1
|
da Costa ALA, Soares MA, Lourenço TGB, Guimarães-Pinto K, Filardy AD, de Oliveira AM, de Luca BG, Magliano DAC, Araujo OMO, Moura L, Lopes RT, Palhares de Miranda AL, Tributino JLM, Vieira Colombo AP. Periodontal pathogen Aggregatibacter actinomycetemcomitans JP2 correlates with colonic leukocytes decrease and gut microbiome imbalance in mice. J Periodontal Res 2024; 59:961-973. [PMID: 38757372 DOI: 10.1111/jre.13288] [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: 01/18/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024]
Abstract
AIM Evidence suggests that translocation of oral pathogens through the oral-gut axis may induce intestinal dysbiosis. This study aimed to evaluate the impact of a highly leukotoxic Aggregatibacter actinomycetemcomitans (Aa) strain on the gut microbiota, intestinal mucosal integrity and immune system in healthy mice. METHODS Eight-week-old male C57BL6 mice were divided into control (n = 16) and JP2 groups (n = 19), which received intragastric gavage with PBS and with a suspension of Aa JP2 (HK921), respectively, twice a week for 4 weeks. Colonic lamina propria, fecal material, serum, gingival tissues, and mandibles were obtained for analyses of leukocyte populations, inflammatory mediators, mucosal integrity, alveolar bone loss, and gut microbiota. Differences between groups for these parameters were examined by non-parametric tests. RESULTS The gut microbial richness and the number of colonic macrophages, neutrophils, and monocytes were significantly lower in Aa JP2-infected mice than in controls (p < .05). In contrast, infected animals showed higher abundance of Clostridiaceae, Lactobacillus taiwanensis, Helicobacter rodentium, higher levels of IL-6 expression in colonic tissues, and higher splenic MPO activity than controls (p < .05). No differences in tight junction expression, serum endotoxin levels, and colonic inflammatory cytokines were observed between groups. Infected animals presented also slightly more alveolar bone loss and gingival IL-6 levels than controls (p < .05). CONCLUSION Based on this model, intragastric administration of Aa JP2 is associated with changes in the gut ecosystem of healthy hosts, characterized by less live/recruited myeloid cells, enrichment of the gut microbiota with pathobionts and decrease in commensals. Negligible levels of colonic pro-inflammatory cytokines, and no signs of mucosal barrier disruption were related to these changes.
Collapse
Affiliation(s)
- André L A da Costa
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Cellular Immunology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana A Soares
- Department of Pharmaceutical Biotechnology, Laboratory of Studies in Experimental Pharmacology, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Talita G B Lourenço
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kamila Guimarães-Pinto
- Cellular Immunology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandra D Filardy
- Cellular Immunology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriana Miranda de Oliveira
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Olga M O Araujo
- Laboratory of Nuclear Instrumentation, Nuclear Engineering Program, Institute Alberto Luiz de Coimbra of Graduate and Research in Engineering, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa Moura
- Laboratory of Nuclear Instrumentation, Nuclear Engineering Program, Institute Alberto Luiz de Coimbra of Graduate and Research in Engineering, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo Tadeu Lopes
- Laboratory of Nuclear Instrumentation, Nuclear Engineering Program, Institute Alberto Luiz de Coimbra of Graduate and Research in Engineering, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Luisa Palhares de Miranda
- Cellular Immunology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge L M Tributino
- Molecular Pharmacology Laboratory, Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula Vieira Colombo
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
2
|
Porter JC, Inshaw J, Solis VJ, Denneny E, Evans R, Temkin MI, De Vasconcelos N, Aramburu IV, Hoving D, Basire D, Crissell T, Guinto J, Webb A, Esmail H, Johnston V, Last A, Rampling T, Lippert L, Helbig ET, Kurth F, Williams B, Flynn A, Lukey PT, Birault V, Papayannopoulos V. Anti-inflammatory therapy with nebulized dornase alfa for severe COVID-19 pneumonia: a randomized unblinded trial. eLife 2024; 12:RP87030. [PMID: 39009040 PMCID: PMC11251720 DOI: 10.7554/elife.87030] [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: 07/17/2024] Open
Abstract
Background Prinflammatory extracellular chromatin from neutrophil extracellular traps (NETs) and other cellular sources is found in COVID-19 patients and may promote pathology. We determined whether pulmonary administration of the endonuclease dornase alfa reduced systemic inflammation by clearing extracellular chromatin. Methods Eligible patients were randomized (3:1) to the best available care including dexamethasone (R-BAC) or to BAC with twice-daily nebulized dornase alfa (R-BAC + DA) for seven days or until discharge. A 2:1 ratio of matched contemporary controls (CC-BAC) provided additional comparators. The primary endpoint was the improvement in C-reactive protein (CRP) over time, analyzed using a repeated-measures mixed model, adjusted for baseline factors. Results We recruited 39 evaluable participants: 30 randomized to dornase alfa (R-BAC +DA), 9 randomized to BAC (R-BAC), and included 60 CC-BAC participants. Dornase alfa was well tolerated and reduced CRP by 33% compared to the combined BAC groups (T-BAC). Least squares (LS) mean post-dexamethasone CRP fell from 101.9 mg/L to 23.23 mg/L in R-BAC +DA participants versus a 99.5 mg/L to 34.82 mg/L reduction in the T-BAC group at 7 days; p=0.01. The anti-inflammatory effect of dornase alfa was further confirmed with subgroup and sensitivity analyses on randomised participants only, mitigating potential biases associated with the use of CC-BAC participants. Dornase alfa increased live discharge rates by 63% (HR 1.63, 95% CI 1.01-2.61, p=0.03), increased lymphocyte counts (LS mean: 1.08 vs 0.87, p=0.02) and reduced circulating cf-DNA and the coagulopathy marker D-dimer (LS mean: 570.78 vs 1656.96 μg/mL, p=0.004). Conclusions Dornase alfa reduces pathogenic inflammation in COVID-19 pneumonia, demonstrating the benefit of cost-effective therapies that target extracellular chromatin. Funding LifeArc, Breathing Matters, The Francis Crick Institute (CRUK, Medical Research Council, Wellcome Trust). Clinical trial number NCT04359654.
