1
|
Kaofai C, Jearanaiwitayakul T, Saisingha K, Limthongkul J, Masrinoul P, Ubol S. Transcriptomic analysis of DENV-2-infected human dermal fibroblasts identified potential mechanisms that suppressed ZIKV replication during sequential coinfection. Virol J 2025; 22:154. [PMID: 40399994 PMCID: PMC12096689 DOI: 10.1186/s12985-025-02769-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/29/2025] [Indexed: 05/23/2025] Open
Abstract
Dengue virus (DENV) and Zika virus (ZIKV) are closely related flaviviruses which are transmitted by the same species of mosquitoes. Due to overlapping geographic distributions and transmission vectors, cases of DENV-ZIKV coinfection have been reported. However, the impact of coinfection on disease outcomes remains unclear. In this study, an in vitro model of DENV-ZIKV coinfection was developed using the primary human dermal fibroblasts (HDFs). The interaction between DENV-2 and ZIKV during sequential coinfection revealed that prior DENV-2 infection significantly suppressed ZIKV RNA accumulation in the culture supernatant. Transcriptomic profile in response to DENV-2 infection suggested three hypothetical pathways that potentially interfere with ZIKV replication. The first mechanism is prior DENV infection drove HDFs into an antiviral state through upregulation of genes involving innate immune response pathways, including PRR signaling, type I and type II IFN signaling, ISG activity, and cytokine/chemokine activity. This state significantly enhanced resistance to subsequent ZIKV infection in both infected cells and uninfected neighboring cells. The second potential pathway is inhibition of viral entry. This was supported by DENV-2-infected HDFs significantly suppressed expression of ZIKV receptor and reduced expression of genes involving in clathrin-mediated endocytosis. This can interfere with entry of ZIKV into host cells. The last possible mechanism is driving cells into cell cycle arrest, as DENV-2 infection downregulated genes related to cell cycle progression, which may hinder ZIKV replication. These findings partly unfold the interplay between DENV and ZIKV at the entry site which may explain the disease outcome of DENV-ZIKV coinfection.
Collapse
Affiliation(s)
- Chernkhwan Kaofai
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Tuksin Jearanaiwitayakul
- Department of Clinical Pathology, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Khwankhao Saisingha
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Jitra Limthongkul
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Promsin Masrinoul
- Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand.
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
2
|
Lin Q, Luo Z, He L, Zhong L, Zeng Q, Zhou Y, Chen Q, Deng X, Song X, Song Q, Song J. Nafamostat mesylate augments survival in rats afflicted by exertional heat stroke. Front Pharmacol 2025; 16:1559181. [PMID: 40444038 PMCID: PMC12119467 DOI: 10.3389/fphar.2025.1559181] [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: 01/12/2025] [Accepted: 04/17/2025] [Indexed: 06/02/2025] Open
Abstract
Objective To evaluate the impact of Nafamostat mesylate (NM) in improving survival outcomes among rats subjected to exertional heat stroke. Methods This study involved a cohort of 45 specific pathogen-free (SPF) male Sprague Dawley (SD) rats. After successfully inducing exertional heat stroke, the rats were randomly divided into three groups: the Control group (Con, n = 15), the Exertional Heat Stroke group (EHS, n = 15), and the Nafamostat Mesylate group (NM, n = 15). A subset of ten rats from each group was selected for a 72-h survival analysis. Three hours following the successful establishment of the model, blood samples were collected under anesthesia for comprehensive analysis. This included routine hematological tests, coagulation assessments, and quantitative proteomics analysis, which were later validated using Parallel Reaction Monitoring (PRM). Additionally, tissue samples were harvested from the brain, heart, lung, kidney, liver, and duodenum of rats in each group for subsequent pathological examination. Results The 72-h survival rate in the NM group was markedly higher than that observed in the EHS group. Pathological assessments indicated a notable reduction in thrombus formation within the brain, lungs, and liver in the NM group when compared to the EHS group. Furthermore, the NM group exhibited an elevated platelet count and a significant reduction in prothrombin time (PT) and activated partial thromboplastin time (APTT) relative to the EHS group. Proteomic profiling identified a total of 1,971 differentially expressed proteins, with 160 proteins being downregulated and 52 upregulated in the NM group as compared to the EHS group. PRM validation confirmed that the NM group significantly dampened the expression levels of key differential proteins, including ribosomal protein P2 (rpLP2), Histone 4c16 (H4c16), neutrophilic granule protein (NGP), and inositol monophosphatase 1 (Impa1), which are implicated in anti-inflammatory responses, suppression of immune-mediated thrombosis, and enhancement of cellular metabolism. Conclusion NM mitigates coagulopathy, alleviates thrombus burden, and improves the 72-h survival rate in EHS rats through the modulation of differentially expressed proteins, specifically rpLP2, H4c16, NGP, and Impa1.
Collapse
Affiliation(s)
- Qingwei Lin
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, China
| | - Zhuqing Luo
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, China
| | - Longping He
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, China
| | - Lincui Zhong
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, China
| | - Qingbo Zeng
- Intensive Care Unit, Nanchang Hongdu Traditional Chinese Medicine Hospital, Nanchang, China
| | - Ye Zhou
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, China
| | - Qi Chen
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, China
| | - Xingping Deng
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, China
| | - Xiaomin Song
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, China
| | - Qing Song
- Department of Critical Care Medicine, Hainan Hospital, Chinese PLA General Hospital, Sanya, China
- Heatstroke Treatment and Research Center of PLA, Sanya, China
| | - Jingchun Song
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, China
- Heatstroke Treatment and Research Center of PLA, Sanya, China
| |
Collapse
|
3
|
Bacaloni S, Agrawal DK. Nutrition, Gut Microbiota, and Epigenetics in the Modulation of Immune Response and Metabolic Health. CARDIOLOGY AND CARDIOVASCULAR MEDICINE 2025; 9:111-124. [PMID: 40443829 PMCID: PMC12121961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Immune system function is intricately shaped by nutritional status, dietary patterns, and gut microbiota composition. Micronutrients such as vitamins A, C, D, E, B-complex, zinc, selenium, iron, and magnesium are critical for maintaining physical barriers, supporting immune cell proliferation, and regulating inflammation. Macronutrients-including proteins, fats, and carbohydrates-also modulate immune responses through their impact on immune metabolism and the gut-immune axis. Epigenetic mechanisms, including DNA methylation, histone modifications, and microRNA expression, mediate the long-term effects of diet on immune function and tolerance. Diet-induced alterations in gut microbiota further influence immune homeostasis via microbial metabolites like short-chain fatty acids. Imbalanced diets, particularly the Western diet, contribute to immune dysregulation, chronic inflammation, and the development of metabolic disorders such as obesity and type 2 diabetes. While plant-based and Mediterranean dietary patterns have shown anti-inflammatory and immunoregulatory benefits, gaps remain in understanding the long-term epigenetic impacts of these diets. This review integrates current knowledge on how nutrition and the microbiome regulate immunity, highlighting future directions for personalized dietary strategies in preventing chronic immune-related conditions.
Collapse
Affiliation(s)
- Sabrin Bacaloni
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| |
Collapse
|
4
|
Caruso R, Caruso V, Rigoli L. Eosinophil cytolysis with or without ETosis in four cases of human gastric cancer: a comparative ultrastructural study. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002309. [PMID: 40291980 PMCID: PMC12022494 DOI: 10.37349/etat.2025.1002309] [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: 11/18/2024] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
The ultrastructural morphology of eosinophil cytolysis and extracellular trap cell death (ETosis) has predominantly been examined in non-neoplastic eosinophil-associated diseases, with a limited investigation in neoplasms. This current electron microscopy study examined the ultrastructural characteristics of eosinophil cytolysis and ETosis across four distinct gastric cancer cases: three cases (cases 1-3) exhibited non-ETotic cytolysis, while one case (case 4) presented eosinophils at various stages of ETosis. In cases 1-3, eosinophil non-ETotic cytolysis was characterized by localized plasma membrane disruption, the presence of free extracellular granules (FEGs), and the maintenance of a round or oval nuclear lobe profile. In case 4, eosinophils were observed in progressive stages of ETosis, arbitrarily subdivided into early, intermediate, and advanced. Although early ETosis and non-ETotic cytolysis exhibited overlapping ultrastructural features, chromatin decondensation and nuclear envelope enlargement were more pronounced in early ETosis. Nuclear envelope disruption, loss of the round or oval nuclear lobe profile (intermediate stage), extracellular DNA trap deposition, and the appearance of Charcot-Leyden crystals (advanced stage) were all distinctive features of ETosis. The findings of this case report confirm previous observations of eosinophil cytolysis with or without ETosis in non-neoplastic diseases and extend them to advanced gastric carcinoma. Since Charcot-Leyden crystals were only seen in case 4, their correlation with ETosis was further supported. In gastric cancer, the release of FEGs during non-ETotic cytolysis and the release of both FEGs and DNA traps during ETotic cytolysis may contribute to the formation of an antitumor microenvironment.
Collapse
Affiliation(s)
- Rosario Caruso
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98123 Messina, Italy
| | - Valerio Caruso
- Clinical and Experimental Medicine Department, University of Pisa, 56100 Pisa, Italy
| | - Luciana Rigoli
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98123 Messina, Italy
| |
Collapse
|
5
|
Frank J, Tehrani L, Gamer J, Van Booven DJ, Ballarin S, Rossman R, Edelstein A, Uppalati S, Reuthebuck A, Collado F, Klimas NG, Nathanson L. Gulf War Illness Induced Sex-Specific Transcriptional Differences Under Stressful Conditions. Int J Mol Sci 2025; 26:3610. [PMID: 40332133 PMCID: PMC12026906 DOI: 10.3390/ijms26083610] [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: 02/15/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Gulf War Illness (GWI) is a multi-symptom disorder affecting 1990-1991 Persian Gulf War veterans and is characterized by post-exertional malaise, neurological symptoms, immune deregulation, and exhaustion. Causation is not understood, and effective diagnostics and therapies have not yet been developed. In this work, we analyzed stress-related, sex-specific transcriptomic shifts in GWI subjects and healthy controls through RNA sequencing of peripheral blood mononuclear cells (PBMCs). Blood samples at baseline (T0), at maximal exertion (T1), and four hours post-exertion (T2) were analyzed. In female subjects with GWI, pathways associated with pro-inflammatory processes were found to be deregulated, and in male GWI subjects, pathways related to IL-12 signaling and lymphocytic activation were deregulated at T1 compared to T0. During recovery from stress, pathways corresponding to immune responses and microglial cell activation were altered in female GWI subjects, and apoptotic signaling changed in males with GWI. Documented sex-specific immune deregulation leads to finding better biomarkers. Targeting sex-specific transcriptomic markers of the disease could lead to new therapies for GWI.
Collapse
Affiliation(s)
- Joshua Frank
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.F.); (F.C.); (N.G.K.)
| | - Lily Tehrani
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (L.T.); (J.G.); (S.B.); (R.R.); (A.E.)
| | - Jackson Gamer
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (L.T.); (J.G.); (S.B.); (R.R.); (A.E.)
| | - Derek J. Van Booven
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33146, USA;
| | - Sarah Ballarin
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (L.T.); (J.G.); (S.B.); (R.R.); (A.E.)
| | - Raquel Rossman
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (L.T.); (J.G.); (S.B.); (R.R.); (A.E.)
| | - Abraham Edelstein
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (L.T.); (J.G.); (S.B.); (R.R.); (A.E.)
| | - Sadhika Uppalati
- Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (S.U.); (A.R.)
| | - Ana Reuthebuck
- Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (S.U.); (A.R.)
| | - Fanny Collado
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.F.); (F.C.); (N.G.K.)
| | - Nancy G. Klimas
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.F.); (F.C.); (N.G.K.)
