1
|
Zhao X, Qiu Y, Liang L, Fu X. Interkingdom signaling between gastrointestinal hormones and the gut microbiome. Gut Microbes 2025; 17:2456592. [PMID: 39851261 PMCID: PMC11776477 DOI: 10.1080/19490976.2025.2456592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/12/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
The interplay between the gut microbiota and gastrointestinal hormones plays a pivotal role in the health of the host and the development of diseases. As a vital component of the intestinal microecosystem, the gut microbiota influences the synthesis and release of many gastrointestinal hormones through mechanisms such as modulating the intestinal environment, producing metabolites, impacting mucosal barriers, generating immune and inflammatory responses, and releasing neurotransmitters. Conversely, gastrointestinal hormones exert feedback regulation on the gut microbiota by modulating the intestinal environment, nutrient absorption and utilization, and the bacterial biological behavior and composition. The distributions of the gut microbiota and gastrointestinal hormones are anatomically intertwined, and close interactions between the gut microbiota and gastrointestinal hormones are crucial for maintaining gastrointestinal homeostasis. Interventions leveraging the interplay between the gut microbiota and gastrointestinal hormones have been employed in the clinical management of metabolic diseases and inflammatory bowel diseases, such as bariatric surgery and fecal microbiota transplantation, offering promising targets for the treatment of dysbiosis-related diseases.
Collapse
Affiliation(s)
- Xinyu Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Ye Qiu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Lanfan Liang
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiangsheng Fu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Chen J, Yu K, Yu X, Zhang R, Chen B. Transcriptomic and physiological analyses reveal the toxic effects of inorganic filters (nZnO and nTiO 2) on scleractinian coral Galaxea fascicularis. ENVIRONMENTAL RESEARCH 2025; 267:120663. [PMID: 39709120 DOI: 10.1016/j.envres.2024.120663] [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: 10/10/2024] [Revised: 12/02/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
The effects of sunscreen on scleractinian corals have garnered widespread attention; however, the toxic effects and mechanisms remain unclear. This study investigated the toxicological effects of two common inorganic filters used in sunscreens, nano zinc oxide and titanium dioxide (nZnO and nTiO₂), on the reef-building coral Galaxea fascicularis, focusing on the phenotypic, physiological, and transcriptomic responses. The results showed that after exposure to 0.8 mg/L of nZnO and 30 mg/L of nTiO₂ for 48 h, all coral polyps exhibited retraction. Zn and Ti ions were detected in coral tissues at concentrations of 67.18 and 24.87 μg/g, respectively, indicating the accumulation of nZnO and nTiO2 in coral tissues. The zooxanthellae density, Fv/Fm, and chlorophyll-a content decreased significantly. The activity of antioxidant enzymes showed an increasing trend. Meanwhile, glutamine synthetase and glutamate dehydrogenase activities exhibited a decreasing trend. The health status of corals was impacted as a result of nZnO and nTiO2 stress. Transcriptomic analysis showed that the toxicity mechanisms of nZnO and nTiO2 differed in corals. Following exposure to nZnO, differentially expressed genes (DEGs) in corals were mainly enriched in signaling pathways related to immune response. The genes related to innate immunity, such as MASP1, MUC5AC, TLRs, and C2, were significantly upregulated, indicating that nZnO exposure induces an innate immune response in corals. Meanwhile, following nTiO2 exposure, the upregulated DEGs were mainly enriched in signaling pathways related to transporter activity. In contrast, the downregulated DEGs were mainly enriched in energy metabolism pathways, indicating that nTiO2 disrupted the energy supply of corals, thereby leading to an increased demand for nutrient transport. This study reveals the toxic effects of nZnO and nTiO2, and their mechanisms of action on scleractinian corals, providing a reference for further assessing the toxicity of sunscreen on corals.
Collapse
Affiliation(s)
- Jian Chen
- Guangxi Laboratory on the Study of Coral Reefs in the South China Sea, Coral Reef Research Center of China, School of Marine Sciences, Guangxi University, Nanning, 530004, China
| | - Kefu Yu
- Guangxi Laboratory on the Study of Coral Reefs in the South China Sea, Coral Reef Research Center of China, School of Marine Sciences, Guangxi University, Nanning, 530004, China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China.
| | - Xiaopeng Yu
- Guangxi Laboratory on the Study of Coral Reefs in the South China Sea, Coral Reef Research Center of China, School of Marine Sciences, Guangxi University, Nanning, 530004, China
| | - Ruijie Zhang
- Guangxi Laboratory on the Study of Coral Reefs in the South China Sea, Coral Reef Research Center of China, School of Marine Sciences, Guangxi University, Nanning, 530004, China
| | - Biao Chen
- Guangxi Laboratory on the Study of Coral Reefs in the South China Sea, Coral Reef Research Center of China, School of Marine Sciences, Guangxi University, Nanning, 530004, China
| |
Collapse
|
3
|
Wang YC, Zhu HH, He LC, Yao YT, Zhang L, Xue XL, Li JY, Zhang L, Song B, Shi CH, Li YS, Gao Y, Yang JH, Xu YM. Proteome Profiling of Serum Reveals Pathological Mechanisms and Biomarker Candidates for Cerebral Small Vessel Disease. Transl Stroke Res 2025:10.1007/s12975-025-01332-6. [PMID: 39934548 DOI: 10.1007/s12975-025-01332-6] [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: 11/12/2024] [Revised: 12/29/2024] [Accepted: 01/18/2025] [Indexed: 02/13/2025]
Abstract
Cerebral small vessel disease (CSVD) is a global brain disorder that is characterized by a series of clinical, neuroimaging, and neuropathological manifestations. However, the molecular pathophysiological mechanisms of CSVD have not been thoroughly investigated. Liquid chromatography-tandem mass spectrometry-based proteomics has broad application prospects in biomedicine. It is used to elucidate disease-related molecular processes and pathophysiological pathways, thus providing an important opportunity to explore the pathophysiological mechanisms of CSVD. Serum samples were obtained from 96 participants (58 with CSVD and 38 controls) consecutively recruited from The First Affiliated Hospital of Zhengzhou University. After removing high-abundance proteins, the serum samples were analyzed using high-resolution mass spectrometry. Bioinformatics methods were used for in-depth analysis of the obtained proteomic data, and the results were verified experimentally. Compared with the control group, 52 proteins were differentially expressed in the sera of the CSVD group. Furthermore, analyses indicated the involvement of these differentially expressed proteins in CSVD through participation in the overactivation of complement and coagulation cascades and dysregulation of insulin-like growth factor-binding proteins. The proteomic biomarker panel identified by the machine learning model combined with clinical features is expected to facilitate the diagnosis of CSVD (AUC = 0.947, 95% CI = 0.895-0.978). The study is the most in-depth study on CSVD proteomics to date and suggests that the overactivation of the complement cascade and the dysregulation of IGFBP on- IGF may be closely correlated with the occurrence and progression of CSVD, offering the potential to develop peripheral blood biomarkers and providing new insights into the biological basis of CSVD.
Collapse
Affiliation(s)
- Yun-Chao Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- National Health Commission Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China
| | - Hang-Hang Zhu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- National Health Commission Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China
| | - Liu-Chang He
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- National Health Commission Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China
| | - Ya-Ting Yao
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China
| | - Lei Zhang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China
| | - Xin-Li Xue
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China
| | - Jing-Yi Li
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China
| | - Li Zhang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China
| | - Bo Song
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China
| | - Chang-He Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China
| | - Yu-Sheng Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China
| | - Yuan Gao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China.
| | - Jing-Hua Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China.
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- National Health Commission Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, China.
- The First Affiliated Hospital of Zhengzhou University, 1 Jian-She East Road, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
4
|
Hanford HE, Price CTD, Uriarte S, Abu Kwaik Y. Inhibition and evasion of neutrophil microbicidal responses by Legionella longbeachae. mBio 2025; 16:e0327424. [PMID: 39679679 PMCID: PMC11796426 DOI: 10.1128/mbio.03274-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
Legionella species evade degradation and proliferate within alveolar macrophages as an essential step for the manifestation of disease. However, most intracellular bacterial pathogens are restricted in neutrophils, which are the first line of innate immune defense against invading pathogens. Bacterial degradation within neutrophils is mediated by the fusion of microbicidal granules to pathogen-containing phagosomes and the generation of reactive oxygen species (ROS) by the phagocyte NADPH oxidase complex. Here, we show that human neutrophils fail to trigger microbicidal processes and, consequently, fail to restrict L. longbeachae. In addition, neutrophils infected with L. longbeachae fail to undergo a robust pro-inflammatory response, such as degranulation and IL-8 production. Here, we identify three strategies employed by L. longbeachae for evading restriction by neutrophils and inhibiting the neutrophil microbicidal response to other bacteria co-inhabiting in the same cell. First, L. longbeachae excludes the cytosolic and membrane-bound subunits of the phagocyte NADPH oxidase complex from its phagosomal membrane independent of the type 4 secretion system (T4SS). Consequently, infected neutrophils fail to generate robust ROS in response to L. longbeachae. Second, L. longbeachae impedes the fusion of azurophilic granules to its phagosome and the phagosomes of bacteria co-inhabiting the same cell through T4SS-independent mechanisms. Third, L. longbeachae protects phagosomes of co-inhabiting bacteria from degradation by ROS through a trans-acting T4SS-dependent mechanism. Collectively, we conclude that L. longbeachae evades restriction by human neutrophils via T4SS-independent mechanisms and utilizes trans-acting T4SS-dependent mechanisms for inhibition of neutrophil ROS generation throughout the cell cytosol. IMPORTANCE Legionella longbeachae is commonly found in soil environments where it interacts with a wide variety of protist hosts and microbial competitors. Upon transmission to humans, L. longbeachae invades and replicates within alveolar macrophages, leading to the manifestation of pneumonia. In addition to alveolar macrophages, neutrophils are abundant immune cells acting as the first line of defense against invading pathogens. While most intracellular bacterial species are killed and degraded by neutrophils, we show that L. longbeachae evades degradation. The pathogen impairs the major neutrophils' microbicidal processes, including the fusion of microbicidal granules to the pathogen-containing vacuole. By inhibiting of assembly of the phagocyte NADPH oxidase complex, the pathogen blocks neutrophils from generating microbicide reactive oxygen species. Overall, L. longbeachae employs unique virulence strategies to evade the major microbicidal processes of neutrophils.
Collapse
Affiliation(s)
- Hannah E. Hanford
- Department of Microbiology and Immunology, College of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Christopher T. D. Price
- Department of Microbiology and Immunology, College of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Silvia Uriarte
- Department of Microbiology and Immunology, College of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology, College of Medicine, University of Louisville, Louisville, Kentucky, USA
- Center for Predictive Medicine, College of Medicine, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
5
|
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
|
6
|
Das D, Thacker H, Priya K, Jain M, Singh S, Rai G. Complement component 5a receptor 1 and leukotriene B4 receptor 1 regulate neutrophil extracellular trap (NET) formation through Rap1a/B-Raf/ERK signaling pathway and their deficiency in term low birth weight newborns leads to deficient NETosis. Int Immunopharmacol 2024; 142:113165. [PMID: 39303536 DOI: 10.1016/j.intimp.2024.113165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Neutrophil extracellular traps (NETs) being one of the predominant activities of neutrophils has become its key defense mechanism owing to its extensive role in inflammation and infection. However, the mechanisms regulating NET formation or NETosis still remains to be better understood. Our earlier whole genome transcriptomic data revealed two G-protein couple receptors (GPCRs) - complement component 5a receptor 1 (C5aR1) and leukotriene B4 receptor 1 (LTB4R1) were downregulated in term low birth weight (tLBW) newborns with deficient NET formation abilities. Neutrophils employ C5aR1 and LTB4R1 for mediating their immune responses, inflammation and antimicrobial activity. Hence, this study was aimed to explore the role of two GPCRs, C5aR1 and LTB4R1 including their downstream signaling molecules in NETs induction and regulation. METHODS The validation of the transcriptomic data for C5aR1 and LTB4R1 was done using quantitative real time PCR. Pharmacological inhibition of C5aR1 and LTB4R1 using W-54011 and LY223982 on neutrophils of adults and newborns' was done to study their impact on NETosis. Extracellular DNA release, Reactive oxygen species (ROS) generation, expression of NET proteins, and signaling molecules downstream to C5aR1 and LTB4R1 were quantified using plate reader based assay, immunofluorescence, and western blotting. Myeloperoxidase (MPO)-DNA quantified by flow cytometry. Knockdown studies using siRNA against C5aR1 and LTB4R1 were done in HL-60 cells derived surrogate neutrophils and expression of downstream molecules of the two GPCRs, C5aR1 and LTB4R1 signaling axis along with NET proteins was quantified by western blotting. RESULTS The expression of C5aR1 and LTB4R1, extracellular DNA, ROS and NET associated proteins (NE, CitH3, PAD4 and MPO) was notably increased upon NET induction in healthy adults and normal birth weight (NBW) newborns' neutrophils. Pharmacological inhibition of these two GPCRs led to substantial reduction in NETosis, extracellular DNA, ROS generation, and expression of NET associated proteins like CitH3, NE, PAD4, MPO along with downstream signaling molecules Rap1a, B-Raf and pERK. Our observations suggest a precise role of C5aR1 and LTB4R1 on induction of NETs via Rap1a/B-Raf/ERK signaling axis. CONCLUSION The C5aR1 and LTB4R1 signaling via Rap1a/B-Raf/ERK axis acts as a signal-relay mechanism to regulate NET formation in neutrophils. Further, C5aR1 and LTB4R1 signaling cascade along with NET-associated proteins are remarkably downregulated in tLBW newborns' neutrophils leading to impaired NETosis in them. Therefore, C5aR1 and LTB4R1 and their signaling molecules could provide an effective therapeutic target for compromised NETosis like tLBW newborns.