Collapse
Affiliation(s)
- Joanna C Porter
- UCL Respiratory, University College LondonLondonUnited Kingdom
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | | | | | - Emma Denneny
- UCL Respiratory, University College LondonLondonUnited Kingdom
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | - Rebecca Evans
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | - Mia I Temkin
- Antimicrobial Defence Lab, The Francis Crick InstituteLondonUnited Kingdom
| | | | | | - Dennis Hoving
- Antimicrobial Defence Lab, The Francis Crick InstituteLondonUnited Kingdom
| | - Donna Basire
- UCL Respiratory, University College LondonLondonUnited Kingdom
| | - Tracey Crissell
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | - Jesusa Guinto
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | - Alison Webb
- University College London Hospitals NHS TrustLondonUnited Kingdom
| | - Hanif Esmail
- University College London Hospitals NHS TrustLondonUnited Kingdom
- National Institute for Health Research, University College London Hospital Biomedical Research CentreLondonUnited Kingdom
| | - Victoria Johnston
- University College London Hospitals NHS TrustLondonUnited Kingdom
- National Institute for Health Research, University College London Hospital Biomedical Research CentreLondonUnited Kingdom
| | - Anna Last
- University College London Hospitals NHS TrustLondonUnited Kingdom
- Clinical Research Department, London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Thomas Rampling
- University College London Hospitals NHS TrustLondonUnited Kingdom
- National Institute for Health Research, University College London Hospital Biomedical Research CentreLondonUnited Kingdom
| | - Lena Lippert
- Charité – Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory MedicineBerlinGermany
| | - Elisa Theresa Helbig
- Charité – Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory MedicineBerlinGermany
| | - Florian Kurth
- Charité – Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory MedicineBerlinGermany
| | - Bryan Williams
- University College London Hospitals NHS TrustLondonUnited Kingdom
- National Institute for Health Research, University College London Hospital Biomedical Research CentreLondonUnited Kingdom
| | | | | | | | | |
Collapse
|
3
|
Zhang X, Zhang Y, Yuan S, Zhang J. The potential immunological mechanisms of sepsis. Front Immunol 2024; 15:1434688. [PMID: 39040114 PMCID: PMC11260823 DOI: 10.3389/fimmu.2024.1434688] [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: 05/18/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Sepsis is described as a life-threatening organ dysfunction and a heterogeneous syndrome that is a leading cause of morbidity and mortality in intensive care settings. Severe sepsis could incite an uncontrollable surge of inflammatory cytokines, and the host immune system's immunosuppression could respond to counter excessive inflammatory responses, characterized by the accumulated anti-inflammatory cytokines, impaired function of immune cells, over-proliferation of myeloid-derived suppressor cells and regulatory T cells, depletion of immune effector cells by different means of death, etc. In this review, we delve into the underlying pathological mechanisms of sepsis, emphasizing both the hyperinflammatory phase and the associated immunosuppression. We offer an in-depth exploration of the critical mechanisms underlying sepsis, spanning from individual immune cells to a holistic organ perspective, and further down to the epigenetic and metabolic reprogramming. Furthermore, we outline the strengths of artificial intelligence in analyzing extensive datasets pertaining to septic patients, showcasing how classifiers trained on various clinical data sources can identify distinct sepsis phenotypes and thus to guide personalized therapy strategies for the management of sepsis. Additionally, we provide a comprehensive summary of recent, reliable biomarkers for hyperinflammatory and immunosuppressive states, facilitating more precise and expedited diagnosis of sepsis.
Collapse
Affiliation(s)
- Xinyu Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yujing Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiying Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiancheng Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Das S, Kaminski TW, Schlegel BT, Bain W, Hu S, Patel A, Kale SL, Chen K, Lee JS, Mallampalli RK, Kagan VE, Rajasundaram D, McVerry BJ, Sundd P, Kitsios GD, Ray A, Ray P. Neutrophils and galectin-3 defend mice from lethal bacterial infection and humans from acute respiratory failure. Nat Commun 2024; 15:4724. [PMID: 38830855 PMCID: PMC11148175 DOI: 10.1038/s41467-024-48796-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: 08/16/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Respiratory infection by Pseudomonas aeruginosa, common in hospitalized immunocompromised and immunocompetent ventilated patients, can be life-threatening because of antibiotic resistance. This raises the question of whether the host's immune system can be educated to combat this bacterium. Here we show that prior exposure to a single low dose of lipopolysaccharide (LPS) protects mice from a lethal infection by P. aeruginosa. LPS exposure trained the innate immune system by promoting expansion of neutrophil and interstitial macrophage populations distinguishable from other immune cells with enrichment of gene sets for phagocytosis- and cell-killing-associated genes. The cell-killing gene set in the neutrophil population uniquely expressed Lgals3, which encodes the multifunctional antibacterial protein, galectin-3. Intravital imaging for bacterial phagocytosis, assessment of bacterial killing and neutrophil-associated galectin-3 protein levels together with use of galectin-3-deficient mice collectively highlight neutrophils and galectin-3 as central players in LPS-mediated protection. Patients with acute respiratory failure revealed significantly higher galectin-3 levels in endotracheal aspirates (ETAs) of survivors compared to non-survivors, galectin-3 levels strongly correlating with a neutrophil signature in the ETAs and a prognostically favorable hypoinflammatory plasma biomarker subphenotype. Taken together, our study provides impetus for harnessing the potential of galectin-3-expressing neutrophils to protect from lethal infections and respiratory failure.