- Department of Veterans Affairs, Miami VA Healthcare System, Geriatric Research Education and Clinical Center (GRECC), Miami, FL 33125, USA
| | - Lubov Nathanson
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.F.); (F.C.); (N.G.K.)
| |
Collapse
|
6
|
Li Y, Yadollahi P, Essien FN, Putluri V, Ambati CSR, Kami Reddy KR, Kamal AHM, Putluri N, Abdurrahman LM, Ruiz Echartea ME, Ernste KJ, Trivedi AJ, Vazquez-Perez J, Hudson WH, Decker WK, Patel R, Osman AA, Kheradmand F, Lai SY, Myers JN, Skinner HD, Coarfa C, Lee K, Jain A, Malovannaya A, Frederick MJ, Sandulache VC. Tobacco smoke exposure is a driver of altered oxidative stress response and immunity in head and neck cancer. J Transl Med 2025; 23:403. [PMID: 40188338 PMCID: PMC11971752 DOI: 10.1186/s12967-025-06258-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: 11/22/2024] [Accepted: 02/17/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Exposomes are critical drivers of carcinogenesis. However, how they modulate tumor behavior remains unclear. Extensive clinical data show cigarette smoke to be a key exposome that promotes aggressive tumors, higher rates of metastasis, reduced response to chemoradiotherapy, and suppressed anti-tumor immunity. We sought to determine whether smoke itself can modulate aggressive tumor behavior in head and neck squamous cell carcinoma (HNSCC) through reprogramming of the cellular reductive state. METHODS Using established human and murine HNSCC cell lines and syngeneic mouse models, we utilized conventional western blotting, steady state and flux metabolomics, RNA sequencing, quantitative proteomics and flow cytometry to analyze the impact of smoke exposure on HNSCC tumor biology and anti-tumor immunity. RESULTS Cigarette smoke persistently activated Nrf2 target genes essential for maintenance of the cellular reductive state and survival under conditions of increased oxidative stress in HNSCC regardless of human papillomavirus (HPV) association. In contrast to e-cigarette vapor, conventional cigarette smoke mobilizes cellular metabolism toward oxidative stress adaptation, resulting in development of cross-resistance to cisplatin. In parallel, smoke exposure modulates expression of PDL1 and the secretory phenotype of HNSCC cells resulting in an altered tumor immune microenvironment (TIME) in syngeneic mouse models and downregulated expression of antigen presentation and costimulatory genes in myeloid cells. CONCLUSION The cigarette smoke exposome is a potent activator of the Nrf2 pathway and appears to be the primary trigger for a tripartite phenotype of aggressive HNSCC consisting of: (1) reduced chemotherapy sensitivity, (2) enhanced metastatic potential and (3) suppressed anti-tumor immunity.
Collapse
Affiliation(s)
- Yang Li
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA
| | - Pedram Yadollahi
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA
| | - Fonma N Essien
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA
| | - Vasanta Putluri
- Advanced Technology Cores, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Chandra Shekar R Ambati
- Advanced Technology Cores, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Karthik Reddy Kami Reddy
- Advanced Technology Cores, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Abu Hena Mostafa Kamal
- Advanced Technology Cores, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nagireddy Putluri
- Advanced Technology Cores, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Lama M Abdurrahman
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA
| | - Maria E Ruiz Echartea
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Keenan J Ernste
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Akshar J Trivedi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | | | - William H Hudson
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Cell Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - William K Decker
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Center for Cell Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Rutulkumar Patel
- Department of Radiation Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Abdullah A Osman
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farrah Kheradmand
- Department of Medicine-Pulmonary, Baylor College of Medicine, Houston, TX, USA
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Stephen Y Lai
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heath D Skinner
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cristian Coarfa
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Kwangwon Lee
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Anna Malovannaya
- Verna and Marrs Mclean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Mitchell J Frederick
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA
| | - Vlad C Sandulache
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, MS: NA102, Houston, TX, 77030, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| |
Collapse
|
7
|
Nosaka N, Borges V, Martinon D, Crother TR, Arditi M, Shimada K. Hypothermia protects against ventilator-induced lung injury by limiting IL-1β release and NETs formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.02.610778. [PMID: 40236184 PMCID: PMC11996356 DOI: 10.1101/2024.09.02.610778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Although mechanical ventilation is a critical intervention for acute respiratory distress syndrome (ARDS), it can trigger an IL-1β-associated complication known as ventilator-induced lung injury. In mice, we found that LPS and high-volume ventilation, LPS-HVV, leads to hypoxemia with neutrophil extracellular traps (NETs) formation in the alveoli. Furthermore, Il1r1 -/- LPS-HVV mice did not develop hypoxemia and had reduced NETs, indicating that IL-1R1 signaling is important for NETs formation and hypoxemia. Therapeutic hypothermia (TH) is known to reduce the release of inflammatory mediators. In LPS-HVV mice, TH (32 °C body temperature) prevented hypoxemia development, reducing albumin leakage, IL-1β, gasdermin D (GSDMD) and NETs formation. We also observed that LPS-primed macrophages, when stimulated at 32°C with ATP or nigericin, release less IL-1β associated with reduced GSDMD cleavage. Thus, hypothermia is an important modulating factor in the NLRP3 inflammasome activation, IL-1β release and NETs formation, preventing LPS-HVV-induced acute respiratory failure.
Collapse
|
8
|
Speziale P, Foster TJ, Arciola CR. The endothelium at the interface between tissues and Staphylococcus aureus in the bloodstream. Clin Microbiol Rev 2025; 38:e0009824. [PMID: 39807893 PMCID: PMC11905367 DOI: 10.1128/cmr.00098-24] [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: 01/16/2025] Open
Abstract
SUMMARYStaphylococcus aureus is a major human pathogen. It can cause many types of infections, in particular bacteremia, which frequently leads to infective endocarditis, osteomyelitis, sepsis, and other debilitating diseases. The development of secondary infections is based on the bacterium's ability to associate with endothelial cells lining blood vessels. The success of endothelial colonization and infection by S. aureus relies on its ability to express a wide array of cell wall-anchored and secreted virulence factors. Establishment of endothelial infection by the pathogen is a multistep process involving adhesion, invasion, extravasation, and dissemination of the bacterium into surrounding tissues. The process is dependent on the type of endothelium in different organs (tissues) and pathogenetic potential of the individual strains. In this review, we report an update on the organization of the endothelium in the vessels, the structure and function of the virulence factors of S. aureus, and the several aspects of bacteria-endothelial cell interactions. After these sections, we will discuss recent advances in understanding the specific mechanisms of infections that develop in the heart, bone and joints, lung, and brain. Finally, we describe how neutrophils bind to endothelial cells, migrate to the site of infection to kill bacteria in the tissues, and how staphylococci counteract neutrophils' actions. Knowledge of the molecular details of S. aureus-endothelial cell interactions will promote the development of new therapeutic strategies and tools to combat this formidable pathogen.
Collapse
Affiliation(s)
- Pietro Speziale
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Pavia, Italy
| | | | - Carla Renata Arciola
- Laboratory of Pathology of Implant Infections, Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
9
|
Yin J, Chi Y, Liu D, Li X, Li X. Unfractionated heparin attenuated histone-induced pulmonary endothelial glycocalyx injury through Ang/Tie2 pathway. J Inflamm (Lond) 2025; 22:9. [PMID: 39962477 PMCID: PMC11834598 DOI: 10.1186/s12950-025-00437-x] [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: 11/23/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
PURPOSE This study aimed to investigate the involvement of angiopoietin (Ang)/Tie2 pathway in mediating pulmonary endothelial glycocalyx injury in histone-induced acute lung injury in mice, and the protective mechanism of unfractionated heparin (UFH). METHODS Twenty-four male C57BL/6 mice (20-25 g), 8-12 weeks old, were randomly divided into control, histone, and histone + UFH groups. The histone (50 mg/kg) was administered via tail vein. UFH (400 U/kg) was administered 1 h after histone injection. The control group was administered by an equal amount of sterile saline solution. The lungs of all groups were harvested 4 h after the injection of histones or sterile saline. RESULTS UFH attenuated histone-induced lung histopathological changes and edema. UFH alleviated pulmonary endothelial injury and glycocalyx shedding by reducing histone-induced low expression of thrombomodulin (TM) and decreased lung syndecan-1 levels. UFH improved histone-induced low mRNA expression of TM, syndecan-1, Ang-1, Tie2 and high expression of heparinase (HPA), Ang-2. CONCLUSION UFH may attenuate histone-induced lung injury and pulmonary endothelial glycocalyx degradation via the Ang/Tie2 pathway.
Collapse
Affiliation(s)
- Jia Yin
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, Liaoning, 110001, China
| | - Yawen Chi
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, Liaoning, 110001, China
| | - Danyan Liu
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, Liaoning, 110001, China
| | - Xinghua Li
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, Liaoning, 110001, China
| | - Xu Li
- Department of Critical Care Medicine, The First Affiliated Hospital, China Medical University, North Nanjing Street 155, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
10
|
Serrat J, Torres-Valle M, De Marco Verissimo C, Siles-Lucas M, González-Miguel J. Binding and cleavage of pro-urokinase by a tegument extract of Fasciola hepatica newly excysted juveniles activate the host fibrinolytic system. Vet Res 2025; 56:20. [PMID: 39856784 PMCID: PMC11762853 DOI: 10.1186/s13567-025-01449-4] [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: 08/22/2024] [Accepted: 11/26/2024] [Indexed: 01/27/2025] Open
Abstract
Plasmin, the final product of fibrinolysis, is a broad-spectrum serine protease that degrades extracellular matrix (ECM) components, a function exploited by multiple pathogens for dissemination purposes. The trematode Fasciola hepatica is the leading cause of fasciolosis, a major disease of livestock and an emerging zoonosis in humans. Infection success depends on the ability of F. hepatica newly excysted juveniles (FhNEJ) to penetrate the host intestinal wall, a process that remains incompletely understood. We have previously shown that FhNEJ are capable of binding plasminogen (PLG), the zymogen of plasmin, on their tegument surface, which leads to plasmin generation in the presence of host-derived PLG activators and subsequent degradation of laminin, a major component of the intestinal ECM. Here, we describe the interaction between a tegument extract of FhNEJ and the precursor of the urokinase-type PLG activator (pro-u-PA). We found that F. hepatica cathepsins B3, L3, enolase and glutathione S-transferase mediate this interaction, suggesting a multifactorial or moonlighting role for these proteins. Additionally, our results revealed that the tegument of FhNEJ contains a protease that is capable of cleaving and activating pro-u-PA into its catalytically active form, which positively impacts the capacity of the parasites to generate plasmin from the host PLG. Collectively, our findings indicate that FhNEJ interact with the host fibrinolytic system at multiple levels, reinforcing the potential of targeting this interaction as a strategy to prevent FhNEJ trans-intestinal migration and infection success.
Collapse
Affiliation(s)
- Judit Serrat
- Laboratory of Helminth Parasites of Zoonotic Importance (ATENEA), Institute of Natural Resources and Agrobiology of Salamanca (IRNASA-CSIC), Salamanca, Spain
| | - María Torres-Valle
- Laboratory of Helminth Parasites of Zoonotic Importance (ATENEA), Institute of Natural Resources and Agrobiology of Salamanca (IRNASA-CSIC), Salamanca, Spain
| | | | - Mar Siles-Lucas
- Laboratory of Helminth Parasites of Zoonotic Importance (ATENEA), Institute of Natural Resources and Agrobiology of Salamanca (IRNASA-CSIC), Salamanca, Spain
| | - Javier González-Miguel
- Laboratory of Helminth Parasites of Zoonotic Importance (ATENEA), Institute of Natural Resources and Agrobiology of Salamanca (IRNASA-CSIC), Salamanca, Spain.
| |
Collapse
|
11
|
Zhang Y, Tan J, Zhao Y, Guan L, Li S. By activating endothelium histone H4 mediates oleic acid-induced acute respiratory distress syndrome. BMC Pulm Med 2025; 25:3. [PMID: 39757148 DOI: 10.1186/s12890-024-03334-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 10/08/2024] [Indexed: 01/07/2025] Open
Abstract
OBJECTIVE This study investigated pathogenic role and mechanism of extracellular histone H4 during oleic acid (OA)-induced acute respiratory distress syndrome (ARDS). METHODS ARDS was induced by intravenous injection of OA in mice, and evaluated by blood gas, pathological analysis, lung edema, and survival rate. Heparan sulfate (HS) degradation was evaluated using immunofluorescence and flow cytometry. The released von Willebrand factor (vWF) was measured using ELISA. P-selectin translocation and neutrophil infiltration were measured via immunohistochemical analysis. Changes in VE-cadherin were measured by western blot. Blocking antibodies against TLRs were used to investigate the signaling pathway. RESULTS Histone H4 in plasma and BALF increased significantly after OA injection. Histone H4 was closely correlated with the OA dose, which determined the ARDS severity. Pretreatment with histone H4 further aggravated pulmonary edema and death rate, while anti-H4 antibody exerted obvious protective effects. Histone H4 directly activated the endothelia. Endothelial activation was evidently manifested as HS degradation, release of vWF, P-selectin translocation, and VE-Cadherin reduction. The synergistic stimulus of activated endothelia was required for effective neutrophil activation by histone H4. Both TLRs and calcium mediated histone H4-induced endothelial activation. CONCLUSIONS Histone H4 is a pro-inflammatory and pro-thrombotic molecule in OA-induced ARDS in mice.
