1
|
Cai C, Wang M, Wang L, Guo J, Wang L, Zhang Y, Wu G, Hua B, Stuart MAC, Guo X, Cao L, Yan Z. Zwitterionic Brush-Grafted Interfacial Bio-Lubricant Evades Complement C3-Mediated Macrophage Phagocytosis for Osteoarthritis Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2501137. [PMID: 40304130 DOI: 10.1002/adma.202501137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/10/2025] [Indexed: 05/02/2025]
Abstract
Administering a bio-lubricant is a promising therapeutic approach for the treatment of osteoarthritis (OA), in particular, if it can both manage symptoms and halt disease progression. However, the clearance of these bio-lubricants mediated by synovial macrophages leads to reduced therapeutic efficiency and adverse inflammatory responses. Herein, it is shown that this process is predominantly mediated by the specific binding of complement C3 (on nanoparticle) and CD11b (on macrophage). More importantly, through a systematic evaluation of various interface modifications, a macrophage-evading nanoparticle strategy is proposed, which not only minimizes friction, but also largely suppresses C3 adsorption. It involves employing a zwitterionic poly-2-methacryloyloxyethyl phosphorylcholine (PMPC) brush layer grafted from a crosslinked gelatin core. In vitro studies demonstrate that such a nanoparticle lubricant can evade macrophage phagocytosis and further prevent the pro-inflammatory M1 polarization and subsequent harmful release of cytokines. In vivo studies show that the designed PMPC brush layer effectively mitigates synovial inflammation, alleviates OA-associated pain, and protects cartilage from degeneration, thus preventing OA progression. These findings clarify the pivotal role of complement C3-mediated macrophage recognition in nanoparticles clearance and offer a promising nanoparticle design strategy to restore joint lubrication.
Collapse
Affiliation(s)
- Chuandong Cai
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Mingwei Wang
- School of Chemical Engineering, State-Key Laboratory of Chemical Engineering and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Department of Dentistry-Regenerative Biomaterials, Radboud University Medical Center, Nijmegen, 6525 EX, The Netherlands
| | - Luman Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
- Department of Immunology, School of Basic Medical Sciences, Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jiangtao Guo
- School of Chemical Engineering, State-Key Laboratory of Chemical Engineering and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Lipeng Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Yingkai Zhang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Guohao Wu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Bingxuan Hua
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Martien A Cohen Stuart
- School of Chemical Engineering, State-Key Laboratory of Chemical Engineering and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Physical Chemistry and Soft Matter, Wageningen University and Research, Wageningen, 6708 WE, The Netherlands
| | - Xuhong Guo
- School of Chemical Engineering, State-Key Laboratory of Chemical Engineering and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, 410205, China
| | - Lu Cao
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Zuoqin Yan
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
- Department of Orthopaedic Surgery, Shanghai Geriatric Medical Center, 2560 Chunshen Road, Shanghai, 201104, China
| |
Collapse
|
2
|
Rosell A, Krygowska AA, Alcón Pérez M, Cuesta C, Voisin MB, de Paz J, Sanz-Fraile H, Rajeeve V, Carreras-González A, Berral-González A, Swinyard O, Gabandé-Rodríguez E, Downward J, Alcaraz J, Anguita J, García-Macías C, De Las Rivas J, Cutillas PR, Castellano Sanchez E. RAS-p110α signalling in macrophages is required for effective inflammatory response and resolution of inflammation. eLife 2025; 13:RP94590. [PMID: 40272400 PMCID: PMC12021417 DOI: 10.7554/elife.94590] [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: 04/25/2025] Open
Abstract
Macrophages are crucial in the body's inflammatory response, with tightly regulated functions for optimal immune system performance. Our study reveals that the RAS-p110α signalling pathway, known for its involvement in various biological processes and tumourigenesis, regulates two vital aspects of the inflammatory response in macrophages: the initial monocyte movement and later-stage lysosomal function. Disrupting this pathway, either in a mouse model or through drug intervention, hampers the inflammatory response, leading to delayed resolution and the development of more severe acute inflammatory reactions in live models. This discovery uncovers a previously unknown role of the p110α isoform in immune regulation within macrophages, offering insight into the complex mechanisms governing their function during inflammation and opening new avenues for modulating inflammatory responses.
Collapse
Affiliation(s)
- Alejandro Rosell
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Agata Adelajda Krygowska
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Marta Alcón Pérez
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Cristina Cuesta
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Mathieu-Benoit Voisin
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Juan de Paz
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Héctor Sanz-Fraile
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de BarcelonaBarcelonaSpain
| | - Vinothini Rajeeve
- Centre for Cancer Genomics and Computational Biology, Cell Signalling and Proteomics Laboratory, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Ana Carreras-González
- Bioinformatics and Functional Genomics, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de SalamancaSalamancaSpain
| | | | - Ottilie Swinyard
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Enrique Gabandé-Rodríguez
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Julian Downward
- Oncogene Biology Laboratory, Francis Crick InstituteLondonUnited Kingdom
| | - Jordi Alcaraz
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de BarcelonaBarcelonaSpain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST)BarcelonaSpain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Lab, CIC bioGUNEDerioSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
- Pathology Unit, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de SalamancaSalamancaSpain
| | - Carmen García-Macías
- Pathology Unit, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de SalamancaSalamancaSpain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de SalamancaSalamancaSpain
| | - Pedro R Cutillas
- Centre for Cancer Genomics and Computational Biology, Cell Signalling and Proteomics Laboratory, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Esther Castellano Sanchez
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| |
Collapse
|
3
|
Zhao H, Lv Y, Xu J, Song X, Wang Q, Zhai X, Ma X, Qiu J, Cui L, Sun Y. The activation of microglia by the complement system in neurodegenerative diseases. Ageing Res Rev 2025; 104:102636. [PMID: 39647582 DOI: 10.1016/j.arr.2024.102636] [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: 11/22/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
Neurodegenerative diseases (NDDs) are a group of neurological disorders characterized by the progressive loss of neuronal structure and function, leading to cognitive and behavioral impairments. Despite significant research advancements, there is currently no definitive cure for NDDs. With global aging on the rise, the burden of these diseases is becoming increasingly severe, highlighting the urgency of understanding their pathogenesis and developing effective therapeutic strategies. Microglia, specialized macrophages in the central nervous system, play a dual role in maintaining neural homeostasis. They are involved in clearing cellular debris and apoptotic cells, but in their activated state, they release inflammatory factors that contribute significantly to neuroinflammation. The complement system (CS), a critical component of the innate immune system, assists in clearing damaged cells and proteins. However, excessive or uncontrolled activation of the CS can lead to chronic neuroinflammation, exacerbating neuronal damage. This review aims to explore the roles of microglia and the CS in the progression of NDDs, with a specific focus on the mechanisms through which the CS activates microglia by modulating mitochondrial function. Understanding these interactions may provide insights into potential therapeutic targets for mitigating neuroinflammation and slowing neurodegeneration.
Collapse
Affiliation(s)
- He Zhao
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Yayun Lv
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Jiasen Xu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Xiaoyu Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Qi Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Xiaoyu Zhai
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Xiaohui Ma
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Jingjing Qiu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China.
| | - Limei Cui
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China.
| | - Yan Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China.
| |
Collapse
|
4
|
Troise D, Allegra C, Cirolla LA, Mercuri S, Infante B, Castellano G, Stallone G. Exploring Potential Complement Modulation Strategies for Ischemia-Reperfusion Injury in Kidney Transplantation. Antioxidants (Basel) 2025; 14:66. [PMID: 39857400 PMCID: PMC11761266 DOI: 10.3390/antiox14010066] [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: 12/07/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
The complement system plays a crucial role in regulating the inflammatory responses in kidney transplantation, potentially contributing to early decline in kidney function. Ischemia-reperfusion injury (IRI) is among the factors affecting graft outcomes and a primary contributor to delayed graft function. Complement activation, particularly the alternative pathway, participates in the pathogenesis of IRI, involving all kidney compartments. In particular, tubular epithelial cells often acquire a dysfunctional phenotype that can exacerbate complement activation and kidney damage. Currently, complement-modulating drugs are under investigation for the treatment of kidney diseases. Many of these drugs have shown potential therapeutic benefits, but no effective clinical treatments for renal IRI have been identified yet. In this review, we will explore drugs that target complement factors, complement receptors, and regulatory proteins, aiming to highlight their potential value in improving the management of renal IRI.
Collapse
Affiliation(s)
- Dario Troise
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Costanza Allegra
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Luciana Antonia Cirolla
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Silvia Mercuri
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Giuseppe Castellano
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, 20122 Milan, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
5
|
Zhang F, Xia Y, Su J, Quan F, Zhou H, Li Q, Feng Q, Lin C, Wang D, Jiang Z. Neutrophil diversity and function in health and disease. Signal Transduct Target Ther 2024; 9:343. [PMID: 39638788 PMCID: PMC11627463 DOI: 10.1038/s41392-024-02049-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/21/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Neutrophils, the most abundant type of granulocyte, are widely recognized as one of the pivotal contributors to the acute inflammatory response. Initially, neutrophils were considered the mobile infantry of the innate immune system, tasked with the immediate response to invading pathogens. However, recent studies have demonstrated that neutrophils are versatile cells, capable of regulating various biological processes and impacting both human health and disease. Cytokines and other active mediators regulate the functional activity of neutrophils by activating multiple receptors on these cells, thereby initiating downstream signal transduction pathways. Dysfunctions in neutrophils and disruptions in neutrophil homeostasis have been implicated in the pathogenesis of numerous diseases, including cancer and inflammatory disorders, often due to aberrant intracellular signaling. This review provides a comprehensive synthesis of neutrophil biological functions, integrating recent advancements in this field. Moreover, it examines the biological roles of receptors on neutrophils and downstream signaling pathways involved in the regulation of neutrophil activity. The pathophysiology of neutrophils in numerous human diseases and emerging therapeutic approaches targeting them are also elaborated. This review also addresses the current limitations within the field of neutrophil research, highlighting critical gaps in knowledge that warrant further investigation. In summary, this review seeks to establish a comprehensive and multidimensional model of neutrophil regulation, providing new perspectives for potential clinical applications and further research.
Collapse
Affiliation(s)
- Fengyuan Zhang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yidan Xia
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiayang Su
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Fushi Quan
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Hengzong Zhou
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qirong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qiang Feng
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Chao Lin
- School of Grain Science and Technology, Jilin Business and Technology College, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China.
| | - Ziping Jiang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China.
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
6
|
Dong Q, Zhou J, Feng M, Kong L, Fang B, Zhang Z. A review of bacterial and osteoclast differentiation in bone infection. Microb Pathog 2024; 197:107102. [PMID: 39505086 DOI: 10.1016/j.micpath.2024.107102] [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: 03/29/2024] [Revised: 10/18/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
Bone infections are characterized by bacterial invasion of the bone microenvironment and subsequent bone structure deterioration. This holds significance because osteoclasts, which are the only cells responsible for bone resorption, are abnormally stimulated during bone infections. Multiple communication factors secreted by bone stromal cells regulate the membrane of osteoclast progenitor cells, thereby maintaining bone homeostasis through the expression of many types of receptors. During infection, the immunoinflammatory response triggered by bacterial invasion and multiple virulence factors of bacterial origin can disrupt osteoclast homeostasis. Therefore, clarifying the pathways through which bacteria affect osteoclasts can offer a theoretical basis for preventing and treating bone infections. This review summarizes studies investigating bone destruction caused by different bacterial infections. In conclusion, bacteria can affect osteoclast metabolic activity through multiple pathways, including direct contact, release of virulence factors, induction of immunoinflammatory responses, influence on bone stromal cell metabolism, and intracellular infections.
Collapse
Affiliation(s)
- Qi Dong
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Jiuqin Zhou
- Department of Infectious Disease of Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Mingzhe Feng
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Lingqiang Kong
- Department of Orthopedics, the Central Hospital Affiliated to Shaoxing University, Shaoxing, 312030, China.
| | - Bin Fang
- Department of Orthopedics, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310000, China.
| | - Zhen Zhang
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China.
| |
Collapse
|
7
|
Colavite PM, Azevedo MDCS, Francisconi CF, Fonseca AC, Tabanez AP, Melchiades JL, Passadori DC, Borrego A, De Franco M, Trombone APF, Garlet GP. Intermediate and Transitory Inflammation Mediate Proper Alveolar Bone Healing Outcome in Contrast to Extreme Low/High Responses: Evidence from Mice Strains Selected for Distinct Inflammatory Phenotypes. BIOLOGY 2024; 13:972. [PMID: 39765639 PMCID: PMC11673754 DOI: 10.3390/biology13120972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025]
Abstract
Alveolar bone healing is influenced by various local and systemic factors, including the local inflammatory response. This study aimed to evaluate the role of inflammatory responsiveness in alveolar bone healing using 8-week-old male and female mice (N = 5/time/group) strains selected for maximum (AIRmax) or minimum (AIRmin) acute inflammatory response carrying distinct homozygous RR/SS Slc11a1 genotypes, namely AIRminRR, AIRminSS, AIRmaxRR, and AIRmaxSS mice. After upper right incisor extraction, bone healing was analyzed at 0, 3, 7, and 14 days using micro-computed tomography, histomorphometry, birefringence, immunohistochemistry, and PCRArray analysis. AIRmaxSS and AIRminRR presented the highest and lowest inflammatory readouts, respectively, associated with lowest repair levels in both strains, while intermediate inflammatory phenotypes observed in AIRminSS and AIRmaxRR were associated with higher repair levels in such strains. The better healing outcomes are associated with intermediate inflammatory cell counts, a balanced expression of pro- and anti-inflammatory cytokines and chemokines, increased expression of growth and osteogenic factors and MSCs markers. Our results demonstrate that extreme high and low inflammatory responses are not ideal for a proper bone repair outcome, while an intermediate and transitory inflammation is associated with a proper alveolar bone healing outcome.