Collapse
Affiliation(s)
- Sudipta Das
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Tomasz W Kaminski
- VERSITI Blood Research Institute and Medical College of Wisconsin, Milwaukee, WI, 53233, USA
| | - Brent T Schlegel
- Department of Pediatrics, Division of Health Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - William Bain
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Veteran's Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA
| | - Sanmei Hu
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Akruti Patel
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Sagar L Kale
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Kong Chen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Janet S Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rama K Mallampalli
- Department of Medicine, The Ohio State University (OSU), Columbus, OH, 43210, USA
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Division of Health Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - Bryan J McVerry
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Prithu Sundd
- VERSITI Blood Research Institute and Medical College of Wisconsin, Milwaukee, WI, 53233, USA
| | - Georgios D Kitsios
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Anuradha Ray
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Prabir Ray
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
5
|
Tran N, Mills EL. Redox regulation of macrophages. Redox Biol 2024; 72:103123. [PMID: 38615489 PMCID: PMC11026845 DOI: 10.1016/j.redox.2024.103123] [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: 01/22/2024] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 04/16/2024] Open
Abstract
Redox signaling, a mode of signal transduction that involves the transfer of electrons from a nucleophilic to electrophilic molecule, has emerged as an essential regulator of inflammatory macrophages. Redox reactions are driven by reactive oxygen/nitrogen species (ROS and RNS) and redox-sensitive metabolites such as fumarate and itaconate, which can post-translationally modify specific cysteine residues in target proteins. In the past decade our understanding of how ROS, RNS, and redox-sensitive metabolites control macrophage function has expanded dramatically. In this review, we discuss the latest evidence of how ROS, RNS, and metabolites regulate macrophage function and how this is dysregulated with disease. We highlight the key tools to assess redox signaling and important questions that remain.
Collapse
Affiliation(s)
- Nhien Tran
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Evanna L Mills
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Munalisa R, Lien TS, Tsai PY, Sun DS, Cheng CF, Wu WS, Li CC, Hu CT, Tsai KW, Lee YL, Chou YC, Chang HH. Restraint Stress-Induced Neutrophil Inflammation Contributes to Concurrent Gastrointestinal Injury in Mice. Int J Mol Sci 2024; 25:5261. [PMID: 38791301 PMCID: PMC11121713 DOI: 10.3390/ijms25105261] [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: 04/16/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Psychological stress increases risk of gastrointestinal tract diseases. However, the mechanism behind stress-induced gastrointestinal injury is not well understood. The objective of our study is to elucidate the putative mechanism of stress-induced gastrointestinal injury and develop an intervention strategy. To achieve this, we employed the restraint stress mouse model, a well-established method to study the pathophysiological changes associated with psychological stress in mice. By orally administering gut-nonabsorbable Evans blue dye and monitoring its plasma levels, we were able to track the progression of gastrointestinal injury in live mice. Additionally, flow cytometry was utilized to assess the viability, death, and inflammatory status of splenic leukocytes, providing insights into the stress-induced impact on the innate immune system associated with stress-induced gastrointestinal injury. Our findings reveal that neutrophils represent the primary innate immune leukocyte lineage responsible for stress-induced inflammation. Splenic neutrophils exhibited elevated expression levels of the pro-inflammatory cytokine IL-1, cellular reactive oxygen species, mitochondrial burden, and cell death following stress challenge compared to other innate immune cells such as macrophages, monocytes, and dendritic cells. Regulated cell death analysis indicated that NETosis is the predominant stress-induced cell death response among other analyzed regulated cell death pathways. NETosis culminates in the formation and release of neutrophil extracellular traps, which play a crucial role in modulating inflammation by binding to pathogens. Treatment with the NETosis inhibitor GSK484 rescued stress-induced neutrophil extracellular trap release and gastrointestinal injury, highlighting the involvement of neutrophil extracellular traps in stress-induced gastrointestinal inflammation. Our results suggest that neutrophil NETosis could serve as a promising drug target for managing psychological stress-induced gastrointestinal injuries.
Collapse
Grants
- 104-2320-B-320 -009 -MY3, 107-2311-B-320-002-MY3, 111-2320-B320-006-MY3, 112-2320-B-320-007 National Science and Technology Council, Taiwan
- TCMMP104-06, TCMMP108-04, TCMMP 111-01, TCAS111-02, TCAS-112-02, TCAS113-04, TCRD112-033, TCRD113-041 Tzu-Chi Medical Foundation
Collapse
Affiliation(s)
- Rina Munalisa
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (T.-S.L.); (P.-Y.T.); (D.-S.S.)
| | - Te-Sheng Lien
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (T.-S.L.); (P.-Y.T.); (D.-S.S.)
| | - Ping-Yeh Tsai
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (T.-S.L.); (P.-Y.T.); (D.-S.S.)
| | - Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (T.-S.L.); (P.-Y.T.); (D.-S.S.)
| | - Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan;
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan;
| | - Wen-Sheng Wu
- Division of General Surgery, Department of Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Chi-Cheng Li
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddha Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Center of Stem Cell & Precision Medicine, Hualien Tzu Chi Hospital, Buddha Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Chi-Tan Hu
- Research Center for Hepatology and Department of Gastroenterology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Kuo-Wang Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan;
| | - Yungling Leo Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan;
- College of Public Health, China Medical University, Taichung 404, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center, Academia Sinica, Taipei 115, Taiwan;
| | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (T.-S.L.); (P.-Y.T.); (D.-S.S.)