Collapse
Affiliation(s)
- Yanlin Zhang
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China.
| | - Jingjin Tan
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Yiran Zhao
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Li Guan
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Shuqiang Li
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
12
|
Oriowo TO, Chrysostomakis I, Martin S, Kukowka S, Brown T, Winkler S, Myers EW, Böhne A, Stange M. A chromosome-level, haplotype-resolved genome assembly and annotation for the Eurasian minnow (Leuciscidae: Phoxinus phoxinus) provide evidence of haplotype diversity. Gigascience 2025; 14:giae116. [PMID: 39877992 PMCID: PMC11775470 DOI: 10.1093/gigascience/giae116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/06/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND In this study, we present an in-depth analysis of the Eurasian minnow (Phoxinus phoxinus) genome, highlighting its genetic diversity, structural variations, and evolutionary adaptations. We generated an annotated haplotype-phased, chromosome-level genome assembly (2n = 50) by integrating high-fidelity (HiFi) long reads and chromosome conformation capture data (Hi-C). RESULTS We achieved a haploid size of 940 megabase pairs (Mbp) for haplome 1 and 929 Mbp for haplome 2 with high scaffold N50 values of 36.4 Mb and 36.6 Mb and BUSCO scores of 96.9% and 97.2%, respectively, indicating a highly complete genome assembly. We detected notable heterozygosity (1.43%) and a high repeat content (approximately 54%), primarily consisting of DNA transposons, which contribute to genome rearrangements and variations. We found substantial structural variations within the genome, including insertions, deletions, inversions, and translocations. These variations affect genes enriched in functions such as dephosphorylation, developmental pigmentation, phagocytosis, immunity, and stress response. In the annotation of protein-coding genes, 30,980 messenger RNAs and 23,497 protein-coding genes were identified with a high completeness score, which further underpins the high contiguity of our genome assemblies. We performed a gene family evolution analysis by comparing our proteome to 10 other teleost species, which identified immune system gene families that prioritize histone-based disease prevention over NB-LRR-related-based immune responses. Additionally, demographic analysis indicates historical fluctuations in the effective population size of P. phoxinus, likely correlating with past climatic changes. CONCLUSIONS This annotated, phased reference genome provides a crucial resource for resolving the taxonomic complexity within the genus Phoxinus and highlights the importance of haplotype-phased assemblies in understanding haplotype diversity in species characterized by high heterozygosity.
Collapse
Affiliation(s)
- Temitope Opeyemi Oriowo
- Leibniz Institute for the Analysis of Biodiversity Change, Museum Koenig Bonn, 53113 Bonn, Germany
| | - Ioannis Chrysostomakis
- Leibniz Institute for the Analysis of Biodiversity Change, Museum Koenig Bonn, 53113 Bonn, Germany
| | - Sebastian Martin
- Leibniz Institute for the Analysis of Biodiversity Change, Museum Koenig Bonn, 53113 Bonn, Germany
| | - Sandra Kukowka
- Leibniz Institute for the Analysis of Biodiversity Change, Museum Koenig Bonn, 53113 Bonn, Germany
| | - Thomas Brown
- Max Planck Institute of Molecular Cell Biology and Genetics, Sequencing and Genotyping, 01307 Dresden, Germany
| | - Sylke Winkler
- Max Planck Institute of Molecular Cell Biology and Genetics, Sequencing and Genotyping, 01307 Dresden, Germany
| | - Eugene W Myers
- Max Planck Institute of Molecular Cell Biology and Genetics, Sequencing and Genotyping, 01307 Dresden, Germany
- Okinawa Institute of Science and Technology, Algorithms for Ecological and Evolutionary Genomics, Okinawa 904-0412, Japan
| | - Astrid Böhne
- Leibniz Institute for the Analysis of Biodiversity Change, Museum Koenig Bonn, 53113 Bonn, Germany
| | - Madlen Stange
- Leibniz Institute for the Analysis of Biodiversity Change, Museum Koenig Bonn, 53113 Bonn, Germany
| |
Collapse
|
13
|
Xu JQ, Zhang WY, Fu JJ, Fang XZ, Gao CG, Li C, Yao L, Li QL, Yang XB, Ren LH, Shu HQ, Peng K, Wu Y, Zhang DY, Qiu Y, Zhou X, Yao YM, Shang Y. Viral sepsis: diagnosis, clinical features, pathogenesis, and clinical considerations. Mil Med Res 2024; 11:78. [PMID: 39676169 PMCID: PMC11648306 DOI: 10.1186/s40779-024-00581-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
Sepsis, characterized as life-threatening organ dysfunction resulting from dysregulated host responses to infection, remains a significant challenge in clinical practice. Despite advancements in understanding host-bacterial interactions, molecular responses, and therapeutic approaches, the mortality rate associated with sepsis has consistently ranged between 10 and 16%. This elevated mortality highlights critical gaps in our comprehension of sepsis etiology. Traditionally linked to bacterial and fungal pathogens, recent outbreaks of acute viral infections, including Middle East respiratory syndrome coronavirus (MERS-CoV), influenza virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), among other regional epidemics, have underscored the role of viral pathogenesis in sepsis, particularly when critically ill patients exhibit classic symptoms indicative of sepsis. However, many cases of viral-induced sepsis are frequently underdiagnosed because standard evaluations typically exclude viral panels. Moreover, these viruses not only activate conventional pattern recognition receptors (PRRs) and retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) but also initiate primary antiviral pathways such as cyclic guanosine monophosphate adenosine monophosphate (GMP-AMP) synthase (cGAS)-stimulator of interferon genes (STING) signaling and interferon response mechanisms. Such activations lead to cellular stress, metabolic disturbances, and extensive cell damage that exacerbate tissue injury while leading to a spectrum of clinical manifestations. This complexity poses substantial challenges for the clinical management of affected cases. In this review, we elucidate the definition and diagnosis criteria for viral sepsis while synthesizing current knowledge regarding its etiology, epidemiology, and pathophysiology, molecular mechanisms involved therein as well as their impact on immune-mediated organ damage. Additionally, we discuss clinical considerations related to both existing therapies and advanced treatment interventions, aiming to enhance the comprehensive understanding surrounding viral sepsis.
Collapse
Affiliation(s)
- Ji-Qian Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wan-Ying Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jia-Ji Fu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiang-Zhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cheng-Gang Gao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chang Li
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lu Yao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qi-Lan Li
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao-Bo Yang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Le-Hao Ren
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hua-Qing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ke Peng
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 43007, China
| | - Ying Wu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School, Wuhan University, Wuhan, 430072, China
| | - Ding-Yu Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yang Qiu
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 43007, China
| | - Xi Zhou
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 43007, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and the Fourth Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China.
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
14
|
Grinat J, Shriever NP, Christophorou MA. Fantastic proteins and where to find them - histones, in the nucleus and beyond. J Cell Sci 2024; 137:jcs262071. [PMID: 39704565 PMCID: PMC11827605 DOI: 10.1242/jcs.262071] [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: 12/21/2024] Open
Abstract
Animal genomes are packaged into chromatin, a highly dynamic macromolecular structure of DNA and histone proteins organised into nucleosomes. This accommodates packaging of lengthy genomic sequences within the physical confines of the nucleus while also enabling precise regulation of access to genetic information. However, histones existed before chromatin and have lesser-known functions beyond genome regulation. Most notably, histones are potent antimicrobial agents, and the release of chromatin to the extracellular space is a defence mechanism nearly as ancient and widespread as chromatin itself. Histone sequences have changed very little throughout evolution, suggesting the possibility that some of their 'non-canonical' functions are at play in parallel or in concert with their genome regulatory functions. In this Review, we take an evolutionary perspective of histone, nuclear chromatin and extracellular chromatin biology and describe the known extranuclear and extracellular functions of histones. We detail molecular mechanisms of chromatin release and extracellular chromatin sensing, and we discuss their roles in physiology and disease. Finally, we present evidence and give a perspective on the potential of extracellular histones to act as bioactive, cell modulatory factors.
Collapse
|
15
|
Baranova IN, Bocharov AV, Vishnyakova TG, Chen Z, Ke Y, Birukova AA, Yuen PST, Tsuji T, Star RA, Birukov KG, Patterson AP, Eggerman TL. Class B Scavenger Receptor CD36 as a Potential Therapeutic Target in Inflammation Induced by Danger-Associated Molecular Patterns. Cells 2024; 13:1992. [PMID: 39682740 PMCID: PMC11640246 DOI: 10.3390/cells13231992] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The class B scavenger receptor CD36 is known to bind and mediate the transport of lipid-related ligands and it functions as a pattern recognition receptor (PRR) for a variety of pathogens, including bacteria and viruses. In this study, we assessed CD36's role as a PRR mediating pro-inflammatory effects of several known Danger-Associated Molecular Patterns (DAMPs) used either as a single preparation or as a combination of DAMPs in the form of total cell/skeletal muscle tissue lysates. Our data demonstrated that multiple DAMPs, including HMGB1, HSPs, histone H3, SAA, and oxPAPC, as well as cell/tissue lysate preparations, induced substantially higher (~7-10-fold) IL-8 cytokine responses in HEK293 cells overexpressing CD36 compared to control WT cells. At the same time, DAMP-induced secretion of IL-6 in bone marrow-derived macrophages (BMDM) from CD36-/- mice was markedly (~2-3 times) reduced, as compared to macrophages from normal mice. Synthetic amphipathic helical peptides (SAHPs), known CD36 ligands, efficiently blocked CD36-dependent inflammatory responses induced by both cell and tissue lysates, HMGB1 and histone H3 in CD36+ cells. IP injection of total cellular lysate preparation induced inflammatory responses that were assessed by the expression of liver and lung pro-inflammatory markers, including IL-6, TNF-α, CD68, and CXCL1, and was reduced by ~50% in CD36-deficient mice compared to normal mice. Our findings demonstrate that CD36 is a PRR contributing to the innate immune response via mediating DAMP-induced inflammatory signaling and highlight the importance of this receptor as a potential therapeutic target in DAMP-associated inflammatory conditions.
Collapse
Affiliation(s)
- Irina N. Baranova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; (A.V.B.); (T.G.V.); (Z.C.); (A.P.P.); (T.L.E.)
| | - Alexander V. Bocharov
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; (A.V.B.); (T.G.V.); (Z.C.); (A.P.P.); (T.L.E.)
| | - Tatyana G. Vishnyakova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; (A.V.B.); (T.G.V.); (Z.C.); (A.P.P.); (T.L.E.)
| | - Zhigang Chen
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; (A.V.B.); (T.G.V.); (Z.C.); (A.P.P.); (T.L.E.)
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (Y.K.); (K.G.B.)
| | - Anna A. Birukova
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Peter S. T. Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (P.S.T.Y.); (T.T.); (R.A.S.)
| | - Takayuki Tsuji
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (P.S.T.Y.); (T.T.); (R.A.S.)
| | - Robert A. Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (P.S.T.Y.); (T.T.); (R.A.S.)