Collapse
Affiliation(s)
- Priscila Maria Colavite
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru CEP 17012-901, SP, Brazil; (P.M.C.); (M.d.C.S.A.); (A.C.F.); (D.C.P.)
| | - Michelle de Campos Soriani Azevedo
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru CEP 17012-901, SP, Brazil; (P.M.C.); (M.d.C.S.A.); (A.C.F.); (D.C.P.)
| | - Carolina Fávaro Francisconi
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru CEP 17012-901, SP, Brazil; (P.M.C.); (M.d.C.S.A.); (A.C.F.); (D.C.P.)
| | - Angélica Cristina Fonseca
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru CEP 17012-901, SP, Brazil; (P.M.C.); (M.d.C.S.A.); (A.C.F.); (D.C.P.)
| | - André Petenucci Tabanez
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru CEP 17012-901, SP, Brazil; (P.M.C.); (M.d.C.S.A.); (A.C.F.); (D.C.P.)
| | - Jéssica Lima Melchiades
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru CEP 17012-901, SP, Brazil; (P.M.C.); (M.d.C.S.A.); (A.C.F.); (D.C.P.)
| | - Daniela Carignatto Passadori
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru CEP 17012-901, SP, Brazil; (P.M.C.); (M.d.C.S.A.); (A.C.F.); (D.C.P.)
| | - Andrea Borrego
- Laboratory of Immunogenetics, Butantan Institute, Secretary of Health, Government of the State of São Paulo, Sao Paulo CEP 05503-900, SP, Brazil; (A.B.); (M.D.F.)
| | - Marcelo De Franco
- Laboratory of Immunogenetics, Butantan Institute, Secretary of Health, Government of the State of São Paulo, Sao Paulo CEP 05503-900, SP, Brazil; (A.B.); (M.D.F.)
- Pasteur Institute, Diagnostic Section, Sao Paulo CEP 01311-000, SP, Brazil
| | | | - Gustavo Pompermaier Garlet
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru CEP 17012-901, SP, Brazil; (P.M.C.); (M.d.C.S.A.); (A.C.F.); (D.C.P.)
| |
Collapse
|
8
|
Yang Y, Guo L, Wei L, Yu J, Zhu S, Li X, Liu J, Liang R, Peng W, Ge F, Zhang J. Da-yuan-yin decoction alleviates ulcerative colitis by inhibiting complement activation, LPS-TLR4/NF-κB signaling pathway and NET formation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118392. [PMID: 38797378 DOI: 10.1016/j.jep.2024.118392] [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: 02/04/2024] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Da-yuan-yin decoction (DYY) is a classical traditional Chinese medicine prescription for ulcerative colitis (UC). AIM OF STUDY This study explored the protective effects and mechanisms of DYY on UC. MATERIALS AND METHODS The mice were fed 2.5% dextran sulfate sodium (DSS) for 7 days to establish UC. On the second day, DYY (0.4 g/kg, 0.8 g/kg, 1.6 g/kg) was orally administered daily for 7 consecutive days. The colon tissues and serum were measured by histopathological examination and biochemical analysis. RESULTS DYY significantly reduced the disease activity index (DAI) and severity of colon shortening and alleviated pathological changes in the colon tissue. DYY restored the protein expression of intestinal tight junction (TJ) protein (ZO-1, occludin and claudin-3). DYY remarkably decreased the level of lipopolysaccharide (LPS), Lactic acid (LA), circulating free DNA (cfDNA), complement (C3, C3a, C3c, C3aR1, C5a and C5aR1) and regulated the levels of inflammatory cytokines in serum. DYY significantly inhibited the expressions of nuclear factor kappa-B p65 (NF-κB p65) and Toll-like receptor 4 (TLR4), citrullinated histone H3 (CitH3) and myeloperoxidase (MPO), reactive oxygen species (ROS) peptidylarginine deiminase 4 (PAD4) and CD 11b, the mRNA levels of PADI4, MPO and ELANE in colon tissues. CONCLUSIONS DYY significantly attenuated DSS-induced UC, which was related with regulating the inflammatory response by the inhibition of complement activation, the LPS-TLR4/NF-κB signaling pathway and neutrophil extracellular traps (NETs) formation. DYY is a potential therapeutic agent for UC.
Collapse
Affiliation(s)
- Yun Yang
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China.
| | - Lengqiu Guo
- Suzhou Vocational Health College, Suzhou, 215009, China
| | - Lan Wei
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Jinghua Yu
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Song Zhu
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Xinyi Li
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Jiangyun Liu
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Rui Liang
- Suzhou Vocational Health College, Suzhou, 215009, China
| | - Wei Peng
- The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| | - Fei Ge
- Department of Gastroenterology, Haian Hospital of Traditional Chinese Medicine, Nantong, 226000, China.
| | - Jian Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
9
|
Shah MM, Layhadi JA, Hourcade DE, Fulton WT, Tan TJ, Dunham D, Chang I, Vel MS, Fernandes A, Lee AS, Liu J, Arunachalam PS, Galli SJ, Boyd SD, Pulendran B, Davis MM, O’Hara R, Park H, Mitchell LM, Akk A, Patterson A, Jerath MR, Monroy JM, Ren Z, Kendall PL, Durham SR, Fedina A, Gibbs BF, Agache I, Chinthrajah S, Sindher SB, Heider A, Akdis CA, Shamji MH, Pham CT, Nadeau KC. Elucidating allergic reaction mechanisms in response to SARS-CoV-2 mRNA vaccination in adults. Allergy 2024; 79:2502-2523. [PMID: 39033312 PMCID: PMC11368657 DOI: 10.1111/all.16231] [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: 03/13/2024] [Revised: 05/31/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND During the COVID-19 pandemic, novel nanoparticle-based mRNA vaccines were developed. A small number of individuals developed allergic reactions to these vaccines although the mechanisms remain undefined. METHODS To understand COVID-19 vaccine-mediated allergic reactions, we enrolled 19 participants who developed allergic events within 2 h of vaccination and 13 controls, nonreactors. Using standard hemolysis assays, we demonstrated that sera from allergic participants induced stronger complement activation compared to nonallergic subjects following ex vivo vaccine exposure. RESULTS Vaccine-mediated complement activation correlated with anti-polyethelyne glycol (PEG) IgG (but not IgM) levels while anti-PEG IgE was undetectable in all subjects. Depletion of total IgG suppressed complement activation in select individuals. To investigate the effects of vaccine excipients on basophil function, we employed a validated indirect basophil activation test that stratified the allergic populations into high and low responders. Complement C3a and C5a receptor blockade in this system suppressed basophil response, providing strong evidence for complement involvement in vaccine-mediated basophil activation. Single-cell multiome analysis revealed differential expression of genes encoding the cytokine response and Toll-like receptor (TLR) pathways within the monocyte compartment. Differential chromatin accessibility for IL-13 and IL-1B genes was found in allergic and nonallergic participants, suggesting that in vivo, epigenetic modulation of mononuclear phagocyte immunophenotypes determines their subsequent functional responsiveness, contributing to the overall physiologic manifestation of vaccine reactions. CONCLUSION These findings provide insights into the mechanisms underlying allergic reactions to COVID-19 mRNA vaccines, which may be used for future vaccine strategies in individuals with prior history of allergies or reactions and reduce vaccine hesitancy.
Collapse
Affiliation(s)
- Mihir M. Shah
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
- These authors contributed equally to this work
| | - Janice A. Layhadi
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
- These authors contributed equally to this work
| | - Dennis E. Hourcade
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
- These authors contributed equally to this work
| | - William T. Fulton
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Tiak Ju Tan
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Diane Dunham
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Iris Chang
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Monica S. Vel
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Andrea Fernandes
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Alexandra S. Lee
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - James Liu
- Stanford Health Library; Stanford, CA, USA
| | - Prabhu S. Arunachalam
- Institute for Immunity, Transplantation and Infection,
Stanford University; Stanford, CA, USA
| | - Stephen J. Galli
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
- Department of Pathology, Stanford University School of
Medicine; Stanford, California, USA
- Department of Microbiology and Immunology, Stanford
University School of Medicine; Stanford, California, USA
| | - Scott D. Boyd
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
- Department of Pathology, Stanford University School of
Medicine; Stanford, California, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection,
Stanford University; Stanford, CA, USA
- Department of Pathology, Stanford University School of
Medicine; Stanford, California, USA
- Department of Microbiology and Immunology, Stanford
University School of Medicine; Stanford, California, USA
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection,
Stanford University; Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford
University School of Medicine; Stanford, California, USA
| | - Ruth O’Hara
- Department of Veteran’s Administration and
Dean’s Office, Stanford University; Stanford, CA, USA
| | - Helen Park
- Veterans Affairs Palo Alto Health Care System; Palo Alto,
CA, USA
| | - Lynne M. Mitchell
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
| | - Antonina Akk
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
| | - Alexander Patterson
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Maya R. Jerath
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Jennifer M. Monroy
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Zhen Ren
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Peggy L. Kendall
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Stephen R. Durham
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Aleksandra Fedina
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Bernhard F Gibbs
- Department of Human Medicine, School of Medicine and
Health Sciences, Carl von Ossietzky University of Oldenburg; Oldenburg,
Germany
- Canterbury Christ Church University, UK
| | - Ioana Agache
- Faculty of Medicine, Transilvania University; Brasov,
Romania
| | | | | | - Anja Heider
- Swiss Institute of Allergy and Asthma Research (SIAF),
University of Zurich; Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF),
University of Zurich; Davos, Switzerland
| | - Mohamed H. Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
- These authors contributed equally to this work
| | - Christine T.N. Pham
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
- These authors contributed equally to this work
| | - Kari C. Nadeau
- Harvard T.H. Chan School of Public Health, Harvard
University; Cambridge, Massachusetts
- These authors contributed equally to this work
| |
Collapse
|
10
|
Dinice L, Esposito G, Cacciamani A, Balzamino BO, Cosimi P, Cafiero C, Ripandelli G, Micera A. TLR2 and TLR4 Are Expressed in Epiretinal Membranes: Possible Links with Vitreous Levels of Complement Fragments and DAMP-Related Proteins. Int J Mol Sci 2024; 25:7732. [PMID: 39062973 PMCID: PMC11276880 DOI: 10.3390/ijms25147732] [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: 05/24/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Previous studies reported the expression of toll-like receptors (TLRs), merely TLR2 and TLR4, and complement fragments (C3a, C5b9) in vitreoretinal disorders. Other than pathogens, TLRs can recognize endogenous products of tissue remodeling as damage-associated molecular pattern (DAMPs). The aim of this study was to confirm the expression of TLR2 and TLR4 in the fibrocellular membranes and vitreal fluids (soluble TLRs) of patients suffering of epiretinal membranes (ERMs) and assess their association with disease severity, complement fragments and inflammatory profiles. Twenty (n = 20) ERMs and twelve (n = 12) vitreous samples were collected at the time of the vitrectomy. Different severity-staged ERMs were processed for: immunolocalization (IF), transcriptomic (RT-PCR) and proteomics (ELISA, IP/WB, Protein Chip Array) analysis. The investigation of targets included TLR2, TLR4, C3a, C5b9, a few selected inflammatory biomarkers (Eotaxin-2, Rantes, Vascular Endothelial Growth Factor (VEGFA), Vascular Endothelial Growth Factor receptor (VEGFR2), Interferon-γ (IFNγ), Interleukin (IL1β, IL12p40/p70)) and a restricted panel of matrix enzymes (Matrix metalloproteinases (MMPs)/Tissue Inhibitor of Metallo-Proteinases (TIMPs)). A reduced cellularity was observed as function of ERM severity. TLR2, TLR4 and myD88 transcripts/proteins were detected in membranes and decreased upon disease severity. The levels of soluble TLR2 and TLR4, as well as C3a, C5b9, Eotaxin-2, Rantes, VEGFA, VEGFR2, IFNγ, IL1β, IL12p40/p70, MMP7 and TIMP2 levels were changed in vitreal samples. Significant correlations were observed between TLRs and complement fragments and between TLRs and some inflammatory mediators. Our findings pointed at TLR2 and TLR4 over-expression at early stages of ERM formation, suggesting the participation of the local immune response in the severity of disease. These activations at the early-stage of ERM formation suggest a potential persistence of innate immune response in the early phases of fibrocellular membrane formation.
Collapse
Affiliation(s)
- Lucia Dinice
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS—Fondazione Bietti, 00184 Rome, Italy; (L.D.); (G.E.); (B.O.B.)
| | - Graziana Esposito
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS—Fondazione Bietti, 00184 Rome, Italy; (L.D.); (G.E.); (B.O.B.)
| | - Andrea Cacciamani
- Surgical Retina Research Unit, IRCCS—Fondazione Bietti, 00184 Rome, Italy; (A.C.); (P.C.); (G.R.)
| | - Bijorn Omar Balzamino
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS—Fondazione Bietti, 00184 Rome, Italy; (L.D.); (G.E.); (B.O.B.)
| | - Pamela Cosimi
- Surgical Retina Research Unit, IRCCS—Fondazione Bietti, 00184 Rome, Italy; (A.C.); (P.C.); (G.R.)
| | - Concetta Cafiero
- Anatomic Pathology Unit, Fabrizio Spaziani Hospital, 03100 Frosinone, Italy;
| | - Guido Ripandelli
- Surgical Retina Research Unit, IRCCS—Fondazione Bietti, 00184 Rome, Italy; (A.C.); (P.C.); (G.R.)
| | - Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS—Fondazione Bietti, 00184 Rome, Italy; (L.D.); (G.E.); (B.O.B.)
| |
Collapse
|
11
|
Alhamdan F, Bayarsaikhan G, Yuki K. Toll-like receptors and integrins crosstalk. Front Immunol 2024; 15:1403764. [PMID: 38915411 PMCID: PMC11194410 DOI: 10.3389/fimmu.2024.1403764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/24/2024] [Indexed: 06/26/2024] Open
Abstract
Immune system recognizes invading microbes at both pathogen and antigen levels. Toll-like receptors (TLRs) play a key role in the first-line defense against pathogens. Major functions of TLRs include cytokine and chemokine production. TLRs share common downstream signaling pathways with other receptors. The crosstalk revolving around TLRs is rather significant and complex, underscoring the intricate nature of immune system. The profiles of produced cytokines and chemokines via TLRs can be affected by other receptors. Integrins are critical heterodimeric adhesion molecules expressed on many different cells. There are studies describing synergetic or inhibitory interplay between TLRs and integrins. Thus, we reviewed the crosstalk between TLRs and integrins. Understanding the nature of the crosstalk could allow us to modulate TLR functions via integrins.