| |
Collapse
|
7
|
de Castilhos J, Tillmanns K, Blessing J, Laraño A, Borisov V, Stein-Thoeringer CK. Microbiome and pancreatic cancer: time to think about chemotherapy. Gut Microbes 2024; 16:2374596. [PMID: 39024520 PMCID: PMC11259062 DOI: 10.1080/19490976.2024.2374596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer characterized by late diagnosis, rapid progression, and a high mortality rate. Its complex biology, characterized by a dense, stromal tumor environment with an immunosuppressive milieu, contributes to resistance against standard treatments like chemotherapy and radiation. This comprehensive review explores the dynamic role of the microbiome in modulating chemotherapy efficacy and outcomes in PDAC. It delves into the microbiome's impact on drug metabolism and resistance, and the interaction between microbial elements, drugs, and human biology. We also highlight the significance of specific bacterial species and microbial enzymes in influencing drug action and the immune response in the tumor microenvironment. Cutting-edge methodologies, including artificial intelligence, low-biomass microbiome analysis and patient-derived organoid models, are discussed, offering insights into the nuanced interactions between microbes and cancer cells. The potential of microbiome-based interventions as adjuncts to conventional PDAC treatments are discussed, paving the way for personalized therapy approaches. This review synthesizes recent research to provide an in-depth understanding of how the microbiome affects chemotherapy efficacy. It focuses on elucidating key mechanisms and identifying existing knowledge gaps. Addressing these gaps is crucial for enhancing personalized medicine and refining cancer treatment strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Juliana de Castilhos
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Katharina Tillmanns
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Jana Blessing
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Arnelyn Laraño
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Vadim Borisov
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Christoph K. Stein-Thoeringer
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| |
Collapse
|
8
|
Zhang H, Zhang J, Lai J, Chen Y, Tian M, Pan G, Yang X, Qi Y. Nanoscale Surface Refinement of CoCrMo Alloy for Artificial Knee Joints via Chemical Mechanical Polishing. MATERIALS (BASEL, SWITZERLAND) 2023; 17:8. [PMID: 38203862 PMCID: PMC10779557 DOI: 10.3390/ma17010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024]
Abstract
In this study, we address the challenge of surface roughness in CoCrMo alloys, typically used in artificial knee joints, which can initiate a cascade of biological responses causing inflammation, osteolysis, joint instability, and increased susceptibility to infection. We propose the application of a chemical mechanical polishing (CMP) technique, using an ecologically responsible slurry composed of 4 wt% SiO2, 0.3 wt% H2O2, 1.0 wt% glycine, and 0.05 wt% benzotriazole. Our innovative approach demonstrated significant improvements, achieving a material removal rate of 30.9 nm/min and reducing the arithmetic mean roughness from 20.76 nm to 0.25 nm, thereby enhancing the nanoscale surface quality of the artificial knee joint alloy. The smoother surface is attributed to a decrease in corrosion potential to 0.18 V and a reduction in corrosion current density from 9.55 µA/cm2 to 4.49 µA/cm2 with the addition of BTA, evidenced by electrochemical tests. Furthermore, the preservation of the phase structure of the CoCrMo alloy, as confirmed by XRD analysis and elemental mapping, ensures the structural integrity of the treated surfaces. These outcomes and our simulation results demonstrate the effectiveness of our CMP method in engineering surface treatments for artificial knee joints to optimize friction behavior and potentially extend their lifespans.
Collapse
Affiliation(s)
- Hanji Zhang
- Department of Orthopedics, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin 300121, China;
- School of Electronics and Information Engineering, Hebei University of Technology, Tianjin 300130, China; (J.Z.); (J.L.); (G.P.)
| | - Jiangliang Zhang
- School of Electronics and Information Engineering, Hebei University of Technology, Tianjin 300130, China; (J.Z.); (J.L.); (G.P.)
| | - Jinghui Lai
- School of Electronics and Information Engineering, Hebei University of Technology, Tianjin 300130, China; (J.Z.); (J.L.); (G.P.)
| | - Yilin Chen
- The Second Clinical College, China Medical University, Shenyang 110001, China;
| | - Mengqiang Tian
- Department of Orthopedics, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin 300121, China;
| | - Guofeng Pan
- School of Electronics and Information Engineering, Hebei University of Technology, Tianjin 300130, China; (J.Z.); (J.L.); (G.P.)
| | - Xueli Yang
- School of Electronics and Information Engineering, Hebei University of Technology, Tianjin 300130, China; (J.Z.); (J.L.); (G.P.)
| | - Yuhang Qi
- Department of Orthopedics, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin 300121, China;
| |
Collapse
|
9
|
Yang W, Wang Y, Tao K, Li R. Metabolite itaconate in host immunoregulation and defense. Cell Mol Biol Lett 2023; 28:100. [PMID: 38042791 PMCID: PMC10693715 DOI: 10.1186/s11658-023-00503-3] [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: 09/06/2023] [Accepted: 10/20/2023] [Indexed: 12/04/2023] Open
Abstract
Metabolic states greatly influence functioning and differentiation of immune cells. Regulating the metabolism of immune cells can effectively modulate the host immune response. Itaconate, an intermediate metabolite derived from the tricarboxylic acid (TCA) cycle of immune cells, is produced through the decarboxylation of cis-aconitate by cis-aconitate decarboxylase in the mitochondria. The gene encoding cis-aconitate decarboxylase is known as immune response gene 1 (IRG1). In response to external proinflammatory stimulation, macrophages exhibit high IRG1 expression. IRG1/itaconate inhibits succinate dehydrogenase activity, thus influencing the metabolic status of macrophages. Therefore, itaconate serves as a link between macrophage metabolism, oxidative stress, and immune response, ultimately regulating macrophage function. Studies have demonstrated that itaconate acts on various signaling pathways, including Keap1-nuclear factor E2-related factor 2-ARE pathways, ATF3-IκBζ axis, and the stimulator of interferon genes (STING) pathway to exert antiinflammatory and antioxidant effects. Furthermore, several studies have reported that itaconate affects cancer occurrence and development through diverse signaling pathways. In this paper, we provide a comprehensive review of the role IRG1/itaconate and its derivatives in the regulation of macrophage metabolism and functions. By furthering our understanding of itaconate, we intend to shed light on its potential for treating inflammatory diseases and offer new insights in this field.