Collapse
Affiliation(s)
- Doli Das
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Hiral Thacker
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Khushbu Priya
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Madhu Jain
- Department of Obstetrics & Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Shambhavi Singh
- Dr. D. Y. Patil Medical College, Navi Mumbai, Maharashtra 400706, India
| | - Geeta Rai
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
7
|
Avdonin PP, Blinova MS, Serkova AA, Komleva LA, Avdonin PV. Immunity and Coagulation in COVID-19. Int J Mol Sci 2024; 25:11267. [PMID: 39457048 PMCID: PMC11508857 DOI: 10.3390/ijms252011267] [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: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Discovered in late 2019, the SARS-CoV-2 coronavirus has caused the largest pandemic of the 21st century, claiming more than seven million lives. In most cases, the COVID-19 disease caused by the SARS-CoV-2 virus is relatively mild and affects only the upper respiratory tract; it most often manifests itself with fever, chills, cough, and sore throat, but also has less-common mild symptoms. In most cases, patients do not require hospitalization, and fully recover. However, in some cases, infection with the SARS-CoV-2 virus leads to the development of a severe form of COVID-19, which is characterized by the development of life-threatening complications affecting not only the lungs, but also other organs and systems. In particular, various forms of thrombotic complications are common among patients with a severe form of COVID-19. The mechanisms for the development of thrombotic complications in COVID-19 remain unclear. Accumulated data indicate that the pathogenesis of severe COVID-19 is based on disruptions in the functioning of various innate immune systems. The key role in the primary response to a viral infection is assigned to two systems. These are the pattern recognition receptors, primarily members of the toll-like receptor (TLR) family, and the complement system. Both systems are the first to engage in the fight against the virus and launch a whole range of mechanisms aimed at its rapid elimination. Normally, their joint activity leads to the destruction of the pathogen and recovery. However, disruptions in the functioning of these innate immune systems in COVID-19 can cause the development of an excessive inflammatory response that is dangerous for the body. In turn, excessive inflammation entails activation of and damage to the vascular endothelium, as well as the development of the hypercoagulable state observed in patients seriously ill with COVID-19. Activation of the endothelium and hypercoagulation lead to the development of thrombosis and, as a result, damage to organs and tissues. Immune-mediated thrombotic complications are termed "immunothrombosis". In this review, we discuss in detail the features of immunothrombosis associated with SARS-CoV-2 infection and its potential underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Pavel V. Avdonin
- Koltzov Institute of Developmental Biology RAS, ul. Vavilova, 26, 119334 Moscow, Russia; (P.P.A.)
| |
Collapse
|
8
|
Martínez-García M, Hernández-Lemus E. The Molecular Comorbidity Network of Periodontal Disease. Int J Mol Sci 2024; 25:10161. [PMID: 39337647 PMCID: PMC11432284 DOI: 10.3390/ijms251810161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Periodontal disease, a multifactorial inflammatory condition affecting the supporting structures of the teeth, has been increasingly recognized for its association with various systemic diseases. Understanding the molecular comorbidities of periodontal disease is crucial for elucidating shared pathogenic mechanisms and potential therapeutic targets. In this study, we conducted comprehensive literature and biological database mining by utilizing DisGeNET2R for extracting gene-disease associations, Romin for integrating and modeling molecular interaction networks, and Rentrez R libraries for accessing and retrieving relevant information from NCBI databases. This integrative bioinformatics approach enabled us to systematically identify diseases sharing associated genes, proteins, or molecular pathways with periodontitis. Our analysis revealed significant molecular overlaps between periodontal disease and several systemic conditions, including cardiovascular diseases, diabetes mellitus, rheumatoid arthritis, and inflammatory bowel diseases. Shared molecular mechanisms implicated in the pathogenesis of these diseases and periodontitis encompassed dysregulation of inflammatory mediators, immune response pathways, oxidative stress pathways, and alterations in the extracellular matrix. Furthermore, network analysis unveiled the key hub genes and proteins (such as TNF, IL6, PTGS2, IL10, NOS3, IL1B, VEGFA, BCL2, STAT3, LEP and TP53) that play pivotal roles in the crosstalk between periodontal disease and its comorbidities, offering potential targets for therapeutic intervention. Insights gained from this integrative approach shed light on the intricate interplay between periodontal health and systemic well-being, emphasizing the importance of interdisciplinary collaboration in developing personalized treatment strategies for patients with periodontal disease and associated comorbidities.
Collapse
Affiliation(s)
- Mireya Martínez-García
- Department of Immunology, National Institute of Cardiology ‘Ignacio Chávez’, Mexico City 14080, Mexico;
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City 14610, Mexico
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
9
|
Kruglikov IL, Scherer PE. Is the endotoxin-complement cascade the major driver in lipedema? Trends Endocrinol Metab 2024; 35:769-780. [PMID: 38688780 PMCID: PMC11387139 DOI: 10.1016/j.tem.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024]
Abstract
Lipedema is a poorly understood disorder of adipose tissue characterized by abnormal but symmetrical deposition of subcutaneous white adipose tissue (WAT) in proximal extremities. Here, we propose that the underlying cause for lipedema could be triggered by a selective accumulation of bacterial lipopolysaccharides (LPS; also known as endotoxin) in gluteofemoral WAT. Together with a malfunctioning complement system, this induces low-grade inflammation in the depot and raises its uncontrollable expansion. Correspondingly, more attention should be paid in future research to the endotoxemia prevalent in patients with lipedema. We would like to propose that proper management of endotoxemia can reduce the progression and even improve the state of disease in patients with lipedema.
Collapse
Affiliation(s)
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA.
| |
Collapse
|
10
|
Schartz ND, Liang HY, Carvalho K, Chu SH, Mendoza-Arvilla A, Petrisko TJ, Gomez-Arboledas A, Mortazavi A, Tenner AJ. C5aR1 antagonism suppresses inflammatory glial responses and alters cellular signaling in an Alzheimer's disease mouse model. Nat Commun 2024; 15:7028. [PMID: 39147742 PMCID: PMC11327341 DOI: 10.1038/s41467-024-51163-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 07/30/2024] [Indexed: 08/17/2024] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in older adults, and the need for effective, sustainable therapeutic targets is imperative. The complement pathway has been proposed as a therapeutic target. C5aR1 inhibition reduces plaque load, gliosis, and memory deficits in animal models, however, the cellular bases underlying this neuroprotection were unclear. Here, we show that the C5aR1 antagonist PMX205 improves outcomes in the Arctic48 mouse model of AD. A combination of single cell and single nucleus RNA-seq analysis of hippocampi derived from males and females identified neurotoxic disease-associated microglia clusters in Arctic mice that are C5aR1-dependent, while microglial genes associated with synapse organization and transmission and learning were overrepresented in PMX205-treated mice. PMX205 also reduced neurotoxic astrocyte gene expression, but clusters associated with protective responses to injury were unchanged. C5aR1 inhibition promoted mRNA-predicted signaling pathways between brain cell types associated with cell growth and repair, while suppressing inflammatory pathways. Finally, although hippocampal plaque load was unaffected, PMX205 prevented deficits in short-term memory in female Arctic mice. In conclusion, C5aR1 inhibition prevents cognitive loss, limits detrimental glial polarization while permitting neuroprotective responses, as well as leaving most protective functions of complement intact, making C5aR1 antagonism an attractive therapeutic strategy for AD.
Collapse
Affiliation(s)
- Nicole D Schartz
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Heidi Y Liang
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Klebea Carvalho
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Shu-Hui Chu
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Adrian Mendoza-Arvilla
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Tiffany J Petrisko
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Angela Gomez-Arboledas
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Ali Mortazavi
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA.
- Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, CA, USA.
| |
Collapse
|
11
|
Bitar L, Puig B, Oertner TG, Dénes Á, Magnus T. Changes in Neuroimmunological Synapses During Cerebral Ischemia. Transl Stroke Res 2024:10.1007/s12975-024-01286-1. [PMID: 39103660 DOI: 10.1007/s12975-024-01286-1] [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: 03/28/2024] [Revised: 06/24/2024] [Accepted: 07/25/2024] [Indexed: 08/07/2024]
Abstract
The direct interplay between the immune and nervous systems is now well established. Within the brain, these interactions take place between neurons and resident glial cells, i.e., microglia and astrocytes, or infiltrating immune cells, influenced by systemic factors. A special form of physical cell-cell interactions is the so-called "neuroimmunological (NI) synapse." There is compelling evidence that the same signaling pathways that regulate inflammatory responses to injury or ischemia also play potent roles in brain development, plasticity, and function. Proper synaptic wiring is as important during development as it is during disease states, as it is necessary for activity-dependent refinement of neuronal circuits. Since the process of forming synaptic connections in the brain is highly dynamic, with constant changes in strength and connectivity, the immune component is perfectly suited for the regulatory task as it is in constant turnover. Many cellular and molecular players in this interaction remain to be uncovered, especially in pathological states. In this review, we discuss and propose possible communication hubs between components of the adaptive and innate immune systems and the synaptic element in ischemic stroke pathology.
Collapse
Affiliation(s)
- Lynn Bitar
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, Hamburg, 20246, Germany
| | - Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, Hamburg, 20246, Germany
| | - Thomas G Oertner
- Institute for Synaptic Physiology, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Tim Magnus
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, Hamburg, 20246, Germany.
| |
Collapse
|
12
|
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
|
13
|
Wu F, Kong H, Xie L, Sokolova IM. Exposure to nanopollutants (nZnO) enhances the negative effects of hypoxia and delays recovery of the mussels' immune system. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 351:124112. [PMID: 38705446 DOI: 10.1016/j.envpol.2024.124112] [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: 02/06/2024] [Revised: 04/03/2024] [Accepted: 05/03/2024] [Indexed: 05/07/2024]
Abstract
Aquatic environments face escalating challenges from multiple stressors like hypoxia and nanoparticle exposure, with impact of these combined stressors on mussel immunity being poorly understood. We investigated the individual and combined effects of short-term and long-term hypoxia and exposure to zinc oxide nanoparticles (nZnO) on immune system of the mussels (Mytilus edulis). Hemocyte functional traits (mortality, adhesion capacity, phagocytosis, lysosomal abundance, and oxidative burst), and transcript levels of immune-related genes involved in pathogen recognition (the Toll-like receptors, the complement system components, and the adaptor proteins MyD88) were assessed. Short-term hypoxia minimally affected hemocyte parameters, while prolonged exposure led to immunosuppression, impacting hemocyte abundance, viability, phagocytosis, and defensin gene expression. Under normoxia, nZnO stimulated immune responses of mussel hemocytes. However, combined nZnO and hypoxia induced more pronounced and rapid immunosuppression than hypoxia alone, indicating a synergistic interaction. nZnO exposure hindered immune parameter recovery during post-hypoxic reoxygenation, suggesting persistent impact. Opposing trends were observed in pathogen-sensing and pathogen-elimination mechanisms, with a positive correlation between pathogen-recognition system activation and hemocyte mortality. These findings underscore a complex relationship and potential conflict between pathogen-recognition ability, immune function, and cell survival in mussel hemocytes under hypoxia and nanopollutant stress, and emphasize the importance of considering multiple stressors in assessing the vulnerability and adaptability of mussel immune system under complex environmental conditions of anthropogenically modified coastal ecosystems.
