1
|
Liu Y, Zhao W, Huang Q, Wan L, Ren Z, Zhang B, Han C, Yang J, Zhang H, Zhang J. Advances in Research on the Release of von Willebrand Factor from Endothelial Cells through the Membrane Attack Complex C5b-9 in Sepsis. J Inflamm Res 2025; 18:6719-6733. [PMID: 40438181 PMCID: PMC12118641 DOI: 10.2147/jir.s520726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 05/13/2025] [Indexed: 06/01/2025] Open
Abstract
Sepsis, a lethal organ dysfunction syndrome driven by aberrant host responses to infection, intertwines excessive inflammatory responses and dysregulated coagulation processes in its pathophysiology. Emerging research reveals the complement terminal membrane attack complex C5b-9 orchestrates ultralarge von Willebrand factor (ULVWF) release from vascular endothelial cells (ECs) through multifaceted mechanisms: C5b-9 compromises EC membrane integrity, activates calcium influx cascades, and provokes NLRP3 inflammasome signaling, triggering massive exocytosis of ULVWF stored within Weibel-Palade bodies (WPBs). When ADAMTS13 activity falters, undegraded ULVWF complexes with platelets to spawn microthrombi, precipitating microvascular occlusion and multiorgan collapse. Strikingly, elevated plasma von Willebrand factor (vWF) antigen levels in sepsis patients correlate robustly with endothelial injury, thrombocytopenia, and mortality-underscoring C5b-9-driven vWF release as a linchpin of septic coagulopathy. Current therapeutic strategies targeting these pathways, including recombinant ADAMTS13 (rhADAMTS13), N-acetylcysteine (NAC), and complement inhibitors like eculizumab, face limitations in clinical translation, necessitating further validation of their efficacy. Additionally, investigating complement regulatory molecules such as CD59 may unlock novel therapeutic avenues. Deciphering the intricate interplay within the C5b-9-vWF axis and advancing precision therapies hold transformative potential for ameliorating sepsis outcomes.
Collapse
Affiliation(s)
- Yi Liu
- Department of Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Weili Zhao
- Laboratory Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Qingqing Huang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Linjun Wan
- Department of Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Zongfang Ren
- Department of Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Bangting Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Chen Han
- Department of Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Jin Yang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Haoling Zhang
- Department of Biomedical Science, Advanced Medical and Dental Institute, University Sains Malaysia, Penang, 13200, Malaysia
| | - Jingjing Zhang
- Fuwai Yunnan Hospital, Chinese Academy Medical Sciences, Kunming, 650000, People’s Republic of China
- Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, 650000, People’s Republic of China
| |
Collapse
|
2
|
Wang X, Chen L, Wei J, Zheng H, Zhou N, Xu X, Deng X, Liu T, Zou Y. The immune system in cardiovascular diseases: from basic mechanisms to therapeutic implications. Signal Transduct Target Ther 2025; 10:166. [PMID: 40404619 PMCID: PMC12098830 DOI: 10.1038/s41392-025-02220-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/22/2024] [Accepted: 03/20/2025] [Indexed: 05/24/2025] Open
Abstract
Immune system plays a crucial role in the physiological and pathological regulation of the cardiovascular system. The exploration history and milestones of immune system in cardiovascular diseases (CVDs) have evolved from the initial discovery of chronic inflammation in atherosclerosis to large-scale clinical studies confirming the importance of anti-inflammatory therapy in treating CVDs. This progress has been facilitated by advancements in various technological approaches, including multi-omics analysis (single-cell sequencing, spatial transcriptome et al.) and significant improvements in immunotherapy techniques such as chimeric antigen receptor (CAR)-T cell therapy. Both innate and adaptive immunity holds a pivotal role in CVDs, involving Toll-like receptor (TLR) signaling pathway, nucleotide-binding oligomerization domain-containing proteins 1 and 2 (NOD1/2) signaling pathway, inflammasome signaling pathway, RNA and DNA sensing signaling pathway, as well as antibody-mediated and complement-dependent systems. Meanwhile, immune responses are simultaneously regulated by multi-level regulations in CVDs, including epigenetics (DNA, RNA, protein) and other key signaling pathways in CVDs, interactions among immune cells, and interactions between immune and cardiac or vascular cells. Remarkably, based on the progress in basic research on immune responses in the cardiovascular system, significant advancements have also been made in pre-clinical and clinical studies of immunotherapy. This review provides an overview of the role of immune system in the cardiovascular system, providing in-depth insights into the physiological and pathological regulation of immune responses in various CVDs, highlighting the impact of multi-level regulation of immune responses in CVDs. Finally, we also discuss pre-clinical and clinical strategies targeting the immune system and translational implications in CVDs.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Liming Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Wei
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Hao Zheng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Ning Zhou
- Department of Cardiovascular Medicine, Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Deng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China.
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Jiangsu, Nanjing, China.
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
- Institutes of Advanced Medical Sciences and Huaihe Hospital, Henan University, Kaifeng, Henan, China.
| |
Collapse
|
3
|
Ahmed H, Fernandes MF, Abbas K, Synowsky SA, Shirran SL, Ajjan RA, Stewart AJ. Quantitative proteomics identifies plasma protein alterations that associate with metabolic and thrombotic profile changes after bariatric surgery. Diabetes Obes Metab 2025; 27:2647-2657. [PMID: 40000393 PMCID: PMC11965011 DOI: 10.1111/dom.16267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025]
Abstract
OBJECTIVE Roux-en-Y gastric bypass (RYGB) surgery has been shown to lead to favourable health outcomes in obese patients. However, the molecular changes that occur and how they relate to clinical measures are poorly understood. Here, we characterise the proteomic alterations that occur in plasma of RYGB patients before and 9 months after surgery using quantitative proteomics. METHODS Plasma proteomics was performed by sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH-MS) to identify and quantify differentially abundant proteins. Relationships between significantly altered proteins and clinical markers were examined. A gene set enrichment analysis was also conducted to identify altered pathways. RESULTS From the proteomic analysis, 27 proteins increased, and 43 proteins decreased in abundance 9 months after surgery, providing insights into the physiological changes that accompany weight loss. Proteins including sex hormone binding globulin (SHBG), inter-alpha-trypsin inhibitor heavy chain 3 (ITIH3) and apolipoprotein D (APOD), which increased in abundance post-surgery, highlight improvements in lipid regulation, insulin sensitivity and inflammation. Proteins involved in coagulation, including α2-macroglobulin, kallikrein-B1, prothrombin, and factor (FX, FXI and FXII), exhibited reduced levels, aligning with a decrease in thrombotic potential. CONCLUSIONS These findings provide a mechanistic understanding of how bariatric surgery leads to systemic changes in metabolic and haemostatic pathways, thus favourably modulating the risk of developing cardiovascular disease.
Collapse
Affiliation(s)
- Hasnain Ahmed
- School of MedicineUniversity of St AndrewsNorth HaughSt AndrewsFifeUK
| | | | - Kazim Abbas
- Renal Transplant UnitRoyal Liverpool University HospitalPrescot StreetLiverpoolMerseysideUK
| | - Silvia A. Synowsky
- Biomedical Sciences Research Complex and School of BiologyUniversity of St AndrewsNorth HaughSt AndrewsFifeUK
| | - Sally L. Shirran
- Biomedical Sciences Research Complex and School of BiologyUniversity of St AndrewsNorth HaughSt AndrewsFifeUK
| | - Ramzi A. Ajjan
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsClarendon WayLeedsYorkshireUK
| | - Alan J. Stewart
- School of MedicineUniversity of St AndrewsNorth HaughSt AndrewsFifeUK
| |
Collapse
|
4
|
Aalinkeel R, Quigg RJ, Alexander J. The complement system and kidney cancer: pathogenesis to clinical applications. J Clin Invest 2025; 135:e188351. [PMID: 40309765 PMCID: PMC12043091 DOI: 10.1172/jci188351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Kidney cancer poses unique clinical challenges because of its resistance to conventional treatments and its tendency to metastasize. The kidney is particularly susceptible to dysfunction of the complement system, an immune network that tumors often exploit. Recent discoveries have highlighted that the complement system not only plays a crucial role in immune surveillance and defense in the circulatory system, but also functions intracellularly and autonomously. This concept has shifted the focus of investigation toward understanding how complement proteins influence cancer progression by regulating the tumor microenvironment (TME), cell signaling, proliferation, metabolism, and the immune response. With the complement system and its inhibitors emerging as a promising new class of immunotherapeutics and potential complement-targeted treatments advancing through development pipelines and clinical trials, this Review provides a timely examination of how harnessing the complement system could lead to effective tumor treatments and how to strategically combine complement inhibitors with other cancer treatments, offering renewed hope in the fight against kidney cancer.
Collapse
|
5
|
Cyranka L, Mariegaard I, Fageräng B, Pérez‐Alós L, Harpf V, Mollnes TE, Garred P, Rosbjerg A. Inhibition of Alternative and Terminal Complement Pathway Components Modulate the Immune Response Against Bacteria and Fungi in Whole Blood. Scand J Immunol 2025; 101:e70030. [PMID: 40387159 PMCID: PMC12087264 DOI: 10.1111/sji.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 03/30/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025]
Abstract
Complement activation plays a critical role in the inflammatory response to Escherichia coli and Aspergillus fumigatus conidia. However, the specific contributions of complement components, including anaphylatoxin receptors, remain unclear. Using an ex vivo lepirudin whole blood model, we examined the activation of all three complement pathways (C4c, C3bc, and sC5b-9) induced by these microbes. We also assessed granulocyte and monocyte receptor expression of CD11b, CD64, C3aR, C5aR1, and C5aR2, along with phagocytosis, leukocyte activation (MPO), and cytokine release. Additionally, we investigated selective inhibition of complement components FD, C3, C5, and C5aR1. Both microbes increased complement activation products (C3bc and sC5-9), CD11b and CD64 expression, MPO release, and proinflammatory cytokines (IL-1β, IL-6, IL-8, and TNF), while decreasing C3aR, C5aR1, and C5aR2 expression. Complement inhibition reduced CD11b and CD64 expression and partially restored C3aR and C5aR1 levels, with minimal effects on C5aR2. FD, C3, and C5 inhibition reduced downstream complement markers, with FD and C3 inhibition also reducing phagocytosis, and only C3 inhibition reducing MPO release. The cytokine response varied by microbe: E. coli triggered higher proinflammatory cytokines, and FD and C3 inhibition generally reduced cytokine release, while C5 inhibition was less effective. Interestingly, A. fumigatus-induced cytokines significantly increased with C5aR1 inhibition, highlighting immune response differences related to C5aR1 signalling in bacterial versus fungal infections. In conclusion, regulation of inflammation through FD, C3, C5, and C5aR1 underscores the immunoregulatory role of the complement system in anti-microbial immune responses.
Collapse
Affiliation(s)
- Leon Cyranka
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631Copenhagen University Hospital ‐ RigshospitaletCopenhagenDenmark
| | - Ida Mariegaard
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631Copenhagen University Hospital ‐ RigshospitaletCopenhagenDenmark
| | - Beatrice Fageräng
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631Copenhagen University Hospital ‐ RigshospitaletCopenhagenDenmark
| | - Laura Pérez‐Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631Copenhagen University Hospital ‐ RigshospitaletCopenhagenDenmark
| | - Verena Harpf
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631Copenhagen University Hospital ‐ RigshospitaletCopenhagenDenmark
| | - Tom Eirik Mollnes
- Department of ImmunologyOslo University Hospital and University of OsloOsloNorway
- Research LaboratoryNordland HospitalBodøNorway
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631Copenhagen University Hospital ‐ RigshospitaletCopenhagenDenmark
| | - Anne Rosbjerg
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631Copenhagen University Hospital ‐ RigshospitaletCopenhagenDenmark
| |
Collapse
|
6
|
Atalay Ekiner S, Gęgotek A, Domingues MR, Domingues P, Skrzydlewska E. The Effects of Lipid Extracts from Microalgae Chlorococcum amblystomatis and Nannochloropsis oceanica on the Proteome of 3D-Cultured Fibroblasts Exposed to UVA Radiation. Antioxidants (Basel) 2025; 14:545. [PMID: 40427427 PMCID: PMC12108275 DOI: 10.3390/antiox14050545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Nannochloropsis oceanica and Chlorococcum amblystomatis exhibit significant potential for protecting skin cells from oxidative stress-induced metabolic dysfunctions, owing to their high bioactive lipid content. This study aimed to evaluate their cytoprotective effects on the ultraviolet A (UVA)-perturbed proteome of 3D-cultured skin fibroblasts, using high-throughput proteomics. Chlorococcum amblystomatis lipid extract promoted a reduction in UVA-induced cytochrome c oxidase subunit 4 isoform 1 and cell death protein 6 levels, alongside the restoration of ferritin light chain expression diminished by UVA. It downregulated the expression of ubiquitin-conjugating enzyme E2 and lactoylglutathione lyase, which were upregulated by UVA. Furthermore, the elevated superoxide dismutase [Mn] mitochondrial levels in the caspase-1 interactome emphasized the lipid extract's role in mitigating oxidative stress-associated chronic inflammation by regulating caspase-1 activity. In addition to this notable redox balance-regulating and cytoprotective activity, conversely, the protein inflammation signaling mediated by UVA was regulated in terms of wound healing potential in the case of Nannochloropsis oceanica lipid extract. Following UVA radiation, it promoted the upregulation of complement component B, thrombospondin-1, MMP1, and fibulin-1. The results revealed that both lipid extracts effectively reversed the UVA-perturbed proteomic profile of fibroblasts, highlighting their therapeutic potential in protecting the skin from UV radiation.
Collapse
Affiliation(s)
- Sinemyiz Atalay Ekiner
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland; (A.G.); (E.S.)
| | - Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland; (A.G.); (E.S.)
| | - Maria Rosário Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; (M.R.D.); (P.D.)
| | - Pedro Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; (M.R.D.); (P.D.)