Collapse
Affiliation(s)
- Fahd Alhamdan
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia, Boston Children’s Hospital, Boston, MA, United States
- Department of Anesthesia and Immunology, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Ganchimeg Bayarsaikhan
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia, Boston Children’s Hospital, Boston, MA, United States
- Department of Anesthesia and Immunology, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia, Boston Children’s Hospital, Boston, MA, United States
- Department of Anesthesia and Immunology, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
12
|
Wei Y, Guo J, Meng T, Gao T, Mai Y, Zuo W, Yang J. The potential application of complement inhibitors-loaded nanosystem for autoimmune diseases via regulation immune balance. J Drug Target 2024; 32:485-498. [PMID: 38491993 DOI: 10.1080/1061186x.2024.2332730] [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: 12/20/2023] [Accepted: 03/14/2024] [Indexed: 03/18/2024]
Abstract
The complement is an important arm of the innate immune system, once activated, the complement system rapidly generates large quantities of protein fragments that are potent mediators of inflammation. Recent studies have shown that over-activated complement is the main proinflammatory system of autoimmune diseases (ADs). In addition, activated complements interact with autoantibodies, immune cells exacerbate inflammation, further worsening ADs. With the increasing threat of ADs to human health, complement-based immunotherapy has attracted wide attention. Nevertheless, efficient and targeted delivery of complement inhibitors remains a significant challenge owing to their inherent poor targeting, degradability, and low bioavailability. Nanosystems offer innovative solutions to surmount these obstacles and amplify the potency of complement inhibitors. This prime aim to present the current knowledge of complement in ADs, analyse the function of complement in the pathogenesis and treatment of ADs, we underscore the current situation of nanosystems assisting complement inhibitors in the treatment of ADs. Considering technological, physiological, and clinical validation challenges, we critically appraise the challenges for successfully translating the findings of preclinical studies of these nanosystem assisted-complement inhibitors into the clinic, and future perspectives were also summarised. (The graphical abstract is by BioRender.).
Collapse
Affiliation(s)
- Yaya Wei
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jueshuo Guo
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Tingting Meng
- Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ting Gao
- Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yaping Mai
- School of Science and Technology Centers, Ningxia Medical University, Yinchuan, China
| | - Wenbao Zuo
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jianhong Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
13
|
Inomata M, Abe M, Kawase Y, Hayashi T, Amano S, Sakagami H. Dectin-1/SYK Activation Induces Antimicrobial Peptide and Negative Regulator of NF-κB Signaling in Human Oral Epithelial Cells. In Vivo 2024; 38:1042-1048. [PMID: 38688646 PMCID: PMC11059888 DOI: 10.21873/invivo.13537] [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: 12/19/2023] [Revised: 02/03/2024] [Accepted: 02/08/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND/AIM Oral epithelial cells serve as the primary defense against microbial exposure in the oral cavity, including the fungus Candida albicans. Dectin-1 is crucial for recognition of β-glucan in fungi. However, expression and function of Dectin-1 in oral epithelial cells remain unclear. MATERIALS AND METHODS We assessed Dectin-1 expression in Ca9-22 (gingiva), HSC-2 (mouth), HSC-3 (tongue), and HSC-4 (tongue) human oral epithelial cells using flow cytometry and real-time polymerase chain reaction. Cell treated with β-glucan-rich zymosan were evaluated using real-time polymerase chain reaction. Phosphorylation of spleen-associated tyrosine kinase (SYK) was analyzed by western blotting. RESULTS Dectin-1 was expressed in all four cell types, with high expression in Ca9-22 and HSC-2. In Ca9-22 cells, exposure to β-glucan-rich zymosan did not alter the mRNA expression of chemokines nor of interleukin (IL)6, IL8, IL1β, IL17A, and IL17F. Zymosan induced the expression of antimicrobial peptides β-defensin-1 and LL-37, but not S100 calcium-binding protein A8 (S100A8) and S100A9. Furthermore, the expression of cylindromatosis (CYLD), a negative regulator of nuclear factor kappa B (NF-κB) signaling, was induced. In HSC-2 cells, zymosan induced the expression of IL17A. The expression of tumor necrosis factor alpha-induced protein 3 (TNFAIP3), a negative regulator of NF-κB signaling, was also induced. Expression of other cytokines and antimicrobial peptides remained unchanged. Zymosan induced phosphorylation of SYK in Ca9-22 cells, as well as NF-κB. CONCLUSION Oral epithelial cells express Dectin-1 and recognize β-glucan, which activates SYK and induces the expression of antimicrobial peptides and negative regulators of NF-κB, potentially maintaining oral homeostasis.
Collapse
Affiliation(s)
- Megumi Inomata
- Division of Microbiology and Immunology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan;
| | - Masayo Abe
- Division of Microbiology and Immunology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Yasuko Kawase
- Division of Microbiology and Immunology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Toru Hayashi
- Department of Anatomy Science, School of Allied Health Sciences, Kitasato University, Kitasato, Japan
| | - Shigeru Amano
- Research Institute of Odontology (M-RIO), Meikai University School of Dentistry, Sakado, Japan
| | - Hiroshi Sakagami
- Research Institute of Odontology (M-RIO), Meikai University School of Dentistry, Sakado, Japan
| |
Collapse
|
14
|
Gurubaran IS. Mitochondrial damage and clearance in retinal pigment epithelial cells. Acta Ophthalmol 2024; 102 Suppl 282:3-53. [PMID: 38467968 DOI: 10.1111/aos.16661] [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: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 03/13/2024]
Abstract
Age-related macular degeneration (AMD) is a devastating eye disease that causes permanent vision loss in the central part of the retina, known as the macula. Patients with such severe visual loss face a reduced quality of life and are at a 1.5 times greater risk of death compared to the general population. Currently, there is no cure for or effective treatment for dry AMD. There are several mechanisms thought to underlie the disease, for example, ageing-associated chronic oxidative stress, mitochondrial damage, harmful protein aggregation and inflammation. As a way of gaining a better understanding of the molecular mechanisms behind AMD and thus developing new therapies, we have created a peroxisome proliferator-activated receptor gamma coactivator 1-alpha and nuclear factor erythroid 2-related factor 2 (PGC1α/NFE2L2) double-knockout (dKO) mouse model that mimics many of the clinical features of dry AMD, including elevated levels of oxidative stress markers, damaged mitochondria, accumulating lysosomal lipofuscin and extracellular drusen-like structures in retinal pigment epithelial cells (RPE). In addition, a human RPE cell-based model was established to examine the impact of non-functional intracellular clearance systems on inflammasome activation. In this study, we found that there was a disturbance in the autolysosomal machinery responsible for clearing mitochondria in the RPE cells of one-year-old PGC1α/NFE2L2-deficient mice. The confocal immunohistochemical analysis revealed an increase in autophagosome marker microtubule-associated proteins 1A/1B light chain 3B (LC3B) as well as multiple mitophagy markers such as PTE-induced putative kinase 1 (PINK1) and E3 ubiquitin ligase (PARKIN), along with signs of damaged mitochondria. However, no increase in autolysosome formation was detected, nor was there a colocalization of the lysosomal marker LAMP2 or the mitochondrial marker, ATP synthase β. There was an upregulation of late autolysosomal fusion Ras-related protein (Rab7) in the perinuclear space of RPE cells, together with autofluorescent aggregates. Additionally, we observed an increase in the numbers of Toll-like receptors 3 and 9, while those of NOD-like receptor 3 were decreased in PGC1α/NFE2L2 dKO retinal specimens compared to wild-type animals. There was a trend towards increased complement component C5a and increased involvement of the serine protease enzyme, thrombin, in enhancing the terminal pathway producing C5a, independent of C3. The levels of primary acute phase C-reactive protein and receptor for advanced glycation end products were also increased in the PGC1α/NFE2L2 dKO retina. Furthermore, selective proteasome inhibition with epoxomicin promoted both nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondrial-mediated oxidative stress, leading to the release of mitochondrial DNA to the cytosol, resulting in potassium efflux-dependent activation of the absent in melanoma 2 (AIM2) inflammasome and the subsequent secretion of interleukin-1β in ARPE-19 cells. In conclusion, the data suggest that there is at least a relative decrease in mitophagy, increases in the amounts of C5 and thrombin and decreased C3 levels in this dry AMD-like model. Moreover, selective proteasome inhibition evoked mitochondrial damage and AIM2 inflammasome activation in ARPE-19 cells.
Collapse
Affiliation(s)
- Iswariyaraja Sridevi Gurubaran
- Department of Medicine, Clinical Medicine Unit, University of Eastern Finland Institute of Clinical Medicine, Kuopio, Northern Savonia, Finland
| |
Collapse
|
15
|
Mattos-Graner RO, Klein MI, Alves LA. The complement system as a key modulator of the oral microbiome in health and disease. Crit Rev Microbiol 2024; 50:138-167. [PMID: 36622855 DOI: 10.1080/1040841x.2022.2163614] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/10/2023]
Abstract
In this review, we address the interplay between the complement system and host microbiomes in health and disease, focussing on oral bacteria known to contribute to homeostasis or to promote dysbiosis associated with dental caries and periodontal diseases. Host proteins modulating complement activities in the oral environment and expression profiles of complement proteins in oral tissues were described. In addition, we highlight a sub-set of bacterial proteins involved in complement evasion and/or dysregulation previously characterized in pathogenic species (or strains), but further conserved among prototypical commensal species of the oral microbiome. Potential roles of these proteins in host-microbiome homeostasis and in the emergence of commensal strain lineages with increased virulence were also addressed. Finally, we provide examples of how commensal bacteria might exploit the complement system in competitive or cooperative interactions within the complex microbial communities of oral biofilms. These issues highlight the need for studies investigating the effects of the complement system on bacterial behaviour and competitiveness during their complex interactions within oral and extra-oral host sites.
Collapse
Affiliation(s)
- Renata O Mattos-Graner
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Sao Paulo, Brazil
| | - Marlise I Klein
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Sao Paulo, Brazil
| | - Lívia Araújo Alves
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Sao Paulo, Brazil
- School of Dentistry, Cruzeiro do Sul University (UNICSUL), Sao Paulo, Brazil
| |
Collapse
|
16
|
Li XX, Fung JN, Clark RJ, Lee JD, Woodruff TM. Cell-intrinsic C5a synergizes with Dectin-1 in macrophages to mediate fungal killing. Proc Natl Acad Sci U S A 2024; 121:e2314627121. [PMID: 38252818 PMCID: PMC10835034 DOI: 10.1073/pnas.2314627121] [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: 08/24/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
The complement factor C5a is a core effector product of complement activation. C5a, acting through its receptors C5aR1 and C5aR2, exerts pleiotropic immunomodulatory functions in myeloid cells, which is vital for host defense against pathogens. Pattern-recognition receptors (PRRs) are similarly expressed by immune cells as detectors of pathogen-associated molecular patterns. Although there is evidence of cross talk between complement and PRR signaling pathways, knowledge of the full potential for C5a-PRR interaction is limited. In this study, we comprehensively investigated how C5a signaling through C5a receptors can modulate diverse PRR-mediated cytokine responses in human primary monocyte-derived macrophages and observed a powerful, concentration-dependent bidirectional effect of C5a on PRR activities. Unexpectedly, C5a synergized with Dectin-1, Mincle, and STING in macrophages to a much greater extent than TLRs. Notably, we also identified that selective Dectin-1 activation using depleted zymosan triggered macrophages to generate cell-intrinsic C5a, which acted on intracellular and cell surface C5aR1, to help sustain mitochondrial ROS generation, up-regulate TNFα production, and enhance fungal killing. This study adds further evidence to the holistic functions of C5a as a central immunomodulator and important orchestrator of pathogen sensing and killing by phagocytes.
Collapse
Affiliation(s)
- Xaria X. Li
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD4072, Australia
| | - Jenny N. Fung
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD4072, Australia
| | - Richard J. Clark
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD4072, Australia
| | - John D. Lee
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD4072, Australia
| | - Trent M. Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD4072, Australia
| |
Collapse
|
17
|
Dahmani M, Zhu JC, Cook JH, Riley SP. Anaphylatoxin signaling activates macrophages to control intracellular Rickettsia proliferation. Microbiol Spectr 2023; 11:e0253823. [PMID: 37855623 PMCID: PMC10714731 DOI: 10.1128/spectrum.02538-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/11/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE Pathogenic Rickettsia species are extremely dangerous bacteria that grow within the cytoplasm of host mammalian cells. In most cases, these bacteria are able to overpower the host cell and grow within the protected environment of the cytoplasm. However, a dramatic conflict occurs when Rickettsia encounter innate immune cells; the bacteria can "win" by taking over the host, or the bacteria can "lose" if the host cell efficiently fights the infection. This manuscript examines how the immune complement system is able to detect the presence of Rickettsia and alert nearby cells. Byproducts of complement activation called anaphylatoxins are signals that "activate" innate immune cells to mount an aggressive defensive strategy. This study enhances our collective understanding of the innate immune reaction to intracellular bacteria and will contribute to future efforts at controlling these dangerous infections.
Collapse
Affiliation(s)
- Mustapha Dahmani
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Jinyi C. Zhu
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Jack H. Cook
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Sean P. Riley
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
- Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| |
Collapse
|
18
|
Ma Y, Zhang K, Wu Y, Fu X, Liang S, Peng M, Guo J, Liu M. Revisiting the relationship between complement and ulcerative colitis. Scand J Immunol 2023; 98:e13329. [PMID: 38441324 DOI: 10.1111/sji.13329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/13/2023] [Accepted: 08/28/2023] [Indexed: 03/07/2024]
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disorder (IBD) characterized by relapsing chronic inflammation of the colon that causes continuous mucosal inflammation. The global incidence of UC is steadily increasing. Immune mechanisms are involved in the pathogenesis of UC, of which complement is shown to play a critical role by inducing local chronic inflammatory responses that promote tissue damage. However, the function of various complement components in the development of UC is complex and even paradoxical. Some components (e.g. C1q, CD46, CD55, CD59, and C6) are shown to safeguard the intestinal barrier and reduce intestinal inflammation, while others (e.g. C3, C5, C5a) can exacerbate intestinal damage and accelerate the development of UC. The complement system was originally thought to function primarily in an extracellular mode; however, recent evidence indicates that it can also act intracellularly as the complosome. The current study provides an overview of current studies on complement and its role in the development of UC. While there are few studies that describe how intracellular complement contributes to UC, we discuss potential future directions based on related publications. We also highlight novel methods that target complement for IBD treatment.