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (Y.K.); (K.G.B.)
| | - Amy P. Patterson
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; (A.V.B.); (T.G.V.); (Z.C.); (A.P.P.); (T.L.E.)
- Office of the Director, Division of Program Coordination, Planning and Strategic Initiatives, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas L. Eggerman
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; (A.V.B.); (T.G.V.); (Z.C.); (A.P.P.); (T.L.E.)
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
Al-Aqtash R, Collier DM. Ionotropic purinergic receptor 7 (P2X7) channel structure and pharmacology provides insight regarding non-nucleotide agonism. Channels (Austin) 2024; 18:2355150. [PMID: 38762911 PMCID: PMC11110710 DOI: 10.1080/19336950.2024.2355150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/10/2024] [Indexed: 05/21/2024] Open
Abstract
P2X7 is a member of the Ionotropic Purinergic Receptor (P2X) family. The P2X family of receptors is composed of seven (P2X1-7), ligand-gated, nonselective cation channels. Changes in P2X expression have been reported in multiple disease models. P2Xs have large complex extracellular domains that function as receptors for a variety of ligands, including endogenous and synthetic agonists and antagonists. ATP is the canonical agonist. ATP affinity ranges from nanomolar to micromolar for most P2XRs, but P2X7 has uniquely poor ATP affinity. In many physiological settings, it may be difficult to achieve the millimolar extracellular ATP concentrations needed for P2X7 channel activation; however, channel function is implicated in pain sensation, immune cell function, cardiovascular disease, cancer, and osteoporosis. Multiple high-resolution P2X7 structures have been solved in apo-, ATP-, and antagonist-bound states. P2X7 structural data reveal distinct allosteric and orthosteric antagonist-binding sites. Both allosteric and orthosteric P2X7 antagonists are well documented to inhibit ATP-evoked channel current. However, a growing body of evidence supports P2X7 activation by non-nucleotide agonists, including extracellular histone proteins and human cathelicidin-derived peptides (LL-37). Interestingly, P2X7 non-nucleotide agonism is not inhibited by allosteric antagonists, but is inhibited by orthosteric antagonists. Herein, we review P2X7 function with a focus on the efficacy of available pharmacology on P2X7 channel current activation by non-nucleotide agonists in effort to understand agonist/antagonist efficacy, and consider the impact of these data on the current understanding of P2X7 in physiology and disease given these limitations of P2X7-selective antagonists and incomplete knockout mouse models.
Collapse
Affiliation(s)
- Rua’a Al-Aqtash
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Daniel M. Collier
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
17
|
Young BD, Williams DE, Bright AJ, Peterson A, Traylor-Knowles N, Rosales SM. Genet identity and season drive gene expression in outplanted Acropora palmata at different reef sites. Sci Rep 2024; 14:29444. [PMID: 39604459 PMCID: PMC11603135 DOI: 10.1038/s41598-024-80479-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: 04/12/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
Coral reefs are experiencing decreases in coral cover due to anthropogenic influences. Coral restoration is addressing this decline by outplanting large volumes of corals onto reef systems. Understanding how outplanted corals react at a transcriptomic level to different outplant locations over time is important, as it will highlight how habitat affects the coral host and influences physiological measures. In this study, the transcriptomic dynamics of four genets of outplanted Acropora palmata were assessed over a year at three reef sites in the Florida Keys. Genet identity was more important than time of sampling or outplant site, with differing levels of baseline immune and protein production the key drivers. Once accounting for genet, enriched growth processes were identified in the winter, and increased survival and immune expression were found in the summer. The effect of the reef site was small, with hypothesized differences in autotrophic versus heterotrophic dependent on outplant depth. We hypothesize that genotype identity is an important consideration for reef restoration, as differing baseline gene expression could play a role in survivorship and growth. Additionally, outplanting during cooler winter months may be beneficial due to higher expression of growth processes, allowing establishment of outplants on the reef system.
Collapse
Affiliation(s)
- Benjamin D Young
- Cooperative Institute of Marine and Atmospheric Science, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, Miami, FL, USA.
- Atlantic Oceanographic and Meteorological Laboratory, National Oceanic and Atmospheric Administration, Miami, FL, USA.
- Department of Marine Biology and Ecology, Rosenstiel School of Marine, Atmospheric and Earth Science, University of Miami, Miami, FL, USA.
| | - Dana E Williams
- Cooperative Institute of Marine and Atmospheric Science, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, Miami, FL, USA
- Southeast Fisheries Science Center, National Oceanic and Atmospheric Administration-National Marine Fisheries Service, Miami, FL, USA
| | - Allan J Bright
- Cooperative Institute of Marine and Atmospheric Science, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, Miami, FL, USA
- Southeast Fisheries Science Center, National Oceanic and Atmospheric Administration-National Marine Fisheries Service, Miami, FL, USA
| | - Annie Peterson
- Cooperative Institute of Marine and Atmospheric Science, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, Miami, FL, USA
- Southeast Fisheries Science Center, National Oceanic and Atmospheric Administration-National Marine Fisheries Service, Miami, FL, USA
| | - Nikki Traylor-Knowles
- Department of Marine Biology and Ecology, Rosenstiel School of Marine, Atmospheric and Earth Science, University of Miami, Miami, FL, USA
| | - Stephane M Rosales
- Cooperative Institute of Marine and Atmospheric Science, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, Miami, FL, USA
- Atlantic Oceanographic and Meteorological Laboratory, National Oceanic and Atmospheric Administration, Miami, FL, USA
| |
Collapse
|
18
|
Arévalo-Jaimes BV, Salinas-Pena M, Ponte I, Jordan A, Roque A, Torrents E. Antimicrobial and antibiofilm activity of human recombinant H1 histones against bacterial infections. mSystems 2024; 9:e0070424. [PMID: 39470247 PMCID: PMC11575268 DOI: 10.1128/msystems.00704-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
Histones possess significant antimicrobial potential, yet their activity against biofilms remains underexplored. Moreover, concerns regarding adverse effects limit their clinical implementation. We investigated the antibacterial efficacy of human recombinant histone H1 subtypes against Pseudomonas aeruginosa PAO1, both planktonic and in biofilms. After the in vitro tests, toxicity and efficacy were assessed in a P. aeruginosa PAO1 infection model using Galleria mellonella larvae. Histones were also evaluated in combination with ciprofloxacin (Cpx) and gentamicin (Gm). Our results demonstrate antimicrobial activity of all three histones against P. aeruginosa PAO1, with H1.0 and H1.4 showing efficacy at lower concentrations. The bactericidal effect was associated with a mechanism of membrane disruption. In vitro studies using static and dynamic models showed that H1.4 had antibiofilm potential by reducing cell biomass. Neither H1.0 nor H1.4 showed toxicity in G. mellonella larvae, and both increased larvae survival when infected with P. aeruginosa PAO1. Although in vitro synergism was observed between ciprofloxacin and H1.0, no improvement over the antibiotic alone was noted in vivo. Differences in antibacterial and antibiofilm activity were attributed to sequence and structural variations among histone subtypes. Moreover, the efficacy of H1.0 and H1.4 was influenced by the presence and strength of the extracellular matrix. These findings suggest histones hold promise for combating acute and chronic infections caused by pathogens such as P. aeruginosa.IMPORTANCEThe constant increase of multidrug-resistant bacteria is a critical global concern. The inefficacy of current therapies to treat bacterial infections is attributed to multiple mechanisms of resistance, including the capacity to form biofilms. Therefore, the identification of novel and safe therapeutic strategies is imperative. This study confirms the antimicrobial potential of three histone H1 subtypes against both Gram-negative and Gram-positive bacteria. Furthermore, histones H1.0 and H1.4 demonstrated in vivo efficacy without associated toxicity in an acute infection model of Pseudomonas aeruginosa PAO1 in Galleria mellonella larvae. The bactericidal effect of these proteins also resulted in biomass reduction of P. aeruginosa PAO1 biofilms. Given the clinical significance of this opportunistic pathogen, our research provides a comprehensive initial evaluation of the efficacy, toxicity, and mechanism of action of a potential new therapeutic approach against acute and chronic bacterial infections.
Collapse
Affiliation(s)
- Betsy Verónica Arévalo-Jaimes
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | | | - Inmaculada Ponte
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Albert Jordan
- Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Alicia Roque
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Eduard Torrents
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Gholipourshahraki T, Bai Z, Shrestha M, Hjelholt A, Hu S, Kjolby M, Rohde PD, Sørensen P. Evaluation of Bayesian Linear Regression models for gene set prioritization in complex diseases. PLoS Genet 2024; 20:e1011463. [PMID: 39495786 PMCID: PMC11563439 DOI: 10.1371/journal.pgen.1011463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/14/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024] Open
Abstract
Genome-wide association studies (GWAS) provide valuable insights into the genetic architecture of complex traits, yet interpreting their results remains challenging due to the polygenic nature of most traits. Gene set analysis offers a solution by aggregating genetic variants into biologically relevant pathways, enhancing the detection of coordinated effects across multiple genes. In this study, we present and evaluate a gene set prioritization approach utilizing Bayesian Linear Regression (BLR) models to uncover shared genetic components among different phenotypes and facilitate biological interpretation. Through extensive simulations and analyses of real traits, we demonstrate the efficacy of the BLR model in prioritizing pathways for complex traits. Simulation studies reveal insights into the model's performance under various scenarios, highlighting the impact of factors such as the number of causal genes, proportions of causal variants, heritability, and disease prevalence. Comparative analyses with MAGMA (Multi-marker Analysis of GenoMic Annotation) demonstrate BLR's superior performance, especially in highly overlapped gene sets. Application of both single-trait and multi-trait BLR models to real data, specifically GWAS summary data for type 2 diabetes (T2D) and related phenotypes, identifies significant associations with T2D-related pathways. Furthermore, comparison between single- and multi-trait BLR analyses highlights the superior performance of the multi-trait approach in identifying associated pathways, showcasing increased statistical power when analyzing multiple traits jointly. Additionally, enrichment analysis with integrated data from various public resources supports our results, confirming significant enrichment of diabetes-related genes within the top T2D pathways resulting from the multi-trait analysis. The BLR model's ability to handle diverse genomic features, perform regularization, conduct variable selection, and integrate information from multiple traits, genders, and ancestries demonstrates its utility in understanding the genetic architecture of complex traits. Our study provides insights into the potential of the BLR model to prioritize gene sets, offering a flexible framework applicable to various datasets. This model presents opportunities for advancing personalized medicine by exploring the genetic underpinnings of multifactorial traits.
Collapse
Affiliation(s)
| | - Zhonghao Bai
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Merina Shrestha
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Astrid Hjelholt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Sile Hu
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, United Kingdom
| | - Mads Kjolby
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Palle Duun Rohde
- Genomic Medicine, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Peter Sørensen
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
20
|
Masnikosa R, Cvetković Z, Pirić D. Tumor Biology Hides Novel Therapeutic Approaches to Diffuse Large B-Cell Lymphoma: A Narrative Review. Int J Mol Sci 2024; 25:11384. [PMID: 39518937 PMCID: PMC11545713 DOI: 10.3390/ijms252111384] [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: 07/14/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a malignancy of immense biological and clinical heterogeneity. Based on the transcriptomic or genomic approach, several different classification schemes have evolved over the years to subdivide DLBCL into clinically (prognostically) relevant subsets, but each leaves unclassified samples. Herein, we outline the DLBCL tumor biology behind the actual and potential drug targets and address the challenges and drawbacks coupled with their (potential) use. Therapeutic modalities are discussed, including small-molecule inhibitors, naked antibodies, antibody-drug conjugates, chimeric antigen receptors, bispecific antibodies and T-cell engagers, and immune checkpoint inhibitors. Candidate drugs explored in ongoing clinical trials are coupled with diverse toxicity issues and refractoriness to drugs. According to the literature on DLBCL, the promise for new therapeutic targets lies in epigenetic alterations, B-cell receptor and NF-κB pathways. Herein, we present putative targets hiding in lipid pathways, ferroptosis, and the gut microbiome that could be used in addition to immuno-chemotherapy to improve the general health status of DLBCL patients, thus increasing the chance of being cured. It may be time to devote more effort to exploring DLBCL metabolism to discover novel druggable targets. We also performed a bibliometric and knowledge-map analysis of the literature on DLBCL published from 2014-2023.