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland; (A.G.); (E.S.)
| |
Collapse
|
7
|
Mahdavi S, Rosychuk K, Jenkins DJA, Percy AJ, Borchers CH, El-Sohemy A. Multiomics Analysis of a Micronutrient-Rich Dietary Pattern and the Aging Genotype 9p21 on the Plasma Proteome of Young Adults. Nutrients 2025; 17:1398. [PMID: 40284261 PMCID: PMC12030164 DOI: 10.3390/nu17081398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Diet is one of the most significant modifiable lifestyle factors influencing human health, contributing to both morbidity and mortality. Genetic variations in the pleiotropic 9p21 risk locus further shape premature aging, disease susceptibility, and have been strongly linked to cardiovascular disease (CVD), metabolic disorders, certain cancers, and neurodegenerative conditions. However, given that this region was discovered based on Genome-Wide Association Studies, the mechanisms by which 9p21 exerts its effects remain poorly understood and its interactions with diet and biomarkers are insufficiently explored. Methods: This study investigated the association between the rs2383206 SNP in 9p21, dietary patterns, and plasma proteomic biomarkers in a multi-ethnic cohort of 1280 young adults from the Toronto Nutrigenomics and Health Study. Participants' dietary intake was assessed using a validated food frequency questionnaire, and dietary patterns were categorized using principal component analysis. Plasma proteomics analyses quantified 54 abundant proteins involved in the cardiometabolic and inflammatory pathways. Genotyping identified individuals who were homozygous for the 9p21 risk allele (GG), known to confer the highest susceptibility risk to premature aging and multiple chronic diseases. Results: A significant interaction was observed between the 9p21 genotype and adherence to a micronutrient-rich Prudent dietary pattern for eight plasma proteins (α1 Antichymotrypsin, Complement C4 β chain, Complement C4 γ chain, Complement C9, Fibrinogen α chain, Hemopexin, and Serum amyloid P-component). However, only Complement C4-γ showed a pattern consistent with the risks associated with the 9p21 genotype and adherence to a Prudent diet. Individuals with the high-risk GG genotype had significantly higher concentrations of Complement C4-γ, but only among those with a low adherence to a Prudent diet. Conclusions: These findings suggest that Prudent dietary patterns rich in micronutrients may counteract genetic-mediated proinflammatory susceptibility by modulating key proteomic biomarkers in young adults, highlighting the potential for tailored dietary interventions to mitigate disease risk. This study also introduces a novel framework for post hoc micronutrient resolution within dietary pattern analysis, offering a new lens to interpret nutrient synergies in gene-diet interaction research.
Collapse
Affiliation(s)
- Sara Mahdavi
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, 6 Queens Park Crescent, Toronto, ON M5S 3H2, Canada; (S.M.); (K.R.); (D.J.A.J.)
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, 677 Huntington Ave, Building B, Room 359, Boston, MA 02115, USA
| | - Katie Rosychuk
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, 6 Queens Park Crescent, Toronto, ON M5S 3H2, Canada; (S.M.); (K.R.); (D.J.A.J.)
| | - David J. A. Jenkins
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, 6 Queens Park Crescent, Toronto, ON M5S 3H2, Canada; (S.M.); (K.R.); (D.J.A.J.)
- Clinical Nutrition and Risk Factor Modification Centre, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
| | - Andrew J. Percy
- Genome BC Proteomics Centre, University of Victoria, Victoria, BC V8Z 7X8, Canada;
- Department of Applications Development, Cambridge Isotope Laboratories, Inc., Tewksbury, MA 01876, USA
| | - Christoph H. Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada;
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H3T 1E2, Canada
- Department of Pathology, McGill University, Montreal, QC H3T 1E2, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC H3T 1E2, Canada
| | - Ahmed El-Sohemy
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, 6 Queens Park Crescent, Toronto, ON M5S 3H2, Canada; (S.M.); (K.R.); (D.J.A.J.)
| |
Collapse
|
8
|
Alhamdan F, Maisat W, Higgins L, Chen Y, Ibla J, Yuki K. The Role of Extracellular-Vesicle-Derived miRNAs in Postoperative Organ Dysfunction in Neonates and Infants Undergoing Congenital Cardiac Surgery: An Exploratory Study. Int J Mol Sci 2025; 26:3837. [PMID: 40332490 PMCID: PMC12028143 DOI: 10.3390/ijms26083837] [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: 02/04/2025] [Revised: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
Despite significant advancements in medical and surgical care, the morbidity and mortality rates of neonates and infants undergoing congenital cardiac surgery remain high. To identify new pathomechanisms associated with postoperative organ dysfunction, extracellular vesicles (EVs) were isolated from plasma from neonates and infants with or without organ dysfunction at three different time points around congenital cardiac surgery, and the EV miRNA expression profiles in the plasma were analyzed. A clear distinction was observed between the organ dysfunction (OD) and non-organ dysfunction (NOD) groups based on their EV miRNA expression profiles. Apoptosis and proinflammatory pathways were consistently upregulated across all time points in the OD group. Complement and coagulation cascades unexpectedly displayed downregulation at the end of the surgery in the OD group, which was verified further at the proteomic level in an independent patient cohort. The neutrophil extracellular trap (NET) formation was enhanced in the OD group across all time points compared to that in the NOD group. As NETs are known to consume complement components, these observed events might be interconnected. A feature selection machine learning method identified miR-200b-5p, miR-4800-5p, miR-363-3p, and miR-483-5p as robustly linked to organ dysfunction following congenital cardiac surgery (accuracy score = 9; SD in accuracy = 0.3162). In conclusion, our study suggested that neonates and infants with postoperative organ dysfunction were associated with enhanced NET formation and complement consumption.
Collapse
Affiliation(s)
- Fahd Alhamdan
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, MA 02115, USA; (F.A.); (W.M.); (J.I.)
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Wiriya Maisat
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, MA 02115, USA; (F.A.); (W.M.); (J.I.)
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
- Department of Anesthesiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 73170, Thailand
| | - LeeAnn Higgins
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; (L.H.); (Y.C.)
| | - Yue Chen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; (L.H.); (Y.C.)
| | - Juan Ibla
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, MA 02115, USA; (F.A.); (W.M.); (J.I.)
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
| | - Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, MA 02115, USA; (F.A.); (W.M.); (J.I.)
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
9
|
Hirose T, Hotta K, Otsuka R, Seino KI. Mechanism and regulation of the complement activity in kidney xenotransplantation. Transplant Rev (Orlando) 2025; 39:100931. [PMID: 40233672 DOI: 10.1016/j.trre.2025.100931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
Xenotransplantation is emerging as one of several potential solutions for addressing organ donor shortages, with significant progress bringing it closer to clinical application. However, challenges remain, particularly concerning complement system dysregulation caused by species differences, as well as xenoantigens and coagulopathy. Complement regulatory proteins expressed on endothelial cells of donor xenografts are less compatible with complement components in recipients. These difficulties contribute to hyperacute rejection, characterized by antibody-mediated complement activation that destroys the graft within 24 h. Moreover, because molecules are incompatible across different species, ischemia-reperfusion injury or infection can easily elicit complement activity via all three pathways, resulting in xenograft loss via complement-mediated vascular injury. Complement activity also stimulate innate and adaptive immune cells. To address this issue, genetic modifications in donor pigs and the development of novel medicines have been tested in preclinical models with promising results. Pigs modified to express human complement-regulating molecules such as CD46, CD55, and CD59 have shown longer kidney xenograft survivals over years in preclinical models with nonhuman primates, paving the way for clinical trials. Anti-complement component agents such as C1 esterase and C5 inhibitors have also been shown to increase xenograft survivals. This review examines the role of the complement system in kidney xenotransplantation, emphasizing new research and clinical trial advancements.
Collapse
Affiliation(s)
- Takayuki Hirose
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan.
| | - Kiyohiko Hotta
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Ryo Otsuka
- Massachusetts General Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - Ken-Ichiro Seino
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
10
|
Ho BHT, Spicer BA, Dunstone MA. Action of the Terminal Complement Pathway on Cell Membranes. J Membr Biol 2025:10.1007/s00232-025-00343-6. [PMID: 40122920 DOI: 10.1007/s00232-025-00343-6] [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: 12/28/2024] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
The complement pathway is one of the most ancient elements of the host's innate response and includes a set of protein effectors that rapidly react against pathogens. The late stages of the complement reaction are broadly categorised into two major outcomes. Firstly, C5a receptors, expressed on membranes of host cells, are activated by C5a to generate pro-inflammatory responses. Secondly, target cells are lysed by a hetero-oligomeric pore known as the membrane attack complex (MAC) that punctures the cellular membrane, causing ion and osmotic flux. Generally, several membrane-bound and soluble inhibitors protect the host membrane from complement damage. This includes inhibitors against the MAC, such as clusterin and CD59. This review addresses the most recent molecular and structural insights behind the activation and modulation of the integral membrane proteins, the C5a receptors (C5aR1 and C5aR2), as well as the regulation of MAC assembly. The second aspect of the review focuses on the molecular basis behind inflammatory diseases that are reflective of failure to regulate the terminal complement effectors. Although each arm is unique in its function, both pathways may share similar outcomes in these diseases. As such, the review outlines potential synergy and crosstalk between C5a receptor activation and MAC-mediated cellular responses.
Collapse
Affiliation(s)
- Bill H T Ho
- Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Bradley A Spicer
- Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Michelle A Dunstone
- Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
11
|
Baumert BO, Maretti-Mira AC, Walker DI, Li Z, Stratakis N, Wang H, Zhao Y, Fischer FC, Jia Q, Valvi D, Bartell SM, Chen C, Inge T, Ryder J, Jenkins T, Sisley S, Xanthakos S, Kleiner DE, Kohli R, Rock S, Eckel SP, La Merrill MA, Aung MM, Salomon MP, McConnell R, Goodrich J, Conti DV, Golden-Mason L, Chatzi L. Integrated Spheroid-to-Population Framework for Evaluating PFHpA-Associated Metabolic Dysfunction and Steatotic Liver Disease. RESEARCH SQUARE 2025:rs.3.rs-5960979. [PMID: 40092438 PMCID: PMC11908348 DOI: 10.21203/rs.3.rs-5960979/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The rising prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), particularly among pediatric populations, requires identification of modifiable risk factors to control disease progression. Per- and polyfluoroalkyl substances (PFAS) have emerged as potential contributors to liver damage; however, their role in the etiology of MASLD remains underexplored. This study aimed to bridge the gap between human epidemiological data and in vitro experimental findings to elucidate the effect of perfluoroheptanoic acid (PFHpA), a short chain, unregulated PFAS congener on MASLD development. Our analysis of the Teen-LABS cohort, a national multi-site study on obese adolescents undergoing bariatric surgery, revealed that doubling of PFHpA plasma levels was associated with an 80% increase in MASLD risk (OR, 1.8; 95% CI: 1.3-2.5) based on liver biospies. To further investigate the underlying mechanisms, we used 3D human liver spheroids and single-cell transcriptomics to assess the effect of PFHpA on hepatic metabolism. Integrative analysis identified dysregulation of common pathways in both human and spheroid models, particularly those involved in innate immunity, inflammation, and lipid metabolism. We applied the latent unknown clustering with integrated data (LUCID) model to assess associations between PFHpA exposure, multiomic signatures, and MASLD risk. Our results identified a proteome profile with significantly higher odds of MASLD (OR = 7.1), whereas a distinct metabolome profile was associated with lower odds (OR = 0.51), highlighting the critical role of protein dysregulation in disease pathogenesis. A translational framework was applied to uncover the molecular mechanisms of PFAS-induced MASLD in a cohort of obese adolescents. Identifying key molecular mechanisms for PFAS-induced MASLD can guide the development of targeted prevention and treatment.
Collapse
Affiliation(s)
- Brittney O. Baumert
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ana C. Maretti-Mira
- USC Research Center for Liver Diseases, Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Douglas I. Walker
- Gangarosa Department of Environmental Health, Rollins School of Public Health, 1518 Clifton Road, NE, Atlanta, GA, United States
| | - Zhenjiang Li
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Nikos Stratakis
- Barcelona Institute for Global Health, ISGlobal, Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Hongxu Wang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yinqi Zhao
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Fabian Christoph Fischer
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, United States
| | - Qiran Jia
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Damaskini Valvi
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Scott M. Bartell
- Department of Environmental and Occupational Health, University of California, Irvine, Irvine, CA, United States
| | - Carmen Chen
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Thomas Inge
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
| | - Justin Ryder
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
| | - Todd Jenkins
- Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Stephanie Sisley
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Stavra Xanthakos
- Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - David E. Kleiner
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, United States
| | - Rohit Kohli
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Sarah Rock
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Sandrah P. Eckel
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Michele A. La Merrill
- Department of Environmental Toxicology, University of California, Davis, CA, United States
| | - Max M. Aung
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Matthew P. Salomon
- USC Research Center for Liver Diseases, Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rob McConnell
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jesse Goodrich
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - David V. Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lucy Golden-Mason
- USC Research Center for Liver Diseases, Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lida Chatzi
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
12
|
Alsubaie N, Abd-Elhakim YM, Mohamed AAR, Khamis T, Metwally MMM, Helmi N, Alnajeebi AM, Alotaibi BS, Albaqami A, Mawkili W, Samak MA, Eissa SA. Exploring the CD3/CD56/TNF-α/Caspase3 pathway in pyrethroid-induced immune dysregulation: curcumin-loaded chitosan nanoparticle intervention. Front Pharmacol 2025; 16:1505432. [PMID: 39981186 PMCID: PMC11840570 DOI: 10.3389/fphar.2025.1505432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/09/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Conflict reports exist on the impact of pyrethroid insecticides on immune function and the probable underlying mechanisms. Methods This study evaluated the effect of an extensively used pyrethroid insecticide, fenpropathrin (FTN) (15 mg/kg b.wt), on the innate and humoral immune components, blood cells, splenic oxidative status, and mRNA expression of CD3, CD20, CD56, CD8, CD4, IL-6, TNF-α, and Caspase3 in a 60-day trial in rats. Besides, the possible defensive effect of curcumin-loaded chitosan nanoparticle (CML-CNP) (50 mg/kg b.wt) was evaluated. Results FTN exposure resulted in hypochromic normocytic anemia, thrombocytosis, leukocytosis, and lymphopenia. Besides, a significant reduction in IgG, not IgM, but increased C3 serum levels was evident in the FTN-exposed rats. Moreover, their splenic tissues displayed a substantial increase in the ROS, MDA, IL-6, and IL-1β content, altered splenic histology, and reduced GPX, GSH, and GSH/GSSG. Furthermore, a substantial upregulation of mRNA expression of splenic CD20, CD56, CD8, CD4, CD3, IL-6, and TNF-α, but downregulation of CD8 was detected in FTN-exposed rats. FTN exposure significantly upregulated splenic Caspase-3 and increased its immunohistochemical expression, along with elevated TNF-α immunoexpression. However, the alterations in immune function, splenic antioxidant status, blood cell populations, and immune-related gene expression were notably restored in the FTN + CML-CNP-treated group. Conclusion The findings of this study highlighted the immunosuppressive effects of FTN and suggested the involvement of many CD cell markers as a potential underlying mechanism. Additionally, the results demonstrated the effectiveness of CML-CNP in mitigating pollutant-induced immune disorders.