Collapse
Affiliation(s)
- Wenchang Yang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaxin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Ruidong Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China.
| |
Collapse
|
10
|
Gao Y, Tian X, Zhang X, Milebe Nkoua GD, Chen F, Liu Y, Chai Y. The roles of tissue-resident macrophages in sepsis-associated organ dysfunction. Heliyon 2023; 9:e21391. [PMID: 38027963 PMCID: PMC10643296 DOI: 10.1016/j.heliyon.2023.e21391] [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: 06/26/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis, a syndrome caused by a dysregulated host response to infection and characterized by life-threatening organ dysfunction, particularly septic shock and sepsis-associated organ dysfunction (SAOD), is a medical emergency associated with high morbidity, high mortality, and long-term sequelae. Tissue-resident macrophages (TRMs) are a subpopulation of macrophages derived primarily from yolk sac progenitors and fetal liver during embryogenesis, located primarily in non-lymphoid tissues in adulthood, capable of local self-renewal independent of hematopoiesis, and developmentally and functionally restricted to the non-lymphoid organs in which they reside. TRMs are the first line of defense against life-threatening conditions such as sepsis, tumor growth, traumatic-associated organ injury, and surgical-associated injury. In the context of sepsis, TRMs can be considered as angels or demons involved in organ injury. Our proposal is that sepsis, septic shock, and SAOD can be attenuated by modulating TRMs in different organs. This review summarizes the pathophysiological mechanisms of TRMs in different organs or tissues involved in the development and progression of sepsis.
Collapse
Affiliation(s)
- Yulei Gao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
- Department of Emergency Medicine, China-Congo Friendship Hospital, Brazzaville, 999059, P. R. Congo
| | - Xin Tian
- Department of Medical Research, Beijing Qiansong Technology Development Company, Beijing, 100193, P. R. China
- Department of Medical Research, Sen Sho Ka Gi Company, Inba-gun, Chiba, 285-0905, Japan
| | - Xiang Zhang
- Department of Emergency Medicine, Rizhao People's Hospital of Shandong Province, Rizhao, 276825, P. R. China
| | | | - Fang Chen
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| | - Yancun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| | - Yanfen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| |
Collapse
|
11
|
Ramessur A, Ambasager B, Valle Aramburu I, Peakman F, Gleason K, Lehmann C, Papayannopoulos V, Coombes RC, Malanchi I. Circulating neutrophils from patients with early breast cancer have distinct subtype-dependent phenotypes. Breast Cancer Res 2023; 25:125. [PMID: 37858168 PMCID: PMC10588170 DOI: 10.1186/s13058-023-01707-3] [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: 05/21/2023] [Accepted: 09/10/2023] [Indexed: 10/21/2023] Open
Abstract
PURPOSE An elevated number of circulating neutrophils is a poor prognostic factor for breast cancer, where evidence of bone marrow cancer-dependent priming is found. However, how early this priming is detectable remains unclear. PATIENTS AND METHODS Here, we investigate changes in circulating neutrophils from newly diagnosed breast cancer patients before any therapeutic interventions. To do this, we assessed their lifespan and their broader intracellular kinase network activation states by using the Pamgene Kinome assay which measures the activity of neutrophil kinases. RESULTS We found sub-type specific L-selectin (CD62L) changes in circulating neutrophils as well as perturbations in their overall global kinase activity. Strikingly, breast cancer patients of different subtypes (HR+, HER2+, triple negative) exhibited distinct neutrophil kinase activity patterns indicating that quantifiable perturbations can be detected in circulating neutrophils from early breast cancer patients, that are sensitive to both hormonal and HER-2 status. We also detected an increase in neutrophils lifespan in cancer patients, independently of tumour subtype. CONCLUSIONS Our results suggest that the tumour-specific kinase activation patterns in circulating neutrophils may be used in conjunction with other markers to identify patients with cancer from those harbouring only benign lesions of the breast. Given the important role neutrophil in breast cancer progression, the significance of this sub-type of specific priming warrants further investigation.
Collapse
Affiliation(s)
- Anisha Ramessur
- Tumour Host Interaction Laboratory, The Francis Crick Institute, London, UK
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Bana Ambasager
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | | | - Freddie Peakman
- Tumour Host Interaction Laboratory, The Francis Crick Institute, London, UK
| | - Kelly Gleason
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | | | | | - Raoul Charles Coombes
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.
| | - Ilaria Malanchi
- Tumour Host Interaction Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
12
|
Papayannopoulos V. NET histones inflame periodontitis. J Exp Med 2023; 220:e20230783. [PMID: 37552469 PMCID: PMC10407781 DOI: 10.1084/jem.20230783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
Microbial dysbiosis triggers inflammatory periodontitis. In this issue of JEM, Kim et al. (2023. J. Exp. Med.https://doi.org/10.1084/jem.20221751) demonstrate that neutrophil extracellular trap histones are the major mediators fueling the pathogenic Th17 inflammation that promotes gum and bone loss in periodontitis.