Collapse
Affiliation(s)
- Yujie Ma
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Kaicheng Zhang
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Yuanyuan Wu
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Xiaoyan Fu
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Shujuan Liang
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Meiyu Peng
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Juntang Guo
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Meifang Liu
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| |
Collapse
|
19
|
Natoli V, Charras A, Hahn G, Hedrich CM. Neuropsychiatric involvement in juvenile-onset systemic lupus erythematosus (jSLE). Mol Cell Pediatr 2023; 10:5. [PMID: 37556020 PMCID: PMC10412509 DOI: 10.1186/s40348-023-00161-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a rare autoimmune/inflammatory disease with significant morbidity and mortality. Approximately 15-20% of SLE patients develop the disease during childhood or adolescence (juvenile-onset SLE/jSLE). Patients with jSLE exhibit more variable and severe disease when compared to patients with disease-onset during adulthood. Neuropsychiatric (NP) involvement is a clinically heterogenous and potentially severe complication. Published reports on the incidence and prevalence of NP-jSLE are scarce, and the exact pathophysiology is poorly understood.This manuscript provides a review of the existing literature, suggesting NP involvement in 13.5-51% of jSLE patients. Among patients with NP-jSLE affecting the CNS, we propose two main subgroups: (i) a chronic progressive, predominantly type 1 interferon-driven form that poorly responds to currently used treatments, and (ii) an acutely aggressive form that usually presents early during the disease that may be primarily mediated by auto-reactive effector lymphocytes. While this hypothesis requires to be tested in large collaborative international cohort studies, it may offer future patient stratification and individualised care.
Collapse
Affiliation(s)
- Valentina Natoli
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Rheumatology, Institute in the Park, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
- Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Università degli Studi di Genova, Genoa, Italy
| | - Amandine Charras
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Gabriele Hahn
- Department of Radiology, Universitätsklinikum Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Christian M Hedrich
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.
- Department of Rheumatology, Institute in the Park, Alder Hey Children's NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
20
|
Shen Y, Gong Z, Zhang S, Cao J, Mao W, Yao Y, Zhao J, Li Q, Liu K, Liu B, Feng S. Besides TLR2 and TLR4, NLRP3 is also involved in regulating Escherichia coli infection-induced inflammatory responses in mice. Int Immunopharmacol 2023; 121:110556. [PMID: 37364329 DOI: 10.1016/j.intimp.2023.110556] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
The host Toll-like Receptor-2 (TLR2) and Toll-like Receptor-4 (TLR4) play critical roles in defense against Escherichia coli (E. coli) infection is well-known. The NLR pyrin domain-containing 3 (NLRP3) inflammasome is also an important candidate during the host-recognized pathogen, while the roles of NLRP3 in the host inflammatory response to E. coli infection remains unclear. This study aimed to explore the roles of NLRP3 in regulating the inflammatory response in E. coli infection-induced mice. Our result indicated that compared to wild-type mice, the TLR2-deficient (TLR2-/-), TLR4-deficient (TLR4-/-), and NLRP3-deficient (NLRP3-/-) mice had significant decrease in liver damage after stimulation with Lipopolysaccharide (LPS, 1 μg/mL), Braun lipoprotein (BLP, 1 μg/mL), or infected by WT E. coli (1 × 107 CFU, MOI 5:1). Meanwhile, compared with wild-type mice, the TNF-α and IL-1β production in serum decreased in TLR2-/-, TLR4-/-, and NLRP3-/- mice after LPS, BLP treatment, or WT E. coli infection. In macrophages from NLRP3-/- mice showed significantly reduced secretion of TNF-α and IL-1β in response to stimulation with LPS, BLP, or WT E. coli infection compared with macrophages from wild-type mice. These results indicate that besides TLR2 and TLR4, NLRP3 also plays a critical role in host inflammatory responses to defense against E. coli infection, and might provide a therapeutic target in combating disease with bacterium infection.
Collapse
Affiliation(s)
- Yuan Shen
- Key Laboratory of Molecular Epidemiology of Chronic Diseases, School of Public Health, Inner Mongolia Medical University, No. 5, Xinhua Street, Hui Min District, 010000, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Zhiguo Gong
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Shuangyi Zhang
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Jinshan Cao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Wei Mao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Yuan Yao
- Department of Neurology, Inner Mongolia People's Hospital, No. 20, Zhaowuda Road, Saihan District, 010017, Hohhot City, China
| | - Jiamin Zhao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Qianru Li
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Kun Liu
- Key Laboratory of Molecular Epidemiology of Chronic Diseases, School of Public Health, Inner Mongolia Medical University, No. 5, Xinhua Street, Hui Min District, 010000, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Bo Liu
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China.
| | - Shuang Feng
- Laboratory of Veterinary Public Health, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China.
| |
Collapse
|
21
|
Franklin ME, Bennett C, Arboite M, Alvarez-Ciara A, Corrales N, Verdelus J, Dietrich WD, Keane RW, de Rivero Vaccari JP, Prasad A. Activation of inflammasomes and their effects on neuroinflammation at the microelectrode-tissue interface in intracortical implants. Biomaterials 2023; 297:122102. [PMID: 37015177 PMCID: PMC10614166 DOI: 10.1016/j.biomaterials.2023.122102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/16/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023]
Abstract
Invasive neuroprosthetics rely on microelectrodes (MEs) to record or stimulate the activity of large neuron assemblies. However, MEs are subjected to tissue reactivity in the central nervous system (CNS) due to the foreign body response (FBR) that contribute to chronic neuroinflammation and ultimately result in ME failure. An endogenous, acute set of mechanisms responsible for the recognition and targeting of foreign objects, called the innate immune response, immediately follows the ME implant-induced trauma. Inflammasomes are multiprotein structures that play a critical role in the initiation of an innate immune response following CNS injuries. The activation of inflammasomes facilitates a range of innate immune response cascades and results in neuroinflammation and programmed cell death. Despite our current understanding of inflammasomes, their roles in the context of neural device implantation remain unknown. In this study, we implanted a non-functional Utah electrode array (UEA) into the rat somatosensory cortex and studied the inflammasome signaling and the corresponding downstream effects on inflammatory cytokine expression and the inflammasome-mediated cell death mechanism of pyroptosis. Our results not only demonstrate the continuous activation of inflammasomes and their contribution to neuroinflammation at the electrode-tissue interface but also reveal the therapeutic potential of targeting inflammasomes to attenuate the FBR in invasive neuroprosthetics.
Collapse
Affiliation(s)
- Melissa E Franklin
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Cassie Bennett
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Maelle Arboite
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | | | - Natalie Corrales
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Jennifer Verdelus
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - W Dalton Dietrich
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA
| | - Robert W Keane
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA
| | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA.
| |
Collapse
|
22
|
Kuhn MB, VandenBerg HS, Reynolds AJ, Carson MD, Warner AJ, LaRue AC, Novince CM, Hathaway-Schrader JD. C3a-C3aR signaling is a novel modulator of skeletal homeostasis. Bone Rep 2023; 18:101662. [PMID: 36860797 PMCID: PMC9969257 DOI: 10.1016/j.bonr.2023.101662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
Osteoimmune studies have identified complement signaling as an important regulator of the skeleton. Specifically, complement anaphylatoxin receptors (i.e., C3aR, C5aR) are expressed on osteoblasts and osteoclasts, implying that C3a and/or C5a may be candidate mediators of skeletal homeostasis. The study aimed to determine how complement signaling influences bone modeling/remodeling in the young skeleton. Female C57BL/6J C3aR-/-C5aR-/- vs. wildtype and C3aR-/- vs. wildtype mice were examined at age 10 weeks. Trabecular and cortical bone parameters were analyzed by micro-CT. In situ osteoblast and osteoclast outcomes were determined by histomorphometry. Osteoblast and osteoclast precursors were assessed in vitro. C3aR-/-C5aR-/- mice displayed an increased trabecular bone phenotype at age 10 weeks. In vitro studies revealed C3aR-/-C5aR-/- vs. wildtype cultures had less bone-resorbing osteoclasts and increased bone-forming osteoblasts, which were validated in vivo. To determine whether C3aR alone was critical for the enhanced skeletal outcomes, wildtype vs. C3aR-/- mice were evaluated for osseous tissue outcomes. Paralleling skeletal findings in C3aR-/-C5aR-/- mice, C3aR-/- vs. wildtype mice had an enhanced trabecular bone volume fraction, which was attributed to increased trabecular number. There was elevated osteoblast activity and suppressed osteoclastic cells in C3aR-/- vs. wildtype mice. Furthermore, primary osteoblasts derived from wildtype mice were stimulated with exogenous C3a, which more profoundly upregulated C3ar1 and the pro-osteoclastic chemokine Cxcl1. This study introduces the C3a/C3aR signaling axis as a novel regulator of the young skeleton.
Collapse
Affiliation(s)
- Megan B. Kuhn
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Hayden S. VandenBerg
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Andrew J. Reynolds
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Matthew D. Carson
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Stomatology-Div. of Periodontics, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Pediatrics-Div. of Endocrinology, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Amy J. Warner
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Stomatology-Div. of Periodontics, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Pediatrics-Div. of Endocrinology, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Amanda C. LaRue
- Research Services, Ralph H. Johnson Department of Veterans Affairs Health Care System, Charleston, SC, USA
- Department of Pathology and Laboratory Medicine, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Chad M. Novince
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Stomatology-Div. of Periodontics, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Pediatrics-Div. of Endocrinology, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Jessica D. Hathaway-Schrader
- Department of Stomatology-Div. of Periodontics, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Research Services, Ralph H. Johnson Department of Veterans Affairs Health Care System, Charleston, SC, USA
- Department of Pathology and Laboratory Medicine, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
23
|
Gu D, Wang H, Yan M, Li Y, Yang S, Shi D, Guo S, Wu L, Liu C. Echinacea purpurea (L.) Moench extract suppresses inflammation by inhibition of C3a/C3aR signaling pathway in TNBS-induced ulcerative colitis rats. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116221. [PMID: 36754188 DOI: 10.1016/j.jep.2023.116221] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/15/2023] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Echinacea purpurea (L.) Moench (EP) is a perennial herbaceous flowering plant, commonly known as purple conical flower. It was widely used to treat skin inflammation and gastrointestinal diseases. AIM OF STUDY Ulcerative colitis (UC) is a chronic and nonspecific inflammatory disease. Recent evidence shows that immune disorders are involved in the pathogenesis of UC. To evaluate the protective effect of Echinacea purpurea (L.) Moench exact (EE) on UC and explore the role of complement system in the treatment of UC. MATERIALS AND METHODS UC model was induced in rats by 2,4,6-trinitrobenzene sulfonic acid (TNBS), and then rats were administered with EE for 10 days. Collect colon tissues for analysis of relevant mechanisms. RESULTS EE could reduce the weight loss and diarrhea of UC rats. In addition, EE could improve the integrity of intestinal epithelial barrier in UC rats. EE inhibited the level of proinflammatory cytokines and promoted the antioxidation. Furthermore, EE suppressed the expression of C3aR, CFB, CD55, TLR4 and NLRP3. CONCLUSION These results indicate that EE may achieve therapeutic effect by inhibiting C3a/C3aR signal pathway, suggesting that EE may be used as a medicinal plant to alleviate UC.
Collapse
Affiliation(s)
- Daxing Gu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| | - Huiting Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| | - Mingen Yan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| | - Yaoxing Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| | - Shijing Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| | - Dayou Shi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Technology Research Center for Traditional Chinese Veterinary Medicine and Nature Medicine, Guangzhou, 510642, China; International Institute of Traditional Chinese Veterinary Medicine, Guangzhou, 510642, China.
| | - Shining Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Technology Research Center for Traditional Chinese Veterinary Medicine and Nature Medicine, Guangzhou, 510642, China; International Institute of Traditional Chinese Veterinary Medicine, Guangzhou, 510642, China.
| | - Li Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Technology Research Center for Traditional Chinese Veterinary Medicine and Nature Medicine, Guangzhou, 510642, China; International Institute of Traditional Chinese Veterinary Medicine, Guangzhou, 510642, China.
| | - Cui Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
24
|
Ghosh M, Rana S. The anaphylatoxin C5a: Structure, function, signaling, physiology, disease, and therapeutics. Int Immunopharmacol 2023; 118:110081. [PMID: 36989901 DOI: 10.1016/j.intimp.2023.110081] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
The complement system is one of the oldest known tightly regulated host defense systems evolved for efficiently functioning cell-based immune systems and antibodies. Essentially, the complement system acts as a pivot between the innate and adaptive arms of the immune system. The complement system collectively represents a cocktail of ∼50 cell-bound/soluble glycoproteins directly involved in controlling infection and inflammation. Activation of the complement cascade generates complement fragments like C3a, C4a, and C5a as anaphylatoxins. C5a is the most potent proinflammatory anaphylatoxin, which is involved in inflammatory signaling in a myriad of tissues. This review provides a comprehensive overview of human C5a in the context of its structure and signaling under several pathophysiological conditions, including the current and future therapeutic applications targeting C5a.