Collapse
Affiliation(s)
- Nawal Alsubaie
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Yasmina M. Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Amany Abdel-Rahman Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed M. M. Metwally
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, King Salman International University, Ras Sidr, Egypt
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Nawal Helmi
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Afnan M. Alnajeebi
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Badriyah S. Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Amirah Albaqami
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, Taif, Saudi Arabia
| | - Wedad Mawkili
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Mai A. Samak
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
- College of Medicine, University of Ha’il, Ha’il, Saudi Arabia
| | - Samar A. Eissa
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Kafrelsheikh University, Kafr ElSheikh, Egypt
| |
Collapse
|
13
|
Hickey TE, Mudunuri U, Hempel HA, Kemp TJ, Roche NV, Talsania K, Sellers BA, Cherry JM, Pinto LA. Proteomic and serologic assessments of responses to mRNA-1273 and BNT162b2 vaccines in human recipient sera. Front Immunol 2025; 15:1502458. [PMID: 39931577 PMCID: PMC11808009 DOI: 10.3389/fimmu.2024.1502458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 02/13/2025] Open
Abstract
Introduction The first vaccines approved against SARS-CoV-2, mRNA-1273 and BNT162b2, utilized mRNA platforms. However, little is known about the proteomic markers and pathways associated with host immune responses to mRNA vaccination. In this proof-of-concept study, sera from male and female vaccine recipients were evaluated for proteomic and immunologic responses 1-month and 6-months following homologous third vaccination. Methods An aptamer-based (7,289 marker) proteomic assay coupled with traditional serology was leveraged to generate a comprehensive evaluation of systemic responsiveness in 64 and 68 healthy recipients of mRNA-1273 and BNT162b2 vaccines, respectively. Results Sera from female recipients of mRNA-1273 showed upregulated indicators of inflammatory and immunological responses at 1-month post-third vaccination, and sera from female recipients of BNT162b2 demonstrated upregulated negative regulators of RNA sensors at 1-month. Sera from male recipients of mRNA-1273 showed no significant upregulation of pathways at 1-month post-third vaccination, though there were multiple significantly upregulated proteomic markers. Sera from male recipients of BNT162b2 demonstrated upregulated markers of immune response to doublestranded RNA and cell-cycle G(2)/M transition at 1-month. Random Forest analysis of proteomic data from pre-third-dose sera identified 85 markers used to develop a model predictive of robust or weaker IgG responses and antibody levels to SARS-CoV-2 spike protein at 6-months following boost; no specific markers were individually predictive of 6-month IgG response. Thirty markers that contributed most to the model were associated with complement cascade and activation; IL-17, TNFR pro-apoptotic, and PI3K signaling; and cell cycle progression. Discussion These results demonstrate the utility of proteomics to evaluate correlates or predictors of serological responses to SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Thomas E. Hickey
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Uma Mudunuri
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Heidi A. Hempel
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Troy J. Kemp
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Nancy V. Roche
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Keyur Talsania
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Brian A. Sellers
- Center for Human Immunology, Inflammation and Autoimmunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - James M. Cherry
- Center for Human Immunology, Inflammation and Autoimmunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Ligia A. Pinto
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| |
Collapse
|
14
|
Homan EA, Gilani A, Rubio-Navarro A, Johnson MA, Schaepkens OM, Cortada E, Pereira de Lima R, Stoll L, Lo JC. Complement 3a receptor 1 on macrophages and Kupffer cells is not required for the pathogenesis of metabolic dysfunction-associated steatotic liver disease. eLife 2025; 13:RP100708. [PMID: 39773465 PMCID: PMC11709426 DOI: 10.7554/elife.100708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
Together with obesity and type 2 diabetes, metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing global epidemic. Activation of the complement system and infiltration of macrophages has been linked to progression of metabolic liver disease. The role of complement receptors in macrophage activation and recruitment in MASLD remains poorly understood. In human and mouse, C3AR1 in the liver is expressed primarily in Kupffer cells, but is downregulated in humans with MASLD compared to obese controls. To test the role of complement 3a receptor (C3aR1) on macrophages and liver resident macrophages in MASLD, we generated mice deficient in C3aR1 on all macrophages (C3aR1-MφKO) or specifically in liver Kupffer cells (C3aR1-KpKO) and subjected them to a model of metabolic steatotic liver disease. We show that macrophages account for the vast majority of C3ar1 expression in the liver. Overall, C3aR1-MφKO and C3aR1-KpKO mice have similar body weight gain without significant alterations in glucose homeostasis, hepatic steatosis and fibrosis, compared to controls on a MASLD-inducing diet. This study demonstrates that C3aR1 deletion in macrophages or Kupffer cells, the predominant liver cell type expressing C3ar1, has no significant effect on liver steatosis, inflammation or fibrosis in a dietary MASLD model.
Collapse
Affiliation(s)
- Edwin A Homan
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Maya A Johnson
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Odin M Schaepkens
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Eric Cortada
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Renan Pereira de Lima
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Lisa Stoll
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - James C Lo
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| |
Collapse
|
15
|
Angius F, Puxeddu S, Zaimi S, Canton S, Nematollahzadeh S, Pibiri A, Delogu I, Alvisi G, Moi ML, Manzin A. SARS-CoV-2 Evolution: Implications for Diagnosis, Treatment, Vaccine Effectiveness and Development. Vaccines (Basel) 2024; 13:17. [PMID: 39852796 PMCID: PMC11769326 DOI: 10.3390/vaccines13010017] [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: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
The COVID-19 pandemic, driven by the rapid evolution of the SARS-CoV-2 virus, presents ongoing challenges to global public health. SARS-CoV-2 is characterized by rapidly evolving mutations, especially in (but not limited to) the spike protein, complicating predictions about its evolutionary trajectory. These mutations have significantly affected transmissibility, immune evasion, and vaccine efficacy, leading to multiple pandemic waves with over half a billion cases and seven million deaths globally. Despite several strategies, from rapid vaccine development and administration to the design and availability of antivirals, including monoclonal antibodies, already having been employed, the persistent circulation of the virus and the emergence of new variants continue to result in high case numbers and fatalities. In the past four years, immense research efforts have contributed much to our understanding of the viral pathogenesis mechanism, the COVID-19 syndrome, and the host-microbe interactions, leading to the development of effective vaccines, diagnostic tools, and treatments. The focus of this review is to provide a comprehensive analysis of the functional impact of mutations on diagnosis, treatments, and vaccine effectiveness. We further discuss vaccine safety in pregnancy and the implications of hybrid immunity on long-term protection against infection, as well as the latest developments on a pan-coronavirus vaccine and nasal formulations, emphasizing the need for continued surveillance, research, and adaptive public health strategies in response to the ongoing SARS-CoV-2 evolution race.
Collapse
Affiliation(s)
- Fabrizio Angius
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvia Puxeddu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvio Zaimi
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Serena Canton
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Sepehr Nematollahzadeh
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Andrea Pibiri
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Ilenia Delogu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Gualtiero Alvisi
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Aldo Manzin
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| |
Collapse
|
16
|
Crosson T, Bhat S, Wang JC, Salaun C, Fontaine E, Roversi K, Herzog H, Rafei M, Blunck R, Talbot S. Cytokines reprogram airway sensory neurons in asthma. Cell Rep 2024; 43:115045. [PMID: 39661516 DOI: 10.1016/j.celrep.2024.115045] [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: 07/30/2024] [Revised: 10/01/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
Nociceptor neurons play a crucial role in maintaining the body's homeostasis by detecting and responding to potential environmental dangers. However, this function can be detrimental during allergic reactions, as vagal nociceptors contribute to immune cell infiltration, bronchial hypersensitivity, and mucus imbalance in addition to causing pain and coughing. Despite this, the specific mechanisms by which nociceptors acquire pro-inflammatory characteristics during allergic reactions are not yet fully understood. In this study, we investigate the changes in the molecular profile of airway nociceptor neurons during allergic airway inflammation and identify the signals driving such reprogramming. Using retrograde tracing and lineage reporting, we identify a specific class of inflammatory vagal nociceptor neurons that exclusively innervate the airways. In the ovalbumin mouse model of allergic airway inflammation, these neurons undergo significant reprogramming characterized by the upregulation of the neuropeptide Y (NPY) receptor Npy1r. A screening of cytokines and neurotrophins reveals that interleukin 1β (IL-1β), IL-13, and brain-derived neurotrophic factor (BDNF) drive part of this reprogramming. IL-13 triggers Npy1r overexpression in nociceptors via the JAK/STAT6 pathway. In parallel, NPY is released into the bronchoalveolar fluid of asthmatic mice, which limits the excitability of nociceptor neurons. Single-cell RNA sequencing of lung immune cells reveals that a cell-specific knockout of NPY1R in nociceptor neurons in asthmatic mice altered T cell infiltration. Opposite findings are observed in asthmatic mice in which nociceptor neurons are chemically ablated. In summary, allergic airway inflammation reprograms airway nociceptor neurons to acquire a pro-inflammatory phenotype, while a compensatory mechanism involving NPY1R limits the activity of nociceptor neurons.
Collapse
Affiliation(s)
- Theo Crosson
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Shreyas Bhat
- Centre Interdisciplinaire sur le Cerveau et l'Apprentissage, Université de Montréal, Montreal, QC, Canada; Département de Physique, Université de Montréal, Montreal, QC, Canada
| | - Jo-Chiao Wang
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Clara Salaun
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Eleanne Fontaine
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Katiane Roversi
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Herbert Herzog
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Moutih Rafei
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Rikard Blunck
- Centre Interdisciplinaire sur le Cerveau et l'Apprentissage, Université de Montréal, Montreal, QC, Canada; Département de Physique, Université de Montréal, Montreal, QC, Canada
| | - Sebastien Talbot
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
17
|
DeVaughn H, Rich HE, Shadid A, Vaidya PK, Doursout MF, Shivshankar P. Complement Immune System in Pulmonary Hypertension-Cooperating Roles of Circadian Rhythmicity in Complement-Mediated Vascular Pathology. Int J Mol Sci 2024; 25:12823. [PMID: 39684535 PMCID: PMC11641342 DOI: 10.3390/ijms252312823] [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: 10/28/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Originally discovered in the 1890s, the complement system has traditionally been viewed as a "compliment" to the body's innate and adaptive immune response. However, emerging data have shown that the complement system is a much more complex mechanism within the body involved in regulating inflammation, gene transcription, attraction of macrophages, and many more processes. Sustained complement activation contributes to autoimmunity and chronic inflammation. Pulmonary hypertension is a disease with a poor prognosis and an average life expectancy of 2-3 years that leads to vascular remodeling of the pulmonary arteries; the pulmonary arteries are essential to host homeostasis, as they divert deoxygenated blood from the right ventricle of the heart to the lungs for gas exchange. This review focuses on direct links between the complement system's involvement in pulmonary hypertension, along with autoimmune conditions, and the reliance on the complement system for vascular remodeling processes of the pulmonary artery. Furthermore, circadian rhythmicity is highlighted as the disrupted homeostatic mechanism in the inflammatory consequences in the vascular remodeling within the pulmonary arteries, which could potentially open new therapeutic cues. The current treatment options for pulmonary hypertension are discussed with clinical trials using complement inhibitors and potential therapeutic targets that impact immune cell functions and complement activation, which could alleviate symptoms and block the progression of the disease. Further research on complement's involvement in interstitial lung diseases and pulmonary hypertension could prove beneficial for our understanding of these various diseases and potential treatment options to prevent vascular remodeling of the pulmonary arteries.
Collapse
Affiliation(s)
- Hunter DeVaughn
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA; (H.D.); (H.E.R.); (A.S.); (P.K.V.)
- Center for Immunology and Autoimmune Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA
| | - Haydn E. Rich
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA; (H.D.); (H.E.R.); (A.S.); (P.K.V.)
| | - Anthony Shadid
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA; (H.D.); (H.E.R.); (A.S.); (P.K.V.)
| | - Priyanka K. Vaidya
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA; (H.D.); (H.E.R.); (A.S.); (P.K.V.)
| | - Marie-Francoise Doursout
- Department of Anesthesiology, Critical Care and Pain Medicine, UTHealth-McGovern Medical School, Houston, TX 77030, USA;
| | - Pooja Shivshankar
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA; (H.D.); (H.E.R.); (A.S.); (P.K.V.)
- Center for Immunology and Autoimmune Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX 77030, USA
| |
Collapse
|
18
|
Oakes A, Liu Y, Dubielecka PM. Complement or insult: the emerging link between complement cascade deficiencies and pathology of myeloid malignancies. J Leukoc Biol 2024; 116:966-984. [PMID: 38836653 PMCID: PMC11531810 DOI: 10.1093/jleuko/qiae130] [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: 02/29/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
The complement cascade is an ancient and highly conserved arm of the immune system. The accumulating evidence highlights elevated activity of the complement cascade in cancer microenvironment and emphasizes its effects on the immune, cancer, and cancer stroma cells, pointing to a role in inflammation-mediated etiology of neoplasms. The role the cascade plays in development, progression, and relapse of solid tumors is increasingly recognized, however its role in hematological malignancies, especially those of myeloid origin, has not been thoroughly assessed and remains obscure. As the role of inflammation and autoimmunity in development of myeloid malignancies is becoming recognized, in this review we focus on summarizing the links that have been identified so far for complement cascade involvement in the pathobiology of myeloid malignancies. Complement deficiencies are primary immunodeficiencies that cause an array of clinical outcomes including an increased risk of a range of infectious as well as local or systemic inflammatory and thrombotic conditions. Here, we discuss the impact that deficiencies in complement cascade initiators, mid- and terminal-components and inhibitors have on the biology of myeloid neoplasms. The emergent conclusions indicate that the links between complement cascade, inflammatory signaling, and the homeostasis of hematopoietic system exist, and efforts should continue to detail the mechanistic involvement of complement cascade in the development and progression of myeloid cancers.