Collapse
Affiliation(s)
- Fangli Wu
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China; Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany; School of Environment, South China Normal University, University Town, Guangzhou, 510006, China
| | - Hui Kong
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany
| | - Lingtian Xie
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China; School of Environment, South China Normal University, University Town, Guangzhou, 510006, China
| | - Inna M Sokolova
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany; Department of Maritime Systems, Interdisciplinary Faculty, University of Rostock, Rostock, Germany.
| |
Collapse
|
14
|
Saleki K, Alijanizadeh P, Javanmehr N, Rezaei N. The role of Toll-like receptors in neuropsychiatric disorders: Immunopathology, treatment, and management. Med Res Rev 2024; 44:1267-1325. [PMID: 38226452 DOI: 10.1002/med.22012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/20/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024]
Abstract
Neuropsychiatric disorders denote a broad range of illnesses involving neurology and psychiatry. These disorders include depressive disorders, anxiety, schizophrenia, bipolar disorder, attention deficit hyperactivity disorder, autism spectrum disorders, headaches, and epilepsy. In addition to their main neuropathology that lies in the central nervous system (CNS), lately, studies have highlighted the role of immunity and neuroinflammation in neuropsychiatric disorders. Toll-like receptors (TLRs) are innate receptors that act as a bridge between the innate and adaptive immune systems via adaptor proteins (e.g., MYD88) and downstream elements; TLRs are classified into 13 families that are involved in normal function and illnesses of the CNS. TLRs expression affects the course of neuropsychiatric disorders, and is influenced during their pharmacotherapy; For example, the expression of multiple TLRs is normalized during the major depressive disorder pharmacotherapy. Here, the role of TLRs in neuroimmunology, treatment, and management of neuropsychiatric disorders is discussed. We recommend longitudinal studies to comparatively assess the cell-type-specific expression of TLRs during treatment, illness progression, and remission. Also, further research should explore molecular insights into TLRs regulation and related pathways.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
- Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Javanmehr
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
15
|
Guo J, Zhang QY, Xu L, Li M, Sun QY. Icariin ameliorates LPS-induced acute lung injury in mice via complement C5a-C5aR1 and TLR4 signaling pathways. Int Immunopharmacol 2024; 131:111802. [PMID: 38467082 DOI: 10.1016/j.intimp.2024.111802] [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: 01/06/2024] [Revised: 02/22/2024] [Accepted: 03/02/2024] [Indexed: 03/13/2024]
Abstract
Acute lung injury (ALI) is an acute respiratory-related progressive disorder, which lacks specific pharmacotherapy. Icariin (ICA) has been shown to be effective in treating ALI. However, the targets and pharmacological mechanisms underlying the effects of ICA in the treatment of ALI are relatively lacking. Based on network pharmacology and molecular docking analyses, the gene functions and potential target pathways of ICA in the treatment of ALI were determined. In addition, the underlying mechanisms of ICA were verified by immunohistochemistry, immunofluorescence, quantitative Real-time PCR, and Western blot in LPS-induced ALI mice. The biological processes targeted by ICA in the treatment of ALI included the pathological changes, inflammatory response, and cell signal transduction. Network pharmacology, molecular docking, and in vivo experimental results revealed that ICA inhibited the complement C5a-C5aR1 axis, TLR4 mediated NF-κB, MAPK, and JAK2-STAT3 signaling pathways related gene and protein expressions, and decreased inflammatory cytokine, chemokine, adhesion molecule expressions, and mitochondrial apoptosis in LPS-induced ALI.
Collapse
Affiliation(s)
- Jing Guo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China; School of Chinese Ethnic Medicine, Guizhou Minzu University, Guiyang 550025, China
| | - Qi-Yun Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Lin Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Min Li
- General Ward, Guizhou Provincial People's Hospital, Guiyang 550002, China.
| | - Qian-Yun Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China.
| |
Collapse
|
16
|
Blackman SA, Miles D, Suresh J, Calve S, Bryant SJ. Cell- and Serum-Derived Proteins Act as DAMPs to Activate RAW 264.7 Macrophage-like Cells on Silicone Implants. ACS Biomater Sci Eng 2024; 10:1418-1434. [PMID: 38319825 PMCID: PMC11316276 DOI: 10.1021/acsbiomaterials.3c01393] [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: 02/08/2024]
Abstract
Protein adsorption after biomaterial implantation is the first stage of the foreign body response (FBR). However, the source(s) of the adsorbed proteins that lead to damaged associated molecular patterns (DAMPs) and induce inflammation have not been fully elucidated. This study examined the effects of different protein sources, cell-derived (from a NIH/3T3 fibroblast cell lysate) and serum-derived (from fetal bovine serum), which were compared to implant-derived proteins (after a 30 min subcutaneous implantation in mice) on activation of RAW 264.7 cells cultured in minimal (serum-free) medium. Both cell-derived and serum-derived protein sources when preadsorbed to either tissue culture polystyrene or medical-grade silicone induced RAW 264.7 cell activation. The combination led to an even higher expression of pro-inflammatory cytokine genes and proteins. Implant-derived proteins on silicone explants induced a rapid inflammatory response that then subsided more quickly and to a greater extent than the studies with in vitro cell-derived or serum-derived protein sources. Proteomic analysis of the implant-derived proteins identified proteins that included cell-derived and serum-derived, but also other proteinaceous sources (e.g., extracellular matrix), suggesting that the latter or nonproteinaceous sources may help to temper the inflammatory response in vivo. These findings indicate that both serum-derived and cell-derived proteins adsorbed to implants can act as DAMPs to drive inflammation in the FBR, but other protein sources may play an important role in controlling inflammation.
Collapse
Affiliation(s)
- Samuel A. Blackman
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
| | - Dalton Miles
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
| | - Joshita Suresh
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, 1111 Engineering Dr, Boulder, CO 80309-0427, USA
- BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
| | - Stephanie J. Bryant
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
- BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
- Materials Science and Engineering Program, University of Colorado Boulder, 4001 Discovery Dr, Boulder, CO 80300-0613, USA
| |
Collapse
|
17
|
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
|
18
|
Djurišić M. Immune receptors and aging brain. Biosci Rep 2024; 44:BSR20222267. [PMID: 38299364 PMCID: PMC10866841 DOI: 10.1042/bsr20222267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 01/08/2024] [Accepted: 01/29/2024] [Indexed: 02/02/2024] Open
Abstract
Aging brings about a myriad of degenerative processes throughout the body. A decrease in cognitive abilities is one of the hallmark phenotypes of aging, underpinned by neuroinflammation and neurodegeneration occurring in the brain. This review focuses on the role of different immune receptors expressed in cells of the central and peripheral nervous systems. We will discuss how immune receptors in the brain act as sentinels and effectors of the age-dependent shift in ligand composition. Within this 'old-age-ligand soup,' some immune receptors contribute directly to excessive synaptic weakening from within the neuronal compartment, while others amplify the damaging inflammatory environment in the brain. Ultimately, chronic inflammation sets up a positive feedback loop that increases the impact of immune ligand-receptor interactions in the brain, leading to permanent synaptic and neuronal loss.
Collapse
Affiliation(s)
- Maja Djurišić
- Departments of Biology, Neurobiology, and Bio-X, Stanford University, Stanford, CA 94305, U.S.A
| |
Collapse
|
19
|
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
|
20
|
da Silveira BP, Barhoumi R, Bray JM, Cole-Pfeiffer HM, Mabry CJ, Burghardt RC, Cohen ND, Bordin AI. Impact of surface receptors TLR2, CR3, and FcγRIII on Rhodococcus equi phagocytosis and intracellular survival in macrophages. Infect Immun 2024; 92:e0038323. [PMID: 38018994 PMCID: PMC10790823 DOI: 10.1128/iai.00383-23] [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: 09/21/2023] [Accepted: 10/26/2023] [Indexed: 11/30/2023] Open
Abstract
The virulence-associated protein A (VapA) produced by virulent Rhodococcus equi allows it to replicate in macrophages and cause pneumonia in foals. It is unknown how VapA interacts with mammalian cell receptors, but intracellular replication of avirulent R. equi lacking vapA can be restored by supplementation with recombinant VapA (rVapA). Our objectives were to determine whether the absence of the surface receptors Toll-like receptor 2 (TLR2), complement receptor 3 (CR3), or Fc gamma receptor III (FcγRIII) impacts R. equi phagocytosis and intracellular replication in macrophages, and whether rVapA restoration of virulence in R. equi is dependent upon these receptors. Wild-type (WT) murine macrophages with TLR2, CR3, or FcγRIII blocked or knocked out (KO) were infected with virulent or avirulent R. equi, with or without rVapA supplementation. Quantitative bacterial culture and immunofluorescence imaging were performed. Phagocytosis of R. equi was not affected by blockade or KO of TLR2 or CR3. Intracellular replication of virulent R. equi was not affected by TLR2, CR3, or FcγRIII blockade or KO; however, avirulent R. equi replicated in TLR2-/- and CR3-/- macrophages but not in WT and FcγRIII-/-. rVapA supplementation did not affect avirulent R. equi phagocytosis but promoted intracellular replication in WT and all KO cells. By demonstrating that TLR2 and CR3 limit replication of avirulent but not virulent R. equi and that VapA-mediated virulence is independent of TLR2, CR3, or FcγRIII, our study provides novel insights into the role of these specific surface receptors in determining the entry and intracellular fate of R. equi.
Collapse
Affiliation(s)
- Bibiana Petri da Silveira
- Department of Large Animal Clinical Sciences, Equine Infectious Disease Laboratory, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Rola Barhoumi
- Department of Veterinary Integrative Biosciences, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Jocelyne M. Bray
- Department of Large Animal Clinical Sciences, Equine Infectious Disease Laboratory, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Hannah M. Cole-Pfeiffer
- Department of Large Animal Clinical Sciences, Equine Infectious Disease Laboratory, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Cory J. Mabry
- Department of Large Animal Clinical Sciences, Equine Infectious Disease Laboratory, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Robert C. Burghardt
- Department of Veterinary Integrative Biosciences, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Noah D. Cohen
- Department of Large Animal Clinical Sciences, Equine Infectious Disease Laboratory, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Angela I. Bordin
- Department of Large Animal Clinical Sciences, Equine Infectious Disease Laboratory, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| |
Collapse
|
21
|
Timkovich AE, Holling GA, Afzali MF, Kisiday J, Santangelo KS. TLR4 antagonism provides short-term but not long-term clinical benefit in a full-depth cartilage defect mouse model. Connect Tissue Res 2024; 65:26-40. [PMID: 37898909 PMCID: PMC11271750 DOI: 10.1080/03008207.2023.2269257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 08/23/2023] [Accepted: 10/05/2023] [Indexed: 10/31/2023]
Abstract
PURPOSE/AIM Cartilage injury and subsequent osteoarthritis (OA) are debilitating conditions affecting millions worldwide. As there are no cures for these ailments, novel therapies are needed to suppress disease pathogenesis. Given that joint injuries are known to produce damage-associated molecular patterns (DAMPs), our central premise is that the Toll-like receptor 4 (TLR4) pathway is a principal driver in the early response to cartilage damage and subsequent pathology. We postulate that TLR4 activation is initiated/perpetuated by DAMPs released following joint damage. Thus, antagonism of the TLR4 pathway immediately after injury may suppress the development of joint surface defects. MATERIALS AND METHODS Two groups were utilized: (1) 8-week-old, male C57BL6 mice treated systemically with a known TLR4 antagonist and (2) mice injected with vehicle control. A full-depth cartilage lesion on the midline of the patellofemoral groove was created in the right knee of each mouse. The left knee was used as a sham surgery control. Gait changes were evaluated over 4 weeks using a quantitative gait analysis system. At harvest, knee joints were processed for pathologic assessment, Nanostring® transcript expression, and immunohistochemistry (IHC). RESULTS Short-term treatment with a TLR4 antagonist at 14-days significantly improved relevant gait parameters; improved cartilage metrics and modified Mankin scores were also seen. Additionally, mRNA expression and IHC showed reduced expression of inflammatory mediators in animals treated with the TLR4 antagonist. CONCLUSIONS Collectively, this work demonstrates that systemic treatment with a TLR4 antagonist is protective to further cartilage damage 14-days post-injury in a murine model of induced disease.