Collapse
Affiliation(s)
- Manaswini Ghosh
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha 752050, India
| | - Soumendra Rana
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha 752050, India.
| |
Collapse
|
25
|
Jamar G, Pisani LP. Inflammatory crosstalk between saturated fatty acids and gut microbiota-white adipose tissue axis. Eur J Nutr 2023; 62:1077-1091. [PMID: 36484808 DOI: 10.1007/s00394-022-03062-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE High-fat diets have different metabolic responses via gut dysbiosis. In this review, we discuss the complex interaction between the intake of long- and medium-chain saturated fatty acids (SFAs), gut microbiota, and white adipose tissue (WAT) dysfunction, particularly focusing on the type of fat. RESULTS The evidence for the impact of dietary SFAs on the gut microbiota-WAT axis has been mostly derived from in vitro and animal models, but there is now also evidence emerging from human studies. Most current reports show that, in response to high long- and medium-chain SFA diets, WAT functions are altered and can be modulated from microbial metabolites in several manners; and it appears to be also modified under conditions of obesity. SFAs overconsumption can reduce bacterial content and disrupt the gut environment. Both long- and medium-chain SFAs may contribute to proinflammatory cytokines release and TLR4 cascade signaling, either by regulation of endotoxemia markers or myristoylated protein. Palmitic and stearic acids have pathological effects on the intestinal epithelium, microbes, and inflammatory and lipogenic WAT profiles. While myristic and lauric acids display somewhat controversial outcomes, from probiotic effects and contribution to weight loss to cardiometabolic alterations from WAT inflammation. CONCLUSION Identifying an interference of distinct types of SFA in the binomial gut microbiota-WAT may elucidate essential mechanisms of metabolic endotoxemia, which may be the key to triggering obesity, innovating the therapeutic tools for this disease.
Collapse
Affiliation(s)
- Giovana Jamar
- Post-Graduate Program in Nutrition, Federal University of São Paulo-UNIFESP, São Paulo, SP, Brazil
- Department of Biosciences, Institute of Health and Society, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo-UNIFESP, Rua Silva Jardim, 136/311, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Luciana Pellegrini Pisani
- Post-Graduate Program in Nutrition, Federal University of São Paulo-UNIFESP, São Paulo, SP, Brazil.
- Department of Biosciences, Institute of Health and Society, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo-UNIFESP, Rua Silva Jardim, 136/311, Vila Mathias, Santos, SP, 11015-020, Brazil.
| |
Collapse
|
26
|
Fontes-Dantas FL, Fernandes GG, Gutman EG, De Lima EV, Antonio LS, Hammerle MB, Mota-Araujo HP, Colodeti LC, Araújo SM, Froz GM, da Silva TN, Duarte LA, Salvio AL, Pires KL, Leon LA, Vasconcelos CCF, Romão L, Savio LEB, Silva JL, da Costa R, Clarke JR, Da Poian AT, Alves-Leon SV, Passos GF, Figueiredo CP. SARS-CoV-2 Spike protein induces TLR4-mediated long-term cognitive dysfunction recapitulating post-COVID-19 syndrome in mice. Cell Rep 2023; 42:112189. [PMID: 36857178 PMCID: PMC9935273 DOI: 10.1016/j.celrep.2023.112189] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/16/2022] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Cognitive dysfunction is often reported in patients with post-coronavirus disease 2019 (COVID-19) syndrome, but its underlying mechanisms are not completely understood. Evidence suggests that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Spike protein or its fragments are released from cells during infection, reaching different tissues, including the CNS, irrespective of the presence of the viral RNA. Here, we demonstrate that brain infusion of Spike protein in mice has a late impact on cognitive function, recapitulating post-COVID-19 syndrome. We also show that neuroinflammation and hippocampal microgliosis mediate Spike-induced memory dysfunction via complement-dependent engulfment of synapses. Genetic or pharmacological blockage of Toll-like receptor 4 (TLR4) signaling protects animals against synapse elimination and memory dysfunction induced by Spike brain infusion. Accordingly, in a cohort of 86 patients who recovered from mild COVID-19, the genotype GG TLR4-2604G>A (rs10759931) is associated with poor cognitive outcome. These results identify TLR4 as a key target to investigate the long-term cognitive dysfunction after COVID-19 infection in humans and rodents.
Collapse
Affiliation(s)
- Fabricia L. Fontes-Dantas
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Gabriel G. Fernandes
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Elisa G. Gutman
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil,Clinical Medicine Post-graduation Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Emanuelle V. De Lima
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leticia S. Antonio
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mariana B. Hammerle
- Clinical Medicine Post-graduation Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Hannah P. Mota-Araujo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lilian C. Colodeti
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Suzana M.B. Araújo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gabrielle M. Froz
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Talita N. da Silva
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Larissa A. Duarte
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil,Clinical Medicine Post-graduation Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Andreza L. Salvio
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil
| | - Karina L. Pires
- Neurology Department, Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro, RJ, Brazil
| | - Luciane A.A. Leon
- Laboratório de Desenvolvimento Tecnológico em Virologia, IOC/FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | - Luciana Romão
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luiz Eduardo B. Savio
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jerson L. Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Robson da Costa
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Julia R. Clarke
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Andrea T. Da Poian
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| | - Soniza V. Alves-Leon
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil,Division of Neurology, Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| | - Giselle F. Passos
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| | - Claudia P. Figueiredo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| |
Collapse
|
27
|
Xia Y, Zhang W, He K, Bai L, Miao Y, Liu B, Zhang X, Jin S, Wu Y. Hydrogen sulfide alleviates lipopolysaccharide-induced myocardial injury through TLR4-NLRP3 pathway. Physiol Res 2023; 72:15-25. [PMID: 36545872 PMCID: PMC10069815 DOI: 10.33549/physiolres.934928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023] Open
Abstract
To investigate the effect of hydrogen sulfide (H2S) on myocardial injury in sepsis-induced myocardial dysfunction (SIMD), male C57BL/6 mice were intraperitoneally injected with lipopolysaccharide (LPS) (10 mg/kg, i.p.) to induce cardiac dysfunction without or with the H2S donor sodium hydrosulfide (NaHS) (50 µmol/kg, i.p.) administration 3 h after LPS injection. Six hours after the LPS injection, echocardiography, cardiac hematoxylin and eosin (HE) staining, myocardial damage and inflammatory biomarkers and Western blot results were analyzed. In mice, the administration of LPS decreased left ventricular ejection fraction (LVEF) by 30 % along with lowered H2S levels (35 % reduction). It was observed that cardiac troponin I (cTnI), tumor necrosis factor-alpha (TNF-alpha), and interleukin-1beta (IL-1beta) levels were all increased (by 0.22-fold, 2000-fold and 0.66-fold respectively). HE staining revealed structural damage and inflammatory cell infiltration in the myocardial tissue after LPS administration. Moreover, after 6 h of LPS treatment, toll-like receptor 4 (TLR4) and nod-like receptor protein 3 (NLRP3) expressions were up-regulated 2.7-fold and 1.6-fold respectively. When compared to the septic mice, NaHS enhanced ventricular function (by 0.19-fold), decreased cTnI, TNF-alpha, and IL-1beta levels (by 11 %, 33 %, and 16 % respectively) and downregulated TLR4 and NLRP3 expressions (by 64 % and 31 % respectively). Furthermore, NaHS did not further improve cardiac function and inflammation in TLR4-/- mice or mice in which NLRP3 activation was inhibited by MCC950, after LPS injection. In conclusion, these findings imply that decreased endogenous H2S promotes the progression of SIMD, whereas exogenous H2S alleviates SIMD by inhibiting inflammation via the TLR4-NLRP3 pathway suppression.
Collapse
Affiliation(s)
- Y Xia
- Department of Physiology, Hebei Medical University, Hebei, China. ;
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Uriarte SM, Hajishengallis G. Neutrophils in the periodontium: Interactions with pathogens and roles in tissue homeostasis and inflammation. Immunol Rev 2023; 314:93-110. [PMID: 36271881 PMCID: PMC10049968 DOI: 10.1111/imr.13152] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Neutrophils are of key importance in periodontal health and disease. In their absence or when they are functionally defective, as occurs in certain congenital disorders, affected individuals develop severe forms of periodontitis in early age. These observations imply that the presence of immune-competent neutrophils is essential to homeostasis. However, the presence of supernumerary or hyper-responsive neutrophils, either because of systemic priming or innate immune training, leads to imbalanced host-microbe interactions in the periodontium that culminate in dysbiosis and inflammatory tissue breakdown. These disease-provoking imbalanced interactions are further exacerbated by periodontal pathogens capable of subverting neutrophil responses to their microbial community's benefit and the host's detriment. This review attempts a synthesis of these findings for an integrated view of the neutrophils' ambivalent role in periodontal disease and, moreover, discusses how some of these concepts underpin the development of novel therapeutic approaches to treat periodontal disease.
Collapse
Affiliation(s)
- Silvia M. Uriarte
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
29
|
Zysset-Burri DC, Morandi S, Herzog EL, Berger LE, Zinkernagel MS. The role of the gut microbiome in eye diseases. Prog Retin Eye Res 2023; 92:101117. [PMID: 36075807 DOI: 10.1016/j.preteyeres.2022.101117] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 02/01/2023]
Abstract
The gut microbiome is a complex ecosystem of microorganisms and their genetic entities colonizing the gastrointestinal tract. When in balanced composition, the gut microbiome is in symbiotic interaction with its host and maintains intestinal homeostasis. It is involved in essential functions such as nutrient metabolism, inhibition of pathogens and regulation of immune function. Through translocation of microbes and their metabolites along the epithelial barrier, microbial dysbiosis induces systemic inflammation that may lead to tissue destruction and promote the onset of various diseases. Using whole-metagenome shotgun sequencing, several studies have shown that the composition and associated functional capacities of the gut microbiome are associated with age-related macular degeneration, retinal artery occlusion, central serous chorioretinopathy and uveitis. In this review, we provide an overview of the current knowledge about the gut microbiome in eye diseases, with a focus on interactions between the microbiome, specific microbial-derived metabolites and the immune system. We explain how these interactions may be involved in the pathogenesis of age-related macular degeneration, retinal artery occlusion, central serous chorioretinopathy and uveitis and guide the development of new therapeutic approaches by microbiome-altering interventions for these diseases.
Collapse
Affiliation(s)
- Denise C Zysset-Burri
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland; Department for BioMedical Research, University of Bern, Murtenstrasse 24, CH-3008, Bern, Switzerland.
| | - Sophia Morandi
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland; Department for BioMedical Research, University of Bern, Murtenstrasse 24, CH-3008, Bern, Switzerland.
| | - Elio L Herzog
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland; Department for BioMedical Research, University of Bern, Murtenstrasse 24, CH-3008, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Mittelstrasse 43, CH-3012, Bern, Switzerland.
| | - Lieselotte E Berger
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland; Department for BioMedical Research, University of Bern, Murtenstrasse 24, CH-3008, Bern, Switzerland.
| | - Martin S Zinkernagel
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland; Department for BioMedical Research, University of Bern, Murtenstrasse 24, CH-3008, Bern, Switzerland.
| |
Collapse
|
30
|
Inomata M, Amano S, Abe M, Hayashi T, Sakagami H. Innate immune response of human periodontal ligament fibroblasts via the Dectin-1/Syk pathway. J Med Microbiol 2022; 71. [PMID: 36748551 DOI: 10.1099/jmm.0.001627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Introduction. A diverse microbiota including fungi exists in the subgingival sites of patients with chronic periodontitis. The cell wall of Candida albicans, the most abundant fungal species, contains β-glucan. Dectin-1 binds β-glucan and participates in fungal recognition.Gap statement. Human periodontal ligament fibroblasts (PDLFs) are present in the periodontal ligament and synthesize immunomodulatory cytokines that influence the local response to infections. However, the expression and role of Dectin-1 in PDLFs have not been explored.Aim. This study aimed to determine if PDLFs express Dectin-1 and induce innate immune responses through Dectin-1 and the signalling molecule Syk.Methodology. The expression of Dectin-1 in PDLFs was determined by flow cytometry, western blotting and confocal microscopy. Real-time PCR and Western blotting were used to determine the immune response of PDLFs stimulated with β-glucan-rich zymosan and C. albicans.Results. Dectin-1 was constitutively expressed in PDLFs. Zymosan induced the expression of cytokines, including IL6, IL1B and IL17A, and the chemokine IL8. Zymosan also induced the expression of the antimicrobial peptide β-defensin-1 (DEFB1). Further, the phosphorylation of Syk and NF-κB occurred upon Dectin-1 activation. Notably, heat-killed C. albicans induced the expression of IL6, IL17A, IL8 and DEFB1, and this activation was suppressed by the Syk inhibitor, R406.Conclusion. These findings indicate that the Dectin-1/Syk pathway induces an innate immune response of PDLFs, which may facilitate the control of oral infections such as candidiasis and periodontitis.
Collapse
Affiliation(s)
- Megumi Inomata
- Division of Microbiology and Immunology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Shigeru Amano
- Research Institute of Odontology (M-RIO), Meikai University School of Dentistry, Sakado, Japan
| | - Masayo Abe
- Division of Microbiology and Immunology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Toru Hayashi
- Department of Anatomy Science, School of Allied Health Sciences, Kitasato University, Kitasato, Japan
| | - Hiroshi Sakagami
- Research Institute of Odontology (M-RIO), Meikai University School of Dentistry, Sakado, Japan
| |
Collapse
|
31
|
Stenson EK, Kendrick J, Dixon B, Thurman JM. The complement system in pediatric acute kidney injury. Pediatr Nephrol 2022; 38:1411-1425. [PMID: 36203104 PMCID: PMC9540254 DOI: 10.1007/s00467-022-05755-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/08/2022] [Accepted: 09/09/2022] [Indexed: 10/24/2022]
Abstract
The complement cascade is an important part of the innate immune system. In addition to helping the body to eliminate pathogens, however, complement activation also contributes to the pathogenesis of a wide range of kidney diseases. Recent work has revealed that uncontrolled complement activation is the key driver of several rare kidney diseases in children, including atypical hemolytic uremic syndrome and C3 glomerulopathy. In addition, a growing body of literature has implicated complement in the pathogenesis of more common kidney diseases, including acute kidney injury (AKI). Complement-targeted therapeutics are in use for a variety of diseases, and an increasing number of therapeutic agents are under development. With the implication of complement in the pathogenesis of AKI, complement-targeted therapeutics could be trialed to prevent or treat this condition. In this review, we discuss the evidence that the complement system is activated in pediatric patients with AKI, and we review the role of complement proteins as biomarkers and therapeutic targets in patients with AKI.