Collapse
Affiliation(s)
- Alissa Oakes
- Department of Medicine, Alpert Medical School, Brown University, 69 Brown St, Providence, RI 02906, USA
- Division of Hematology/Oncology, Rhode Island Hospital, 69 Brown St, Providence, RI 02906, USA
- Therapeutic Sciences Graduate program, Brown University, 69 Brown St, Providence, RI 02906, USA
| | - Yuchen Liu
- Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, 22. S. Greene St., Baltimore, MD 21201-1595, USA
| | - Patrycja M Dubielecka
- Department of Medicine, Alpert Medical School, Brown University, 69 Brown St, Providence, RI 02906, USA
- Division of Hematology/Oncology, Rhode Island Hospital, 69 Brown St, Providence, RI 02906, USA
- Therapeutic Sciences Graduate program, Brown University, 69 Brown St, Providence, RI 02906, USA
- Legorreta Cancer Center, Brown University, One Hoppin St., Coro West, Suite 5.01, Providence, RI 02903, USA
| |
Collapse
|
19
|
Domingos IF, Carvalho LB, Lodeiro C, Gerivaz R, Prag G, Micaglio E, Muchtar E, Santos HM, Capelo JL. Dithiothreitol-based protein equalisation in the context of multiple myeloma: Enhancing proteomic analysis and therapeutic insights. Talanta 2024; 279:126589. [PMID: 39116730 DOI: 10.1016/j.talanta.2024.126589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/10/2024]
Abstract
In this study, we employed the dithiothreitol-based protein equalisation technique and analytical proteomics to better understand myeloma diseases by comparing the proteomes of pellets and supernatants formed upon application of DTT on serum samples. The number of unique proteins found in pellets was 252 for healthy individuals and 223 for multiple myeloma patients. The comparison of these proteomes showed 97 dysregulated proteins. The unique proteins found for supernatants were 264 for healthy individuals and 235 for multiple myeloma patients. The comparison of these proteomes showed 87 dysregulated proteins. The analytical proteomic comparison of both groups of dysregulated proteins is translated into parallel dysregulated pathways, including chaperone-mediated autophagy and protein folding, addressing potential therapeutic interventions. Future research endeavours in personalised medicine should prioritize refining analytical proteomic methodologies using serum dithiothreitol-based protein equalisation to explore innovative therapeutic strategies. We highlight the advanced insights gained from this analytical strategy in studying multiple myeloma, emphasising its complex nature and the critical role of personalised, targeted analytical techniques in enhancing therapeutic efficacy in personalised medicine.
Collapse
Affiliation(s)
- Ines F Domingos
- BIOSCOPE Research Group, LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; PROTEOMASS Scientific Society, Praceta Jerónimo Dias, 2825-466., Caparica, Portugal
| | - Luis B Carvalho
- BIOSCOPE Research Group, LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; PROTEOMASS Scientific Society, Praceta Jerónimo Dias, 2825-466., Caparica, Portugal
| | - Carlos Lodeiro
- BIOSCOPE Research Group, LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; PROTEOMASS Scientific Society, Praceta Jerónimo Dias, 2825-466., Caparica, Portugal
| | - Rita Gerivaz
- Serviço de Hematologia, Hospital Garcia de Orta, Almada, Portugal
| | - Gali Prag
- School of Neurobiology, Biochemistry and Biophysics, the George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Israel
| | - Emanuele Micaglio
- Department of Arrhythmology, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097, Milan, Italy
| | - Eli Muchtar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Hugo M Santos
- BIOSCOPE Research Group, LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; PROTEOMASS Scientific Society, Praceta Jerónimo Dias, 2825-466., Caparica, Portugal; Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| | - Jose L Capelo
- BIOSCOPE Research Group, LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; PROTEOMASS Scientific Society, Praceta Jerónimo Dias, 2825-466., Caparica, Portugal.
| |
Collapse
|
20
|
Stocker BW, LaCroix IS, Erickson C, Gallagher LT, Ramser BJ, Thielen O, Hallas W, Mitra S, Moore EE, Hansen K, D'Alessandro A, Silliman CC, Cohen MJ. Trauma patients with type O blood exhibit unique multiomics signature with decreased lectin pathway of complement levels. J Trauma Acute Care Surg 2024; 97:753-763. [PMID: 38745347 PMCID: PMC11502284 DOI: 10.1097/ta.0000000000004367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
BACKGROUND Patients with type O blood may have an increased risk of hemorrhagic complications because of lower baseline levels of von Willebrand factor and factor VIII, but the transition to a mortality difference in trauma is less clear. We hypothesized that type O trauma patients will have differential proteomic and metabolomic signatures in response to trauma beyond von Willebrand factor and factor VIII alone. METHODS Patients meeting the highest level of trauma activation criteria were prospectively enrolled. Blood samples were collected upon arrival to the emergency department. Proteomic and metabolomic (multiomics) analyses of these samples were performed using liquid chromatography-mass spectrometry. Demographic, clinical, and multiomics data were compared between patients with type O blood versus all other patients. RESULTS There were 288 patients with multiomics data; 146 (51%) had type O blood. Demographics, injury patterns, and initial vital signs and laboratory measurements were not different between groups. Type O patients had increased lengths of stay (7 vs. 6 days, p = 0.041) and a trend toward decreased mortality secondary to traumatic brain injury compared with other causes (traumatic brain injury, 44.4% vs. 87.5%; p = 0.055). Type O patients had decreased levels of mannose-binding lectin and mannose-binding lectin-associated serine proteases 1 and 2, which are required for the initiation of the lectin pathway of complement activation. Type O patients also had metabolite differences signifying energy metabolism and mitochondrial dysfunction. CONCLUSION Blood type O patients have a unique multiomics signature, including decreased levels of proteins required to activate the lectin complement pathway. This may lead to overall decreased levels of complement activation and decreased systemic inflammation in the acute phase, possibly leading to a survival advantage, especially in traumatic brain injury. However, this may later impair healing. Future work will need to confirm these associations, and animal studies are needed to test therapeutic targets. LEVEL OF EVIDENCE Prognostic and Epidemiological; Level IV.
Collapse
Affiliation(s)
- Benjamin W Stocker
- From the Department of Surgery (B.W.S., L.T.G., B.J.R., O.T., W.H., S.M., E.E.M., C.C.S., M.J.C.), and Department of Biochemistry and Molecular Genetics (I.S.L., C.E., K.H., A.D.), School of Medicine, University of Colorado Anschutz Medical Campus, Aurora; Department of Surgery (E.E.M.), Ernest E Moore Shock Trauma Center, Denver Health Medical Center; Vitalant Research Institute (C.C.S.), Denver; and Department of Pediatrics (C.C.S.), School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
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
|
22
|
Homan EA, Gilani A, Rubio-Navarro A, Johnson MA, Schaepkens OM, Cortada E, de Lima RP, Stoll L, Lo JC. Complement 3a Receptor 1 on Macrophages and Kupffer cells is not required for the Pathogenesis of Metabolic Dysfunction-Associated Steatotic Liver Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.26.24309550. [PMID: 38978661 PMCID: PMC11230319 DOI: 10.1101/2024.06.26.24309550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Together with obesity and type 2 diabetes, metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing global epidemic. Activation of the complement system and infiltration of macrophages has been linked to progression of metabolic liver disease. The role of complement receptors in macrophage activation and recruitment in MASLD remains poorly understood. In human and mouse, C3AR1 in the liver is expressed primarily in Kupffer cells, but is downregulated in humans with MASLD compared to obese controls. To test the role of complement 3a receptor (C3aR1) on macrophages and liver resident macrophages in MASLD, we generated mice deficient in C3aR1 on all macrophages (C3aR1-MφKO) or specifically in liver Kupffer cells (C3aR1-KpKO) and subjected them to a model of metabolic steatotic liver disease. We show that macrophages account for the vast majority of C3ar1 expression in the liver. Overall, C3aR1-MφKO and C3aR1-KpKO mice have similar body weight gain without significant alterations in glucose homeostasis, hepatic steatosis and fibrosis, compared to controls on a MASLD-inducing diet. This study demonstrates that C3aR1 deletion in macrophages or Kupffer cells, the predominant liver cell type expressing C3aR1, has no significant effect on liver steatosis, inflammation or fibrosis in a dietary MASLD model.
Collapse
Affiliation(s)
- Edwin A. Homan
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Maya A. Johnson
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Odin M. Schaepkens
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Eric Cortada
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Renan Pereira de Lima
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Lisa Stoll
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - James C. Lo
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| |
Collapse
|
23
|
Crosson T, Bhat S, Wang JC, Salaun C, Fontaine E, Roversi K, Herzog H, Rafei M, Blunck R, Talbot S. Cytokines reprogram airway sensory neurons in asthma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.26.525731. [PMID: 39345572 PMCID: PMC11429693 DOI: 10.1101/2023.01.26.525731] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Nociceptor neurons play a crucial role in maintaining the body's homeostasis by detecting and responding to potential dangers in the environment. However, this function can be detrimental during allergic reactions, since vagal nociceptors can contribute to immune cell infiltration, bronchial hypersensitivity, and mucus imbalance, in addition to causing pain and coughing. Despite this, the specific mechanisms by which nociceptors acquire pro-inflammatory characteristics during allergic reactions are not yet fully understood. In this study, we aimed to investigate the molecular profile of airway nociceptor neurons during allergic airway inflammation and identify the signals driving such reprogramming. Using retrograde tracing and lineage reporting, we identified a unique class of inflammatory vagal nociceptor neurons that exclusively innervate the airways. In the ovalbumin mouse model of airway inflammation, these neurons undergo significant reprogramming characterized by the upregulation of the NPY receptor Npy1r. A screening of cytokines and neurotrophins revealed that IL-1β, IL-13 and BDNF drive part of this reprogramming. IL-13 triggered Npy1r overexpression in nociceptors via the JAK/STAT6 pathway. In parallel, sympathetic neurons and macrophages release NPY in the bronchoalveolar fluid of asthmatic mice, which limits the excitability of nociceptor neurons. Single-cell RNA sequencing of lung immune cells has revealed that a cell-specific knockout of Npy1r in nociceptor neurons in asthmatic mice leads to an increase in airway inflammation mediated by T cells. Opposite findings were observed in asthmatic mice in which nociceptor neurons were chemically ablated. In summary, allergic airway inflammation reprograms airway nociceptor neurons to acquire a pro-inflammatory phenotype, while a compensatory mechanism involving NPY1R limits nociceptor neurons' activity.
Collapse
Affiliation(s)
- Théo Crosson
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Shreyas Bhat
- Centre Interdisciplinaire sur le Cerveau et l’Apprentissage, Université de Montréal, Canada
- Département de Physique, Université de Montréal, Canada
| | - Jo-Chiao Wang
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Clara Salaun
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Eleanne Fontaine
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Katiane Roversi
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | | | - Moutih Rafei
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Rikard Blunck
- Centre Interdisciplinaire sur le Cerveau et l’Apprentissage, Université de Montréal, Canada
- Département de Physique, Université de Montréal, Canada
| | - Sebastien Talbot
- Department of Physiology and Pharmacology, Karolinska Institutet. Sweden
- Department of Biomedical and Molecular Sciences, Queen’s University. Canada
| |
Collapse
|
24
|
Lujan E, Zhang I, Garon AC, Liu F. The Interactions of the Complement System with Human Cytomegalovirus. Viruses 2024; 16:1171. [PMID: 39066333 PMCID: PMC11281448 DOI: 10.3390/v16071171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/02/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The complement system is an evolutionarily ancient component of innate immunity that serves as an important first line of defense against pathogens, including viruses. In response to infection, the complement system can be activated by three distinct yet converging pathways (classical, lectin, and alternative) capable of engaging multiple antiviral host responses to confront acute, chronic, and recurrent viral infections. Complement can exert profound antiviral effects via multiple mechanisms including the induction of inflammation and chemotaxis to sites of infection, neutralization/opsonization of viruses and virally infected cells, and it can even shape adaptive immune responses. With millions of years of co-evolution and the ability to establish life-long infections, herpesviruses have evolved unique mechanisms to counter complement-mediated antiviral defenses, thus enabling their survival and replication within humans. This review aims to comprehensively summarize how human herpesviruses engage with the complement system and highlight our understanding of the role of complement in human cytomegalovirus (HCMV) infection, immunity, and viral replication. Herein we describe the novel and unorthodox roles of complement proteins beyond their roles in innate immunity and discuss gaps in knowledge and future directions of complement and HCMV research.
Collapse
Affiliation(s)
- Eduardo Lujan
- Program in Comparative Biochemistry, University of California, Berkeley, CA 94720, USA
| | - Isadora Zhang
- School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Andrea Canto Garon
- Program in Comparative Biochemistry, University of California, Berkeley, CA 94720, USA
| | - Fenyong Liu
- Program in Comparative Biochemistry, University of California, Berkeley, CA 94720, USA
- School of Public Health, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
25
|
Tobias J, Steinberger P, Wilkinson J, Klais G, Kundi M, Wiedermann U. SARS-CoV-2 Vaccines: The Advantage of Mucosal Vaccine Delivery and Local Immunity. Vaccines (Basel) 2024; 12:795. [PMID: 39066432 PMCID: PMC11281395 DOI: 10.3390/vaccines12070795] [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: 06/12/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Immunity against respiratory pathogens is often short-term, and, consequently, there is an unmet need for the effective prevention of such infections. One such infectious disease is coronavirus disease 19 (COVID-19), which is caused by the novel Beta coronavirus SARS-CoV-2 that emerged around the end of 2019. The World Health Organization declared the illness a pandemic on 11 March 2020, and since then it has killed or sickened millions of people globally. The development of COVID-19 systemic vaccines, which impressively led to a significant reduction in disease severity, hospitalization, and mortality, contained the pandemic's expansion. However, these vaccines have not been able to stop the virus from spreading because of the restricted development of mucosal immunity. As a result, breakthrough infections have frequently occurred, and new strains of the virus have been emerging. Furthermore, SARS-CoV-2 will likely continue to circulate and, like the influenza virus, co-exist with humans. The upper respiratory tract and nasal cavity are the primary sites of SARS-CoV-2 infection and, thus, a mucosal/nasal vaccination to induce a mucosal response and stop the virus' transmission is warranted. In this review, we present the status of the systemic vaccines, both the approved mucosal vaccines and those under evaluation in clinical trials. Furthermore, we present our approach of a B-cell peptide-based vaccination applied by a prime-boost schedule to elicit both systemic and mucosal immunity.