Collapse
Affiliation(s)
- Ariel E. Timkovich
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - G. Aaron Holling
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Maryam F. Afzali
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - John Kisiday
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Kelly S. Santangelo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
22
|
Hissen KL, He W, Wu G, Criscitiello MF. Immunonutrition: facilitating mucosal immune response in teleost intestine with amino acids through oxidant-antioxidant balance. Front Immunol 2023; 14:1241615. [PMID: 37841275 PMCID: PMC10570457 DOI: 10.3389/fimmu.2023.1241615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/24/2023] [Indexed: 10/17/2023] Open
Abstract
Comparative animal models generate fundamental scientific knowledge of immune responses. However, these studies typically are conducted in mammals because of their biochemical and physiological similarity to humans. Presently, there has been an interest in using teleost fish models to study intestinal immunology, particularly intestinal mucosa immune response. Instead of targeting the pathogen itself, a preferred approach for managing fish health is through nutrient supplementation, as it is noninvasive and less labor intensive than vaccine administrations while still modulating immune properties. Amino acids (AAs) regulate metabolic processes, oxidant-antioxidant balance, and physiological requirements to improve immune response. Thus, nutritionists can develop sustainable aquafeeds through AA supplementation to promote specific immune responses, including the intestinal mucosa immune system. We propose the use of dietary supplementation with functional AAs to improve immune response by discussing teleost fish immunology within the intestine and explore how oxidative burst is used as an immune defense mechanism. We evaluate immune components and immune responses in the intestine that use oxidant-antioxidant balance through potential selection of AAs and their metabolites to improve mucosal immune capacity and gut integrity. AAs are effective modulators of teleost gut immunity through oxidant-antioxidant balance. To incorporate nutrition as an immunoregulatory means in teleost, we must obtain more tools including genomic, proteomic, nutrition, immunology, and macrobiotic and metabonomic analyses, so that future studies can provide a more holistic understanding of the mucosal immune system in fish.
Collapse
Affiliation(s)
- Karina L. Hissen
- Comparative Immunogenetics Laboratory Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Wenliang He
- Amino Acid Laboratory, Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Guoyao Wu
- Amino Acid Laboratory, Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Michael F. Criscitiello
- Comparative Immunogenetics Laboratory Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
23
|
Jayaprakash Demirel K, Wu R, Neves Guimaraes A, Demirel I. The role of NLRP3 in regulating gingival epithelial cell responses evoked by Aggregatibacter actinomycetemcomitans. Cytokine 2023; 169:156316. [PMID: 37541072 DOI: 10.1016/j.cyto.2023.156316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/18/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) has myriads of virulence factors among which leukotoxin provides A. actinomycetemcomitans with the advantage to thrive in the surrounding hostile environment and evade host immune defences. The NLRP3 inflammasome has been associated with periodontal disease development. However, our understanding of the involvement of caspase-1, caspase-4, and NLRP3 in the release of IL-1β and other inflammatory mediators from gingival epithelial cells during a A. actinomycetemcomitans infection is limited. The aim of this study was to investigate how the inflammasome-associated proteins caspase-1, caspase-4 and NLRP3 regulate the immune response of gingival epithelial cells during a A. actinomycetemcomitans infection. Human gingival epithelial cells (Ca9-22) deficient in NLRP3, caspase-1 or caspase-4 were created using CRISPR/Cas9. Gingival epithelial cells were stimulated with the A. actinomycetemcomitans low-leukotoxic strain NCTC9710 or the highly leukotoxic JP2 strain HK 165 for 6, 12 and 24 h. The results showed that the JP2 strain HK1651 induced higher IL-1β and IL-1RA release and mediated more epithelial cell death compared to the NCTC9710 strain. These findings were found to be capsase-1, caspase-4 and NLRP3-dependant. A targeted protein analysis of inflammation-related proteins showed that the expression of 37 proteins were identified as being significantly altered after HK1651 infection compared to unstimulated Cas9 and NLRP3-deficient cells. Of the 37 proteins, 23 of these inflammation-related proteins released by NLRP3-deficient cells differed significantly compared to Cas9 cells after infection. This suggests that NLRP3 has a broad effect on the inflammatory response in gingival epithelial cells.
Collapse
Affiliation(s)
- Kartheyaene Jayaprakash Demirel
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine and Health, Örebro University, Örebro, Sweden; Department of Odontological Research, Public Dental Service, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
| | - Rongrong Wu
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Alessandra Neves Guimaraes
- Department of Odontological Research, Public Dental Service, Faculty of Medicine and Health, Örebro University, Örebro, Sweden; Department of Periodontology and Implantology, Public Dental Service, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Isak Demirel
- School of Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
24
|
Schartz ND, Liang HY, Carvalho K, Chu SH, Mendoza-Arvilla A, Petrisko TJ, Gomez-Arboledas A, Mortazavi A, Tenner AJ. C5aR1 antagonism suppresses inflammatory glial gene expression and alters cellular signaling in an aggressive Alzheimer's model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.22.554306. [PMID: 37662399 PMCID: PMC10473603 DOI: 10.1101/2023.08.22.554306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in older adults, and the need for effective, sustainable therapeutic targets is imperative. Pharmacologic inhibition of C5aR1 reduces plaque load, gliosis and memory deficits in animal models. However, the cellular basis underlying this neuroprotection and which processes were the consequence of amyloid reduction vs alteration of the response to amyloid were unclear. In the Arctic model, the C5aR1 antagonist PMX205 did not reduce plaque load, but deficits in short-term memory in female mice were prevented. Hippocampal single cell and single nucleus RNA-seq clusters revealed C5aR1 dependent and independent gene expression and cell-cell communication. Microglial clusters containing neurotoxic disease-associated microglial genes were robustly upregulated in Arctic mice and drastically reduced with PMX205 treatment, while genes in microglia clusters that were overrepresented in the Arctic-PMX205 vs Arctic group were associated with synapse organization and transmission and learning. PMX205 treatment also reduced some A-1 astrocyte genes. In spite of changes in transcript levels, overall protein levels of some reactive glial markers were relatively unchanged by C5aR1 antagonism, as were clusters associated with protective responses to injury. C5aR1 inhibition promoted signaling pathways associated with cell growth and repair, such as TGFβ and FGF, in Arctic mice, while suppressing inflammatory pathways including PROS, Pecam1, and EPHA. In conclusion, pharmacologic C5aR1 inhibition prevents cognitive loss, limits microglial polarization to a detrimental inflammatory state and permits neuroprotective responses, as well as leaving protective functions of complement intact, making C5aR1 antagonism an attractive therapeutic strategy for individuals with AD.
Collapse
Affiliation(s)
- Nicole D. Schartz
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
| | - Heidi Y. Liang
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA 92697
| | - Klebea Carvalho
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA 92697
| | - Shu-Hui Chu
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
| | - Adrian Mendoza-Arvilla
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
| | - Tiffany J. Petrisko
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
| | - Angela Gomez-Arboledas
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
| | - Ali Mortazavi
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA 92697
| | - Andrea J. Tenner
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697
- Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, CA 92697
| |
Collapse
|
25
|
Gould S, Templin MV. Off target toxicities and links with physicochemical properties of medicinal products, including antibiotics, oligonucleotides, lipid nanoparticles (with cationic and/or anionic charges). Data review suggests an emerging pattern. Toxicol Lett 2023; 384:14-29. [PMID: 37454775 DOI: 10.1016/j.toxlet.2023.07.011] [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: 01/10/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Toxicology is an essential part of any drug development plan. Circumnavigating the risk of failure because of a toxicity issue can be a challenge, and failure in late development is extremely costly. To identify potential risks, it requires more than just understanding the biological target. The toxicologist needs to consider a compound's structure, it's physicochemical properties (including the impact of the overall formulation), as well as the biological target (e.g., receptor interactions). Understanding the impact of the physicochemical properties can be used to predict potential toxicities in advance by incorporating key endpoints in early screening strategies and/or used to compare toxicity profiles across lead candidates. This review discussed the risks of off-target and/or non-specific toxicities that may be associated with the physicochemical properties of compounds, especially those carrying dominant positive or negative charges, including amphiphilic small molecules, peptides, oligonucleotides and lipids/liposomes/lipid nanoparticles. The latter of which are being seen more and more in drug development, including the recent Covid pandemic, where mRNA and lipid nanoparticle technology is playing more of a role in vaccine development. The translation between non-clinical and clinical data is also considered, questioning how a physicochemical driven toxicity may be more universal across species, which means that such toxicity may be reassuringly translatable between species and as such, this information may also be considered as a support to the 3 R's, particularly in the early screening stages of a drug development plan.
Collapse
|
26
|
Maisat W, Han X, Koutsogiannaki S, Soriano SG, Yuki K. Differential effects of dexmedetomidine on Gram-positive and Gram-negative bacterial killing and phagocytosis. Int Immunopharmacol 2023; 120:110327. [PMID: 37201408 PMCID: PMC10330683 DOI: 10.1016/j.intimp.2023.110327] [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: 03/07/2023] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
Dexmedetomidine is a commonly used sedative in perioperative and intensive care settings with purported immunomodulatory properties. Since its effects on immune functions against infections have not been extensively studied, we tested the effects of dexmedetomidine on Gram-positive [Staphylococcus aureus and Enterococcus faecalis] and Gram-negative bacteria [Escherichia coli], and on effector functions of human monocytes THP-1 cells against them. We evaluated phagocytosis, reactive oxygen species (ROS) formation, and CD11b activation, and performed RNA sequencing analyses. Our study revealed that dexmedetomidine improved Gram-positive but mitigated Gram-negative bacterial phagocytosis and killing in THP-1 cells. The attenuation of Toll-like receptor 4 (TLR4) signaling by dexmedetomidine was previously reported. Thus, we tested TLR4 inhibitor TAK242. Similar to dexmedetomidine, TAK242 reduced E. coli phagocytosis but enhanced CD11b activation. The reduced TLR4 response potentially increases CD11b activation and ROS generation and subsequently enhances Gram-positive bacterial killing. Conversely, dexmedetomidine may inhibit the TLR4-signaling pathway and mitigate the alternative phagocytosis pathway induced by TLR4 activation through LPS-mediated Gram-negative bacteria, resulting in worsened bacterial loads. We also examined another α2 adrenergic agonist, xylazine. Because xylazine did not affect bacterial clearance, we proposed that dexmedetomidine may have an off-target effect on bacterial killing process, potentially involving crosstalk between CD11b and TLR4. Despite its potential to attenuate inflammation, we provide a novel insight into potential risks of dexmedetomidine use during Gram-negative infections, highlighting the differential effect of dexmedetomidine on Gram-positive and Gram-negative bacteria.
Collapse
Affiliation(s)
- Wiriya Maisat
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA; Department of Anesthesiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Xiaohui Han
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA
| | - Sophia Koutsogiannaki
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA; Department of Immunology, Harvard Medical School, Boston, USA
| | - Sulpicio G Soriano
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA
| | - Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA; Department of Immunology, Harvard Medical School, Boston, USA.
| |
Collapse
|
27
|
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
|
28
|
Bohlson SS, Tenner AJ. Complement in the Brain: Contributions to Neuroprotection, Neuronal Plasticity, and Neuroinflammation. Annu Rev Immunol 2023; 41:431-452. [PMID: 36750318 DOI: 10.1146/annurev-immunol-101921-035639] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The complement system is an ancient collection of proteolytic cascades with well-described roles in regulation of innate and adaptive immunity. With the convergence of a revolution in complement-directed clinical therapeutics, the discovery of specific complement-associated targetable pathways in the central nervous system, and the development of integrated multi-omic technologies that have all emerged over the last 15 years, precision therapeutic targeting in Alzheimer disease and other neurodegenerative diseases and processes appears to be within reach. As a sensor of tissue distress, the complement system protects the brain from microbial challenge as well as the accumulation of dead and/or damaged molecules and cells. Additional more recently discovered diverse functions of complement make it of paramount importance to design complement-directed neurotherapeutics such that the beneficial roles in neurodevelopment, adult neural plasticity, and neuroprotective functions of the complement system are retained.