Collapse
Affiliation(s)
- Erin K. Stenson
- grid.430503.10000 0001 0703 675XSection of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado School of Medicine, 13121 E 17th Avenue, MS8414, Aurora, CO 80045 USA
| | - Jessica Kendrick
- grid.430503.10000 0001 0703 675XDivision of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO USA
| | - Bradley Dixon
- grid.430503.10000 0001 0703 675XRenal Section, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO USA
| | - Joshua M. Thurman
- grid.430503.10000 0001 0703 675XDivision of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO USA
| |
Collapse
|
32
|
Oleuropein ameliorated lung ischemia-reperfusion injury by inhibiting TLR4 signaling cascade in alveolar macrophages. Transpl Immunol 2022; 74:101664. [PMID: 35809814 DOI: 10.1016/j.trim.2022.101664] [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: 05/13/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 11/23/2022]
Abstract
Lung ischemia-reperfusion (I/R) injury is a common postoperative complication in patients with lung transplantation, pulmonary embolism, and cardiopulmonary bypass. Lung I/R injury is a sterile inflammatory process that leads to lung dysfunction, and is an important cause of patient death. Effectively alleviating lung I/R injury can thus improve the prognosis of patients. In this study, we created a mouse model of lung I/R injury by transient unilateral left pulmonary artery occlusion. 6-8 weeks male C57BL/6 mice were randomly assigned to four groups: Sham, I/R, I/R + oleuropein (OLE) and OLE. OLE (50 mg/kg) was orally 24 h and 30 min before anesthesia. Measurement of lung pathohistological, isolated alveolar macrophages (AMs), inflammatory mediators, TLR4 and its downstream factors (MyD88, NF-κB) were performed. We then evaluated the ability of oleuropein (OLE) to ameliorate I/R-induced lung injury and explored the possible molecular mechanisms. OLE ameliorated I/R-induced lung injury and edema and decreased inflammatory factors in lung tissue and bronchoalveolar lavage fluid. This protection required toll-like receptor 4 (TLR4). OLE significantly inhibited I/R-induced expression of TLR4 and its downstream factors in lung tissue and alveolar macrophages. In addition, hypoxia-inducible factor 1α protein accumulated in TLR4-mediated lung I/R injury, and further induced the production of inflammatory factors. Collectively, these data suggest that OLE ameliorates I/R-induced lung injury. The mechanism responsible for its protective effect may involve inhibition of the I/R-induced inflammatory response by downregulating the TLR4 signaling cascade in AMs.
Collapse
|
33
|
Leonel TB, Gabrili JJM, Squaiella-Baptistão CC, Woodruff TM, Lambris JD, Tambourgi DV. Bothrops jararaca Snake Venom Inflammation Induced in Human Whole Blood: Role of the Complement System. Front Immunol 2022; 13:885223. [PMID: 35720304 PMCID: PMC9201114 DOI: 10.3389/fimmu.2022.885223] [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: 02/27/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical manifestations of envenomation by Bothrops species are complex and characterized by prominent local effects that can progress to tissue loss, physical disability, or amputation. Systemic signs can also occur, such as hemorrhage, coagulopathy, shock, and acute kidney failure. The rapid development of local clinical manifestations is accompanied by the presence of mediators of the inflammatory process originating from tissues damaged by the bothropic venom. Considering the important role that the complement system plays in the inflammatory response, in this study, we analyzed the action of Bothrops jararaca snake venom on the complement system and cell surface receptors involved in innate immunity using an ex vivo human whole blood model. B. jararaca venom was able to induce activation of the complement system in the human whole blood model and promoted a significant increase in the production of anaphylatoxins C3a/C3a-desArg, C4a/C4a-desArg, C5a/C5a-desArg and sTCC. In leukocytes, the venom of B. jararaca reduced the expression of CD11b, CD14 and C5aR1. Inhibition of the C3 component by Cp40, an inhibitor of C3, resulted in a reduction of C3a/C3a-desArg, C5a/C5a-desArg and sTCC to basal levels in samples stimulated with the venom. Exposure to B. jararaca venom induced the production of inflammatory cytokines and chemokines such as TNF-α, IL-8/CXCL8, MCP-1/CCL2 and MIG/CXCL9 in the human whole blood model. Treatment with Cp40 promoted a significant reduction in the production of TNF-α, IL-8/CXCL8 and MCP-1/CCL2. C5aR1 inhibition with PMX205 also promoted a reduction of TNF-α and IL-8/CXCL8 to basal levels in the samples stimulated with venom. In conclusion, the data presented here suggest that the activation of the complement system promoted by the venom of the snake B. jararaca in the human whole blood model significantly contributes to the inflammatory process. The control of several inflammatory parameters using Cp40, an inhibitor of the C3 component, and PMX205, a C5aR1 antagonist, indicates that complement inhibition may represent a potential therapeutic tool in B. jararaca envenoming.
Collapse
Affiliation(s)
| | | | | | - Trent M. Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | |
Collapse
|
34
|
Gaboriaud C, Lorvellec M, Rossi V, Dumestre-Pérard C, Thielens NM. Complement System and Alarmin HMGB1 Crosstalk: For Better or Worse. Front Immunol 2022; 13:869720. [PMID: 35572583 PMCID: PMC9095977 DOI: 10.3389/fimmu.2022.869720] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/04/2022] [Indexed: 12/21/2022] Open
Abstract
Our immune system responds to infectious (PAMPs) and tissue damage (DAMPs) signals. The complement system and alarmin High-Mobility Group Box 1 (HMGB1) are two powerful soluble actors of human host defense and immune surveillance. These systems involve molecular cascades and amplification loops for their signaling or activation. Initially activated as alarm raising systems, their function can be finally switched towards inflammation resolution, where they sustain immune maturation and orchestrate repair mechanisms, opening the way back to homeostasis. However, when getting out of control, these defense systems can become deleterious and trigger serious cellular and tissue damage. Therefore, they can be considered as double-edged swords. The close interaction between the complement and HMGB1 pathways is described here, as well as their traditional and non-canonical roles, their functioning at different locations and their independent and collective impact in different systems both in health and disease. Starting from these systems and interplay at the molecular level (when elucidated), we then provide disease examples to better illustrate the signs and consequences of their roles and interaction, highlighting their importance and possible vicious circles in alarm raising and inflammation, both individually or in combination. Although this integrated view may open new therapeutic strategies, future challenges have to be faced because of the remaining unknowns regarding the molecular mechanisms underlying the fragile molecular balance which can drift towards disease or return to homeostasis, as briefly discussed at the end.
Collapse
Affiliation(s)
| | | | | | - Chantal Dumestre-Pérard
- Univ. Grenoble Alpes, CEA, CNRS, IBS, Grenoble, France
- Laboratoire d’Immunologie, Pôle de Biologie, CHU Grenoble Alpes, Grenoble, France
| | | |
Collapse
|
35
|
Dias BT, Goundry A, Vivarini AC, Costa TFR, Mottram JC, Lopes UG, Lima APCA. Toll-Like Receptor- and Protein Kinase R-Induced Type I Interferon Sustains Infection of Leishmania donovani in Macrophages. Front Immunol 2022; 13:801182. [PMID: 35154115 PMCID: PMC8831251 DOI: 10.3389/fimmu.2022.801182] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/06/2022] [Indexed: 12/27/2022] Open
Abstract
Leishmania donovani is a protozoan parasite that causes visceral leishmaniasis, provoking liver and spleen tissue destruction that is lethal unless treated. The parasite replicates in macrophages and modulates host microbicidal responses. We have previously reported that neutrophil elastase (NE) is required to sustain L. donovani intracellular growth in macrophages through the induction of interferon beta (IFN-β). Here, we show that the gene expression of IFN-β by infected macrophages was reduced by half when TLR4 was blocked by pre-treatment with neutralizing antibodies or in macrophages from tlr2-/- mice, while the levels in macrophages from myd88-/- mice were comparable to those from wild-type C57BL/6 mice. The neutralization of TLR4 in tlr2-/- macrophages completely abolished induction of IFN-β gene expression upon parasite infection, indicating an additive role for both TLRs. Induction of type I interferon (IFN-I), OASL2, SOD1, and IL10 gene expression by L. donovani was completely abolished in macrophages from NE knock-out mice (ela2-/-) or from protein kinase R (PKR) knock-out mice (pkr-/-), and in C57BL/6 macrophages infected with transgenic L. donovani expressing the inhibitor of serine peptidase 2 (ISP2). Parasite intracellular growth was impaired in pkr-/- macrophages but was fully restored by the addition of exogenous IFN-β, and parasite burdens were reduced in the spleen of pkr-/- mice at 7 days, as compared to the 129Sv/Ev background mice. Furthermore, parasites were unable to grow in macrophages lacking TLR3, which correlated with lack of IFN-I gene expression. Thus, L. donovani engages innate responses in infected macrophages via TLR2, TLR4, and TLR3, via downstream PKR, to induce the expression of pro-survival genes in the host cell, and guarantee parasite intracellular development.
Collapse
Affiliation(s)
- Bruna T. Dias
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amy Goundry
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aislan C. Vivarini
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tatiana F. R. Costa
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jeremy C. Mottram
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| | - Ulisses G. Lopes
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula C. A. Lima
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Ana Paula C. A. Lima,
| |
Collapse
|
36
|
Complement as a powerful "influencer" in the brain during development, adulthood and neurological disorders. Adv Immunol 2021; 152:157-222. [PMID: 34844709 DOI: 10.1016/bs.ai.2021.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The complement system was long considered as only a powerful effector arm of the immune system that, while critically protective, could lead to inflammation and cell death if overactivated, even in the central nervous system (CNS). However, in the past decade it has been recognized as playing critical roles in key physiological processes in the CNS, including neurogenesis and synaptic remodeling in the developing and adult brain. Inherent in these processes are the interactions with cells in the brain, and the cascade of interactions and functional consequences that ensue. As a result, investigations of therapeutic approaches for both suppressing excessive complement driven neurotoxicity and aberrant sculpting of neuronal circuits, require broad (and deep) knowledge of the functional activities of multiple components of this highly evolved and regulated system to avoid unintended negative consequences in the clinic. Advances in basic science are beginning to provide a roadmap for translation to therapeutics, with both small molecule and biologics. Here, we present examples of the critical roles of proper complement function in the development and sculpting of the nervous system, and in enabling rapid protection from infection and clearance of dying cells. Microglia are highlighted as important command centers that integrate signals from the complement system and other innate sensors that are programed to provide support and protection, but that direct detrimental responses to aberrant activation and/or regulation of the system. Finally, we present promising research areas that may lead to effective and precision strategies for complement targeted interventions to promote neurological health.
Collapse
|
37
|
Hasturk H, Hajishengallis G, Lambris JD, Mastellos DC, Yancopoulou D. Phase 2a clinical trial of complement C3 inhibitor AMY-101 in adults with periodontal inflammation. J Clin Invest 2021; 131:152973. [PMID: 34618684 DOI: 10.1172/jci152973] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/05/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Gingivitis and periodontitis are prevalent inflammatory diseases of the periodontal tissues. Current treatments are often ineffective or do not prevent disease recurrence. Uncontrolled complement activation and resulting chronic gingival inflammation is a hallmark of periodontal diseases. We determined efficacy and safety of a complement 3-targeted therapeutic, AMY-101, locally administered in adults with periodontal inflammation. METHODS Thirty-two patients with gingival inflammation were enrolled into a randomized, placebo-controlled, double-blind, split-mouth design phase 2a trial, after dose-escalation study to select safe and effective dose with additional 8 patients. Half of the mouth was randomly assigned to AMY-101 (0.1mg/site) or placebo injections at sites of inflammation, administered on days 0, 7 and 14 and evaluated for safety and efficacy outcomes at days 28, 60 and 90. The primary efficacy outcome was change in gingival inflammation, measured by modified gingival index (MGI), and secondary outcomes included changes in bleeding-on-probing (BOP), amount of plaque, pocket depth, clinical attachment level, and gingival crevicular fluid levels of matrix metalloproteinases (MMPs) over 90 days. RESULTS A once-per-week intragingival injection of AMY-101 for 3 weeks was safe and well-tolerated in all participants resulting in significant (P<0.001) reductions in clinical indices measuring gingival inflammation (MGI and BOP). AMY-101 significantly (P<0.05) reduced MMP-8 and MMP-9 levels, indicators of inflammatory tissue destruction. These therapeutic effects persisted for at least 3 months post-treatment. CONCLUSION AMY-101 causes significant and sustainable reduction in gingival inflammation without adverse events and merits further investigation for the treatment of periodontitis and other oral or peri-implant inflammatory conditions. TRIAL REGISTRATION ClinicalTrials.gov: NCT03694444. FUNDING Amyndas Pharmaceuticals. Amyndas contributed to the design and conducts of the clinical trial and in the writing of the manuscript.
Collapse
Affiliation(s)
- Hatice Hasturk
- Center for Clinical and Translational Research, The Forsyth Institute, Cambridge, United States of America
| | - George Hajishengallis
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, United States of America
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Dimitrios C Mastellos
- Division of Biodiagnostic Sciences and Technologies, National Center for Scientific Research 'Demokritos', Athens, Greece
| | | |
Collapse
|
38
|
Thomas KT, Zakharenko SS. MicroRNAs in the Onset of Schizophrenia. Cells 2021; 10:2679. [PMID: 34685659 PMCID: PMC8534348 DOI: 10.3390/cells10102679] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/30/2021] [Accepted: 10/02/2021] [Indexed: 12/14/2022] Open
Abstract
Mounting evidence implicates microRNAs (miRNAs) in the pathology of schizophrenia. These small noncoding RNAs bind to mRNAs containing complementary sequences and promote their degradation and/or inhibit protein synthesis. A single miRNA may have hundreds of targets, and miRNA targets are overrepresented among schizophrenia-risk genes. Although schizophrenia is a neurodevelopmental disorder, symptoms usually do not appear until adolescence, and most patients do not receive a schizophrenia diagnosis until late adolescence or early adulthood. However, few studies have examined miRNAs during this critical period. First, we examine evidence that the miRNA pathway is dynamic throughout adolescence and adulthood and that miRNAs regulate processes critical to late neurodevelopment that are aberrant in patients with schizophrenia. Next, we examine evidence implicating miRNAs in the conversion to psychosis, including a schizophrenia-associated single nucleotide polymorphism in MIR137HG that is among the strongest known predictors of age of onset in patients with schizophrenia. Finally, we examine how hemizygosity for DGCR8, which encodes an obligate component of the complex that synthesizes miRNA precursors, may contribute to the onset of psychosis in patients with 22q11.2 microdeletions and how animal models of this disorder can help us understand the many roles of miRNAs in the onset of schizophrenia.