Collapse
Affiliation(s)
- Joshua Tobias
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Joy Wilkinson
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gloria Klais
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Kundi
- Department of Environmental Health, Center for Public Health, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
26
|
Jiang F, Lei C, Chen Y, Zhou N, Zhang M. The complement system and diabetic retinopathy. Surv Ophthalmol 2024; 69:575-584. [PMID: 38401574 DOI: 10.1016/j.survophthal.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
Diabetic retinopathy (DR) is one of the common microvascular complications of diabetes mellitus and is the main cause of visual impairment in diabetic patients. The pathogenesis of DR is still unclear. The complement system, as an important component of the innate immune system in addition to defending against the invasion of foreign microorganisms, is involved in the occurrence and development of DR through 3 widely recognized complement activation pathways, the complement regulatory system, and many other pathways. Molecules such as C3a, C5a, and membrane attacking complex, as important molecules of the complement system, are involved in the pathologenesus of DR, either through direct damaging effects or by activating cells (microglia, macroglia, etc.) in the retinal microenvironment to contribute to the pathological damage of DR indirectly. We review the integral association of the complement system and DR to further understand the pathogenesis of DR and possibly provide a new strategy for itstreatment.
Collapse
Affiliation(s)
- Feipeng Jiang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Macular Disease Research Laboratory, West China Hospital, Sichuan University, China
| | - Chunyan Lei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Macular Disease Research Laboratory, West China Hospital, Sichuan University, China
| | - Yingying Chen
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Macular Disease Research Laboratory, West China Hospital, Sichuan University, China
| | - Nenghua Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Meixia Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China; Macular Disease Research Laboratory, West China Hospital, Sichuan University, China.
| |
Collapse
|
27
|
Silawal S, Gesslein M, Willauschus M, Schulze-Tanzil G. In Vitro Investigation of Pulsed Electromagnetic Field Stimulation (PEMF) with MAGCELL ® ARTHRO on the Regulatory Expression of Soluble and Membrane-Bound Complement Factors and Inflammatory Cytokines in Immortalized Synovial Fibroblasts. J Pers Med 2024; 14:701. [PMID: 39063955 PMCID: PMC11277808 DOI: 10.3390/jpm14070701] [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: 05/24/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Pulsed electromagnetic field stimulation (PEMF) is gaining more attention as a non-invasive arthritis treatment. In our study, immortalized synovial fibroblasts (K4IM) derived from a non-arthritic donor were exposed to MAGCELL® ARTHRO, a PEMF device, with 105 mT intensity, 8 Hz frequency, and 2 × 2.5 min sessions conducted thrice with a 1 h interval, to understand the underlying mechanism in regard to the complement system. Additionally, tumor necrosis factor (TNFα, 10 ng/mL) pre-treatment prior to PEMF stimulation, as well as 3-day versus 6-day stimulation, were compared. Gene expression of C4b binding protein-alpha and -beta (C4BPα, C4BPβ), complement factor (CF)-H, CFI, CD55, CD59, Interleukin (IL-6) and TNFα was analyzed. Immunofluorescence staining of CD55, CD59, and Ki67 was conducted. Results showed the absence of C4BPα gene expression, but C4BPβ was present. One and three days of PEMF stimulation caused no significant changes. However, after six days, there was a significant increase in CD55, CFH, and CD59 gene expression, indicating cytoprotective effects. Conversely, IL-6 gene expression increased after six days of stimulation and even after a single session in TNFα pre-stimulated cells, indicating a pro-inflammatory effect. PEMF's ambivalent, i.e., enhancing complement regulatory proteins and pro-inflammatory cytokines, highlights its complexity at the molecular level.
Collapse
Affiliation(s)
- Sandeep Silawal
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany;
| | - Markus Gesslein
- Department of Orthopedics and Traumatology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Breslauer Str. 201, 90471 Nuremberg, Germany; (M.G.); (M.W.)
| | - Maximilian Willauschus
- Department of Orthopedics and Traumatology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Breslauer Str. 201, 90471 Nuremberg, Germany; (M.G.); (M.W.)
| | - Gundula Schulze-Tanzil
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany;
| |
Collapse
|
28
|
Lim JJ, Goedken M, Jin Y, Gu H, Cui JY. Single-cell transcriptomics unveiled that early life BDE-99 exposure reprogrammed the gut-liver axis to promote a proinflammatory metabolic signature in male mice at late adulthood. Toxicol Sci 2024; 200:114-136. [PMID: 38648751 PMCID: PMC11199921 DOI: 10.1093/toxsci/kfae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are legacy flame retardants that bioaccumulate in the environment. The gut microbiome is an important regulator of liver functions including xenobiotic biotransformation and immune regulation. We recently showed that neonatal exposure to polybrominated diphenyl ether-99 (BDE-99), a human breast milk-enriched PBDE congener, up-regulated proinflammation-related and down-regulated drug metabolism-related genes predominantly in males in young adulthood. However, the persistence of this dysregulation into late adulthood, differential impact among hepatic cell types, and the involvement of the gut microbiome from neonatal BDE-99 exposure remain unknown. To address these knowledge gaps, male C57BL/6 mouse pups were orally exposed to corn oil (10 ml/kg) or BDE-99 (57 mg/kg) once daily from postnatal days 2-4. At 15 months of age, neonatal BDE-99 exposure down-regulated xenobiotic and lipid-metabolizing enzymes and up-regulated genes involved in microbial influx in hepatocytes. Neonatal BDE-99 exposure also increased the hepatic proportion of neutrophils and led to a predicted increase of macrophage migration inhibitory factor signaling. This was associated with decreased intestinal tight junction protein (Tjp) transcripts, altered gut environment, and dysregulation of inflammation-related metabolites. ScRNA-seq using germ-free (GF) mice demonstrated the necessity of a normal gut microbiome in maintaining hepatic immune tolerance. Microbiota transplant to GF mice using large intestinal microbiome from adults neonatally exposed to BDE-99 down-regulated Tjp transcripts and up-regulated several cytokines in large intestine. In conclusion, neonatal BDE-99 exposure reprogrammed cell type-specific gene expression and cell-cell communication in liver towards proinflammation, and this may be partly due to the dysregulated gut environment.
Collapse
Affiliation(s)
- Joe Jongpyo Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105, USA
- Environmental Health and Microbiome Research Center (EHMBRACE), Seattle, Washington 98105, USA
| | - Michael Goedken
- Rutgers Research Pathology Services, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Yan Jin
- Center for Translational Science, Florida International University, Port St Lucie, Florida 34987, USA
| | - Haiwei Gu
- Center for Translational Science, Florida International University, Port St Lucie, Florida 34987, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105, USA
- Environmental Health and Microbiome Research Center (EHMBRACE), Seattle, Washington 98105, USA
| |
Collapse
|
29
|
Hassan N, Krieg T, Kopp A, Bach AD, Kröger N. Challenges and Pitfalls of Research Designs Involving Magnesium-Based Biomaterials: An Overview. Int J Mol Sci 2024; 25:6242. [PMID: 38892430 PMCID: PMC11172609 DOI: 10.3390/ijms25116242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Magnesium-based biomaterials hold remarkable promise for various clinical applications, offering advantages such as reduced stress-shielding and enhanced bone strengthening and vascular remodeling compared to traditional materials. However, ensuring the quality of preclinical research is crucial for the development of these implants. To achieve implant success, an understanding of the cellular responses post-implantation, proper model selection, and good study design are crucial. There are several challenges to reaching a safe and effective translation of laboratory findings into clinical practice. The utilization of Mg-based biomedical devices eliminates the need for biomaterial removal surgery post-healing and mitigates adverse effects associated with permanent biomaterial implantation. However, the high corrosion rate of Mg-based implants poses challenges such as unexpected degradation, structural failure, hydrogen evolution, alkalization, and cytotoxicity. The biocompatibility and degradability of materials based on magnesium have been studied by many researchers in vitro; however, evaluations addressing the impact of the material in vivo still need to be improved. Several animal models, including rats, rabbits, dogs, and pigs, have been explored to assess the potential of magnesium-based materials. Moreover, strategies such as alloying and coating have been identified to enhance the degradation rate of magnesium-based materials in vivo to transform these challenges into opportunities. This review aims to explore the utilization of Mg implants across various biomedical applications within cellular (in vitro) and animal (in vivo) models.
Collapse
Affiliation(s)
- Nourhan Hassan
- Department of Plastic, Reconstructive and Aesthetic Surgery, University Hospital Cologne, 50937 Cologne, Germany
- Institute for Laboratory Animal Science and Experimental Surgery, University of Aachen Medical Center, Faculty of Medicine, RWTH-Aachen University, 52074 Aachen, Germany
- Biotechnology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Thomas Krieg
- Translational Matrix Biology, Medical Faculty, University of Cologne, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, 50937 Cologne, Germany
| | | | - Alexander D. Bach
- Department of Plastic, Aesthetic and Hand Surgery, St. Antonius Hospital Eschweiler, 52249 Eschweiler, Germany
| | - Nadja Kröger
- Institute for Laboratory Animal Science and Experimental Surgery, University of Aachen Medical Center, Faculty of Medicine, RWTH-Aachen University, 52074 Aachen, Germany
- Department of Plastic, Aesthetic and Hand Surgery, St. Antonius Hospital Eschweiler, 52249 Eschweiler, Germany
| |
Collapse
|
30
|
Wei X, Tu Y, Bu S, Guo G, Wang H, Wang Z. Unraveling the Intricate Web: Complement Activation Shapes the Pathogenesis of Sepsis-Induced Coagulopathy. J Innate Immun 2024; 16:337-353. [PMID: 38815564 PMCID: PMC11249610 DOI: 10.1159/000539502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Sepsis-associated coagulopathy specifically refers to widespread systemic coagulation activation accompanied by a high risk of hemorrhage and organ damage, which in severe cases manifests as disseminated intravascular coagulation (DIC), or even develops into multiple organ dysfunction syndrome (MODS). The complement system and the coagulation system as the main columns of innate immunity and hemostasis, respectively, undergo substantial activation after sepsis. SUMMARY Dysfunction of the complement, coagulation/fibrinolytic cascades caused by sepsis leads to "thromboinflammation," which ultimately amplifies the systemic inflammatory response and accelerates the development of MODS. Recent studies have revealed that massive activation of the complement system exacerbates sepsis-induced coagulation and even results in DIC, which suggests that inhibition of complement activation may have therapeutic potential in the treatment of septic coagulopathy. KEY MESSAGES Sepsis-associated thrombosis involves the upregulation or activation of procoagulant factors, down-regulation or inactivation of anticoagulant factors, and impairment of the fibrinolytic mechanism. This review aims to summarize the latest literature and analyze the underlying molecular mechanisms of the activation of the complement system on the abnormal coagulation cascades in sepsis.
Collapse
Affiliation(s)
- Xin Wei
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ye Tu
- Department of Pharmacy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shuhong Bu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guimei Guo
- Department of Pediatric Nephrology and Rheumatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongbin Wang
- Master Program of Pharmaceutical Scieneces College of Graduate Studies, Department of Pharmaceutical and Biomedical Sciences College of Pharmacy, Department of Basic Science College of Medicine, California Northstate University, Elk Grove, CA, USA
| | - Zhibin Wang
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China
| |
Collapse
|
31
|
Khurana N, Watkins K, Ghatak D, Staples J, Hubbard O, Yellepeddi V, Watt K, Ghandehari H. Reducing hydrophobic drug adsorption in an in-vitro extracorporeal membrane oxygenation model. Eur J Pharm Biopharm 2024; 198:114261. [PMID: 38490349 PMCID: PMC11186434 DOI: 10.1016/j.ejpb.2024.114261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/04/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Extracorporeal membrane oxygenation (ECMO) is a life-saving cardiopulmonary bypass technology for critically ill patients with heart and lung failure. Patients treated with ECMO receive a range of drugs that are used to treat underlying diseases and critical illnesses. However, the dosing guidelines for these drugs used in ECMO patients are unclear. Mortality rate for patients on ECMO exceeds 40% partly due to inaccurate dosing information, caused in part by the adsorption of drugs in the ECMO circuit and its components. These drugs range in hydrophobicity, electrostatic interactions, and pharmacokinetics. Propofol is commonly administered to ECMO patients and is known to have high adsorption rates to the circuit components due to its hydrophobicity. To reduce adsorption onto the circuit components, we used micellar block copolymers (Poloxamer 188TM and Poloxamer 407TM) and liposomes tethered with poly(ethylene glycol) to encapsulate propofol, provide a hydrophilic shell and prevent its adsorption. Size, polydispersity index (PDI), and zeta potential of the delivery systems were characterized by dynamic light scattering, and encapsulation efficiency was characterized using High Performance Liquid Chromatography (HPLC). All delivery systems used demonstrated colloidal stability at physiological conditions for seven days, cytocompatibility with a human leukemia monocytic cell line, i.e., THP-1 cells, and did not activate the complement pathway in human plasma. We demonstrated a significant reduction in adsorption of propofol in an in-vitro ECMO model upon encapsulation in micelles and liposomes. These results show promise in reducing the adsorption of hydrophobic drugs to the ECMO circuits by encapsulation in nanoscale structures tethered with hydrophilic polymers on the surface.