Collapse
|
13
|
Gander-Bui HTT, Schläfli J, Baumgartner J, Walthert S, Genitsch V, van Geest G, Galván JA, Cardozo C, Graham Martinez C, Grans M, Muth S, Bruggmann R, Probst HC, Gabay C, Freigang S. Targeted removal of macrophage-secreted interleukin-1 receptor antagonist protects against lethal Candida albicans sepsis. Immunity 2023; 56:1743-1760.e9. [PMID: 37478856 DOI: 10.1016/j.immuni.2023.06.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 03/02/2023] [Accepted: 06/27/2023] [Indexed: 07/23/2023]
Abstract
Invasive fungal infections are associated with high mortality rates, and the lack of efficient treatment options emphasizes an urgency to identify underlying disease mechanisms. We report that disseminated Candida albicans infection is facilitated by interleukin-1 receptor antagonist (IL-1Ra) secreted from macrophages in two temporally and spatially distinct waves. Splenic CD169+ macrophages release IL-1Ra into the bloodstream, impeding early neutrophil recruitment. IL-1Ra secreted by monocyte-derived tissue macrophages further impairs pathogen containment. Therapeutic IL-1Ra neutralization restored the functional competence of neutrophils, corrected maladapted hyper-inflammation, and eradicated the otherwise lethal infection. Conversely, augmentation of macrophage-secreted IL-1Ra by type I interferon severely aggravated disease mortality. Our study uncovers how a fundamental immunoregulatory mechanism mediates the high disease susceptibility to invasive candidiasis. Furthermore, interferon-stimulated IL-1Ra secretion may exacerbate fungal dissemination in human patients with secondary candidemia. Macrophage-secreted IL-1Ra should be considered as an additional biomarker and potential therapeutic target in severe systemic candidiasis.
Collapse
Affiliation(s)
- Hang Thi Thuy Gander-Bui
- Division of Experimental Pathology, Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Joëlle Schläfli
- Division of Experimental Pathology, Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | - Johanna Baumgartner
- Division of Experimental Pathology, Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Sabrina Walthert
- Division of Experimental Pathology, Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | - Vera Genitsch
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | - Geert van Geest
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, 3012 Bern, Switzerland
| | - José A Galván
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | - Carmen Cardozo
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | | | - Mona Grans
- Institute for Immunology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Sabine Muth
- Institute for Immunology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, 3012 Bern, Switzerland
| | | | - Cem Gabay
- Division of Rheumatology, Department of Medicine, University Hospital of Geneva, 1211 Geneva, Switzerland
| | - Stefan Freigang
- Division of Experimental Pathology, Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland.
| |
Collapse
|
14
|
Bee GCW, Lokken-Toyli KL, Yeung ST, Rodriguez L, Zangari T, Anderson EE, Ghosh S, Rothlin CV, Brodin P, Khanna KM, Weiser JN. Age-dependent differences in efferocytosis determine the outcome of opsonophagocytic protection from invasive pathogens. Immunity 2023; 56:1255-1268.e5. [PMID: 37059107 PMCID: PMC10330046 DOI: 10.1016/j.immuni.2023.03.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/22/2022] [Accepted: 03/21/2023] [Indexed: 04/16/2023]
Abstract
In early life, susceptibility to invasive infection skews toward a small subset of microbes, whereas other pathogens associated with diseases later in life, including Streptococcus pneumoniae (Spn), are uncommon among neonates. To delineate mechanisms behind age-dependent susceptibility, we compared age-specific mouse models of invasive Spn infection. We show enhanced CD11b-dependent opsonophagocytosis by neonatal neutrophils improved protection against Spn during early life. The augmented function of neonatal neutrophils was mediated by higher CD11b surface expression at the population level due to dampened efferocytosis, which also resulted in more CD11bhi "aged" neutrophils in peripheral blood. Dampened efferocytosis during early life could be attributed to the lack of CD169+ macrophages in neonates and reduced systemic expressions of multiple efferocytic mediators, including MerTK. On experimentally impairing efferocytosis later in life, CD11bhi neutrophils increased and protection against Spn improved. Our findings reveal how age-dependent differences in efferocytosis determine infection outcome through the modulation of CD11b-driven opsonophagocytosis and immunity.
Collapse
Affiliation(s)
- Gavyn Chern Wei Bee
- Department of Microbiology, New York University Grossman School of Medicine, New York, USA.
| | - Kristen L Lokken-Toyli
- Department of Microbiology, New York University Grossman School of Medicine, New York, USA
| | - Stephen T Yeung
- Department of Microbiology, New York University Grossman School of Medicine, New York, USA
| | - Lucie Rodriguez
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Tonia Zangari
- Department of Microbiology, New York University Grossman School of Medicine, New York, USA
| | - Exene E Anderson
- Department of Microbiology, New York University Grossman School of Medicine, New York, USA
| | - Sourav Ghosh
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA; Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Carla V Rothlin
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Petter Brodin
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Department of Immunology & Inflammation, Imperial College London, London, UK
| | - Kamal M Khanna
- Department of Microbiology, New York University Grossman School of Medicine, New York, USA; Laura & Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Jeffrey N Weiser
- Department of Microbiology, New York University Grossman School of Medicine, New York, USA.
| |
Collapse
|
15
|
She H, Tan L, Yang R, Zheng J, Wang Y, Du Y, Peng X, Li Q, Lu H, Xiang X, Hu Y, Liu L, Li T. Identification of featured necroptosis-related genes and imbalanced immune infiltration in sepsis via machine learning. Front Genet 2023; 14:1158029. [PMID: 37091800 PMCID: PMC10117955 DOI: 10.3389/fgene.2023.1158029] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
Background: The precise diagnostic and prognostic biological markers were needed in immunotherapy for sepsis. Considering the role of necroptosis and immune cell infiltration in sepsis, differentially expressed necroptosis-related genes (DE-NRGs) were identified, and the relationship between DE-NRGs and the immune microenvironment in sepsis was analyzed.Methods: Machine learning algorithms were applied for screening hub genes related to necroptosis in the training cohort. CIBERSORT algorithms were employed for immune infiltration landscape analysis. Then, the diagnostic value of these hub genes was verified by the receiver operating characteristic (ROC) curve and nomogram. In addition, consensus clustering was applied to divide the septic patients into different subgroups, and quantitative real-time PCR was used to detect the mRNA levels of the hub genes between septic patients (SP) (n = 30) and healthy controls (HC) (n = 15). Finally, a multivariate prediction model based on heart rate, temperature, white blood count and 4 hub genes was established.Results: A total of 47 DE-NRGs were identified between SP and HC and 4 hub genes (BACH2, GATA3, LEF1, and BCL2) relevant to necroptosis were screened out via multiple machine learning algorithms. The high diagnostic value of these hub genes was validated by the ROC curve and Nomogram model. Besides, the immune scores, correlation analysis and immune cell infiltrations suggested an immunosuppressive microenvironment in sepsis. Septic patients were divided into 2 clusters based on the expressions of hub genes using consensus clustering, and the immune microenvironment landscapes and immune function between the 2 clusters were significantly different. The mRNA levels of the 4 hub genes significantly decreased in SP as compared with HC. The area under the curve (AUC) was better in the multivariate prediction model than in other indicators.Conclusion: This study indicated that these necroptosis hub genes might have great potential in prognosis prediction and personalized immunotherapy for sepsis.