Collapse
Affiliation(s)
- Suzanne S Bohlson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA; ,
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA; ,
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, California, USA
| |
Collapse
|
29
|
Kubelkova K, Bostik V, Joshi L, Macela A. Innate Immune Recognition, Integrated Stress Response, Infection, and Tumorigenesis. BIOLOGY 2023; 12:biology12040499. [PMID: 37106700 PMCID: PMC10135864 DOI: 10.3390/biology12040499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Engagement of PRRs in recognition of PAMPs or DAMPs is one of the processes that initiates cellular stress. These sensors are involved in signaling pathways leading to induction of innate immune processes. Signaling initiated by PRRs is associated with the activation of MyD88-dependent signaling pathways and myddosome formation. MyD88 downstream signaling depends upon the context of signaling initiation, the cell (sub)type and the microenvironment of signal initiation. Recognition of PAMPs or DAMPs through PRRs activates the cellular autonomous defence mechanism, which orchestrates the cell responses to resolve specific insults at the single cell level. In general, stressed endoplasmic reticulum is directly linked with the induction of autophagy and initiation of mitochondrial stress. These processes are regulated by the release of Ca2+ from ER stores accepted by mitochondria, which respond through membrane depolarization and the production of reactive oxygen species generating signals leading to inflammasome activation. In parallel, signaling from PRRs initiates the accumulation of misfolded or inappropriately post-translationally modified proteins in the ER and triggers a group of conserved emergency rescue pathways known as unfolded protein response. The cell-autonomous effector mechanisms have evolutionarily ancient roots and were gradually specialized for the defence of specific cell (sub)types. All of these processes are common to the innate immune recognition of microbial pathogens and tumorigenesis as well. PRRs are active in both cases. Downstream are activated signaling pathways initiated by myddosomes, translated by the cellular autonomous defence mechanism, and finalized by inflammasomes.
Collapse
Affiliation(s)
- Klara Kubelkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
- Correspondence:
| | - Vanda Bostik
- Department of Epidemiology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Lokesh Joshi
- Glycoscience Group, National Centre for Biomedical Engineering Science, University of Galway, H91 W2TY Galway, Ireland
| | - Ales Macela
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| |
Collapse
|
30
|
Xu Z, Hou XF, Feng CM, Zheng L, Xu DX, Zhao H, Fu L. The association between serum complement C3a and severity in patients with community-acquired pneumonia. Front Immunol 2023; 14:1034233. [PMID: 36776834 PMCID: PMC9911530 DOI: 10.3389/fimmu.2023.1034233] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Background A few studies found that the complement system may be involved in the onset and progression of community-acquired pneumonia (CAP). However, the role of the complement system in CAP was obscure. The goal of this study was to analyze the association of serum complement C3a with CAP severity scores based on a cross-sectional study. Methods All 190 CAP patients and 95 control subjects were enrolled. Demographic information and clinical data were extracted. Peripheral blood samples were collected on admission. Results Serum complement C3a on admission was elevated in CAP patients compared with healthy subjects. The level of complement C3a was gradually elevated in parallel with CAP severity scores (CURB-65, CRB-65, PSI, SMART-COP, and CURXO). Complement C3a was positively correlated with blood routine parameters, renal function markers, and inflammatory cytokines in CAP patients. Furthermore, multivariate linear and logistic regression models found that serum complement C3a on admission was positively associated with CAP severity scores. Mechanistic research suggested that complement system inhibition alleviated Streptococcus pneumoniae-induced upregulation of IL-1β, TNF-α, IL-6, and CRP in MLE-12 cells. Conclusions Serum complement C3a on admission is positively associated with the severity of CAP patients. Inhibiting complement system attenuates S. pneumoniae-elevated secretion of inflammatory cytokines in pulmonary epithelial cells, indicating that complement C3a is involved in the pathophysiology of CAP. Serum complement C3a may serve as an earlier diagnostic biomarker for CAP.
Collapse
Affiliation(s)
- Zheng Xu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,Department of Respiratory and Critical Care Medicine, The Sixth People’s Hospital of Fuyang, Anhui, China
| | - Xue-Feng Hou
- School of Pharmacy, Drug Research and Development Center, Wannan Medical College, Wuhu, Anhui, China
| | - Chun-Mei Feng
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ling Zheng
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, Anhui, China,*Correspondence: Lin Fu, ; Hui Zhao, ; De-Xiang Xu,
| | - Hui Zhao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,*Correspondence: Lin Fu, ; Hui Zhao, ; De-Xiang Xu,
| | - Lin Fu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,Department of Toxicology, Anhui Medical University, Hefei, Anhui, China,*Correspondence: Lin Fu, ; Hui Zhao, ; De-Xiang Xu,
| |
Collapse
|
31
|
van Erp IAM, Michailidou I, van Essen TA, van der Jagt M, Moojen W, Peul WC, Baas F, Fluiter K. Tackling Neuroinflammation After Traumatic Brain Injury: Complement Inhibition as a Therapy for Secondary Injury. Neurotherapeutics 2023; 20:284-303. [PMID: 36222978 PMCID: PMC10119357 DOI: 10.1007/s13311-022-01306-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2022] [Indexed: 11/30/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality, sensorimotor morbidity, and neurocognitive disability. Neuroinflammation is one of the key drivers causing secondary brain injury after TBI. Therefore, attenuation of the inflammatory response is a potential therapeutic goal. This review summarizes the most important neuroinflammatory pathophysiology resulting from TBI and the clinical trials performed to attenuate neuroinflammation. Studies show that non-selective attenuation of the inflammatory response, in the early phase after TBI, might be detrimental and that there is a gap in the literature regarding pharmacological trials targeting specific pathways. The complement system and its crosstalk with the coagulation system play an important role in the pathophysiology of secondary brain injury after TBI. Therefore, regaining control over the complement cascades by inhibiting overshooting activation might constitute useful therapy. Activation of the complement cascade is an early component of neuroinflammation, making it a potential target to mitigate neuroinflammation in TBI. Therefore, we have described pathophysiological aspects of complement inhibition and summarized animal studies targeting the complement system in TBI. We also present the first clinical trial aimed at inhibition of complement activation in the early days after brain injury to reduce the risk of morbidity and mortality following severe TBI.
Collapse
Affiliation(s)
- Inge A M van Erp
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and HaGa Hospital, Leiden and The Hague, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, The Netherlands.
| | - Iliana Michailidou
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas A van Essen
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and HaGa Hospital, Leiden and The Hague, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, The Netherlands
| | - Mathieu van der Jagt
- Department of Intensive Care Adults, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Wouter Moojen
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and HaGa Hospital, Leiden and The Hague, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, The Netherlands
| | - Wilco C Peul
- University Neurosurgical Center Holland, Leiden University Medical Center, Haaglanden Medical Center and HaGa Hospital, Leiden and The Hague, Albinusdreef 2, J-11-R-83, 2333 ZA, Leiden, The Netherlands
| | - Frank Baas
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Kees Fluiter
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
32
|
Wang H, Ideguchi H, Kajikawa T, Mastellos DC, Lambris JD, Hajishengallis G. Complement Is Required for Microbe-Driven Induction of Th17 and Periodontitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1370-1378. [PMID: 36028293 PMCID: PMC9530003 DOI: 10.4049/jimmunol.2200338] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/27/2022] [Indexed: 12/29/2022]
Abstract
In both mice and humans, complement and Th17 cells have been implicated in periodontitis, an oral microbiota-driven inflammatory disease associated with systemic disorders. A recent clinical trial showed that a complement C3 inhibitor (AMY-101) causes sustainable resolution of periodontal inflammation, the main effector of tissue destruction in this oral disease. Although both complement and Th17 are required for periodontitis, it is uncertain how these immune components cooperate in disease development. In this study, we dissected the complement-Th17 relationship in the setting of ligature-induced periodontitis (LIP), a model that previously established that microbial dysbiosis drives Th17 cell expansion and periodontal bone loss. Complement was readily activated in the periodontal tissue of LIP-subjected mice but not when the mice were placed on broad-spectrum antibiotics. Microbiota-induced complement activation generated critical cytokines, IL-6 and IL-23, which are required for Th17 cell expansion. These cytokines as well as Th17 accumulation and IL-17 expression were significantly suppressed in LIP-subjected C3-deficient mice relative to wild-type controls. As IL-23 has been extensively studied in periodontitis, we focused on IL-6 and showed that LIP-induced IL-17 and bone loss required intact IL-6 receptor signaling in the periodontium. LIP-induced IL-6 was predominantly produced by gingival epithelial cells that upregulated C3a receptor upon LIP challenge. Experiments in human gingival epithelial cells showed that C3a upregulated IL-6 production in cooperation with microbial stimuli that upregulated C3a receptor expression in ERK1/2- and JNK-dependent manner. In conclusion, complement links the periodontal microbiota challenge to Th17 cell accumulation and thus integrates complement- and Th17-driven immunopathology in periodontitis.
Collapse
Affiliation(s)
- Hui Wang
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA
| | - Hidetaka Ideguchi
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Pathophysiology-Periodontal Science, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tetsuhiro Kajikawa
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Dimitrios C Mastellos
- Division of Biodiagnostic Science and Technologies, The Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research "Demokritos," Athens, Greece; and
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA;
| |
Collapse
|
33
|
Krieg C, Weber LM, Fosso B, Marzano M, Hardiman G, Olcina MM, Domingo E, El Aidy S, Mallah K, Robinson MD, Guglietta S. Complement downregulation promotes an inflammatory signature that renders colorectal cancer susceptible to immunotherapy. J Immunother Cancer 2022; 10:e004717. [PMID: 36137652 PMCID: PMC9511657 DOI: 10.1136/jitc-2022-004717] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND AND AIMS The role of inflammatory immune responses in colorectal cancer (CRC) development and response to therapy is a matter of intense debate. While inflammation is a known driver of CRC, inflammatory immune infiltrates are a positive prognostic factor in CRC and predispose to response to immune checkpoint blockade (ICB) therapy. Unfortunately, over 85% of CRC cases are primarily unresponsive to ICB due to the absence of an immune infiltrate, and even the cases that show an initial immune infiltration can become refractory to ICB. The identification of therapy supportive immune responses in the field has been partially hindered by the sparsity of suitable mouse models to recapitulate the human disease. In this study, we aimed to understand how the dysregulation of the complement anaphylatoxin C3a receptor (C3aR), observed in subsets of patients with CRC, affects the immune responses, the development of CRC, and response to ICB therapy. METHODS We use a comprehensive approach encompassing analysis of publicly available human CRC datasets, inflammation-driven and newly generated spontaneous mouse models of CRC, and multiplatform high-dimensional analysis of immune responses using microbiota sequencing, RNA sequencing, and mass cytometry. RESULTS We found that patients' regulation of the complement C3aR is associated with epigenetic modifications. Specifically, downregulation of C3ar1 in human CRC promotes a tumor microenvironment characterized by the accumulation of innate and adaptive immune cells that support antitumor immunity. In addition, in vivo studies in our newly generated mouse model revealed that the lack of C3a in the colon activates a microbiota-mediated proinflammatory program which promotes the development of tumors with an immune signature that renders them responsive to the ICB therapy. CONCLUSIONS Our findings reveal that C3aR may act as a previously unrecognized checkpoint to enhance antitumor immunity in CRC. C3aR can thus be exploited to overcome ICB resistance in a larger group of patients with CRC.
Collapse
Affiliation(s)
- Carsten Krieg
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center Charleston, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lukas M Weber
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Bruno Fosso
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Bari, Italy
| | - Marinella Marzano
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Bari, Italy
| | - Gary Hardiman
- School of Biological Sciences and Institute for Global Food Security, Queens University of Belfast, Belfast, UK
| | - Monica M Olcina
- Institute of Radiation Oncology, Medical Research Council Oxford Institute for Radiation Oncology, Oxford, UK
| | - Enric Domingo
- Institute of Radiation Oncology, Medical Research Council Oxford Institute for Radiation Oncology, Oxford, UK
| | - Sahar El Aidy
- Host-microbe Metabolic Interactions, Microbiology, University of Groningen, Groningen, The Netherlands
| | - Khalil Mallah
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Mark D Robinson
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Silvia Guglietta
- Hollings Cancer Center Charleston, Medical University of South Carolina, Charleston, South Carolina, USA
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
34
|
Hensler E, Petros H, Gray CC, Chung CS, Ayala A, Fallon EA. The Neonatal Innate Immune Response to Sepsis: Checkpoint Proteins as Novel Mediators of This Response and as Possible Therapeutic/Diagnostic Levers. Front Immunol 2022; 13:940930. [PMID: 35860251 PMCID: PMC9289477 DOI: 10.3389/fimmu.2022.940930] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/07/2022] [Indexed: 11/23/2022] Open
Abstract
Sepsis, a dysfunctional immune response to infection leading to life-threatening organ injury, represents a significant global health issue. Neonatal sepsis is disproportionately prevalent and has a cost burden of 2-3 times that of adult patients. Despite this, no widely accepted definition for neonatal sepsis or recommendations for management exist and those created for pediatric patients are significantly limited in their applicability to this unique population. This is in part due to neonates' reliance on an innate immune response (which is developmentally more prominent in the neonate than the immature adaptive immune response) carried out by dysfunctional immune cells, including neutrophils, antigen-presenting cells such as macrophages/monocytes, dendritic cells, etc., natural killer cells, and innate lymphoid regulatory cell sub-sets like iNKT cells, γδ T-cells, etc. Immune checkpoint inhibitors are a family of proteins with primarily suppressive/inhibitory effects on immune and tumor cells and allow for the maintenance of self-tolerance. During sepsis, these proteins are often upregulated and are thought to contribute to the long-term immunosuppression seen in adult patients. Several drugs targeting checkpoint inhibitors, including PD-1 and PD-L1, have been developed and approved for the treatment of various cancers, but no such therapeutics have been approved for the management of sepsis. In this review, we will comparatively discuss the role of several checkpoint inhibitor proteins, including PD-1, PD-L1, VISTA, and HVEM, in the immune response to sepsis in both adults and neonates, as well as posit how they may uniquely propagate their actions through the neonatal innate immune response. We will also consider the possibility of leveraging these proteins in the clinical setting as potential therapeutics/diagnostics that might aid in mitigating neonatal septic morbidity/mortality.