Collapse
Affiliation(s)
- Nitish Khurana
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA; Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Kamiya Watkins
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA
| | - Debika Ghatak
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA
| | - Jane Staples
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA
| | - Oliver Hubbard
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Venkata Yellepeddi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA; Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Kevin Watt
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA; Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Hamidreza Ghandehari
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
32
|
Rudraprasad D, K V, Nirmal J, Ali MH, Joseph J. Complement Cascade 8 - Alpha and Calpain-2 in Extracellular Vesicles of Human Vitreous as Biomarkers of Infectious Endophthalmitis. Transl Vis Sci Technol 2024; 13:14. [PMID: 38767905 PMCID: PMC11114616 DOI: 10.1167/tvst.13.5.14] [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: 07/04/2023] [Accepted: 04/09/2024] [Indexed: 05/22/2024] Open
Abstract
Purpose Extracellular vesicles (EVs) are messenger pigeons of the cells that communicate about cellular microenvironment. In this study, we evaluated the expression of C8α and calpain-2 in EVs from vitreous of patients with bacterial endophthalmitis to assess its utility as a diagnostic marker. Methods EVs were isolated from vitreous of patients with bacterial endophthalmitis (culture positive and culture negative) and noninfectious control by exosome isolation reagent and characterized, and the levels of C8α and calpain-2 was assessed by enzyme-linked immunosorbent assay in isolated EVs and direct vitreous. The receiver operating characteristic curve was generated to assess the diagnostic performance. Results Scanning electron microscopy (SEM) and dynamic light scattering (DLS) confirmed the presence of EVs having a diameter (nm) of 275.2 ± 93, 92 ± 22, and 77.28 ± 12 in culture-positive (CP), culture-negative (CN), and control respectively. The expression level (ng/mL) of C8α in the EVs obtained from CP was 144 ± 22 and CN was 31.2 ± 9.8, which was significantly higher (P < 0.01) than control 3.7 ± 2.4. Interestingly, C8α is not expressed directly in the vitreous of CN and controls. Calpain-2 was significantly downregulated (P ≤ 0.0001) in CP (0.94 ± 0.16) and CN (0.70 ± 0.14) than control. The sensitivity and specificity of 1 for C8α and calpain-2 in the EVs implied that its diagnostic accuracy was significant. Conclusions This study showed that the EV proteins C8α and calpain-2 could be suitable diagnostic markers for endophthalmitis. However, the presence of C8α in the EVs of CN samples but not in direct vitreous promises EVs as the future of diagnostics. Translational Relevance Expression levels of EV-calpain-2 and EV-C8α could diagnose CN bacterial endophthalmitis.
Collapse
Affiliation(s)
- Dhanwini Rudraprasad
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Velmurugan K
- Department of pharmacy, BITS Pilani, Hyderabad, Telangana, India
| | - Jayabalan Nirmal
- Department of pharmacy, BITS Pilani, Hyderabad, Telangana, India
| | - Md. Hasnat Ali
- Center for Biostatistics and Epidemiology, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Joveeta Joseph
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India
- Ramoji Foundation Centre of Ocular Infections, LV Prasad Eye Institute, Hyderabad, Telangana, India
| |
Collapse
|
33
|
Sakarin S, Rungsipipat A, Roytrakul S, Jaresitthikunchai J, Phaonakrop N, Charoenlappanit S, Thaisakun S, Surachetpong S. Phosphoproteomics analysis of serum from dogs affected with pulmonary hypertension secondary to degenerative mitral valve disease. PeerJ 2024; 12:e17186. [PMID: 38708342 PMCID: PMC11067895 DOI: 10.7717/peerj.17186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/11/2024] [Indexed: 05/07/2024] Open
Abstract
Pulmonary hypertension (PH), a common complication in dogs affected by degenerative mitral valve disease (DMVD), is a progressive disorder characterized by increased pulmonary arterial pressure (PAP) and pulmonary vascular remodeling. Phosphorylation of proteins, impacting vascular function and cell proliferation, might play a role in the development and progression of PH. Unlike gene or protein studies, phosphoproteomic focuses on active proteins that function as end-target proteins within signaling cascades. Studying phosphorylated proteins can reveal active contributors to PH development. Early diagnosis of PH is crucial for effective management and improved clinical outcomes. This study aimed to identify potential serum biomarkers for diagnosing PH in dogs affected with DMVD using a phosphoproteomic approach. Serum samples were collected from healthy control dogs (n = 28), dogs with DMVD (n = 24), and dogs with DMVD and PH (n = 29). Phosphoproteins were enriched from the serum samples and analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Data analysis was performed to identify uniquely expressed phosphoproteins in each group and differentially expressed phosphoproteins among groups. Phosphoproteomic analysis revealed nine uniquely expressed phosphoproteins in the serum of dogs in the DMVD+PH group and 15 differentially upregulated phosphoproteins in the DMVD+PH group compared to the DMVD group. The phosphoproteins previously implicated in PH and associated with pulmonary arterial remodeling, including small nuclear ribonucleoprotein G (SNRPG), alpha-2-macroglobulin (A2M), zinc finger and BTB domain containing 42 (ZBTB42), hemopexin (HPX), serotransferrin (TRF) and complement C3 (C3), were focused on. Their unique expression and differential upregulation in the serum of DMVD dogs with PH suggest their potential as biomarkers for PH diagnosis. In conclusion, this phosphoproteomic study identified uniquely expressed and differentially upregulated phosphoproteins in the serum of DMVD dogs with PH. Further studies are warranted to validate the diagnostic utility of these phosphoproteins.
Collapse
Affiliation(s)
- Siriwan Sakarin
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand, Bangkok, Thailand
| | - Anudep Rungsipipat
- Center of Excellence for Companion Animal Cancer, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand, Bangkok, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand, Bangkok, Thailand
| | - Janthima Jaresitthikunchai
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand, Bangkok, Thailand
| | - Narumon Phaonakrop
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand, Bangkok, Thailand
| | - Sawanya Charoenlappanit
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand, Bangkok, Thailand
| | - Siriwan Thaisakun
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand, Bangkok, Thailand
| | - Sirilak Surachetpong
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand, Bangkok, Thailand
| |
Collapse
|
34
|
Yuan Y, Cui Y, Zhao D, Yuan Y, Zhao Y, Li D, Jiang X, Zhao G. Complement networks in gene-edited pig xenotransplantation: enhancing transplant success and addressing organ shortage. J Transl Med 2024; 22:324. [PMID: 38566098 PMCID: PMC10986007 DOI: 10.1186/s12967-024-05136-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
The shortage of organs for transplantation emphasizes the urgent need for alternative solutions. Xenotransplantation has emerged as a promising option due to the greater availability of donor organs. However, significant hurdles such as hyperacute rejection and organ ischemia-reperfusion injury pose major challenges, largely orchestrated by the complement system, and activated immune responses. The complement system, a pivotal component of innate immunity, acts as a natural barrier for xenotransplantation. To address the challenges of immune rejection, gene-edited pigs have become a focal point, aiming to shield donor organs from human immune responses and enhance the overall success of xenotransplantation. This comprehensive review aims to illuminate strategies for regulating complement networks to optimize the efficacy of gene-edited pig xenotransplantation. We begin by exploring the impact of the complement system on the effectiveness of xenotransplantation. Subsequently, we delve into the evaluation of key complement regulators specific to gene-edited pigs. To further understand the status of xenotransplantation, we discuss preclinical studies that utilize gene-edited pigs as a viable source of organs. These investigations provide valuable insights into the feasibility and potential success of xenotransplantation, offering a bridge between scientific advancements and clinical application.
Collapse
Affiliation(s)
- Yinglin Yuan
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuanyuan Cui
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Dayue Zhao
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuan Yuan
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanshuang Zhao
- Department of Pharmacy, The People's Hospital of Leshan, Leshan, China
| | - Danni Li
- Department of Pharmacy, Longquanyi District of Chengdu Maternity & Child Health Care Hospital, Chengdu, China
| | - Xiaomei Jiang
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Gaoping Zhao
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
35
|
Kaur J, Sharma A, Passi G, Dey P, Khajuria A, Alajangi HK, Jaiswal PK, Barnwal RP, Singh G. Nanomedicine at the Pulmonary Frontier: Immune-Centric Approaches for Respiratory Disease Treatment. Immunol Invest 2024; 53:295-347. [PMID: 38206610 DOI: 10.1080/08820139.2023.2298398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Respiratory diseases (RD) are a group of common ailments with a rapidly increasing global prevalence, posing a significant threat to humanity, especially the elderly population, and imposing a substantial burden on society and the economy. RD represents an unmet medical need that requires the development of viable pharmacotherapies. While various promising strategies have been devised to advance potential treatments for RD, their implementation has been hindered by difficulties in drug delivery, particularly in critically ill patients. Nanotechnology offers innovative solutions for delivering medications to the inflamed organ sites, such as the lungs. Although this approach is enticing, delivering nanomedicine to the lungs presents complex challenges that require sophisticated techniques. In this context, we review the potential of novel nanomedicine-based immunomodulatory strategies that could offer therapeutic benefits in managing this pressing health condition.
Collapse
Affiliation(s)
- Jatinder Kaur
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Akanksha Sharma
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Gautam Passi
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Piyush Dey
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Akhil Khajuria
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Hema Kumari Alajangi
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Pradeep Kumar Jaiswal
- Department of Biochemistry and Biophysics, Texas A & M University, College Station, Texas, USA
| | | | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
36
|
Fagbohun OF, Thilakarathna WPDW, Zhou J, Lehmann C, Jiao G, Rupasinghe HPV. Sea Cucumber and Blueberry Extracts Suppress Inflammation and Reduce Acute Lung Injury through the Regulation of NF-κB/MAPK/JNK Signaling Pathway in Lipopolysaccharide-Treated C57BL/6 Mice. Molecules 2024; 29:1511. [PMID: 38611791 PMCID: PMC11013731 DOI: 10.3390/molecules29071511] [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: 02/11/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Acute lung injury (ALI) represents a life-threatening condition with high morbidity and mortality despite modern mechanical ventilators and multiple pharmacological strategies. Therefore, there is a need to develop efficacious interventions with minimal side effects. The anti-inflammatory activities of sea cucumber (Cucumaria frondosa) and wild blueberry (Vaccinium angustifolium) extracts have been reported recently. However, their anti-inflammatory activities and the mechanism of action against ALI are not fully elucidated. Thus, the present study aims to understand the mechanism of the anti-inflammatory activity of sea cucumber and wild blueberry extracts in the context of ALI. Experimental ALI was induced via intranasal lipopolysaccharide (LPS) instillation in C57BL/6 mice and the anti-inflammatory properties were determined by cytokine analysis, histological examination, western blot, and qRT-PCR. The results showed that oral supplementation of sea cucumber extracts repressed nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, thereby downregulating the expression of interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF) in the lung tissue and in the plasma. Wild blueberry extracts also suppressed the expression of IL-4. Furthermore, the combination of sea cucumber and wild blueberry extracts restrained MAPK signaling pathways by prominent attenuation of phosphorylation of NF-κB, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK) while the levels of pro-inflammatory cytokines were significantly suppressed. Moreover, there was a significant and synergistic reduction in varying degrees of ALI lesions such as distorted parenchyma, increased alveoli thickness, lymphocyte and neutrophil infiltrations, fibrin deposition, pulmonary emphysema, pneumonia, intra-alveolar hemorrhage, and edema. The anti-inflammatory effect of the combination of sea cucumber and wild blueberry extracts is associated with suppressing MAPK and NF-κB signaling pathways, thereby significantly reducing cytokine storm in LPS-induced experimental ALI.
Collapse
Affiliation(s)
- Oladapo F. Fagbohun
- Department Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada; (O.F.F.); (W.P.D.W.T.)
- Department of Biology, Center for Agriculture and Sciences, Wilmington College, 1870 Quaker Way, Wilmington, OH 45177, USA
| | - Wasitha P. D. W. Thilakarathna
- Department Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada; (O.F.F.); (W.P.D.W.T.)
| | - Juan Zhou
- Departments of Anaesthesia, Pain Management and Perioperative Medicine, Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (J.Z.); (C.L.)
| | - Christian Lehmann
- Departments of Anaesthesia, Pain Management and Perioperative Medicine, Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (J.Z.); (C.L.)
| | - Guangling Jiao
- Department of Process Engineering and Applied Science, Faculty of Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - H. P. Vasantha Rupasinghe
- Department Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada; (O.F.F.); (W.P.D.W.T.)
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
37
|
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
|
38
|
Nguyen VD, Hughes TR, Zhou Y. From complement to complosome in non-alcoholic fatty liver disease: When location matters. Liver Int 2024; 44:316-329. [PMID: 38010880 DOI: 10.1111/liv.15796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/21/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a growing public health threat and becoming the leading cause of liver transplantation. Nevertheless, no approved specific treatment is currently available for NAFLD. The pathogenesis of NAFLD is multifaceted and not yet fully understood. Accumulating evidence suggests a significant role of the complement system in the development and progression of NAFLD. Here, we provide an overview of the complement system, incorporating the novel concept of complosome, and summarise the up-to-date evidence elucidating the association between complement dysregulation and the pathogenesis of NAFLD. In this process, the extracellular complement system is activated through various pathways, thereby directly contributing to, or working together with other immune cells in the disease development and progression. We also introduce the complosome and assess the evidence that implicates its potential influence in NAFLD through its direct impact on hepatocytes or non-parenchymal liver cells. Additionally, we expound upon how complement system and the complosome may exert their effects in relation with hepatic zonation in NAFLD. Furthermore, we discuss the potential therapeutic implications of targeting the complement system, extracellularly and intracellularly, for NAFLD treatment. Finally, we present future perspectives towards a better understanding of the complement system's contribution to NAFLD.
Collapse
Affiliation(s)
- Van-Dien Nguyen
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Timothy R Hughes
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - You Zhou
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
39
|
Kolahi Azar H, Imanpour A, Rezaee H, Ezzatifar F, Zarei-Behjani Z, Rostami M, Azami M, Behestizadeh N, Rezaei N. Mesenchymal stromal cells and CAR-T cells in regenerative medicine: The homing procedure and their effective parameters. Eur J Haematol 2024; 112:153-173. [PMID: 37254607 DOI: 10.1111/ejh.14014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 06/01/2023]
Abstract
Mesenchymal stromal cells (MSCs) and chimeric antigen receptor (CAR)-T cells are two core elements in cell therapy procedures. MSCs have significant immunomodulatory effects that alleviate inflammation in the tissue regeneration process, while administration of specific chemokines and adhesive molecules would primarily facilitate CAR-T cell trafficking into solid tumors. Multiple parameters affect cell homing, including the recipient's age, the number of cell passages, proper cell culture, and the delivery method. In addition, several chemokines are involved in the tumor microenvironment, affecting the homing procedure. This review discusses parameters that improve the efficiency of cell homing and significant cell therapy challenges. Emerging comprehensive mechanistic strategies such as non-systemic and systemic homing that revealed a significant role in cell therapy remodeling were also reviewed. Finally, the primary implications for the development of combination therapies that incorporate both MSCs and CAR-T cells for cancer treatment were discussed.