Collapse
Affiliation(s)
- Han She
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, China
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Lei Tan
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, China
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Ruibo Yang
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jie Zheng
- School of Medicine, Chongqing University, Chongqing, China
| | - Yi Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, China
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yuanlin Du
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaoyong Peng
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, China
| | - Qinghui Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, China
| | - Haibin Lu
- Department of Intensive Care Unit, Daping Hospital, Army Medical University, Chongqing, China
| | - Xinming Xiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi Hu
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, China
- *Correspondence: Yi Hu, ; Liangming Liu, ; Tao Li,
| | - Liangming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, China
- *Correspondence: Yi Hu, ; Liangming Liu, ; Tao Li,
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, China
- *Correspondence: Yi Hu, ; Liangming Liu, ; Tao Li,
| |
Collapse
|
16
|
Gungabeesoon J, Gort-Freitas NA, Kiss M, Bolli E, Messemaker M, Siwicki M, Hicham M, Bill R, Koch P, Cianciaruso C, Duval F, Pfirschke C, Mazzola M, Peters S, Homicsko K, Garris C, Weissleder R, Klein AM, Pittet MJ. A neutrophil response linked to tumor control in immunotherapy. Cell 2023; 186:1448-1464.e20. [PMID: 37001504 PMCID: PMC10132778 DOI: 10.1016/j.cell.2023.02.032] [Citation(s) in RCA: 101] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/10/2023] [Accepted: 02/24/2023] [Indexed: 04/01/2023]
Abstract
Neutrophils accumulate in solid tumors, and their abundance correlates with poor prognosis. Neutrophils are not homogeneous, however, and could play different roles in cancer therapy. Here, we investigate the role of neutrophils in immunotherapy, leading to tumor control. We show that successful therapies acutely expanded tumor neutrophil numbers. This expansion could be attributed to a Sellhi state rather than to other neutrophils that accelerate tumor progression. Therapy-elicited neutrophils acquired an interferon gene signature, also seen in human patients, and appeared essential for successful therapy, as loss of the interferon-responsive transcription factor IRF1 in neutrophils led to failure of immunotherapy. The neutrophil response depended on key components of anti-tumor immunity, including BATF3-dependent DCs, IL-12, and IFNγ. In addition, we found that a therapy-elicited systemic neutrophil response positively correlated with disease outcome in lung cancer patients. Thus, we establish a crucial role of a neutrophil state in mediating effective cancer therapy.
Collapse
Affiliation(s)
- Jeremy Gungabeesoon
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | | | - Máté Kiss
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - Evangelia Bolli
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - Marius Messemaker
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA; Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marie Siwicki
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Mehdi Hicham
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - Ruben Bill
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - Peter Koch
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Chiara Cianciaruso
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - Florent Duval
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - Christina Pfirschke
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Michael Mazzola
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Solange Peters
- Service of Medical Oncology, Department of Oncology, CHUV, Lausanne, Switzerland; Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Krisztian Homicsko
- AGORA Cancer Research Center, Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne, Switzerland; Department of Oncology, CHUV, Lausanne, Switzerland; Swiss Cancer Center Leman, Lausanne, Switzerland
| | - Christopher Garris
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA; Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA; Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne, Switzerland; Swiss Cancer Center Leman, Lausanne, Switzerland.
| |
Collapse
|
17
|
Adrover JM, McDowell SAC, He XY, Quail DF, Egeblad M. NETworking with cancer: The bidirectional interplay between cancer and neutrophil extracellular traps. Cancer Cell 2023; 41:505-526. [PMID: 36827980 DOI: 10.1016/j.ccell.2023.02.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/09/2023] [Accepted: 02/01/2023] [Indexed: 02/25/2023]
Abstract
Neutrophils are major effectors and regulators of the immune system. They play critical roles not only in the eradication of pathogens but also in cancer initiation and progression. Conversely, the presence of cancer affects neutrophil activity, maturation, and lifespan. By promoting or repressing key neutrophil functions, cancer cells co-opt neutrophil biology to their advantage. This co-opting includes hijacking one of neutrophils' most striking pathogen defense mechanisms: the formation of neutrophil extracellular traps (NETs). NETs are web-like filamentous extracellular structures of DNA, histones, and cytotoxic granule-derived proteins. Here, we discuss the bidirectional interplay by which cancer stimulates NET formation, and NETs in turn support disease progression. We review how vascular dysfunction and thrombosis caused by neutrophils and NETs underlie an elevated risk of death from cardiovascular events in cancer patients. Finally, we propose therapeutic strategies that may be effective in targeting NETs in the clinical setting.