Collapse
Affiliation(s)
- Emily Hensler
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Providence, RI, United States,Graduate Program in Biotechnology, Brown University, Providence, RI, United States
| | - Habesha Petros
- Graduate Program in Biotechnology, Brown University, Providence, RI, United States
| | - Chyna C. Gray
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Providence, RI, United States,Graduate Program in Biotechnology, Brown University, Providence, RI, United States
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Providence, RI, United States,Graduate Program in Biotechnology, Brown University, Providence, RI, United States
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Providence, RI, United States,Graduate Program in Biotechnology, Brown University, Providence, RI, United States,*Correspondence: Alfred Ayala,
| | - Eleanor A. Fallon
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Providence, RI, United States,Graduate Program in Biotechnology, Brown University, Providence, RI, United States
| |
Collapse
|
35
|
Mendell JR, Connolly AM, Lehman KJ, Griffin DA, Khan SZ, Dharia SD, Quintana-Gallardo L, Rodino-Klapac LR. Testing preexisting antibodies prior to AAV gene transfer therapy: rationale, lessons and future considerations. Mol Ther Methods Clin Dev 2022; 25:74-83. [PMID: 35356756 PMCID: PMC8933338 DOI: 10.1016/j.omtm.2022.02.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Given the increasing number of gene transfer therapy studies either completed or underway, there is growing attention to the importance of preexisting adaptive immunity to the viral vectors used. The recombinant viral vectors developed for gene transfer therapy share structural features with naturally occurring wild-type virus. Antibodies generated against viral vectors obtained through a previous exposure to wild-type virus can potentially compromise transgene expression by blocking transduction, thereby limiting the therapeutic efficacy of the gene transfer therapy; they may also pose potential safety concerns. Therefore, systemic gene transfer delivery requires testing patients for preexisting antibodies. Two different assays have been used: (1) binding assays that focus on total antibodies (both neutralizing and non-neutralizing) and (2) neutralizing assays that detect neutralizing antibodies. In this review we focus on adeno-associated virus-based gene therapies, describing the immune response that occurs to naturally occurring adeno-associated viruses, the implications for patients with this exposure, the assays used to detect preexisting immune responses, and strategies to circumvent preexisting adaptive immunity to expand the patient base that could benefit from such therapies.
Collapse
Affiliation(s)
- Jerry R. Mendell
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics and Neurology, The Ohio State University, Columbus, OH 43205, USA
| | - Anne M. Connolly
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics and Neurology, The Ohio State University, Columbus, OH 43205, USA
| | - Kelly J. Lehman
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | | | - Sohrab Z. Khan
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | - Sachi D. Dharia
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | | | - Louise R. Rodino-Klapac
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics and Neurology, The Ohio State University, Columbus, OH 43205, USA
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| |
Collapse
|
36
|
Alshareef M, Mallah K, Vasas T, Alawieh A, Borucki D, Couch C, Cutrone J, Shope C, Eskandari R, Tomlinson S. A Role of Complement in the Pathogenic Sequelae of Mouse Neonatal Germinal Matrix Hemorrhage. Int J Mol Sci 2022; 23:2943. [PMID: 35328364 PMCID: PMC8954718 DOI: 10.3390/ijms23062943] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/02/2022] [Accepted: 03/06/2022] [Indexed: 11/29/2022] Open
Abstract
Germinal matrix hemorrhage (GMH) is a devastating disease of infancy that results in intraventricular hemorrhage, post-hemorrhagic hydrocephalus (PHH), periventricular leukomalacia, and neurocognitive deficits. There are no curative treatments and limited surgical options. We developed and characterized a mouse model of GMH based on the injection of collagenase into the subventricular zone of post-natal pups and utilized the model to investigate the role of complement in PHH development. The site-targeted complement inhibitor CR2Crry, which binds deposited C3 complement activation products, localized specifically in the brain following its systemic administration after GMH. Compared to vehicle, CR2Crry treatment reduced PHH and lesion size, which was accompanied by decreased perilesional complement deposition, decreased astrocytosis and microgliosis, and the preservation of dendritic and neuronal density. Complement inhibition also improved survival and weight gain, and it improved motor performance and cognitive outcomes measured in adolescence. The progression to PHH, neuronal loss, and associated behavioral deficits was linked to the microglial phagocytosis of complement opsonized neurons, which was reversed with CR2Crry treatment. Thus, complement plays an important role in the pathological sequelae of GMH, and complement inhibition represents a novel therapeutic approach to reduce the disease progression of a condition for which there is currently no treatment outside of surgical intervention.
Collapse
Affiliation(s)
- Mohammed Alshareef
- Department of Neurological Surgery, Medical University of South Carolina, 301 CSB, Charleston, SC 29425, USA;
| | - Khalil Mallah
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425, USA; (K.M.); (C.C.)
| | - Tyler Vasas
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; (T.V.); (D.B.); (J.C.); (C.S.)
| | - Ali Alawieh
- Department of Neurological Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Davis Borucki
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; (T.V.); (D.B.); (J.C.); (C.S.)
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Christine Couch
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425, USA; (K.M.); (C.C.)
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jonathan Cutrone
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; (T.V.); (D.B.); (J.C.); (C.S.)
| | - Chelsea Shope
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; (T.V.); (D.B.); (J.C.); (C.S.)
| | - Ramin Eskandari
- Department of Neurological Surgery, Medical University of South Carolina, 301 CSB, Charleston, SC 29425, USA;
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425, USA; (K.M.); (C.C.)
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB 204, MSC 504, Charleston, SC 29425, USA; (K.M.); (C.C.)
- Ralph Johnson VA Medical Center, Charleston, SC 29401, USA
| |
Collapse
|
37
|
Hayatdavoudi P, Hosseini M, Hajali V, Hosseini A, Rajabian A. The role of astrocytes in epileptic disorders. Physiol Rep 2022; 10:e15239. [PMID: 35343625 PMCID: PMC8958496 DOI: 10.14814/phy2.15239] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/27/2022] [Accepted: 03/09/2022] [Indexed: 04/17/2023] Open
Abstract
Epilepsy affects about 1% of the population and approximately 30% of epileptic patients are resistant to current antiepileptic drugs. As a hallmark in epileptic tissue, many of the epileptic patients show changes in glia morphology and function. There are characteristic changes in different types of glia in different epilepsy models. Some of these changes such as astrogliosis are enough to provoke epileptic seizures. Astrogliosis is well known in mesial temporal lobe epilepsy (MTLE), the most common form of refractory epilepsy. A better understanding of astrocytes alterations could lead to novel and efficient pharmacological approaches for epilepsy. In this review, we present the alterations of astrocyte morphology and function and present some instances of targeting astrocytes in seizure and epilepsy.
Collapse
Affiliation(s)
- Parichehr Hayatdavoudi
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
- Department of PhysiologyFaculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research CenterMashhad University of Medical SciencesMashhadIran
| | - Vahid Hajali
- Department of NeuroscienceFaculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Azar Hosseini
- Pharmacological Research Center of Medicinal PlantsMashhad University of Medical SciencesMashhadIran
- Department of PharmacologyFaculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Arezoo Rajabian
- Department of Internal MedicineFaculty of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
38
|
Abstract
A favorable outcome of the COVID-19 crisis might be achieved with massive vaccination. The proposed vaccines contain several different vaccine active principles (VAP), such as inactivated virus, antigen, mRNA, and DNA, which are associated with either standard adjuvants or nanomaterials (NM) such as liposomes in Moderna's and BioNTech/Pfizer's vaccines. COVID-19 vaccine adjuvants may be chosen among liposomes or other types of NM composed for example of graphene oxide, carbon nanotubes, micelles, exosomes, membrane vesicles, polymers, or metallic NM, taking inspiration from cancer nano-vaccines, whose adjuvants may share some of their properties with those of viral vaccines. The mechanisms of action of nano-adjuvants are based on the facilitation by NM of targeting certain regions of immune interest such as the mucus, lymph nodes, and zones of infection or blood irrigation, the possible modulation of the type of attachment of the VAP to NM, in particular VAP positioning on the NM external surface to favor VAP presentation to antigen presenting cells (APC) or VAP encapsulation within NM to prevent VAP degradation, and the possibility to adjust the nature of the immune response by tuning the physico-chemical properties of NM such as their size, surface charge, or composition. The use of NM as adjuvants or the presence of nano-dimensions in COVID-19 vaccines does not only have the potential to improve the vaccine benefit/risk ratio, but also to reduce the dose of vaccine necessary to reach full efficacy. It could therefore ease the overall spread of COVID-19 vaccines within a sufficiently large portion of the world population to exit the current crisis.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, 75005 Paris, France. .,Nanobacterie SARL, 36 Boulevard Flandrin, 75116, Paris, France.,Institute of Anatomy, UZH University of Zurich, Instiute of Anatomy, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|
39
|
Neves GWP, Wong SSW, Aimanianda V, Simenel C, Guijarro JI, Walls C, Willment JA, Gow NAR, Munro CA, Brown GD, Lopes-Bezerra LM. Complement-Mediated Differential Immune Response of Human Macrophages to Sporothrix Species Through Interaction With Their Cell Wall Peptidorhamnomannans. Front Immunol 2021; 12:749074. [PMID: 34867977 PMCID: PMC8636148 DOI: 10.3389/fimmu.2021.749074] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/25/2021] [Indexed: 12/23/2022] Open
Abstract
In this study, the human immune response mechanisms against Sporothrix brasiliensis and Sporothrix schenckii, two causative agents of human and animal sporotrichosis, were investigated. The interaction of S. brasiliensis and S. schenckii with human monocyte-derived macrophages (hMDMs) was shown to be dependent on the thermolabile serum complement protein C3, which facilitated the phagocytosis of Sporothrix yeast cells through opsonization. The peptidorhamnomannan (PRM) component of the cell walls of these two Sporothrix yeasts was found to be one of their surfaces exposed pathogen-associated molecular pattern (PAMP), leading to activation of the complement system and deposition of C3b on the Sporothrix yeast surfaces. PRM also showed direct interaction with CD11b, the specific component of the complement receptor-3 (CR3). Furthermore, the blockade of CR3 specifically impacted the interleukin (IL)-1β secretion by hMDM in response to both S. brasiliensis and S. schenckii, suggesting that the host complement system plays an essential role in the inflammatory immune response against these Sporothrix species. Nevertheless, the structural differences in the PRMs of the two Sporothrix species, as revealed by NMR, were related to the differences observed in the host complement activation pathways. Together, this work reports a new PAMP of the cell surface of pathogenic fungi playing a role through the activation of complement system and via CR3 receptor mediating an inflammatory response to Sporothrix species.