Collapse
Affiliation(s)
- Hanieh Kolahi Azar
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Aylar Imanpour
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hanieh Rezaee
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ezzatifar
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Molecular and Cell Biology Research Center, Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zeinab Zarei-Behjani
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, Advanced School of Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammadreza Rostami
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Food Science and Nutrition Group (FSAN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmoud Azami
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Behestizadeh
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, 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
|
40
|
Orach J, Hemshekhar M, Rider CF, Spicer V, Lee AH, Yuen ACY, Mookherjee N, Carlsten C. Concentration-dependent alterations in the human plasma proteome following controlled exposure to diesel exhaust. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 342:123087. [PMID: 38061431 DOI: 10.1016/j.envpol.2023.123087] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023]
Abstract
Traffic-related air pollution (TRAP) exposure is associated with systemic health effects, which can be studied using blood-based markers. Although we have previously shown that high TRAP concentrations alter the plasma proteome, the concentration-response relationship between blood proteins and TRAP is unexplored in controlled human exposure studies. We aimed to identify concentration-dependent plasma markers of diesel exhaust (DE), a model of TRAP. Fifteen healthy non-smokers were enrolled into a double-blinded, crossover study where they were exposed to filtered air (FA) and DE at 20, 50 and 150 μg/m3 PM2.5 for 4h, separated by ≥ 4-week washouts. We collected blood at 24h post-exposure and used label-free mass spectrometry to quantify proteins in plasma. Proteins exhibiting a concentration-response, as determined by linear mixed effects models (LMEMs), were assessed for pathway enrichment using WebGestalt. Top candidates, identified by sparse partial least squares discriminant analysis and LMEMs, were confirmed using enzyme-linked immunoassays. Thereafter, we assessed correlations between proteins that showed a DE concentration-response and acute inflammatory endpoints, forced expiratory volume in 1 s (FEV1) and methacholine provocation concentration causing a 20% drop in FEV1 (PC20). DE exposure was associated with concentration-dependent alterations in 45 proteins, which were enriched in complement pathways. Of the 9 proteins selected for confirmatory immunoassays, based on complementary bioinformatic approaches to narrow targets and availability of high-quality assays, complement factor I (CFI) exhibited a significant concentration-dependent decrease (-0.02 μg/mL per μg/m3 of PM2.5, p = 0.04). Comparing to FA at discrete concentrations, CFI trended downward at 50 (-2.14 ± 1.18, p = 0.08) and significantly decreased at 150 μg/m3 PM2.5 (-2.93 ± 1.18, p = 0.02). CFI levels were correlated with FEV1, PC20 and nasal interleukin (IL)-6 and IL-1β. This study details concentration-dependent alterations in the plasma proteome following DE exposure at concentrations relevant to occupational and community settings. CFI shows a robust concentration-response and association with established measures of airway function and inflammation.
Collapse
Affiliation(s)
- Juma Orach
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, Vancouver Coastal Health Research Institute, The University of British Columbia, British Columbia, Vancouver, V5Z1W9, Canada
| | - Mahadevappa Hemshekhar
- Manitoba Center for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Manitoba, Winnipeg, R3E 3P4, Canada
| | - Christopher Francis Rider
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, Vancouver Coastal Health Research Institute, The University of British Columbia, British Columbia, Vancouver, V5Z1W9, Canada
| | - Victor Spicer
- Manitoba Center for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Manitoba, Winnipeg, R3E 3P4, Canada
| | - Amy H Lee
- Molecular Biology and Biochemistry, Department of Molecular Biology and Biochemistry, Simon Fraser University, British Columbia, Burnaby, V5A 1S6, Canada
| | - Agnes Che Yan Yuen
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, Vancouver Coastal Health Research Institute, The University of British Columbia, British Columbia, Vancouver, V5Z1W9, Canada
| | - Neeloffer Mookherjee
- Manitoba Center for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Manitoba, Winnipeg, R3E 3P4, Canada; Department of Immunology, University of Manitoba, Manitoba, Winnipeg, R3E 0T5, Canada
| | - Chris Carlsten
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, Vancouver Coastal Health Research Institute, The University of British Columbia, British Columbia, Vancouver, V5Z1W9, Canada.
| |
Collapse
|
41
|
Miwa T, Sato S, Golla M, Song WC. Expansion of Anticomplement Therapy Indications from Rare Genetic Disorders to Common Kidney Diseases. Annu Rev Med 2024; 75:189-204. [PMID: 37669567 DOI: 10.1146/annurev-med-042921-102405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Complement constitutes a major part of the innate immune system. The study of complement in human health has historically focused on infection risks associated with complement protein deficiencies; however, recent interest in the field has focused on overactivation of complement as a cause of immune injury and the development of anticomplement therapies to treat human diseases. The kidneys are particularly sensitive to complement injury, and anticomplement therapies for several kidney diseases have been investigated. Overactivation of complement can result from loss-of-function mutations in complement regulators; gain-of-function mutations in key complement proteins such as C3 and factor B; or autoantibody production, infection, or tissue stresses, such as ischemia and reperfusion, that perturb the balance of complement activation and regulation. Here, we provide a high-level review of the status of anticomplement therapies, with an emphasis on the transition from rare diseases to more common kidney diseases.
Collapse
Affiliation(s)
- Takashi Miwa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; , , ,
| | - Sayaka Sato
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; , , ,
| | - Madhu Golla
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; , , ,
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; , , ,
| |
Collapse
|
42
|
Detsika MG, Palamaris K, Dimopoulou I, Kotanidou A, Orfanos SE. The complement cascade in lung injury and disease. Respir Res 2024; 25:20. [PMID: 38178176 PMCID: PMC10768165 DOI: 10.1186/s12931-023-02657-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/26/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND The complement system is an important arm of immune defense bringing innate and adaptive immunity. Although originally regarded as a major complementary defense mechanism against pathogens, continuously emerging evidence has uncovered a central role of this complex system in several diseases including lung pathologies. MAIN BODY Complement factors such as anaphylatoxins C3a and C5a, their receptors C3aR, C5aR and C5aR2 as well as complement inhibitory proteins CD55, CD46 and CD59 have been implicated in pathologies such as the acute respiratory distress syndrome, pneumonia, chronic obstructive pulmonary disease, asthma, interstitial lung diseases, and lung cancer. However, the exact mechanisms by which complement factors induce these diseases remain unclear. Several complement-targeting monoclonal antibodies are reported to treat lung diseases. CONCLUSIONS The complement system contributes to the progression of the acute and chronic lung diseases. Better understanding of the underlying mechanisms will provide groundwork to develop new strategy to target complement factors for treatment of lung diseases.
Collapse
Affiliation(s)
- M G Detsika
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, 3, Ploutarchou St., 10675, Athens, Greece.
| | - K Palamaris
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - I Dimopoulou
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, 3, Ploutarchou St., 10675, Athens, Greece
| | - A Kotanidou
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, 3, Ploutarchou St., 10675, Athens, Greece
| | - S E Orfanos
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, 3, Ploutarchou St., 10675, Athens, Greece.
| |
Collapse
|
43
|
Babbar R, Kaur A, Vanya, Arora R, Gupta JK, Wal P, Tripathi AK, Koparde AA, Goyal P, Ramniwas S, Gulati M, Behl T. Impact of Bioactive Compounds in the Management of Various Inflammatory Diseases. Curr Pharm Des 2024; 30:1880-1893. [PMID: 38818920 DOI: 10.2174/0113816128299615240513174041] [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: 01/25/2024] [Revised: 04/02/2024] [Accepted: 04/16/2024] [Indexed: 06/01/2024]
Abstract
Inflammation is an individual's physiological response to a sequence of physical, chemical, or infectious stressors acting mainly to provide localized protection. Although inflammation is a protective and thus beneficial process, its excess or prolonged action can be harmful to the body. An increasing number of the population worldwide are changing their lifestyles, which leads to a rise in inflammatory diseases, such as atherosclerosis, angina pectoris, myocardial infarction, ulcerative colitis, cancer, and many more. Their treatment is based majorly on the pharmacological approach. However, natural products or bioactive compounds are of great significance in inflammation therapy because they show minimum side effects and maximum bioavailability. Therefore, it is critical to investigate bioactive substances that can modify target functions associated with oxidative stress defense and might be used to achieve various health benefits. This review accentuates the essence of bioactive chemicals used in the treatment of inflammation and other inflammatory illnesses. These bioactive compounds can be of any origin, such as plants, animals, bacteria, fungi, marine invertebrates, etc. Bioactive compounds derived from plant sources, such as glycyrrhizin, lignans, lycopene, resveratrol, indoles, and phenolic and polyphenolic compounds, work mainly by reducing oxidative stress and thereby preventing various inflammatory disorders. A large diversity of these anti-inflammatory bioactive compounds has also been discovered in marine environments, giving rise to an increase in the interest of various scientists in marine invertebrates and microbes. The vast diversity of microbes found in the marine environment represents an enormous supply to extract novel compounds, such as from bacteria, cyanobacteria, fungi, algae, microalgae, tiny invertebrates, etc. In the present review, an attempt has been made to summarize such novel bioactive compounds that help prevent inflammatory responses via different mechanisms of action.
Collapse
Affiliation(s)
- Ritchu Babbar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Arpanpreet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vanya
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Pranay Wal
- Department of Pharmacy, Pranveer Singh Institute of Technology, Kanpur, Uttar Pradesh 209305, India
| | | | - Akshada Amit Koparde
- Department of Pharmaceutical Chemistry, Krishna Vishwa Vidyapeeth, Krishna Institute of Pharmacy, Malkapur, Karad 415110, Maharashtra, India
| | - Pradeep Goyal
- Department of Pharmacology, Saraswati College of Pharmacy, Gharuan, Mohali, Punjab, India
| | - Seema Ramniwas
- University Centre for Research and Development, University of Biotechnology, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India
- ARCCIM, Faculty of Health, University of Technology, Sydney, Ultimo, NSW 2007, Australia
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| |
Collapse
|
44
|
Qiu W, Chen F, Feng X, Shang J, Luo X, Chen Y. Potential role of inflammaging mediated by the complement system in enlarged facial pores. J Cosmet Dermatol 2024; 23:27-32. [PMID: 37555304 DOI: 10.1111/jocd.15956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/17/2023] [Accepted: 08/01/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND Enlarged facial pores are a common cosmetic concern of the skin, rather than a disease, and have not received much attention from dermatologists in recent years. Consequently, progress in understanding their pathogenesis has been limited, and current cosmetic solutions have limitations. Given that the complement system has regained interest as a key player in chronic inflammatory skin conditions, various mechanisms involving this system are being investigated. OBJECTIVE We aimed to shed light on the mechanism underlying enlarged facial pores by examining the role of the complement system in skin. METHODS We conducted a comprehensive literature search utilizing various academic databases including PubMed, Web of Science, and Google Scholar. Employing keywords such as "complement system," "inflammation," "facial pores," "enlarged," and "mechanisms," we compiled a selection of relevant studies. These studies provided a comprehensive understanding of the intricate mechanisms underlying the relationship between the "complement system" and "inflammation" within the context of facial pore enlargement. RESULTS Our findings suggest that inflammaging mediated by complement activation may be a critical player in the formation of enlarged facial pores. Specifically, overactivation of the complement system leading to the accumulation of complement fragments could be a major contributor to this process. Notably, the complement system in skin may be involved in a range of skin issues, including aging. CONCLUSION Modulating the complement system presents a promising avenue for future research in improving skin health. Further basic and clinical research is necessary to validate these findings, but we hope that this study can serve as a theoretical foundation for the development of targeted cosmetics.
Collapse
Affiliation(s)
- Wei Qiu
- Beijing Underproved Medical Technology Co., LTD., Beijing, China
| | - Feng Chen
- Beijing Underproved Medical Technology Co., LTD., Beijing, China
| | - Xiaoyue Feng
- Beijing Underproved Medical Technology Co., LTD., Beijing, China
| | - Jianli Shang
- Beijing Underproved Medical Technology Co., LTD., Beijing, China
| | - Xingyi Luo
- Beijing Underproved Medical Technology Co., LTD., Beijing, China
| | - Yong Chen
- Beijing Underproved Medical Technology Co., LTD., Beijing, China
| |
Collapse
|
45
|
Ren Q, Liu Z, Wu L, Yin G, Xie X, Kong W, Zhou J, Liu S. C/EBPβ: The structure, regulation, and its roles in inflammation-related diseases. Biomed Pharmacother 2023; 169:115938. [PMID: 38000353 DOI: 10.1016/j.biopha.2023.115938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 11/26/2023] Open
Abstract
Inflammation, a mechanism of the human body, has been implicated in many diseases. Inflammatory responses include the release of inflammatory mediators by activating various signaling pathways. CCAAT/enhancer binding protein β (C/EBPβ), a transcription factor in the C/EBP family, contains the leucine zipper (bZIP) domain. The expression of C/EBPβ is mediated at the transcriptional and post-translational levels, such as phosphorylation, acetylation, methylation, and SUMOylation. C/EBPβ has been involved in inflammatory responses by mediating several signaling pathways, such as MAPK/NF-κB and IL-6/JAK/STAT3 pathways. C/EBPβ plays an important role in the pathological development of inflammation-related diseases, such as osteoarthritis, pneumonia, hepatitis, inflammatory bowel diseases, and rheumatoid arthritis. Here, we comprehensively discuss the structure and biological effects of C/EBPβ and its role in inflammatory diseases.