Collapse
Affiliation(s)
- Romana Masnikosa
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| | - Zorica Cvetković
- Department of Hematology, Clinical Hospital Centre Zemun, Vukova 9, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, 11000 Belgrade, Serbia
| | - David Pirić
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| |
Collapse
|
21
|
Li Y, Yadollahi P, Essien F, Putluri V, Chandra S, Kami Reddy KR, Kamal A, Putluri N, Abdurrahman LM, Ruiz-Echartea E, Ernste K, Trivedi A, Vazquez-Perez J, Hudson WH, Decker W, Patel R, Osman AA, Kheradmand F, Lai SY, Myers JN, Skinner HD, Coarfa C, Lee K, Jain A, Malovannaya A, Frederick MJ, Sandulache VC. Tobacco smoke exposure is a driver of altered oxidative stress response and immunity in head and neck cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618907. [PMID: 39484602 PMCID: PMC11526855 DOI: 10.1101/2024.10.17.618907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Purpose Exposomes are critical drivers of carcinogenesis. However, how they modulate tumor behavior remains unclear. Extensive clinical data link cigarette smoke as a key exposome that promotes aggressive tumors, higher rates of metastasis, reduced response to chemoradiotherapy, and suppressed anti-tumor immunity. We sought to determine whether smoke itself can modulate aggressive tumor behavior in head and neck squamous cell carcinoma (HNSCC) through reprogramming the cellular reductive state. Experimental design Using established human and murine HNSCC cell lines and syngeneic mouse models, we utilized conventional western blotting, steady state and flux metabolomics, RNA sequencing, quantitative proteomics and flow cytometry to analyze the impact of smoke exposure on HNSCC tumor biology. Results Cigarette smoke persistently activated Nrf2 target genes essential for maintenance of the cellular reductive state and survival under conditions of increased oxidative stress in HNSCC regardless of HPV status. In contrast to e-cigarette vapor, conventional cigarette smoke mobilizes cellular metabolism toward oxidative stress adaptation, resulting in development of cross-resistance to cisplatin. In parallel, smoke exposure modulates both expression of PDL1 and the secretory phenotype of HNSCC cells through activation of NF-κB resulting in an altered tumor immune microenvironment (TIME) in syngeneic mouse models and altered PBMC differentiation that includes downregulated expression of antigen presentation and costimulatory genes in myeloid cells. Conclusion Cigarette smoke exposome is a potent activator of the Nrf2 pathway and is a likely primary trigger for the tripartite phenotype of aggressive HNSCC consisting of: 1) reduced chemotherapy sensitivity, 2) enhanced metastatic potential and 3) suppressed anti-tumor immunity. Statement of significance The smoke exposome drives aggressive tumor behavior, treatment resistance and suppressed immunity through coordinated metabolic reprogramming. Successfully targeting this adaptation is critical to improving survival in smokers with head and neck cancer.
Collapse
|
22
|
Brummerhop AS, Lee CT, Weltman R, Tribble GD, van der Hoeven R, Chiu Y, Hong J, Wang BY. Synergistic effects of antimicrobial components of the human-derived composite amnion-chorion membrane on bacterial growth. Front Cell Infect Microbiol 2024; 14:1472737. [PMID: 39435187 PMCID: PMC11491435 DOI: 10.3389/fcimb.2024.1472737] [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: 07/29/2024] [Accepted: 09/17/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction The human-derived amnion-chorion membrane (ACM) has endogenous antimicrobial properties, which are important for preventing the colonization and survival of oral bacteria on exposed membranes. This project aimed to decipher the underlying mechanism by identifying the components of ACM that confer antibacterial properties. In addition, the antimicrobial efficacy of these identified components on oral bacteria was assessed. Methods Four antimicrobial proteins, histone H2A/H2B, cathelicidin LL-37, lactoferrin, and lysozyme, were identified via mass spectrometry in ACM. These proteins were then assessed for their efficacy in killing Streptococcus gordonii Challis. Log-phased bacterial cells were cultured with the commercially available proteins that were identified in ACM, either individually or in combination, at different concentrations. After incubation for 8 or 24 hours, the bacteria were stained with a live/dead viability kit and analyzed via confocal microscopy. Results The combination of these proteins effectively killed S. gordonii in a dose-dependent fashion after 8 or 24 hours of incubation. When each protein was tested individually, it killed S. gordonii at a much lower efficacy relative to the combinations. The synergistic effects of the antimicrobial protein combinations were also observed in both the viable cell count recovery and minimum inhibitory concentration assays. Discussion By shedding light on the mechanisms in the ACM's antimicrobial property, this study may raise more awareness of the potential benefit of utilization of a membrane with endogenous antimicrobial properties in regeneration surgeries.
Collapse
Affiliation(s)
- Alexandra Su Brummerhop
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| | - Chun-Teh Lee
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| | - Robin Weltman
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
- Department of Clinical Sciences, University of Nevada School of Dental Medicine, Las Vegas, NV, United States
| | - Gena D. Tribble
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| | - Ransome van der Hoeven
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA, United States
- Department of Periodontics, University of Iowa College of Dentistry, Iowa City, IA, United States
| | - Yulun Chiu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jianming Hong
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| | - Bing-Yan Wang
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| |
Collapse
|
23
|
Bellomo R, Patava J, Van Lancker R, Layios N, Peetermans M, Plummer M, Attou R, McNamara R, Udy A, Wibrow B, Deane A, Litton E, Tanudji M, Su F, Zhong Z, Shi L, Ning L. A dose-adjusted, open-label, pilot study of the safety, tolerability, and pharmacokinetics of STC3141 in critically ill patients with sepsis. Pharmacol Res Perspect 2024; 12:e70015. [PMID: 39401149 PMCID: PMC11472796 DOI: 10.1002/prp2.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/21/2024] [Accepted: 09/01/2024] [Indexed: 10/17/2024] Open
Abstract
Increased circulating histones correlate with sepsis severity and are a potential therapeutic target. Pre-clinical studies showed benefit with a histone-neutralizing polyanion molecule (STC3141). We aimed to investigate the safety, tolerability, and pharmacokinetics of STC3141 in critically ill patients with sepsis. We studied 26 patients with sepsis divided into four cohorts of one, five, ten, and ten subjects, respectively. We conducted a dose-adjusted, open-label study to determine the safety, tolerability, and pharmacokinetics of STC3141 administered as an IV infusion for up to 72 h, with rate adjusted to estimated creatinine clearance. Four steady-state concentrations were targeted. Twenty of the 26 subjects (77%) in the study experienced at least one adverse event (AE). The most frequently reported study drug-related AE was a mildly prolonged aPTT (four events). Only one AE (pulmonary hemorrhage) led to discontinuation of the drug. After excluding patients receiving renal replacement therapy (RRT) patients, clearance ranged from 3.3 to 4.2 L/h across cohorts and was essentially completely renal in nature. Half-life values ranged from 5 to 7 h. The mean (±SD) terminal half-life for non-RRT subjects and for whom it was possible to calculate was approximately 9 (±4.77) h but increased to 19 (±7.94) h for subjects on RRT. Overall, 18 (69.2%) patients completed the study to day eight in the ICU, and 22 (84.6%) survived to 28 days. STC3141 administration appeared to have an acceptable degree of safety and tolerability and expected pharmacokinetics. Cautious, larger randomized efficacy trials in sepsis appear justified.
Collapse
Affiliation(s)
| | - John Patava
- Grand Medical Pty LtdSt LeonardsNew South WalesAustralia
| | | | - Nathalie Layios
- C.H.U Liège, Sart TilmanDomaine Universitaire du Sart Tilman—B35Liège 1Belgium
| | | | - Mark Plummer
- Royal Adelaide HospitalAdelaideSouth AustraliaAustralia
| | | | | | - Andrew Udy
- Alfred HospitalMelbourneVictoriaAustralia
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Bradley Wibrow
- Sir Charles Gairdner HospitalNedlandsWestern AustraliaAustralia
| | - Adam Deane
- Department of Critical Care, Melbourne Medical SchoolUniversity of MelbourneParkvilleVictoriaAustralia
| | - Edward Litton
- Fiona Stanley HospitalMurdochWestern AustraliaAustralia
| | - Marcel Tanudji
- Grand Medical Pty LtdSt LeonardsNew South WalesAustralia
| | - Fuhong Su
- Grand Medical Pty LtdSt LeonardsNew South WalesAustralia
| | - Zhang Zhong
- Grand Medical Pty LtdSt LeonardsNew South WalesAustralia
| | - Linda Shi
- Grand Medical Pty LtdSt LeonardsNew South WalesAustralia
| | - Li Ning
- Grand Medical Pty LtdSt LeonardsNew South WalesAustralia
| |
Collapse
|
24
|
Gézsi A, Antal P. GNN4DM: a graph neural network-based method to identify overlapping functional disease modules. Bioinformatics 2024; 40:btae573. [PMID: 39321259 PMCID: PMC11639141 DOI: 10.1093/bioinformatics/btae573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 09/27/2024] Open
Abstract
MOTIVATION Identifying disease modules within molecular interaction networks is an essential exploratory step in computational biology, offering insights into disease mechanisms and potential therapeutic targets. Traditional methods often struggle with the inherent complexity and overlapping nature of biological networks, and they are limited in effectively leveraging the vast amount of available genomic data and biological knowledge. This limitation underscores the need for more effective, automated approaches to integrate these rich data sources. RESULTS In this work, we propose GNN4DM, a novel graph neural network-based structured model that automates the discovery of overlapping functional disease modules. GNN4DM effectively integrates network topology with genomic data to learn the representations of the genes corresponding to functional modules and align these with known biological pathways for enhanced interpretability. Following the DREAM benchmark evaluation setting and extending with three independent data sources (GWAS Atlas, FinnGen, and DisGeNET), we show that GNN4DM performs better than several state-of-the-art methods in detecting biologically meaningful modules. Moreover, we demonstrate the method's applicability by discovering two novel multimorbidity modules significantly enriched across a diverse range of seemingly unrelated diseases. AVAILABILITY AND IMPLEMENTATION Source code, all training data, and all identified disease modules are freely available for download at https://github.com/gezsi/gnn4dm. GNN4DM was implemented in Python.
Collapse
Affiliation(s)
- András Gézsi
- Department of Artificial Intelligence and Systems Engineering, Budapest University of Technology and Economics, Budapest H-1117, Hungary
| | - Péter Antal
- Department of Artificial Intelligence and Systems Engineering, Budapest University of Technology and Economics, Budapest H-1117, Hungary
| |
Collapse
|
25
|
Song Y, Wu Y, Ding F, Li S, Shen Y, Yang B, Tang X, Ren L, Deng L, Jin X, Yan Y. The Preventive and Therapeutic Effects of Acute and Severe Inflammatory Disorders with Heparin and Heparinoid. Biomolecules 2024; 14:1078. [PMID: 39334845 PMCID: PMC11430252 DOI: 10.3390/biom14091078] [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: 07/18/2024] [Revised: 08/18/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Systematic inflammatory response syndrome (SIRS) and the accompanying sepsis pose a huge threat to human health worldwide. Heparin is a part of the standard supportive care for the disease. However, the molecular mechanism is not fully understood yet, and the potential signaling pathways that play key roles have not yet been elucidated. In this paper, the main findings regarding the molecular mechanisms associated with the beneficial effects of heparin, including inhibiting HMGB-1-driven inflammation reactions, histone-induced toxicity, thrombo-inflammatory response control and the new emerging mechanisms are concluded. To set up the link between the preclinical research and the clinical effects, the outcomes of the clinical trials are summarized. Then, the structure and function relationship of heparin is discussed. By providing an updated analysis of the above results, the paper highlights the feasibility of heparin as a possible alternative for sepsis prophylaxis and therapy.