Collapse
Affiliation(s)
- Gabriela W. P. Neves
- Cell Biology Department, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | | | - Catherine Simenel
- Institut Pasteur, Biological NMR and HDX-MS Technological Platform, CNRS UMR3528, Paris, France
| | - J. Iñaki Guijarro
- Institut Pasteur, Biological NMR and HDX-MS Technological Platform, CNRS UMR3528, Paris, France
| | - Catriona Walls
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Janet A. Willment
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom,Medical Research Council Centre for Medical Mycology at the University of Exeter, Exeter, United Kingdom
| | - Neil A. R. Gow
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom,Medical Research Council Centre for Medical Mycology at the University of Exeter, Exeter, United Kingdom
| | - Carol A. Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Gordon D. Brown
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom,Medical Research Council Centre for Medical Mycology at the University of Exeter, Exeter, United Kingdom
| | - Leila M. Lopes-Bezerra
- Cell Biology Department, Rio de Janeiro State University, Rio de Janeiro, Brazil,Biomedical Institute and Technology and Innovation Center (CIETEC), São Paulo University, São Paulo, Brazil,*Correspondence: Leila M. Lopes-Bezerra,
| |
Collapse
|
40
|
McGee HM, Marciscano AE, Campbell AM, Monjazeb AM, Kaech SM, Teijaro JR. Parallels Between the Antiviral State and the Irradiated State. J Natl Cancer Inst 2021; 113:969-979. [PMID: 33252657 PMCID: PMC8502484 DOI: 10.1093/jnci/djaa190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/07/2020] [Accepted: 11/16/2020] [Indexed: 01/12/2023] Open
Abstract
Improved understanding of host antiviral defense and antitumor immunity have elucidated molecular pathways important to both processes. During viral infection, RNA or DNA in the host cell serves as a danger signal that initiates the antiviral response. Recent studies have elucidated similarities in the signaling pathways activated by viruses and the signaling pathways induced by tumor DNA that is released into the cytoplasm of irradiated tumor cells. Both the host defense to viral infection and the sterile inflammation provoked by radiotherapy induce a type I interferon response that is necessary for pathogen control and immune-mediated tumor control, respectively. These findings have led to the hypothesis that radiotherapy employs a form of viral mimicry.
Collapse
Affiliation(s)
- Heather M McGee
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ariel E Marciscano
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Allison M Campbell
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Arta M Monjazeb
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - John R Teijaro
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
41
|
Liu Q, Shkirkova K, Lamorie-Foote K, Connor M, Patel A, Babadjouni R, Huuskonen M, Montagne A, Baertsch H, Zhang H, Chen JC, Mack WJ, Walcott BP, Zlokovic BV, Sioutas C, Morgan TE, Finch CE, Mack WJ. Air Pollution Particulate Matter Exposure and Chronic Cerebral Hypoperfusion and Measures of White Matter Injury in a Murine Model. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:87006. [PMID: 34424052 PMCID: PMC8382048 DOI: 10.1289/ehp8792] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 05/19/2023]
Abstract
BACKGROUND Exposure to ambient air pollution particulate matter (PM) is associated with increased risk of dementia and accelerated cognitive loss. Vascular contributions to cognitive impairment are well recognized. Chronic cerebral hypoperfusion (CCH) promotes neuroinflammation and blood-brain barrier weakening, which may augment neurotoxic effects of PM. OBJECTIVES This study examined interactions of nanoscale particulate matter (nPM; fine particulate matter with aerodynamic diameter ≤ 200 nm ) and CCH secondary to bilateral carotid artery stenosis (BCAS) in a murine model to produce white matter injury. Based on other air pollution interactions, we predicted synergies of nPM with BCAS. METHODS nPM was collected using a particle sampler near a Los Angeles, California, freeway. Mice were exposed to 10 wk of reaerosolized nPM or filtered air (FA) for 150 h. CCH was induced by BCAS surgery. Mice (C57BL/6J males) were randomized to four exposure paradigms: a) FA, b) nPM, c) FA + BCAS , and d) nPM + BCAS . Behavioral outcomes, white matter injury, glial cell activation, inflammation, and oxidative stress were assessed. RESULTS The joint nPM + BCAS group exhibited synergistic effects on white matter injury (2.3× the additive nPM and FA + BCAS scores) with greater loss of corpus callosum volume on T2 magnetic resonance imaging (MRI) (30% smaller than FA group). Histochemical analyses suggested potential microglial-specific inflammatory responses with synergistic effects on corpus callosum C5 immunofluorescent density and whole brain nitrate concentrations (2.1× and 3.9× the additive nPM and FA + BCAS effects, respectively) in the joint exposure group. Transcriptomic responses (RNA-Seq) showed greater impact of nPM + BCAS than individual additive effects, consistent with changes in proinflammatory pathways. Although nPM exposure alone did not alter working memory, the nPM + BCAS cohort demonstrated impaired working memory when compared to the FA + BCAS group. DISCUSSION Our data suggest that nPM and CCH contribute to white matter injury in a synergistic manner in a mouse model. Adverse neurological effects may be aggravated in a susceptible population exposed to air pollution. https://doi.org/10.1289/EHP8792.
Collapse
Affiliation(s)
- Qinghai Liu
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Kristina Shkirkova
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Krista Lamorie-Foote
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Michelle Connor
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Arati Patel
- Department of Neurological Surgery, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Robin Babadjouni
- Department of Neurological Surgery, Cedars-Sinai, Los Angeles, California, USA
| | - Mikko Huuskonen
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, California, USA
| | - Axel Montagne
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, California, USA
| | - Hans Baertsch
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Hongqiao Zhang
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - Jiu-Chiuan Chen
- Department of Preventative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA
| | - Wendy J. Mack
- Department of Preventative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA
| | - Brian P. Walcott
- Department of Neurosurgery, Northshore Neurological Institute, Evanston, Illinois, USA
| | - Berislav V. Zlokovic
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, California, USA
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Todd E. Morgan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - Caleb E. Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - William J. Mack
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
42
|
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
|
43
|
Abendaño N, Esparza-Baquer A, Bernales I, Reina R, de Andrés D, Jugo BM. Gene Expression Profiling Reveals New Pathways and Genes Associated with Visna/Maedi Viral Disease. Animals (Basel) 2021; 11:ani11061785. [PMID: 34203742 PMCID: PMC8232142 DOI: 10.3390/ani11061785] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/02/2021] [Accepted: 06/06/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Visna/Maedi is a disease caused by a small ruminant lentivirus (SRLV), with different symptoms in adult sheep such as pneumonia, arthritis, encephalitis and mastitis. SRLV infection in sheep is widespread across the world, with Europe showing the highest individual prevalence. There is currently no effective treatment for SRLV infections and, due to their constant changes, effective vaccine development has been and is still challenging. The dynamics of the sheep immune response to these virus infections is unclear, and changes in gene expression can help to explain the processes occurring in infected sheep. In this study, a gene expression microarray was used to identify the differentially expressed genes in infected and diseased sheep by comparing animals with different serologic statuses and with the presence of VM-characteristic clinical lesions in the lungs. The expression profile analysis revealed many interesting genes that may be associated with the viral infection process (such as OXT and a number of genes implicated in the Toll Like Receptors signaling network and complement pathway). This work improves our understanding of the sheep immune response against SRLVs. Abstract Visna/Maedi virus (VMV) is a lentivirus that infects the cells of the monocyte/macrophage lineage in sheep, goats and wild ruminants. Infection with VMV causes a multisystemic inflammatory disorder, which includes pneumonia, encephalitis, mastitis or arthritis. The immune response to VMV infection is complex, and the infection and pathogenesis of this virus are not totally characterized yet. In this work, a gene expression microarray was used to identify the differentially expressed genes in VMV infection and disease development by comparing sheep with different serologic status and with presence of VM-characteristic clinical lesions. The expression profile analysis has revealed many interesting genes that may be associated with the viral infection process. Among them, the OXT gene appeared significantly up-regulated, so the oxytocin-secreting system could play an essential role in VM disease. Moreover, some of the most significantly enriched functions in up-regulated genes appeared the complement pathway, which (in combination with the Toll-like receptor signaling network) could compose a mechanism in the VMV pathogenesis. Identifying the host genetic factors associated with VMV infection can be applied to develop strategies for preventing infection and develop effective vaccines that lead to therapeutic treatments.
Collapse
Affiliation(s)
- Naiara Abendaño
- Genetics, Physical Anthropology and Animal Physiology Department, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (N.A.); (A.E.-B.)
| | - Aitor Esparza-Baquer
- Genetics, Physical Anthropology and Animal Physiology Department, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (N.A.); (A.E.-B.)
| | - Irantzu Bernales
- Gene Expression Unit, Genomics Facility of General Research Services (SGIker), Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
| | - Ramsés Reina
- Instituto de Agrobiotecnología (CSIC-Gobierno de Navarra), 31192 Mutilva Baja, Spain; (R.R.); (D.d.A.)
| | - Damián de Andrés
- Instituto de Agrobiotecnología (CSIC-Gobierno de Navarra), 31192 Mutilva Baja, Spain; (R.R.); (D.d.A.)
| | - Begoña M. Jugo
- Genetics, Physical Anthropology and Animal Physiology Department, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (N.A.); (A.E.-B.)
- Correspondence:
| |
Collapse
|
44
|
Pinocembrin alleviates ulcerative colitis in mice via regulating gut microbiota, suppressing TLR4/MD2/NF-κB pathway and promoting intestinal barrier. Biosci Rep 2021; 40:225839. [PMID: 32687156 PMCID: PMC7391130 DOI: 10.1042/bsr20200986] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/24/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022] Open
Abstract
Pinocembrin, a plant-derived flavonoid, has a variety of pharmacological activities, including anti-infection, anti-cancer, anti-inflammation, cardiovascular protection, etc. However, the mechanism of pinocembrin on the anti-colitis efficacy remains elusive and needs further investigation. Here, we reported that pinocembrin eased the severity of dextran sulfate sodium (DSS)-induced colitis in mice by suppressing the abnormal activation of toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signal pathway in vivo. In addition, the gut microbiota was disordered in DSS colitis mice, which was associated with a significant decrease in microbiota diversity and a great shift in bacteria profiles; however, pinocembrin treatment improved the imbalance of gut microbiota and made it similar to that in normal mice. On the other hand, in vitro, pinocembrin down-regulated the TLR4/NF-κB signaling cascades in lipopolysaccharide (LPS)-stimulated macrophages. At the upstream level, pinocembrin competitively inhibited the binding of LPS to myeloid differentiation protein 2 (MD2), thereby blocking the formation of receptor multimer TLR4/MD2·LPS. Furthermore, pinocembrin dose-dependently promoted the expression of tight junction proteins (ZO-1, Claudin-1, Occludin and JAM-A) in Caco-2 cells. In conclusion, our work presented evidence that pinocembrin attenuated DSS-induced colitis in mouse, at least in part, via regulating intestinal microbiota, inhibiting the over-activation of TLR4/MD2/NF-κB signaling pathway, and improving the barriers of intestine.
Collapse
|
45
|
Rajasekaran S, Chitraa T, Dilip Chand Raja S, Raveendran M, Sharon Miracle N, Sri Vijayanand KS, Ajoy Prasad S, Rishi Mugesh K. Subclinical infection can be an initiator of inflammaging leading to degenerative disk disease: evidence from host-defense response mechanisms. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2021; 30:2586-2604. [PMID: 33835272 DOI: 10.1007/s00586-021-06826-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/06/2021] [Accepted: 03/20/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE There is considerable controversy on the role of genetics, mechanical and environmental factors, and, recently, on subclinical infection in triggering inflammaging leading to disk degeneration. The present study investigated sequential molecular events in the host, analyzing proteome level changes that will reveal triggering factors of inflammaging and degeneration. METHODS Ten MRI normal disks (ND) from braindead organ donors and 17 degenerated disks (DD) from surgery were subjected to in-gel-based label-free ESI-LC-MS/MS analysis. Bacterial-responsive host-defense response proteins/pathways leading to Inflammaging were identified and compared between ND and DD. RESULTS Out of the 263 well-established host-defense response proteins (HDRPs), 243 proteins were identified, and 64 abundantly expressed HDRPs were analyzed further. Among the 21 HDRPs common to both ND and DD, complement factor 3 (C3) and heparan sulfate proteoglycan 2 (HSPG2) were significantly upregulated, and lysozyme (LYZ), superoxide dismutase 3 (SOD3), phospholipase-A2 (PLA2G2A), and tissue inhibitor of metalloproteinases 3 (TIMP-3) were downregulated in DD. Forty-two specific HDRPs mainly, complement proteins, apolipoproteins, and antimicrobial proteins involved in the complement cascade, neutrophil degranulation, and oxidative-stress regulation pathways representing an ongoing host response to subclinical infection and uncontrolled inflammation were identified in DD. Protein-Protein interaction analysis revealed cross talk between most of the expressed HDRPs, adding evidence to bacterial presence and stimulation of these defense pathways. CONCLUSIONS The predominance of HDRPs involved in complement cascades, neutrophil degranulation, and oxidative-stress regulation indicated an ongoing infection mediated inflammatory process in DD. Our study has documented increasing evidence for bacteria's role in triggering the innate immune system leading to chronic inflammation and degenerative disk disease.