Collapse
Affiliation(s)
- Qun Ren
- Department of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Zhaowen Liu
- Department of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Longhuo Wu
- Department of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Guoqiang Yin
- Ganzhou People's Hospital Affiliated to Nanchang University, Ganzhou 341000, China
| | - Xunlu Xie
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Weihao Kong
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Shiwei Liu
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| |
Collapse
|
46
|
Rahmouni M, De Marco L, Spadoni JL, Tison M, Medina-Santos R, Labib T, Noirel J, Tamouza R, Limou S, Delaneau O, Fellay J, Bensussan A, Le Clerc S, McLaren PJ, Zagury JF. The HLA-B*57:01 allele corresponds to a very large MHC haploblock likely explaining its massive effect for HIV-1 elite control. Front Immunol 2023; 14:1305856. [PMID: 38146367 PMCID: PMC10749428 DOI: 10.3389/fimmu.2023.1305856] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/10/2023] [Indexed: 12/27/2023] Open
Abstract
Introduction We have reanalyzed the genomic data of the International Collaboration for the Genomics of HIV (ICGH), centering on HIV-1 Elite Controllers. Methods We performed a genome-wide Association Study comparing 543 HIV Elite Controllers with 3,272 uninfected controls of European descent. Using the latest database for imputation, we analyzed 35,552 Single Nucleotide Polymorphisms (SNPs) within the Major Histocompatibility Complex (MHC) region. Results Our analysis identified 2,626 SNPs significantly associated (p<5. 10-8) with elite control of HIV-1 infection, including well-established MHC signals such as the rs2395029-G allele which tags HLA-B*57:01. A thorough investigation of SNPs in linkage disequilibrium with rs2395029 revealed an extensive haploblock spanning 1.9 megabases in the MHC region tagging HLA-B*57:01, comprising 379 SNP alleles impacting 72 genes. This haploblock contains damaging variations in proteins like NOTCH4 and DXO and is also associated with a strong differential pattern of expression of multiple MHC genes such as HLA-B, MICB, and ZBTB12. The study was expanded to include two cohorts of seropositive African-American individuals, where a haploblock tagging the HLA-B*57:03 allele was similarly associated with control of viral load. The mRNA expression profile of this haploblock in African Americans closely mirrored that in the European cohort. Discussion These findings suggest that additional molecular mechanisms beyond the conventional antigen-presenting role of class I HLA molecules may contribute to the observed influence of HLA-B*57:01/B*57:03 alleles on HIV-1 elite control. Overall, this study has uncovered a large haploblock associated with HLA-B*57 alleles, providing novel insights into their massive effect on HIV-1 elite control.
Collapse
Affiliation(s)
- Myriam Rahmouni
- Laboratoire Génomique, Bioinformatique, et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, Paris, France
| | - Lorenzo De Marco
- Laboratoire Génomique, Bioinformatique, et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, Paris, France
| | - Jean-Louis Spadoni
- Laboratoire Génomique, Bioinformatique, et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, Paris, France
| | - Maxime Tison
- Laboratoire Génomique, Bioinformatique, et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, Paris, France
| | - Raissa Medina-Santos
- Laboratoire Génomique, Bioinformatique, et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, Paris, France
| | - Taoufik Labib
- Laboratoire Génomique, Bioinformatique, et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, Paris, France
| | - Josselin Noirel
- Laboratoire Génomique, Bioinformatique, et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, Paris, France
| | - Ryad Tamouza
- Université Paris Est Créteil, INSERM U955, IMRB, Laboratoire Neuro-Psychiatrie translationnelle, Créteil, France
| | - Sophie Limou
- Nantes Université, Ecole Centrale Nantes, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), Nantes, France
| | - Olivier Delaneau
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Jacques Fellay
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Sigrid Le Clerc
- Laboratoire Génomique, Bioinformatique, et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, Paris, France
| | - Paul J. McLaren
- Sexually Transmitted and Blood-Borne Infections Division at JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratory Branch, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Jean-François Zagury
- Laboratoire Génomique, Bioinformatique, et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, Paris, France
| |
Collapse
|
47
|
Yang Y, Zhao C, Chen B, Yu X, Zhou Y, Ni D, Zhang X, Zhang J, Ling X, Zhang Z, Huo R. Follicular fluid C3a-peptide promotes oocyte maturation through F-actin aggregation. BMC Biol 2023; 21:285. [PMID: 38066646 PMCID: PMC10709936 DOI: 10.1186/s12915-023-01760-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Immature cumulus-oocyte complexes are retrieved to obtain mature oocytes by in vitro maturation (IVM), a laboratory tool in reproductive medicine to obtain mature oocytes. Unfortunately, the efficiency of IVM is not satisfactory. To circumvent this problem, we therefore intended to commence with the composition of ovarian follicular fluid (FF), an important microenvironment influencing oocyte growth. It is well known that FF has a critical role in oocyte development and maturation. However, the components in human FF remain largely unknown, particularly with regard to small molecular peptides. RESULTS In current study, the follicular fluid derived from human mature and immature follicles were harvested. The peptide profiles of FF were further investigated by using combined ultrafiltration and LC-MS/MS. The differential peptides were preliminary determined by performing differentially expressed analysis. Human and mouse oocyte culture were used to verify the influence of differential peptides on oocyte development. Constructing plasmids, cell transfecting, Co-IP, PLA etc. were used to reveal the detail molecular mechanism. The results from differentially expressed peptide as well as cultured human and mouse oocytes analyses showed that highly conserved C3a-peptide, a cleavage product of complement C3a, definitely affected oocytes development. Intriguingly, C3a-peptide possessed a novel function that promoted F-actin aggregation and spindle migration, raised the percentage of oocytes at the MII stage, without increasing the chromosome aneuploidy ratio, especially in poor-quality oocytes. These effects of C3a-peptide were attenuated by C3aR morpholino inhibition, suggesting that C3a-peptide affected oocytes development by collaborating with its classical receptor, C3aR. Specially, we found that C3aR co-localized to the spindle with β-tubulin to recruit F-actin toward the spindle and subcortical region of the oocytes through specific binding to MYO10, a key regulator for actin organization, spindle morphogenesis and positioning in oocytes. CONCLUSIONS Our results provide a new perspective for improving IVM culture systems by applying FF components and also provide molecular insights into the physiological function of C3a-peptide, its interaction with C3aR, and their roles in enabling meiotic division of oocytes.
Collapse
Affiliation(s)
- Ye Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Chun Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
| | - Beili Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Xiaoning Yu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Yuxi Zhou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
| | - Danyu Ni
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
| | - Xiaolan Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
| | - Junqiang Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
| | - Xiufeng Ling
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China.
| | - Zhiguo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Anhui, China.
| | - Ran Huo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China.
- Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School,, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
48
|
Erb C, Reinehr S, Theiss C, Dick HB, Joachim SC. HSP27 induced glaucomatous damage in mice of young and advanced age. Front Cell Neurosci 2023; 17:1257297. [PMID: 37744880 PMCID: PMC10513106 DOI: 10.3389/fncel.2023.1257297] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Age-related diseases such as glaucoma, a leading cause of blindness, are having an upward trend due to an aging society. In glaucoma, some patients display altered antibody profiles and increased antibody titers, for example against heat shock protein 27 (HSP27). An intravitreal injection of HSP27 leads to glaucoma-like damage in rats. We now aimed to investigate if aged mice are more prone to this damage than younger ones. Methods We intravitreally injected HSP27 into young (1-2 months) and aged (7-8 months) mice to compare glaucomatous damage. Respective age-matched controls received PBS. Not injected eyes served as naive controls. Results Optical coherence tomography 4 weeks after injection showed no changes in retinal thickness in all groups at both ages. Cell counts and RT-qPCR revealed a significant reduction in RGC numbers in HSP27 mice at both ages. Comparing aged and young HSP27 mice, no differences in Rbpms and Pou4f1 (RGCs) expression was detected, while the Tubb3 expression (neuronal cells) was significantly upregulated in aged HSP27 animals. Neither microglia/macrophages nor (resident) microglia counts revealed significant differences in HSP27 mice at both ages. Nevertheless, increased relative Iba1 and Tmem119 expression was detected in young and aged HSP27 mice. Aged HSP27 mice displayed a significantly lower Iba1 expression than young ones, whereas Cd68 levels were upregulated. A larger GFAP+ area and an upregulation of GFAP expression in HSP27 animals of both ages indicated a macrogliosis. Also, elevated Il1b and Nos2 expression levels were observed in young and aged HSP27 mice. However, only Il1b levels were upregulated when comparing 7-8 months to 1-2 months old animals. A larger HSP25+ area was seen in aged HSP27 animals, while Hspb2 expression levels were downregulated in both HSP27 groups. The aged HSP27 group displayed an upregulated Hspb2 expression compared to young mice. Furthermore, a higher optic nerve degeneration score was noted in young and aged HSP27 groups. Discussion These findings indicate that an intravitreal injection of HSP27 led to RGC loss accompanied by inflammation. Age-dependent effects (7-8 months vs. 1-2 months) were not very prominent. The results suggest a potential role of extracellular HSP27 in the development of glaucoma.
Collapse
Affiliation(s)
- Clivia Erb
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Carsten Theiss
- Institute of Anatomy, Department of Cytology, Ruhr-University Bochum, Bochum, Germany
| | - H. Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
49
|
Lipsa D, Magrì D, Della Camera G, La Spina R, Cella C, Garmendia-Aguirre I, Mehn D, Ruiz-Moreno A, Fumagalli F, Calzolai L, Gioria S. Differences in Physico-Chemical Properties and Immunological Response in Nanosimilar Complex Drugs: The Case of Liposomal Doxorubicin. Int J Mol Sci 2023; 24:13612. [PMID: 37686418 PMCID: PMC10487543 DOI: 10.3390/ijms241713612] [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: 07/14/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
This study aims to highlight the impact of physicochemical properties on the behaviour of nanopharmaceuticals and how much carrier structure and physiochemical characteristics weigh on the effects of a formulation. For this purpose, two commercially available nanosimilar formulations of Doxil and their respective carriers were compared as a case study. Although the two formulations were "similar", we detected different toxicological effects (profiles) in terms of in vitro toxicity and immunological responses at the level of cytokines release and complement activation (iC3b fragment), that could be correlated with the differences in the physicochemical properties of the formulations. Shedding light on nanosimilar key quality attributes of liposome-based materials and the need for an accurate characterization, including investigation of the immunological effects, is of fundamental importance considering their great potential as delivery system for drugs, genes, or vaccines and the growing market demand.
Collapse
Affiliation(s)
- Dorelia Lipsa
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (D.L.); (D.M.); (G.D.C.); (R.L.S.); (C.C.); (I.G.-A.); (D.M.); (A.R.-M.); (F.F.); (L.C.)
| | - Davide Magrì
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (D.L.); (D.M.); (G.D.C.); (R.L.S.); (C.C.); (I.G.-A.); (D.M.); (A.R.-M.); (F.F.); (L.C.)
| | - Giacomo Della Camera
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (D.L.); (D.M.); (G.D.C.); (R.L.S.); (C.C.); (I.G.-A.); (D.M.); (A.R.-M.); (F.F.); (L.C.)
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), 80131 Naples, Italy
| | - Rita La Spina
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (D.L.); (D.M.); (G.D.C.); (R.L.S.); (C.C.); (I.G.-A.); (D.M.); (A.R.-M.); (F.F.); (L.C.)
| | - Claudia Cella
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (D.L.); (D.M.); (G.D.C.); (R.L.S.); (C.C.); (I.G.-A.); (D.M.); (A.R.-M.); (F.F.); (L.C.)
| | - Irantzu Garmendia-Aguirre
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (D.L.); (D.M.); (G.D.C.); (R.L.S.); (C.C.); (I.G.-A.); (D.M.); (A.R.-M.); (F.F.); (L.C.)
| | - Dora Mehn
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (D.L.); (D.M.); (G.D.C.); (R.L.S.); (C.C.); (I.G.-A.); (D.M.); (A.R.-M.); (F.F.); (L.C.)
| | - Ana Ruiz-Moreno
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (D.L.); (D.M.); (G.D.C.); (R.L.S.); (C.C.); (I.G.-A.); (D.M.); (A.R.-M.); (F.F.); (L.C.)
| | - Francesco Fumagalli
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (D.L.); (D.M.); (G.D.C.); (R.L.S.); (C.C.); (I.G.-A.); (D.M.); (A.R.-M.); (F.F.); (L.C.)
| | - Luigi Calzolai
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (D.L.); (D.M.); (G.D.C.); (R.L.S.); (C.C.); (I.G.-A.); (D.M.); (A.R.-M.); (F.F.); (L.C.)
| | - Sabrina Gioria
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (D.L.); (D.M.); (G.D.C.); (R.L.S.); (C.C.); (I.G.-A.); (D.M.); (A.R.-M.); (F.F.); (L.C.)
| |
Collapse
|
50
|
Hestekin CN, Pakkaner E, Hestekin JA, De Souza LS, Chowdhury PP, Marçal JL, Moore J, Hesse SA, Takacs CJ, Tassone CJ, Dachavaram SS, Crooks PA, Williams K, Kurtz I. High flux novel polymeric membrane for renal applications. Sci Rep 2023; 13:11703. [PMID: 37474512 PMCID: PMC10359412 DOI: 10.1038/s41598-023-37765-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 06/27/2023] [Indexed: 07/22/2023] Open
Abstract
Biocompatibility and the ability to mediate the appropriate flux of ions, urea, and uremic toxins between blood and dialysate components are key parameters for membranes used in dialysis. Oxone-mediated TEMPO-oxidized cellulose nanomaterials have been demonstrated to be excellent additives in the production and tunability of ultrafiltration and dialysis membranes. In the present study, nanocellulose ionic liquid membranes (NC-ILMs) were tested in vitro and ex vivo. An increase in flux of up to two orders of magnitude was observed with increased rejection (about 99.6%) of key proteins compared to that of polysulfone (PSf) and other commercial membranes. NC-ILMs have a sharper molecular weight cut-off than other phase inversion polymeric membranes, allowing for high throughput of urea and a uremic toxin surrogate and limited passage of proteins in dialysis applications. Superior anti-fouling properties were also observed for the NC-ILMs, including a > 5-h operation time with no systemic anticoagulation in blood samples. Finally, NC-ILMs were found to be biocompatible in rat ultrafiltration and dialysis experiments, indicating their potential clinical utility in dialysis and other blood filtration applications. These superior properties may allow for a new class of membranes for use in a wide variety of industrial applications, including the treatment of patients suffering from renal disease.
Collapse
Affiliation(s)
- Christa N Hestekin
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA.
| | - Efecan Pakkaner
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Jamie A Hestekin
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Leticia Santos De Souza
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Partha Pratim Chowdhury
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Juliana Louzada Marçal
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - John Moore
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Sarah A Hesse
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, CA, 94025, USA
| | - Christopher J Takacs
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, CA, 94025, USA
| | - Christopher J Tassone
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, CA, 94025, USA
| | - Soma Shekar Dachavaram
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Kate Williams
- St. Francis Animal Hospital, 121 Virginia Street, Springdale, AR, 72764, USA
| | - Ira Kurtz
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Brain Research Institute, University of California, Los Angeles, CA, 90095, USA
| |
Collapse
|