Collapse
Affiliation(s)
- Ying Song
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Yuxiang Wu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Fangfang Ding
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Shuo Li
- Medi-X Pingshan, Southern University of Science and Technology, Shenzhen 518118, China
| | - Yaojia Shen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Bingyan Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Xinran Tang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Lige Ren
- Shenzhen Hepalink Pharmaceutical Group Co., Ltd., Shenzhen 518057, China
| | - Lirong Deng
- Shenzhen Hepalink Pharmaceutical Group Co., Ltd., Shenzhen 518057, China
| | - Xuewen Jin
- Shenzhen Hepalink Pharmaceutical Group Co., Ltd., Shenzhen 518057, China
| | - Yishu Yan
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
26
|
Ma X, Li J, Li M, Qi G, Wei L, Zhang D. Nets in fibrosis: Bridging innate immunity and tissue remodeling. Int Immunopharmacol 2024; 137:112516. [PMID: 38906006 DOI: 10.1016/j.intimp.2024.112516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
Fibrosis, a complex pathological process characterized by excessive deposition of extracellular matrix components, leads to tissue scarring and dysfunction. Emerging evidence suggests that neutrophil extracellular traps (NETs), composed of DNA, histones, and antimicrobial proteins, significantly contribute to fibrotic diseases pathogenesis. This review summarizes the process of NETs production, molecular mechanisms, and related diseases, and outlines the cellular and molecular mechanisms associated with fibrosis. Subsequently, this review comprehensively summarizes the current understanding of the intricate interplay between NETs and fibrosis across various organs, including the lung, liver, kidney, skin, and heart. The mechanisms by which NETs contribute to fibrogenesis, including their ability to promote inflammation, induce epithelial-mesenchymal transition (EMT), activate fibroblasts, deposit extracellular matrix (ECM) components, and trigger TLR4 signaling were explored. This review aimed to provide insights into the complex relationship between NETs and fibrosis via a comprehensive analysis of existing reports, offering novel perspectives for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Xueni Ma
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jipin Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Muyang Li
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Guoqing Qi
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lina Wei
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
27
|
Li P, Liu M, He WM. Integrated Transcriptomic Analysis Reveals Reciprocal Interactions between SARS-CoV-2 Infection and Multi-Organ Dysfunction, Especially the Correlation of Renal Failure and COVID-19. Life (Basel) 2024; 14:960. [PMID: 39202702 PMCID: PMC11355357 DOI: 10.3390/life14080960] [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: 06/11/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
The COVID-19 pandemic, which is caused by the SARS-CoV-2 virus, has resulted in extensive health challenges globally. While SARS-CoV-2 primarily targets the respiratory system, clinical studies have revealed that it could also affect multiple organs, including the heart, kidneys, liver, and brain, leading to severe complications. To unravel the intricate molecular interactions between the virus and host tissues, we performed an integrated transcriptomic analysis to investigate the effects of SARS-CoV-2 on various organs, with a particular focus on the relationship between renal failure and COVID-19. A comparative analysis showed that SARS-CoV-2 triggers a systemic immune response in the brain, heart, and kidney tissues, characterized by significant upregulation of cytokine and chemokine secretion, along with enhanced migration of lymphocytes and leukocytes. A weighted gene co-expression network analysis demonstrated that SARS-CoV-2 could also induce tissue-specific transcriptional profiling. More importantly, single-cell sequencing revealed that COVID-19 patients with renal failure exhibited lower metabolic activity in lung epithelial and B cells, with reduced ligand-receptor interactions, especially CD226 and ICAM, suggesting a compromised immune response. A trajectory analysis revealed that COVID-19 patients with renal failure exhibited less mature alveolar type 1 cells. Furthermore, these patients showed potential fibrosis in the hearts, liver, and lung increased extracellular matrix remodeling activities. However, there was no significant metabolic dysregulation in the liver of COVID-19 patients with renal failure. Candidate drugs prediction by Drug Signatures database and LINCS L1000 Antibody Perturbations Database underscored the importance of considering multi-organ effects in COVID-19 management and highlight potential therapeutic strategies, including targeting viral entry and replication, controlling tissue fibrosis, and alleviating inflammation.
Collapse
Affiliation(s)
- Pai Li
- Capricorn Partner, 3000 Leuven, Belgium
| | - Meng Liu
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Wei-Ming He
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| |
Collapse
|
28
|
Dhahri H, Saintilnord WN, Chandler D, Fondufe-Mittendorf YN. Beyond the Usual Suspects: Examining the Role of Understudied Histone Variants in Breast Cancer. Int J Mol Sci 2024; 25:6788. [PMID: 38928493 PMCID: PMC11203562 DOI: 10.3390/ijms25126788] [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: 05/21/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
The incorporation of histone variants has structural ramifications on nucleosome dynamics and stability. Due to their unique sequences, histone variants can alter histone-histone or histone-DNA interactions, impacting the folding of DNA around the histone octamer and the overall higher-order structure of chromatin fibers. These structural modifications alter chromatin compaction and accessibility of DNA by transcription factors and other regulatory proteins to influence gene regulatory processes such as DNA damage and repair, as well as transcriptional activation or repression. Histone variants can also generate a unique interactome composed of histone chaperones and chromatin remodeling complexes. Any of these perturbations can contribute to cellular plasticity and the progression of human diseases. Here, we focus on a frequently overlooked group of histone variants lying within the four human histone gene clusters and their contribution to breast cancer.
Collapse
Affiliation(s)
- Hejer Dhahri
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA or (H.D.); (W.N.S.)
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA;
| | - Wesley N. Saintilnord
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA or (H.D.); (W.N.S.)
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA;
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- The Edison Family Center of Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Darrell Chandler
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA;
| | | |
Collapse
|
29
|
Ocaña-Paredes B, Rivera-Orellana S, Ramírez-Sánchez D, Montalvo-Guerrero J, Freire MP, Espinoza-Ferrao S, Altamirano-Colina A, Echeverría-Espinoza P, Ramos-Medina MJ, Echeverría-Garcés G, Granda-Moncayo D, Jácome-Alvarado A, Andrade MG, López-Cortés A. The pharmacoepigenetic paradigm in cancer treatment. Front Pharmacol 2024; 15:1381168. [PMID: 38720770 PMCID: PMC11076712 DOI: 10.3389/fphar.2024.1381168] [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: 02/02/2024] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Abstract
Epigenetic modifications, characterized by changes in gene expression without altering the DNA sequence, play a crucial role in the development and progression of cancer by significantly influencing gene activity and cellular function. This insight has led to the development of a novel class of therapeutic agents, known as epigenetic drugs. These drugs, including histone deacetylase inhibitors, histone acetyltransferase inhibitors, histone methyltransferase inhibitors, and DNA methyltransferase inhibitors, aim to modulate gene expression to curb cancer growth by uniquely altering the epigenetic landscape of cancer cells. Ongoing research and clinical trials are rigorously evaluating the efficacy of these drugs, particularly their ability to improve therapeutic outcomes when used in combination with other treatments. Such combination therapies may more effectively target cancer and potentially overcome the challenge of drug resistance, a significant hurdle in cancer therapy. Additionally, the importance of nutrition, inflammation control, and circadian rhythm regulation in modulating drug responses has been increasingly recognized, highlighting their role as critical modifiers of the epigenetic landscape and thereby influencing the effectiveness of pharmacological interventions and patient outcomes. Epigenetic drugs represent a paradigm shift in cancer treatment, offering targeted therapies that promise a more precise approach to treating a wide spectrum of tumors, potentially with fewer side effects compared to traditional chemotherapy. This progress marks a step towards more personalized and precise interventions, leveraging the unique epigenetic profiles of individual tumors to optimize treatment strategies.
Collapse
Affiliation(s)
- Belén Ocaña-Paredes
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | | | - David Ramírez-Sánchez
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | | | - María Paula Freire
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | | | | | | | - María José Ramos-Medina
- German Cancer Research Center (DKFZ), Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Gabriela Echeverría-Garcés
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática, Instituto Nacional de Investigación en Salud Pública “Leopoldo Izquieta Pérez”, Quito, Ecuador
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago, Chile
| | | | - Andrea Jácome-Alvarado
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - María Gabriela Andrade
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| |
Collapse
|
30
|
Weinzapfel EN, Fedder-Semmes KN, Sun ZW, Keogh MC. Beyond the tail: the consequence of context in histone post-translational modification and chromatin research. Biochem J 2024; 481:219-244. [PMID: 38353483 PMCID: PMC10903488 DOI: 10.1042/bcj20230342] [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: 12/30/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
The role of histone post-translational modifications (PTMs) in chromatin structure and genome function has been the subject of intense debate for more than 60 years. Though complex, the discourse can be summarized in two distinct - and deceptively simple - questions: What is the function of histone PTMs? And how should they be studied? Decades of research show these queries are intricately linked and far from straightforward. Here we provide a historical perspective, highlighting how the arrival of new technologies shaped discovery and insight. Despite their limitations, the tools available at each period had a profound impact on chromatin research, and provided essential clues that advanced our understanding of histone PTM function. Finally, we discuss recent advances in the application of defined nucleosome substrates, the study of multivalent chromatin interactions, and new technologies driving the next era of histone PTM research.
Collapse
|
31
|
Sernoskie SC, Bonneil É, Thibault P, Jee A, Uetrecht J. Involvement of Extracellular Vesicles in the Proinflammatory Response to Clozapine: Implications for Clozapine-Induced Agranulocytosis. J Pharmacol Exp Ther 2024; 388:827-845. [PMID: 38262745 DOI: 10.1124/jpet.123.001970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/27/2023] [Accepted: 12/08/2023] [Indexed: 01/25/2024] Open
Abstract
Most idiosyncratic drug reactions (IDRs) appear to be immune-mediated, but mechanistic events preceding severe reaction onset remain poorly defined. Damage-associated molecular patterns (DAMPs) may contribute to both innate and adaptive immune phases of IDRs, and changes in extracellular vesicle (EV) cargo have been detected post-exposure to several IDR-associated drugs. To explore the hypothesis that EVs are also a source of DAMPs in the induction of the immune response preceding drug-induced agranulocytosis, the proteome and immunogenicity of clozapine- (agranulocytosis-associated drug) and olanzapine- (non-agranulocytosis-associated drug) exposed EVs were compared in two preclinical models: THP-1 macrophages and Sprague-Dawley rats. Compared with olanzapine, clozapine induced a greater increase in the concentration of EVs enriched from both cell culture media and rat serum. Moreover, treatment of drug-naïve THP-1 cells with clozapine-exposed EVs induced an inflammasome-dependent response, supporting a potential role for EVs in immune activation. Proteomic and bioinformatic analyses demonstrated an increased number of differentially expressed proteins with clozapine that were enriched in pathways related to inflammation, myeloid cell chemotaxis, wounding, transforming growth factor-β signaling, and negative regulation of stimuli response. These data indicate that, although clozapine and olanzapine exposure both alter the protein cargo of EVs, clozapine-exposed EVs carry mediators that exhibit significantly greater immunogenicity. Ultimately, this supports the working hypothesis that drugs associated with a risk of IDRs induce cell stress, release of proinflammatory mediators, and early immune activation that precedes severe reaction onset. Further studies characterizing EVs may elucidate biomarkers that predict IDR risk during development of drug candidates. SIGNIFICANCE STATEMENT: This work demonstrates that clozapine, an idiosyncratic drug-induced agranulocytosis (IDIAG)-associated drug, but not olanzapine, a safer structural analogue, induces an acute proinflammatory response and increases extracellular vesicle (EV) release in two preclinical models. Moreover, clozapine-exposed EVs are more immunogenic, as measured by their ability to activate inflammasomes, and contain more differentially expressed proteins, highlighting a novel role for EVs during the early immune response to clozapine and enhancing our mechanistic understanding of IDIAG and other idiosyncratic reactions.