Collapse
Affiliation(s)
- Kristen T. Thomas
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Stanislav S. Zakharenko
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
39
|
Ma H, Chen Y, Yu M, Chen X, Qi L, Wei S, Fan Q, Xu Q, Zhan M, Sha Z. Immune role of the complement component 6 gene and its associated novel miRNA, miR-727, in half-smooth tongue sole (Cynoglossus semilaevis). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 123:104156. [PMID: 34077766 DOI: 10.1016/j.dci.2021.104156] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 06/12/2023]
Abstract
The complement component 6 (C6) gene is a component of the membrane attack complex (MAC), which causes rapid lytic destruction of bacteria. MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene stability, including that of immune genes. However, current research on the function of C6 and its regulation by miRNAs is lacking. In the present study, we identified and characterized C6 and a novel miRNA, miR-727 (designated CsC6 and Cse-miR-727, respectively), of the half-smooth tongue sole (Cynoglossus semilaevis) that responded to infection with Vibrio anguillarum, a Gram-negative pathogen of marine fish. The full-length cDNA of CsC6 contained a 256 bp 5' untranslated region (5'-UTR), a 2820 bp open reading frame (ORF) encoding 939 amino acids, and a 205 bp 3'-UTR. SMART analysis showed that CsC6 contains typical C6 domains, including three TSP1 domains, one LDLa domain, one MACPF domain, two CCP domains and two FIMAC domains. CsC6 and Cse-miR-727 are widely expressed in the 13 tissues of half-smooth tongue sole, and their expression in immune tissues is significantly changed after V. anguillarum infection, generally showing an inverse trend. We confirmed that CsC6 was the target gene of Cse-miR-727 using the dual luciferase reporter assay and that Cse-miR-727 regulated CsC6 at the protein level using quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting. The hepatic expression levels of not only the MAC components C7, C8α, C8β, C8γ and C9 but also the MAPKs, NF-κβ, AP-1, IL1β, IL6 and TNFα, which are involved in many signaling pathways, changed significantly in half-smooth tongue sole following stimulation with the Cse-miR-727 agomir and inhibitor. This evidence suggested that CsC6 could be mediated by Cse-miR-727 to affect MAC assembly and immune signaling pathways in half-smooth tongue soles. To our best knowledge, this study is the first to investigate the regulatory mechanism and immune response of complement genes mediated by miRNAs in fish.
Collapse
Affiliation(s)
- Hui Ma
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Yadong Chen
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, 266071, China
| | - Mengjun Yu
- College of Fisheries and Life Sciences, Dalian Ocean University, Dalian, 116023, China
| | - Xuejie Chen
- College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 200000, China
| | - Longjiang Qi
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Shu Wei
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Qingxin Fan
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Qian Xu
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Min Zhan
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Zhenxia Sha
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
40
|
Hajishengallis G, Hasturk H, Lambris JD. C3-targeted therapy in periodontal disease: moving closer to the clinic. Trends Immunol 2021; 42:856-864. [PMID: 34483038 PMCID: PMC8487962 DOI: 10.1016/j.it.2021.08.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023]
Abstract
Complement plays a key role in immunosurveillance and homeostasis. When dysregulated or overactivated, complement can become a pathological effector, as seen in several inflammatory disorders, including periodontal disease. Recently, clinical correlative studies and preclinical mechanistic investigations have collectively demonstrated that complement is hyperactivated during periodontitis and that targeting its central component (C3) provides therapeutic benefit in nonhuman primates (NHPs). The preclinical efficacy of a C3-targeted drug candidate combined with excellent safety and pharmacokinetic profiles supported its use in a recent Phase IIa clinical study in which C3 inhibition resolved gingival inflammation in patients with periodontal disease. We posit that C3-targeted intervention might represent a novel and transformative host-modulation therapy meriting further investigation in Phase III clinical trials for the treatment of periodontitis.
Collapse
Affiliation(s)
- George Hajishengallis
- University of Pennsylvania, Penn Dental Medicine, Department of Basic and Translational Sciences, Philadelphia, PA, USA.
| | - Hatice Hasturk
- The Forsyth Institute, Center for Clinical and Translational Research, Cambridge, MA, USA.
| | - John D Lambris
- University of Pennsylvania, Perelman School of Medicine, Department of Pathology and Laboratory Medicine, Philadelphia, USA.
| |
Collapse
|
41
|
Sarkar A, Galasiti Kankanamalage AC, Zhang Q, Cheng H, Sivaprakasam P, Naglich J, Xie C, Gangwar S, Boger DL. Synthesis, structure-activity relationship studies and evaluation of a TLR 3/8/9 agonist and its analogues. Med Chem Res 2021; 30:1377-1385. [PMID: 34421287 DOI: 10.1007/s00044-021-02736-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A comprehensive SAR study of a putative TLR 3/8/9 agonist was conducted. Despite the excitement surrounding the potential of the first small molecule TLR3 agonist with a compound that additionally displayed agonist activity for TLR8 and TLR9, compound 1 displayed disappointing activity in our hands, failing to match the potency (EC50) reported and displaying only a low efficacy for the extent of stimulated NF-κB activation and release. The evaluation of >75 analogs of 1, many of which constitute minor modifications in the structure, failed to identify any that displayed significant activity and none that exceeded the modest activity found for 1.
Collapse
Affiliation(s)
- Anindya Sarkar
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, USA
| | - Anushka C Galasiti Kankanamalage
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, USA
| | - Qian Zhang
- Bristol Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, USA
| | - Heng Cheng
- Bristol Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, USA
| | - Prasanna Sivaprakasam
- Bristol Myers Squibb Research & Development, PO Box 4000, Princeton, New Jersey 08543 USA
| | - Joseph Naglich
- Bristol Myers Squibb Research & Development, PO Box 4000, Princeton, New Jersey 08543 USA
| | - Chunshan Xie
- Bristol Myers Squibb Research & Development, PO Box 4000, Princeton, New Jersey 08543 USA
| | - Sanjeev Gangwar
- Bristol Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, USA
| | - Dale L Boger
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
42
|
Zipris D. Visceral Adipose Tissue: A New Target Organ in Virus-Induced Type 1 Diabetes. Front Immunol 2021; 12:702506. [PMID: 34421908 PMCID: PMC8371384 DOI: 10.3389/fimmu.2021.702506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes (T1D) is a proinflammatory pathology that leads to the specific destruction of insulin producing β-cells and hyperglycaemia. Much of the knowledge about type 1 diabetes (T1D) has focused on mechanisms of disease progression such as adaptive immune cells and the cytokines that control their function, whereas mechanisms linked with the initiation of the disease remain unknown. It has been hypothesized that in addition to genetics, environmental factors play a pivotal role in triggering β-cell autoimmunity. The BioBreeding Diabetes Resistant (BBDR) and LEW1.WR1 rats have been used to decipher the mechanisms that lead to virus-induced T1D. Both animals develop β-cell inflammation and hyperglycemia upon infection with the parvovirus Kilham Rat Virus (KRV). Our earlier in vitro and in vivo studies indicated that KRV-induced innate immune upregulation early in the disease course plays a causal role in triggering β-cell inflammation and destruction. Furthermore, we recently found for the first time that infection with KRV induces inflammation in visceral adipose tissue (VAT) detectable as early as day 1 post-infection prior to insulitis and hyperglycemia. The proinflammatory response in VAT is associated with macrophage recruitment, proinflammatory cytokine and chemokine upregulation, endoplasmic reticulum (ER) and oxidative stress responses, apoptosis, and downregulation of adipokines and molecules that mediate insulin signaling. Downregulation of inflammation suppresses VAT inflammation and T1D development. These observations are strikingly reminiscent of data from obesity and type 2 diabetes (T2D) in which VAT inflammation is believed to play a causal role in disease mechanisms. We propose that VAT inflammation and dysfunction may be linked with the mechanism of T1D progression.
Collapse
Affiliation(s)
- Danny Zipris
- Innate Biotechnologies LLC, Denver, CO, United States
| |
Collapse
|
43
|
Kwon SH, Park JE, Cho YH, Lee JS. Effect of Vibrio-Derived Extracellular Protease vEP-45 on the Blood Complement System. BIOLOGY 2021; 10:biology10080798. [PMID: 34440030 PMCID: PMC8389632 DOI: 10.3390/biology10080798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/09/2021] [Accepted: 08/15/2021] [Indexed: 11/16/2022]
Abstract
Vibrio vulnificus is a pathogenic bacterium that can causes wound infections and fetal septicemia. We have reported that V. vulnificus ATCC29307 produces an extracellular zinc-metalloprotease (named vEP-45). Our previous results showed that vEP-45 can convert prothrombin to active thrombin and also activate the plasma kallikrein/kinin system. In this study, the effect of vEP-45 on the activation of the complement system was examined. We found that vEP-45 could proteolytically convert the key complement precursor molecules, including C3, C4, and C5, to their corresponding active forms (e.g., C3a, C3b, C4a, C4b, and C5a) in vitro cleavage assays. C5b production from C5 cleavage mediated by vEP-45 was not observed, whereas the level of C5a was increased in a dose-dependent manner compared to that of the non-treated control. The cleavage of the complement proteins in human plasma by vEP-45 was also confirmed via Western blotting. Furthermore, vEP-45 could convert C3 and C5 to active C3a and C5a as a proinflammatory mediator, while no cleavage of C4 was observed. These results suggest that vEP-45 can activate the complement system involved in innate immunity through an alternative pathway.
Collapse
Affiliation(s)
- So Hyun Kwon
- Department of Biomedical Science, College of Natural Sciences and Public Health and Safety, Chosun University, Gwangju 61452, Korea; (S.H.K.); (J.E.P.); (Y.H.C.)
| | - Jung Eun Park
- Department of Biomedical Science, College of Natural Sciences and Public Health and Safety, Chosun University, Gwangju 61452, Korea; (S.H.K.); (J.E.P.); (Y.H.C.)
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Korea
| | - Yeong Hee Cho
- Department of Biomedical Science, College of Natural Sciences and Public Health and Safety, Chosun University, Gwangju 61452, Korea; (S.H.K.); (J.E.P.); (Y.H.C.)
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Korea
| | - Jung Sup Lee
- Department of Biomedical Science, College of Natural Sciences and Public Health and Safety, Chosun University, Gwangju 61452, Korea; (S.H.K.); (J.E.P.); (Y.H.C.)
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Korea
- Correspondence: ; Tel.: +82-62-230-6665
| |
Collapse
|
44
|
Oxidative Stress and Mitochondrial Damage in Dry Age-Related Macular Degeneration Like NFE2L2/PGC-1α -/- Mouse Model Evoke Complement Component C5a Independent of C3. BIOLOGY 2021; 10:biology10070622. [PMID: 34356477 PMCID: PMC8301195 DOI: 10.3390/biology10070622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022]
Abstract
Aging-associated chronic oxidative stress and inflammation are known to be involved in various diseases, e.g., age-related macular degeneration (AMD). Previously, we reported the presence of dry AMD-like signs, such as elevated oxidative stress, dysfunctional mitophagy and the accumulation of detrimental oxidized materials in the retinal pigment epithelial (RPE) cells of nuclear factor erythroid 2-related factor 2, and a peroxisome proliferator-activated receptor gamma coactivator 1-alpha (NFE2L2/PGC1α) double knockout (dKO) mouse model. Here, we investigated the dynamics of inflammatory markers in one-year-old NFE2L2/PGC1α dKO mice. Immunohistochemical analysis revealed an increase in levels of Toll-like receptors 3 and 9, while those of NOD-like receptor 3 were decreased in NFE2L2/PGC1α dKO retinal specimens as compared to wild type animals. Further analysis showed a trend towards an increase in complement component C5a independent of component C3, observed to be tightly regulated by complement factor H. Interestingly, we found that thrombin, a serine protease enzyme, was involved in enhancing the terminal pathway producing C5a, independent of C3. We also detected an increase in primary acute phase C-reactive protein and receptor for advanced glycation end products in NFE2L2/PGC1α dKO retina. Our main data show C5 and thrombin upregulation together with decreased C3 levels in this dry AMD-like model. In general, the retina strives to mount an orchestrated inflammatory response while attempting to maintain tissue homeostasis and resolve inflammation.
Collapse
|
45
|
Tang Q, Hao Y, Wang L, Lu C, Li M, Si Z, Wu X, Lu Z. Characterization of a bacterial strain Lactobacillus paracasei LP10266 recovered from an endocarditis patient in Shandong, China. BMC Microbiol 2021; 21:183. [PMID: 34134621 PMCID: PMC8210379 DOI: 10.1186/s12866-021-02253-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/31/2021] [Indexed: 02/06/2023] Open
Abstract
Background Lactobacilli are often recognized as beneficial partners in human microbial environments. However, lactobacilli also cause diseases in human, e.g. infective endocarditis (IE), septicaemia, rheumatic vascular disease, and dental caries. Therefore, the identification of potential pathogenic traits associated with lactobacilli will facilitate the prevention and treatment of the diseases caused by lactobacilli. Herein, we investigated the genomic traits and pathogenic potential of a novel bacterial strain Lactobacillus paracasei LP10266 which has caused a case of IE. We isolated L. paracasei LP10266 from an IE patient’s blood to perform high-throughput sequencing and compared the genome of strain LP10266 with those of closely related lactobacilli to determine genes associated with its infectivity. We performed the antimicrobial susceptibility testing on strain LP10266. We assessed its virulence by mouse lethality and serum bactericidal assays as well as its serum complement- and platelet-activating ability. The biofilm formation and adherence of strain LP10266 were also studied. Results Phylogenetic analysis revealed that strain LP10266 was allied with L. casei and L. paracasei. Genomic studies revealed two spaCBA pilus clusters and one novel exopolysaccharides (EPS) cluster in strain LP10266, which was sensitive to ampicillin, penicillin, levofloxacin, and imipenem, but resistant to cefuroxime, cefazolin, cefotaxime, meropenem, and vancomycin. Strain LP10266 was nonfatal and sensitive to serum, capable of activating complement 3a and terminal complement complex C5b-9 (TCC). Strain LP10266 could not induce platelet aggregation but displayed a stronger biofilm formation ability and adherence to human vascular endothelial cells (HUVECs) compared to the standard control strain L. paracasei ATCC25302. Conclusion The genome of a novel bacterial strain L. paracasei LP10266 was sequenced. Our results based on various types of assays consistently revealed that L. paracasei LP10266 was a potential pathogen to patients with a history of cardiac disease and inguinal hernia repair. Strain LP10266 showed strong biofilm formation ability and adherence, enhancing the awareness of L. paracasei infections. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-021-02253-8.
