathological processes. For example, NLRP3 participates in the development and expansion of HSPCs, and their release from bone marrow into the peripheral blood has been implicated in certain hematological disorders including various types of leukemia. In addition, accumulating evidence indicates that activation of NLRP3 plays a pivotal role in the development of transplant complications in patients receiving hematopoietic stem cell transplantation (HSCT) including graft versus host disease, severe infections, and transplant-related mortality. The majority of these complications are triggered by the severe tissue damage derived from the conditioning regimens utilized in HSCT which, in turn, activates NLRP3 and, ultimately, promotes the release of proinflammatory cytokines such as IL-1β and IL-18. Here, we summarize the implications of NLRP3 in HSCT with an emphasis on the involvement of this inflammasome component in transplant complications.
Collapse
|
19
|
Gastaldi S, Boscaro V, Gianquinto E, Sandall CF, Giorgis M, Marini E, Blua F, Gallicchio M, Spyrakis F, MacDonald JA, Bertinaria M. Chemical Modulation of the 1-(Piperidin-4-yl)-1,3-dihydro-2 H-benzo[d]imidazole-2-one Scaffold as a Novel NLRP3 Inhibitor. Molecules 2021; 26:molecules26133975. [PMID: 34209843 PMCID: PMC8271538 DOI: 10.3390/molecules26133975] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
In the search for new chemical scaffolds able to afford NLRP3 inflammasome inhibitors, we used a pharmacophore-hybridization strategy by combining the structure of the acrylic acid derivative INF39 with the 1-(piperidin-4-yl)1,3-dihydro-2H-benzo[d]imidazole-2-one substructure present in HS203873, a recently identified NLRP3 binder. A series of differently modulated benzo[d]imidazole-2-one derivatives were designed and synthesised. The obtained compounds were screened in vitro to test their ability to inhibit NLRP3-dependent pyroptosis and IL-1β release in PMA-differentiated THP-1 cells stimulated with LPS/ATP. The selected compounds were evaluated for their ability to reduce the ATPase activity of human recombinant NLRP3 using a newly developed assay. From this screening, compounds 9, 13 and 18, able to concentration-dependently inhibit IL-1β release in LPS/ATP-stimulated human macrophages, emerged as the most promising NLRP3 inhibitors of the series. Computational simulations were applied for building the first complete model of the NLRP3 inactive state and for identifying possible binding sites available to the tested compounds. The analyses led us to suggest a mechanism of protein–ligand binding that might explain the activity of the compounds.
Collapse
Affiliation(s)
- Simone Gastaldi
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Torino, Italy; (S.G.); (V.B.); (E.G.); (M.G.); (E.M.); (F.B.); (M.G.); (F.S.)
| | - Valentina Boscaro
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Torino, Italy; (S.G.); (V.B.); (E.G.); (M.G.); (E.M.); (F.B.); (M.G.); (F.S.)
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Torino, Italy; (S.G.); (V.B.); (E.G.); (M.G.); (E.M.); (F.B.); (M.G.); (F.S.)
| | - Christina F. Sandall
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada; (C.F.S.); (J.A.M.)
| | - Marta Giorgis
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Torino, Italy; (S.G.); (V.B.); (E.G.); (M.G.); (E.M.); (F.B.); (M.G.); (F.S.)
| | - Elisabetta Marini
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Torino, Italy; (S.G.); (V.B.); (E.G.); (M.G.); (E.M.); (F.B.); (M.G.); (F.S.)
| | - Federica Blua
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Torino, Italy; (S.G.); (V.B.); (E.G.); (M.G.); (E.M.); (F.B.); (M.G.); (F.S.)
| | - Margherita Gallicchio
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Torino, Italy; (S.G.); (V.B.); (E.G.); (M.G.); (E.M.); (F.B.); (M.G.); (F.S.)
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Torino, Italy; (S.G.); (V.B.); (E.G.); (M.G.); (E.M.); (F.B.); (M.G.); (F.S.)
| | - Justin A. MacDonald
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada; (C.F.S.); (J.A.M.)
| | - Massimo Bertinaria
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Torino, Italy; (S.G.); (V.B.); (E.G.); (M.G.); (E.M.); (F.B.); (M.G.); (F.S.)
- Correspondence: ; Tel.: +39-011-6707146
| |
Collapse
|