Collapse
Affiliation(s)
- Samantha Christine Sernoskie
- Departments of Pharmaceutical Sciences, Faculty of Pharmacy (S.C.S., J.U.) and Pharmacology and Toxicology, Temerty Faculty of Medicine (A.J., J.U.), University of Toronto, Toronto, Ontario; and Institute for Research in Immunology and Cancer (É.B., P.T.) and Department of Chemistry (P.T.), University of Montreal, Montreal, Quebec
| | - Éric Bonneil
- Departments of Pharmaceutical Sciences, Faculty of Pharmacy (S.C.S., J.U.) and Pharmacology and Toxicology, Temerty Faculty of Medicine (A.J., J.U.), University of Toronto, Toronto, Ontario; and Institute for Research in Immunology and Cancer (É.B., P.T.) and Department of Chemistry (P.T.), University of Montreal, Montreal, Quebec
| | - Pierre Thibault
- Departments of Pharmaceutical Sciences, Faculty of Pharmacy (S.C.S., J.U.) and Pharmacology and Toxicology, Temerty Faculty of Medicine (A.J., J.U.), University of Toronto, Toronto, Ontario; and Institute for Research in Immunology and Cancer (É.B., P.T.) and Department of Chemistry (P.T.), University of Montreal, Montreal, Quebec
| | - Alison Jee
- Departments of Pharmaceutical Sciences, Faculty of Pharmacy (S.C.S., J.U.) and Pharmacology and Toxicology, Temerty Faculty of Medicine (A.J., J.U.), University of Toronto, Toronto, Ontario; and Institute for Research in Immunology and Cancer (É.B., P.T.) and Department of Chemistry (P.T.), University of Montreal, Montreal, Quebec
| | - Jack Uetrecht
- Departments of Pharmaceutical Sciences, Faculty of Pharmacy (S.C.S., J.U.) and Pharmacology and Toxicology, Temerty Faculty of Medicine (A.J., J.U.), University of Toronto, Toronto, Ontario; and Institute for Research in Immunology and Cancer (É.B., P.T.) and Department of Chemistry (P.T.), University of Montreal, Montreal, Quebec
| |
Collapse
|
32
|
Cicchinelli S, Pignataro G, Gemma S, Piccioni A, Picozzi D, Ojetti V, Franceschi F, Candelli M. PAMPs and DAMPs in Sepsis: A Review of Their Molecular Features and Potential Clinical Implications. Int J Mol Sci 2024; 25:962. [PMID: 38256033 PMCID: PMC10815927 DOI: 10.3390/ijms25020962] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Sepsis is a serious organ dysfunction caused by a dysregulated immune host reaction to a pathogen. The innate immunity is programmed to react immediately to conserved molecules, released by the pathogens (PAMPs), and the host (DAMPs). We aimed to review the molecular mechanisms of the early phases of sepsis, focusing on PAMPs, DAMPs, and their related pathways, to identify potential biomarkers. We included studies published in English and searched on PubMed® and Cochrane®. After a detailed discussion on the actual knowledge of PAMPs/DAMPs, we analyzed their role in the different organs affected by sepsis, trying to elucidate the molecular basis of some of the most-used prognostic scores for sepsis. Furthermore, we described a chronological trend for the release of PAMPs/DAMPs that may be useful to identify different subsets of septic patients, who may benefit from targeted therapies. These findings are preliminary since these pathways seem to be strongly influenced by the peculiar characteristics of different pathogens and host features. Due to these reasons, while initial findings are promising, additional studies are necessary to clarify the potential involvement of these molecular patterns in the natural evolution of sepsis and to facilitate their transition into the clinical setting.
Collapse
Affiliation(s)
- Sara Cicchinelli
- Department of Emergency, S.S. Filippo e Nicola Hospital, 67051 Avezzano, Italy;
| | - Giulia Pignataro
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Stefania Gemma
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Andrea Piccioni
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Domitilla Picozzi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Veronica Ojetti
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Francesco Franceschi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| |
Collapse
|
33
|
Galieva A, Egorov A, Malogolovkin A, Brovin A, Karabelsky A. RNA-Seq Analysis of Trans-Differentiated ARPE-19 Cells Transduced by AAV9-AIPL1 Vectors. Int J Mol Sci 2023; 25:197. [PMID: 38203368 PMCID: PMC10778816 DOI: 10.3390/ijms25010197] [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: 11/18/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Inherited retinal disorders (IRD) have become a primary focus of gene therapy research since the success of adeno-associated virus-based therapeutics (voretigene neparvovec-rzyl) for Leber congenital amaurosis type 2 (LCA2). Dozens of monogenic IRDs could be potentially treated with a similar approach using an adeno-associated virus (AAV) to transfer a functional gene into the retina. Here, we present the results of the design, production, and in vitro testing of the AAV serotype 9 (AAV9) vector carrying the codon-optimized (co) copy of aryl hydrocarbon receptor-interacting protein like-1 (AIPL1) as a possible treatment for LCA4. The pAAV-AIPL1co was able to successfully transduce retinal pigment epithelium cells (ARPE-19) and initiate the expression of human AIPL1. Intriguingly, cells transduced with AAV9-AIPL1co showed much less antiviral response than AAV9-AIPL1wt (wild-type AIPL1) transduced. RNA-sequencing (RNA-seq) analysis of trans-differentiated ARPE-19 cells transduced with AAV9-AIPL1co demonstrated significant differences in the expression of genes involved in the innate immune response. In contrast, AAV9-AIPL1wt induced the prominent activation of multiple interferon-stimulated genes. The key part of the possible regulatory molecular mechanism is the activation of dsRNA-responsive antiviral oligoadenylate synthetases, and a significant increase in the level of histone coding genes' transcripts overrepresented in RNA-seq data (i.e., H1, H2A, H2B, H3, and H4). The RNA-seq data suggests that AAV9-AIPL1co exhibiting less immunogenicity than AAV9-AIPL1wt can be used for potency testing, using relevant animal models to develop future therapeutics for LCA4.
Collapse
Affiliation(s)
- Alima Galieva
- Gene Therapy Department, Science Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (A.G.); (A.M.); (A.B.)
| | - Alexander Egorov
- Gene Therapy Department, Science Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (A.G.); (A.M.); (A.B.)
| | - Alexander Malogolovkin
- Gene Therapy Department, Science Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (A.G.); (A.M.); (A.B.)
- Molecular Virology Laboratory, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Andrew Brovin
- Gene Therapy Department, Science Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (A.G.); (A.M.); (A.B.)
| | - Alexander Karabelsky
- Gene Therapy Department, Science Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (A.G.); (A.M.); (A.B.)
| |
Collapse
|
34
|
Castellanos FX, Moreno-Santillán D, Hughes GM, Paulat NS, Sipperly N, Brown AM, Martin KR, Poterewicz GM, Lim MCW, Russell AL, Moore MS, Johnson MG, Corthals AP, Ray DA, Dávalos LM. The evolution of antimicrobial peptides in Chiroptera. Front Immunol 2023; 14:1250229. [PMID: 37822944 PMCID: PMC10562630 DOI: 10.3389/fimmu.2023.1250229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Abstract
High viral tolerance coupled with an extraordinary regulation of the immune response makes bats a great model to study host-pathogen evolution. Although many immune-related gene gains and losses have been previously reported in bats, important gene families such as antimicrobial peptides (AMPs) remain understudied. We built an exhaustive bioinformatic pipeline targeting the major gene families of defensins and cathelicidins to explore AMP diversity and analyze their evolution and distribution across six bat families. A combination of manual and automated procedures identified 29 AMP families across queried species, with α-, β-defensins, and cathelicidins representing around 10% of AMP diversity. Gene duplications were inferred in both α-defensins, which were absent in five species, and three β-defensin gene subfamilies, but cathelicidins did not show significant shifts in gene family size and were absent in Anoura caudifer and the pteropodids. Based on lineage-specific gains and losses, we propose diet and diet-related microbiome evolution may determine the evolution of α- and β-defensins gene families and subfamilies. These results highlight the importance of building species-specific libraries for genome annotation in non-model organisms and shed light on possible drivers responsible for the rapid evolution of AMPs. By focusing on these understudied defenses, we provide a robust framework for explaining bat responses to pathogens.
Collapse
Affiliation(s)
| | - Diana Moreno-Santillán
- Department of Integrative Biology, University of California, Berkeley, CA, United States
| | - Graham M. Hughes
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - Nicole S. Paulat
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Nicolette Sipperly
- Department of Ecology and Evolution, Stony Brook University, Stony Brook, NY, United States
| | - Alexis M. Brown
- Department of Ecology and Evolution, Stony Brook University, Stony Brook, NY, United States
| | - Katherine R. Martin
- Department of Biology, University of Central Florida, Orlando, FL, United States
| | - Gregory M. Poterewicz
- Department of Ecology and Evolution, Stony Brook University, Stony Brook, NY, United States
| | - Marisa C. W. Lim
- Department of Ecology and Evolution, Stony Brook University, Stony Brook, NY, United States
| | - Amy L. Russell
- Department of Biology, Grand Valley State University, Allendale, MI, United States
| | - Marianne S. Moore
- College of Science and Mathematics, University of the Virgin Islands, St. Thomas, VI, United States
| | - Matthew G. Johnson
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Angelique P. Corthals
- Department of Sciences, John Jay College of Criminal Justice, New York, NY, United States
| | - David A. Ray
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Liliana M. Dávalos
- Department of Ecology and Evolution, Stony Brook University, Stony Brook, NY, United States
- Consortium for Inter-Disciplinary Environmental Research, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
35
|
Li X, Xiao S, Filipczak N, Yalamarty SSK, Shang H, Zhang J, Zheng Q. Role and Therapeutic Targeting Strategies of Neutrophil Extracellular Traps in Inflammation. Int J Nanomedicine 2023; 18:5265-5287. [PMID: 37746050 PMCID: PMC10516212 DOI: 10.2147/ijn.s418259] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Neutrophil extracellular traps (NETs) are large DNA reticular structures secreted by neutrophils and decorated with histones and antimicrobial proteins. As a key mechanism for neutrophils to resist microbial invasion, NETs play an important role in the killing of microorganisms (bacteria, fungi, and viruses). Although NETs are mostly known for mediating microbial killing, increasing evidence suggests that excessive NETs induced by stimulation of physical and chemical components, microorganisms, and pathological factors can exacerbate inflammation and organ damage. This review summarizes the induction and role of NETs in inflammation and focuses on the strategies of inhibiting NETosis and the mechanisms involved in pathogen evasion of NETs. Furthermore, herbal medicine inhibitors and nanodelivery strategies improve the efficiency of inhibition of excessive levels of NETs.
Collapse
Affiliation(s)
- Xiang Li
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Shanghua Xiao
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
| | | | - Hongming Shang
- Department of Biochemistry & Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - Jing Zhang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| |
Collapse
|
36
|
Vasudevan SO, Behl B, Rathinam VA. Pyroptosis-induced inflammation and tissue damage. Semin Immunol 2023; 69:101781. [PMID: 37352727 PMCID: PMC10598759 DOI: 10.1016/j.smim.2023.101781] [Citation(s) in RCA: 110] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 06/25/2023]
Abstract
Pyroptosis is a programmed necrotic cell death executed by gasdermins, a family of pore-forming proteins. The cleavage of gasdermins by specific proteases enables their pore-forming activity. The activation of the prototype member of the gasdermin family, gasdermin D (GSDMD), is linked to innate immune monitoring by inflammasomes. Additional gasdermins such as GSDMA, GSDMB, GSDMC, and GSDME are activated by inflammasome-independent mechanisms. Pyroptosis is emerging as a key host defense strategy against pathogens. However, excessive pyroptosis causes cytokine storm and detrimental inflammation leading to tissue damage and organ dysfunction. Consequently, dysregulated pyroptotic responses contribute to the pathogenesis of various diseases, including sepsis, atherosclerosis, acute respiratory distress syndrome, and neurodegenerative disorders. This review will discuss the inflammatory consequences of pyroptosis and the mechanisms of pyroptosis-induced tissue damage and disease pathogenesis.