Collapse
Affiliation(s)
- S Rajasekaran
- Department of Orthopaedics and Spine Surgery, Ganga Hospital, 313, Mettupalayam road, Coimbatore, India.
| | - Tangavel Chitraa
- Ganga Research Centre, No 91, Mettupalayam road, Coimbatore, 641030, India
| | - S Dilip Chand Raja
- Department of Orthopaedics and Spine Surgery, Ganga Hospital, 313, Mettupalayam road, Coimbatore, India
| | - M Raveendran
- Department of Plant Biotechnology, Tamil Nadu Agricultural University, Coimbatore, 641003, India
| | | | - K S Sri Vijayanand
- Department of Orthopaedics and Spine Surgery, Ganga Hospital, 313, Mettupalayam road, Coimbatore, India
| | - Shetty Ajoy Prasad
- Department of Orthopaedics and Spine Surgery, Ganga Hospital, 313, Mettupalayam road, Coimbatore, India
| | - Kanna Rishi Mugesh
- Department of Orthopaedics and Spine Surgery, Ganga Hospital, 313, Mettupalayam road, Coimbatore, India
| |
Collapse
|
46
|
Montay-Gruel P, Markarian M, Allen BD, Baddour JD, Giedzinski E, Jorge PG, Petit B, Bailat C, Vozenin MC, Limoli C, Acharya MM. Ultra-High-Dose-Rate FLASH Irradiation Limits Reactive Gliosis in the Brain. Radiat Res 2021; 194:636-645. [PMID: 32853387 DOI: 10.1667/rade-20-00067.1] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 06/18/2020] [Indexed: 12/20/2022]
Abstract
Encephalic radiation therapy delivered at a conventional dose rate (CONV, 0.1-2.0 Gy/min) elicits a variety of temporally distinct damage signatures that invariably involve persistent indications of neuroinflammation. Past work has shown an involvement of both the innate and adaptive immune systems in modulating the central nervous system (CNS) radiation injury response, where elevations in astrogliosis, microgliosis and cytokine signaling define a complex pattern of normal tissue toxicities that never completely resolve. These side effects constitute a major limitation in the management of CNS malignancies in both adult and pediatric patients. The advent of a novel ultra-high dose-rate irradiation modality termed FLASH radiotherapy (FLASH-RT, instantaneous dose rates ≥106 Gy/s; 10 Gy delivered in 1-10 pulses of 1.8 µs) has been reported to minimize a range of normal tissue toxicities typically concurrent with CONV exposures, an effect that has been coined the "FLASH effect." Since the FLASH effect has now been found to significantly limit persistent inflammatory signatures in the brain, we sought to further elucidate whether changes in astrogliosis might account for the differential dose-rate response of the irradiated brain. Here we report that markers selected for activated astrogliosis and immune signaling in the brain (glial fibrillary acidic protein, GFAP; toll-like receptor 4, TLR4) are expressed at reduced levels after FLASH irradiation compared to CONV-irradiated animals. Interestingly, while FLASH-RT did not induce astrogliosis and TLR4, the expression level of complement C1q and C3 were found to be elevated in both FLASH and CONV irradiation modalities compared to the control. Although functional outcomes in the CNS remain to be cross-validated in response to the specific changes in protein expression reported, the data provide compelling evidence that distinguishes the dose-rate response of normal tissue injury in the irradiated brain.
Collapse
Affiliation(s)
- Pierre Montay-Gruel
- Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| | - Mineh Markarian
- Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| | - Barrett D Allen
- Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| | - Jabra D Baddour
- Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| | - Patrik Goncalves Jorge
- Laboratory of Radiation Oncology, Department of Radiation Oncology. Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Benoît Petit
- Laboratory of Radiation Oncology, Department of Radiation Oncology. Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Claude Bailat
- Laboratory of Radiation Oncology, Department of Radiation Oncology. Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Department of Radiation Oncology. Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Charles Limoli
- Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| | - Munjal M Acharya
- Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| |
Collapse
|
47
|
Corrêa T, Feltes BC, Gonzalez EA, Baldo G, Matte U. Network Analysis Reveals Proteins Associated with Aortic Dilatation in Mucopolysaccharidoses. Interdiscip Sci 2021; 13:34-43. [PMID: 33475959 DOI: 10.1007/s12539-020-00406-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 11/25/2020] [Accepted: 12/03/2020] [Indexed: 06/12/2023]
Abstract
Mucopolysaccharidoses are caused by a deficiency of enzymes involved in the degradation of glycosaminoglycans. Heart diseases are a significant cause of morbidity and mortality in MPS patients, even in conditions in which enzyme replacement therapy is available. In this sense, cardiovascular manifestations, such as heart hypertrophy, cardiac function reduction, increased left ventricular chamber, and aortic dilatation, are among the most frequent. However, the downstream events which influence the heart dilatation process are unclear. Here, we employed systems biology tools together with transcriptomic data to investigate new elements that may be involved in aortic dilatation in Mucopolysaccharidoses syndrome. We identified candidate genes involved in biological processes related to inflammatory responses, deposition of collagen, and lipid accumulation in the cardiovascular system that may be involved in aortic dilatation in the Mucopolysaccharidoses I and VII. Furthermore, we investigated the molecular mechanisms of losartan treatment in Mucopolysaccharidoses I mice to underscore how this drug acts to prevent aortic dilation. Our data indicate that the association between the TGF-b signaling pathway, Fos, and Col1a1 proteins can play an essential role in aortic dilation's pathophysiology and its subsequent improvement by losartan treatment.
Collapse
Affiliation(s)
- Thiago Corrêa
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Porto Alegre, 90035-903, Brazil
- Postgraduation Program on Genetics and Molecular Biology, Federal University of Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Bruno César Feltes
- Institute of Informatics, Federal University of Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Esteban Alberto Gonzalez
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Porto Alegre, 90035-903, Brazil
- Postgraduation Program on Genetics and Molecular Biology, Federal University of Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Guilherme Baldo
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Porto Alegre, 90035-903, Brazil
- Postgraduation Program on Genetics and Molecular Biology, Federal University of Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Ursula Matte
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Porto Alegre, 90035-903, Brazil.
- Postgraduation Program on Genetics and Molecular Biology, Federal University of Rio Grande Do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
48
|
Keshari RS, Silasi R, Popescu NI, Regmi G, Chaaban H, Lambris JD, Lupu C, Mollnes TE, Lupu F. CD14 inhibition improves survival and attenuates thrombo-inflammation and cardiopulmonary dysfunction in a baboon model of Escherichia coli sepsis. J Thromb Haemost 2021; 19:429-443. [PMID: 33174372 PMCID: PMC8312235 DOI: 10.1111/jth.15162] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/02/2020] [Accepted: 10/26/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND During sepsis, gram-negative bacteria induce robust inflammation primarily via lipopolysacharride (LPS) signaling through TLR4, a process that involves the glycosylphosphatidylinositol (GPI)-anchored receptor CD14 transferring LPS to the Toll-like receptor 4/myeloid differentiation factor 2 (TLR4/MD-2) complex. Sepsis also triggers the onset of disseminated intravascular coagulation and consumptive coagulopathy. OBJECTIVES We investigated the effect of CD14 blockade on sepsis-induced coagulopathy, inflammation, organ dysfunction, and mortality. METHODS We used a baboon model of lethal Escherichia (E) coli sepsis to study two experimental groups (n = 5): (a) E coli challenge; (b) E coli challenge plus anti-CD14 (23G4) inhibitory antibody administered as an intravenous bolus 30 minutes before the E coli. RESULTS Following anti-CD14 treatment, two animals reached the 7-day end-point survivor criteria, while three animals had a significantly prolonged survival as compared to the non-treated animals that developed multiple organ failure and died within 30 hours. Anti-CD14 reduced the activation of coagulation through inhibition of tissue factor-dependent pathway, especially in the survivors, and enhanced the fibrinolysis due to strong inhibition of plasminogen activator inhibitor 1. The treatment prevented the robust complement activation induced by E coli, as shown by significantly decreased C3b, C5a, and sC5b-9. Vital signs, organ function biomarkers, bacteria clearance, and leukocyte and fibrinogen consumption were all improved at varying levels. Anti-CD14 reduced neutrophil activation, cell death, LPS levels, and pro-inflammatory cytokines (tumor necrosis factor, interleukin (IL)-6, IL-1β, IL-8, interferon gamma, monocyte chemoattractant protein-1), more significantly in the survivors than non-surviving animals. CONCLUSIONS Our results highlight the crosstalk between coagulation/fibrinolysis, inflammation, and complement systems and suggest a protective role of anti-CD14 treatment in E coli sepsis.
Collapse
Affiliation(s)
- Ravi S. Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Narcis I. Popescu
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Girija Regmi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Hala Chaaban
- Department of Pediatrics, Neonatal and Perinatal Section, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - John D. Lambris
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cristina Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Tom E. Mollnes
- Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, Oslo, Norway
- Research Laboratory Nordland Hospital, K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Bodo, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Departments of Cell Biology, Pathology and Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
49
|
Yasmin H, Saha S, Butt MT, Modi RK, George AJT, Kishore U. SARS-CoV-2: Pathogenic Mechanisms and Host Immune Response. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:99-134. [PMID: 34661893 DOI: 10.1007/978-3-030-67452-6_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an enveloped, positive-sense RNA coronavirus responsible for the COVID-19 pandemic. Since December 2019, coronavirus disease 2019 (COVID-19) has affected more than 127 million people, 2.7 million deaths globally (as per WHO dashboard, dated 31 March, 2020), the virus is capable of transmitting from human to human via inhalation of infected respiratory droplets or aerosols or contact with infected fomites. Clinically, patients with COVID-19 present with severe respiratory distress syndrome, which is very similar to the presentation of other respiratory viral infections. A huge variation in the host response exists, with the resulting symptoms varying from mild to moderate. Comorbidities such as cardiovascular disease, hypertension, diabetes, coagulation dysfunction, stroke, malignant tumor and multiple organ dysfunction syndrome, as well as age and sex, are associated with severe COVID-19 cases. So far, no targeted therapies have been developed to treat this disease and existing drugs are being investigated for repurposing. This chapter discusses the epidemiology, clinical features of COVID-19, pathogenesis and the innate and adaptive immune response mounted by the host to the SARS-CoV-2 infection. A deeper understanding of the host-pathogen interaction is fundamental to the development of a vaccine.
Collapse
Affiliation(s)
- Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Sudipta Saha
- Amity Institute of Physiology and Allied Sciences, Amity University Campus, Noida, Uttar Pradesh, India
| | - Mariam Tariq Butt
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | - Rishab Kumar Modi
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Andrew J T George
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK.
| |
Collapse
|
50
|
Schartz ND, Tenner AJ. The good, the bad, and the opportunities of the complement system in neurodegenerative disease. J Neuroinflammation 2020; 17:354. [PMID: 33239010 PMCID: PMC7690210 DOI: 10.1186/s12974-020-02024-8] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
The complement cascade is a critical effector mechanism of the innate immune system that contributes to the rapid clearance of pathogens and dead or dying cells, as well as contributing to the extent and limit of the inflammatory immune response. In addition, some of the early components of this cascade have been clearly shown to play a beneficial role in synapse elimination during the development of the nervous system, although excessive complement-mediated synaptic pruning in the adult or injured brain may be detrimental in multiple neurogenerative disorders. While many of these later studies have been in mouse models, observations consistent with this notion have been reported in human postmortem examination of brain tissue. Increasing awareness of distinct roles of C1q, the initial recognition component of the classical complement pathway, that are independent of the rest of the complement cascade, as well as the relationship with other signaling pathways of inflammation (in the periphery as well as the central nervous system), highlights the need for a thorough understanding of these molecular entities and pathways to facilitate successful therapeutic design, including target identification, disease stage for treatment, and delivery in specific neurologic disorders. Here, we review the evidence for both beneficial and detrimental effects of complement components and activation products in multiple neurodegenerative disorders. Evidence for requisite co-factors for the diverse consequences are reviewed, as well as the recent studies that support the possibility of successful pharmacological approaches to suppress excessive and detrimental complement-mediated chronic inflammation, while preserving beneficial effects of complement components, to slow the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Nicole D. Schartz
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| | - Andrea J. Tenner
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Neurobiology and Behavior, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| |
Collapse
|