Collapse
Affiliation(s)
- Qi Tang
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Yingying Hao
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China.,Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Lu Wang
- Department of Dermatology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, Shandong, China
| | - Chao Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ming Li
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Zaifeng Si
- Department of Clinical Laboratory, Dezhou Traditional Chinese Medicine Hospital, Dezhou, 253000, Shandong, China
| | - Xiaoben Wu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
46
|
Wang CW, Hao Y, Di Gianfilippo R, Sugai J, Li J, Gong W, Kornman KS, Wang HL, Kamada N, Xie Y, Giannobile WV, Lei YL. Machine learning-assisted immune profiling stratifies peri-implantitis patients with unique microbial colonization and clinical outcomes. Theranostics 2021; 11:6703-6716. [PMID: 34093848 PMCID: PMC8171076 DOI: 10.7150/thno.57775] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/31/2021] [Indexed: 12/15/2022] Open
Abstract
Rationale: The endemic of peri-implantitis affects over 25% of dental implants. Current treatment depends on empirical patient and site-based stratifications and lacks a consistent risk grading system. Methods: We investigated a unique cohort of peri-implantitis patients undergoing regenerative therapy with comprehensive clinical, immune, and microbial profiling. We utilized a robust outlier-resistant machine learning algorithm for immune deconvolution. Results: Unsupervised clustering identified risk groups with distinct immune profiles, microbial colonization dynamics, and regenerative outcomes. Low-risk patients exhibited elevated M1/M2-like macrophage ratios and lower B-cell infiltration. The low-risk immune profile was characterized by enhanced complement signaling and higher levels of Th1 and Th17 cytokines. Fusobacterium nucleatum and Prevotella intermedia were significantly enriched in high-risk individuals. Although surgery reduced microbial burden at the peri-implant interface in all groups, only low-risk individuals exhibited suppression of keystone pathogen re-colonization. Conclusion: Peri-implant immune microenvironment shapes microbial composition and the course of regeneration. Immune signatures show untapped potential in improving the risk-grading for peri-implantitis.
Collapse
Affiliation(s)
- Chin-Wei Wang
- Department of Periodontics and Oral Medicine, the University of Michigan School of Dentistry, Ann Arbor, MI 48109
| | - Yuning Hao
- Department of Computational Mathematics, Science, and Engineering, Michigan State University, East Lansing, MI 48823
| | - Riccardo Di Gianfilippo
- Department of Periodontics and Oral Medicine, the University of Michigan School of Dentistry, Ann Arbor, MI 48109
| | - James Sugai
- Department of Periodontics and Oral Medicine, the University of Michigan School of Dentistry, Ann Arbor, MI 48109
| | - Jiaqian Li
- Department of Periodontics and Oral Medicine, the University of Michigan School of Dentistry, Ann Arbor, MI 48109
| | - Wang Gong
- Department of Periodontics and Oral Medicine, the University of Michigan School of Dentistry, Ann Arbor, MI 48109
| | - Kenneth S. Kornman
- Department of Periodontics and Oral Medicine, the University of Michigan School of Dentistry, Ann Arbor, MI 48109
| | - Hom-Lay Wang
- Department of Periodontics and Oral Medicine, the University of Michigan School of Dentistry, Ann Arbor, MI 48109
| | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, the University of Michigan Medical School, Ann Arbor, MI 48105
- Rogel Cancer Center, the University of Michigan, Ann Arbor, MI 48105
| | - Yuying Xie
- Department of Computational Mathematics, Science, and Engineering, Michigan State University, East Lansing, MI 48823
| | - William V. Giannobile
- Department of Periodontics and Oral Medicine, the University of Michigan School of Dentistry, Ann Arbor, MI 48109
- Department of Biomedical Engineering, College of Engineering & Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
- Current Affiliation: Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yu Leo Lei
- Department of Periodontics and Oral Medicine, the University of Michigan School of Dentistry, Ann Arbor, MI 48109
- Rogel Cancer Center, the University of Michigan, Ann Arbor, MI 48105
| |
Collapse
|
47
|
Fu YL, Harrison RE. Microbial Phagocytic Receptors and Their Potential Involvement in Cytokine Induction in Macrophages. Front Immunol 2021; 12:662063. [PMID: 33995386 PMCID: PMC8117099 DOI: 10.3389/fimmu.2021.662063] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
Phagocytosis is an essential process for the uptake of large (>0.5 µm) particulate matter including microbes and dying cells. Specialized cells in the body perform phagocytosis which is enabled by cell surface receptors that recognize and bind target cells. Professional phagocytes play a prominent role in innate immunity and include macrophages, neutrophils and dendritic cells. These cells display a repertoire of phagocytic receptors that engage the target cells directly, or indirectly via opsonins, to mediate binding and internalization of the target into a phagosome. Phagosome maturation then proceeds to cause destruction and recycling of the phagosome contents. Key subsequent events include antigen presentation and cytokine production to alert and recruit cells involved in the adaptive immune response. Bridging the innate and adaptive immunity, macrophages secrete a broad selection of inflammatory mediators to orchestrate the type and magnitude of an inflammatory response. This review will focus on cytokines produced by NF-κB signaling which is activated by extracellular ligands and serves a master regulator of the inflammatory response to microbes. Macrophages secrete pro-inflammatory cytokines including TNFα, IL1β, IL6, IL8 and IL12 which together increases vascular permeability and promotes recruitment of other immune cells. The major anti-inflammatory cytokines produced by macrophages include IL10 and TGFβ which act to suppress inflammatory gene expression in macrophages and other immune cells. Typically, macrophage cytokines are synthesized, trafficked intracellularly and released in response to activation of pattern recognition receptors (PRRs) or inflammasomes. Direct evidence linking the event of phagocytosis to cytokine production in macrophages is lacking. This review will focus on cytokine output after engagement of macrophage phagocytic receptors by particulate microbial targets. Microbial receptors include the PRRs: Toll-like receptors (TLRs), scavenger receptors (SRs), C-type lectin and the opsonic receptors. Our current understanding of how macrophage receptor stimulation impacts cytokine production is largely based on work utilizing soluble ligands that are destined for endocytosis. We will instead focus this review on research examining receptor ligation during uptake of particulate microbes and how this complex internalization process may influence inflammatory cytokine production in macrophages.
Collapse
Affiliation(s)
- Yan Lin Fu
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada
| | - Rene E. Harrison
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada
| |
Collapse
|
48
|
Wang X, Sahu KK, Cerny J. Coagulopathy, endothelial dysfunction, thrombotic microangiopathy and complement activation: potential role of complement system inhibition in COVID-19. J Thromb Thrombolysis 2021; 51:657-662. [PMID: 33063256 PMCID: PMC7561230 DOI: 10.1007/s11239-020-02297-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/26/2020] [Indexed: 12/18/2022]
Abstract
Coronavirus disease-2019 (COVID-19) is a rapidly evolving health crisis caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 is a novel disease entity and we are in a learning phase with regards to the pathogenesis, disease manifestations, and therapeutics. In addition to the primary lung injury, many patients especially the ones with moderate to severe COVID-19 display evidence of endothelial damage, complement activation, which leads to a pro-coagulable state. While there are still missing links in our understanding, the interplay of endothelium, complement system activation, and immune response to the SARS-CoV-2 virus is a surprisingly major factor in COVID-19 pathogenesis. One could envision COVID-19 becoming a novel hematological syndrome. This review is to discuss the available literature with regards to the involvement of the complement system, and coagulation cascade and their interaction with endothelium.
Collapse
Affiliation(s)
- Xin Wang
- Department of Medicine, UMass Memorial Health Care, University of Massachusetts Medical School, Worcester, MA USA
| | - Kamal Kant Sahu
- Department of Internal Medicine, Saint Vincent Hospital, Worcester, MA 01608 USA
| | - Jan Cerny
- Department of Medicine - Hematology, and Oncology, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA USA
| |
Collapse
|
49
|
Gao M, Dong Q, Wang W, Yang Z, Guo L, Lu Y, Ding B, Chen L, Zhang J, Xu R. Induced neural stem cell grafts exert neuroprotection through an interaction between Crry and Akt in a mouse model of closed head injury. Stem Cell Res Ther 2021; 12:128. [PMID: 33579360 PMCID: PMC7881465 DOI: 10.1186/s13287-021-02186-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 01/24/2021] [Indexed: 12/26/2022] Open
Abstract
Background Recently, growing evidence has indicated an important role of the complement system, a crucial component of immunity, in mediating neuroinflammation and promoting neuronal apoptosis following closed head injury (CHI). We previously reported that transplanted induced neural stem cells (iNSCs) pre-treated with CHI mouse serum could enhance complement receptor type 1-related protein y (Crry) expression and ameliorate complement-mediated damage in mouse CHI models. However, the mechanism underlying the elevated levels of Crry expression remains elusive. Methods CHI models were established using a standardized weight-drop device. We collected CHI mouse serum at 12 h post-trauma. RT-QPCR assay, western blot analysis, complement deposition assay, Akt inhibition assay, flow cytometry, cell transplantation, and functional assay were utilized to clarify the mechanism of Crry expression in iNSCs receiving CHI mouse serum treatment. Results We observed dramatic increases in the levels of Crry expression and Akt activation in iNSCs receiving CHI mouse serum treatment. Remarkably, Akt inhibition led to the reduction of Crry expression in iNSCs. Intriguingly, the treatment of iNSC-derived neurons with recombinant complement receptor 2-conjugated Crry (CR2-Crry), which inhibits all complement pathways, substantially enhanced Crry expression and Akt activation in neurons after CHI mouse serum treatment. In subsequent in vitro experiments of pre-treatment of iNSCs with CR2-Crry, we observed significant increases in the levels of Crry expression and Akt activation in iNSCs and iNSC-derived astrocytes and neurons post-treatment with CHI mouse serum. Additionally, an in vivo study showed that intracerebral-transplanted iNSCs pre-treated with CR2-Crry markedly enhanced Crry expression in neurons and protected neurons from complement-dependent damage in the brains of CHI mice. Conclusion INSCs receiving CR2-Crry pre-treatment increased the levels of Crry expression in iNSCs and iNSC-derived astrocytes and neurons and attenuated complement-mediated injury following CHI. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02186-z.
Collapse
Affiliation(s)
- Mou Gao
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.,Department of Neurosurgery, The PLA General Hospital, Beijing, 100853, China
| | - Qin Dong
- Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing, 100038, China
| | - Wenjia Wang
- Department of ENT-HN, Hainan Hospital of PLA General Hospital, Sanya, 572013, China
| | - Zhijun Yang
- Department of Neurosurgery, The Seventh Medical Center, The PLA General Hospital, Beijing, 100700, China
| | - Lili Guo
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yingzhou Lu
- Department of Obstetrics, Fu Xing Hospital, Capital Medical University, Beijing, 100038, China
| | - Boyun Ding
- Department of Neurosurgery, The Seventh Medical Center, The PLA General Hospital, Beijing, 100700, China
| | - Lihua Chen
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianning Zhang
- Department of Neurosurgery, The PLA General Hospital, Beijing, 100853, China.
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China. .,Department of Neurosurgery, The Seventh Medical Center, The PLA General Hospital, Beijing, 100700, China.
| |
Collapse
|
50
|
Mommert S, Doenni L, Szudybill P, Zoeller C, Beyer FH, Werfel T. C3a and Its Receptor C3aR Are Detectable in Normal Human Epidermal Keratinocytes and Are Differentially Regulated via TLR3 and LL37. J Innate Immun 2021; 13:164-178. [PMID: 33445177 DOI: 10.1159/000512547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/25/2020] [Indexed: 12/20/2022] Open
Abstract
To study the molecular interplay between TLRs and complement representing ancient danger-sensing mechanisms, we examined the regulation of the C3a/anaphylatoxin C3a receptor (C3aR) axis in normal human epidermal keratinocytes (NHEKs) by treatment with different TLR ligands. Protein staining followed by flow cytometry revealed highly constitutive intracellular expression levels of the C3aR in NHEKs. Stimulation with Poly I:C up-regulated C3aR mRNA and intra- and extracellular expression in NHEKs which showed functional relevance by up-regulating CXCL10 and down-regulating C3 expression in response to C3a. mRNA and protein levels of C3 and protease cathepsin L (CTSL) that can cleave C3 were up-regulated by the TLR3 ligand Poly I:C. Enhanced intracellular expression levels of the biologically active C3 fragment (C3a), in response to TLR3 stimulation were also detectable in NHEKs. Cathelicidin antimicrobial peptide LL-37 potentiated Poly I:C-induced C3aR, C3, and CTSL up-regulation. In conclusion, we point to a role of TLR3 to promote up-regulation of C3aR, C3, and CTSL expression levels and generation of C3a. Our data provide evidence that local generation and activation of complement components as described for T cells or myeloid cells represent a scenario which may take place in a similar way in NHEKs.
Collapse
Affiliation(s)
- Susanne Mommert
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany,
| | - Lisa Doenni
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Phillip Szudybill
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Christoph Zoeller
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Frerk Hinnerk Beyer
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| |
Collapse
|