Collapse
Affiliation(s)
- Swathy O Vasudevan
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | | | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA.
| |
Collapse
|
37
|
McMahon‑Cole H, Johnson A, Sadat Aghamiri S, Helikar T, Crawford LB. Modeling and Remodeling the Cell: How Digital Twins and HCMV Can Elucidate the Complex Interactions of Viral Latency, Epigenetic Regulation, and Immune Responses. CURRENT CLINICAL MICROBIOLOGY REPORTS 2023; 10:141-151. [PMID: 37901689 PMCID: PMC10601359 DOI: 10.1007/s40588-023-00201-w] [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] [Accepted: 07/12/2023] [Indexed: 10/31/2023]
Abstract
Purpose of Review Human cytomegalovirus (HCMV), while asymptomatic in most, causes significant complications during fetal development, following transplant or in immunosuppressed individuals. The host-virus interactions regulating viral latency and reactivation and viral control of the cellular environment (immune regulation, differentiation, epigenetics) are highly complex. Understanding these processes is essential to controlling infection and can be leveraged as a novel approach for understanding basic cell biology. Recent Findings Immune digital twins (IDTs) are digital simulations integrating knowledge of human immunology, physiology, and patient-specific clinical data to predict individualized immune responses and targeted treatments. Recent studies used IDTs to elucidate mechanisms of T cells, dendritic cells, and epigenetic control-all key to HCMV biology. Summary Here, we discuss how leveraging the unique biology of HCMV and IDTs will clarify immune response dynamics, host-virus interactions, and viral latency and reactivation and serve as a powerful IDT-validation platform for individualized and holistic health management.
Collapse
Affiliation(s)
- Hana McMahon‑Cole
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Alicia Johnson
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Sara Sadat Aghamiri
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Lindsey B. Crawford
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Center for Virology, Lincoln, NE, USA
- Nebraska Center for Integrated Biomolecular Communication, Lincoln, NE, USA
| |
Collapse
|
38
|
Patel BV, Lee TML, O'Dea K. CLUSTERINg Circulating Histones in Sepsis. Am J Respir Crit Care Med 2023; 208:125-127. [PMID: 37311246 PMCID: PMC10395490 DOI: 10.1164/rccm.202305-0935ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/07/2023] [Indexed: 06/15/2023] Open
Affiliation(s)
- Brijesh V Patel
- Division of Anaesthetics, Pain Medicine and Intensive Care Imperial College London London, United Kingdom
- Department of Adult Critical Care Royal Brompton Hospital London, United Kingdom
| | - Teresa M L Lee
- Anaesthetics, Pain Medicine, and Intensive Care Imperial College London London, United Kingdom
| | - Kieran O'Dea
- Anaesthetics, Pain Medicine, and Intensive Care Imperial College London London, United Kingdom
| |
Collapse
|
39
|
Agidigbi TS, Kwon HK, Knight JR, Zhao D, Lee FY, Oh I. Transcriptomic identification of genes expressed in invasive S. aureus diabetic foot ulcer infection. Front Cell Infect Microbiol 2023; 13:1198115. [PMID: 37434783 PMCID: PMC10332306 DOI: 10.3389/fcimb.2023.1198115] [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: 04/03/2023] [Accepted: 05/25/2023] [Indexed: 07/13/2023] Open
Abstract
Introduction Infection in diabetic foot ulcers (DFUs) is one of the major complications associated with patients with diabetes. Staphylococcus aureus is the most common offending pathogen in patients with infected DFU. Previous studies have suggested the application of species-specific antibodies against S. aureus for diagnosis and monitoring treatment response. Early and accurate identification of the main pathogen is critical for management of DFU infection. Understanding the host immune response against species-specific infection may facilitate diagnosis and may suggest potential intervention options to promote healing infected DFUs. We sought to investigate evolving host transcriptome associated with surgical treatment of S. aureus- infected DFU. Methods This study compared the transcriptome profile of 21 patients with S. aureus- infected DFU who underwent initial foot salvage therapy with irrigation and debridement followed by intravenous antibiotic therapy. Blood samples were collected at the recruitment (0 weeks) and 8 weeks after therapy to isolate peripheral blood mononuclear cells (PBMCs). We analyzed the PBMC expression of transcriptomes at two different time points (0 versus 8 weeks). Subjects were further divided into two groups at 8 weeks: healed (n = 17, 80.95%) versus non-healed (n = 4, 19.05%) based on the wound healing status. DESeq2 differential gene analysis was performed. Results and discussion An increased expression of IGHG1, IGHG2, IGHG3, IGLV3-21, and IGLV6-57 was noted during active infection at 0 weeks compared with that at 8 weeks. Lysine- and arginine-rich histones (HIST1H2AJ, HIST1H2AL, HIST1H2BM, HIST1H3B, and HIST1H3G) were upregulated at the initial phase of active infection at 0 weeks. CD177 and RRM2 were also upregulated at the initial phase of active infection (0 weeks) compared with that at 8 weeks of follow-up. Genes of heat shock protein members (HSPA1A, HSPE1, and HSP90B1) were high in not healed patients compared with that in healed patients 8 weeks after therapy. The outcome of our study suggests that the identification of genes evolution based on a transcriptomic profiling could be a useful tool for diagnosing infection and assessing severity and host immune response to therapies.
Collapse
Affiliation(s)
- Taiwo Samuel Agidigbi
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
| | - Hyuk-Kwon Kwon
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
- Division of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - James R. Knight
- Yale Center for Genome Analysis, Department of Genetics, Yale School of Medicine, New Haven, CT, United States
| | - Dejian Zhao
- Yale Center for Genome Analysis, Department of Genetics, Yale School of Medicine, New Haven, CT, United States
| | - Francis Y. Lee
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
| | - Irvin Oh
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
40
|
Yang T, Yu J, Ahmed T, Nguyen K, Nie F, Zan R, Li Z, Han P, Shen H, Zhang X, Takayama S, Song Y. Synthetic neutrophil extracellular traps dissect bactericidal contribution of NETs under regulation of α-1-antitrypsin. SCIENCE ADVANCES 2023; 9:eadf2445. [PMID: 37115934 PMCID: PMC10146876 DOI: 10.1126/sciadv.adf2445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/30/2023] [Indexed: 05/03/2023]
Abstract
Deciphering the complex interplay of neutrophil extracellular traps (NETs) with the surrounding environment is a challenge with notable clinical implications. To bridge the gap in knowledge, we report our findings on the antibacterial activity against Pseudomonas aeruginosa of synthetic NET-mimetic materials composed of nanofibrillated DNA-protein complexes. Our synthetic system makes component-by-component bottom-up analysis of NET protein effects possible. When the antimicrobial enzyme neutrophil elastase (NE) is incorporated into the bactericidal DNA-histone complexes, the resulting synthetic NET-like structure exhibits an unexpected reduction in antimicrobial activity. This critical immune function is rescued upon treatment with alpha-1-antitrypsin (AAT), a physiological tissue-protective protease inhibitor. This suggests a direct causal link between AAT inhibition of NE and preservation of histone-mediated antimicrobial activity. These results help better understand the complex and, at times, contradictory observations of in vivo antimicrobial effects of NETs and AAT by excluding neutrophil, cytokine, and chemoattractant contributions.
Collapse
Affiliation(s)
- Ting Yang
- School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA 30332, USA
| | - Jinlong Yu
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Tasdiq Ahmed
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA 30332, USA
| | - Katherine Nguyen
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA 30332, USA
| | - Fang Nie
- Renji Hospital affiliated to Shanghai Jiao Tong University, Shanghai 200127, China
| | - Rui Zan
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai 200032, China
| | - Zhiwei Li
- Renji Hospital affiliated to Shanghai Jiao Tong University, Shanghai 200127, China
| | - Pei Han
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hao Shen
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiaonong Zhang
- School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai 200032, China
| | - Shuichi Takayama
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA 30332, USA
| | - Yang Song
- School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
41
|
Kubelkova K, Bostik V, Joshi L, Macela A. Innate Immune Recognition, Integrated Stress Response, Infection, and Tumorigenesis. BIOLOGY 2023; 12:biology12040499. [PMID: 37106700 PMCID: PMC10135864 DOI: 10.3390/biology12040499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Engagement of PRRs in recognition of PAMPs or DAMPs is one of the processes that initiates cellular stress. These sensors are involved in signaling pathways leading to induction of innate immune processes. Signaling initiated by PRRs is associated with the activation of MyD88-dependent signaling pathways and myddosome formation. MyD88 downstream signaling depends upon the context of signaling initiation, the cell (sub)type and the microenvironment of signal initiation. Recognition of PAMPs or DAMPs through PRRs activates the cellular autonomous defence mechanism, which orchestrates the cell responses to resolve specific insults at the single cell level. In general, stressed endoplasmic reticulum is directly linked with the induction of autophagy and initiation of mitochondrial stress. These processes are regulated by the release of Ca2+ from ER stores accepted by mitochondria, which respond through membrane depolarization and the production of reactive oxygen species generating signals leading to inflammasome activation. In parallel, signaling from PRRs initiates the accumulation of misfolded or inappropriately post-translationally modified proteins in the ER and triggers a group of conserved emergency rescue pathways known as unfolded protein response. The cell-autonomous effector mechanisms have evolutionarily ancient roots and were gradually specialized for the defence of specific cell (sub)types. All of these processes are common to the innate immune recognition of microbial pathogens and tumorigenesis as well. PRRs are active in both cases. Downstream are activated signaling pathways initiated by myddosomes, translated by the cellular autonomous defence mechanism, and finalized by inflammasomes.
Collapse
Affiliation(s)
- Klara Kubelkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
- Correspondence:
| | - Vanda Bostik
- Department of Epidemiology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Lokesh Joshi
- Glycoscience Group, National Centre for Biomedical Engineering Science, University of Galway, H91 W2TY Galway, Ireland
| | - Ales Macela
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| |
Collapse
|
42
|
Li Y, Chen Y, Yang T, Chang K, Deng N, Zhao W, Su B. Targeting circulating high mobility group box-1 and histones by extracorporeal blood purification as an immunomodulation strategy against critical illnesses. Crit Care 2023; 27:77. [PMID: 36855150 PMCID: PMC9972334 DOI: 10.1186/s13054-023-04382-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
Both high mobility group box-1 (HMGB1) and histones are major damage-associated molecular patterns (DAPMs) that mediate lethal systemic inflammation, activation of the complement and coagulation system, endothelial injury and multiple organ dysfunction syndrome in critical illnesses. Although accumulating evidence collectively shows that targeting HMGB1 or histones by their specific antibodies or inhibitors could significantly mitigate aberrant immune responses in multiple critically ill animal models, routine clinical use of such agents is still not recommended by any guideline. In contrast, extracorporeal blood purification, which has been widely used to replace dysfunctional organs and remove exogenous or endogenous toxins in intensive care units, may also exert an immunomodulatory effect by eliminating inflammatory mediators such as cytokines, endotoxin, HMGB1 and histones in patients with critical illnesses. In this review, we summarize the multiple immunopathological roles of HMGB1 and histones in mediating inflammation, immune thrombosis and organ dysfunction and discuss the rationale for the removal of these DAMPs using various hemofilters. The latest preclinical and clinical evidence for the use of extracorporeal blood purification to improve the clinical outcome of critically ill patients by targeting circulating HMGB1 and histones is also gathered.
Collapse
Affiliation(s)
- Yupei Li
- grid.13291.380000 0001 0807 1581Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Chen
- grid.13291.380000 0001 0807 1581State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Tinghang Yang
- grid.13291.380000 0001 0807 1581Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Kaixi Chang
- grid.13291.380000 0001 0807 1581Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Ningyue Deng
- grid.13291.380000 0001 0807 1581Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Weifeng Zhao
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, China. .,Med-X Center for Materials, Sichuan University, Chengdu, China.
| | - Baihai Su
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China. .,Med-X Center for Materials, Sichuan University, Chengdu, China. .,Med+ Biomaterial Institute of West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|