Collapse
Affiliation(s)
- Jose M Adrover
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Sheri A C McDowell
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada; Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Xue-Yan He
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada; Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
18
|
Yeung ST, Ovando LJ, Russo AJ, Rathinam VA, Khanna KM. CD169+ macrophage intrinsic IL-10 production regulates immune homeostasis during sepsis. Cell Rep 2023; 42:112171. [PMID: 36867536 PMCID: PMC10123955 DOI: 10.1016/j.celrep.2023.112171] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 09/23/2022] [Accepted: 02/10/2023] [Indexed: 03/04/2023] Open
Abstract
Macrophages facilitate critical functions in regulating pathogen clearance and immune homeostasis in tissues. The remarkable functional diversity exhibited by macrophage subsets is dependent on tissue environment and the nature of the pathological insult. Our current knowledge of the mechanisms that regulate the multifaceted counter-inflammatory responses mediated by macrophages remains incomplete. Here, we report that CD169+ macrophage subsets are necessary for protection under excessive inflammatory conditions. We show that in the absence of these macrophages, even under mild septic conditions, mice fail to survive and exhibit increased production of inflammatory cytokines. Mechanistically, CD169+ macrophages control inflammatory responses via interleukin-10 (IL-10), as CD169+ macrophage-specific deletion of IL-10 was lethal during septic conditions, and recombinant IL-10 treatment reduced lipopolysaccharide (LPS)-induced lethality in mice lacking CD169+ macrophages. Collectively, our findings show a pivotal homeostatic role for CD169+ macrophages and suggest they may serve as an important target for therapy under damaging inflammatory conditions.
Collapse
Affiliation(s)
- Stephen T Yeung
- Department of Microbiology, New York University Langone School of Medicine, New York, NY 10016, USA
| | - Luis J Ovando
- Department of Microbiology, New York University Langone School of Medicine, New York, NY 10016, USA
| | - Ashley J Russo
- Department of Immunology, UConn Health School of Medicine, Farmington, CT 06032, USA
| | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, Farmington, CT 06032, USA
| | - Kamal M Khanna
- Department of Microbiology, New York University Langone School of Medicine, New York, NY 10016, USA; Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
19
|
Aramburu IV, Hoving D, Vernardis SI, Tin MCF, Ioannou M, Temkin MI, De Vasconcelos NM, Demichev V, Helbig ET, Lippert L, Stahl K, White M, Radbruch H, Ihlow J, Horst D, Chiesa ST, Deanfield JE, David S, Bode C, Kurth F, Ralser M, Papayannopoulos V. Functional proteomic profiling links deficient DNA clearance with increased mortality in individuals with severe COVID-19 pneumonia. Immunity 2022; 55:2436-2453.e5. [PMID: 36462503 PMCID: PMC9671605 DOI: 10.1016/j.immuni.2022.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 09/01/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
The factors that influence survival during severe infection are unclear. Extracellular chromatin drives pathology, but the mechanisms enabling its accumulation remain elusive. Here, we show that in murine sepsis models, splenocyte death interferes with chromatin clearance through the release of the DNase I inhibitor actin. Actin-mediated inhibition was compensated by upregulation of DNase I or the actin scavenger gelsolin. Splenocyte death and neutrophil extracellular trap (NET) clearance deficiencies were prevalent in individuals with severe COVID-19 pneumonia or microbial sepsis. Activity tracing by plasma proteomic profiling uncovered an association between low NET clearance and increased COVID-19 pathology and mortality. Low NET clearance activity with comparable proteome associations was prevalent in healthy donors with low-grade inflammation, implicating defective chromatin clearance in the development of cardiovascular disease and linking COVID-19 susceptibility to pre-existing conditions. Hence, the combination of aberrant chromatin release with defects in protective clearance mechanisms lead to poor survival outcomes.
Collapse
Affiliation(s)
| | - Dennis Hoving
- The Francis Crick Institute, Antimicrobial Defence Laboratory, London, UK
| | - Spyros I Vernardis
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London, UK
| | - Martha C F Tin
- The Francis Crick Institute, Antimicrobial Defence Laboratory, London, UK
| | - Marianna Ioannou
- The Francis Crick Institute, Antimicrobial Defence Laboratory, London, UK
| | - Mia I Temkin
- The Francis Crick Institute, Antimicrobial Defence Laboratory, London, UK
| | | | - Vadim Demichev
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London, UK
| | - Elisa Theresa Helbig
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
| | - Lena Lippert
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
| | - Klaus Stahl
- Department of Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Hannover, Germany
| | - Matthew White
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London, UK
| | - Helena Radbruch
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neuropathology, Charitéplatz 1, 10117 Berlin, Germany
| | - Jana Ihlow
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany
| | - David Horst
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany
| | - Scott T Chiesa
- Institute of Cardiovascular Science, University College London, London, UK
| | - John E Deanfield
- Institute of Cardiovascular Science, University College London, London, UK
| | - Sascha David
- Institute for Intensive Care Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Christian Bode
- Department of Anaesthesiology and Critical Care, University Hospital Bonn, Bonn, Germany
| | - Florian Kurth
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
| | - Markus Ralser
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London, UK; Charité - Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | | |
Collapse
|
20
|
Li X, Ye Y, Peng K, Zeng Z, Chen L, Zeng Y. Histones: The critical players in innate immunity. Front Immunol 2022; 13:1030610. [PMID: 36479112 PMCID: PMC9720293 DOI: 10.3389/fimmu.2022.1030610] [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: 08/29/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
The highly conserved histones in different species seem to represent a very ancient and universal innate host defense system against microorganisms in the biological world. Histones are the essential part of nuclear matter and act as a control switch for DNA transcription. However, histones are also found in the cytoplasm, cell membranes, and extracellular fluid, where they function as host defenses and promote inflammatory responses. In some cases, extracellular histones can act as damage-associated molecular patterns (DAMPs) and bind to pattern recognition receptors (PRRs), thereby triggering innate immune responses and causing initial organ damage. Histones and their fragments serve as antimicrobial peptides (AMPs) to directly eliminate bacteria, viruses, fungi, and parasites in vitro and in vivo. Histones are also involved in phagocytes-related innate immune response as components of neutrophil extracellular traps (NETs), neutrophil activators, and plasminogen receptors. In addition, as a considerable part of epigenetic regulation, histone modifications play a vital role in regulating the innate immune response and expression of corresponding defense genes. Here, we review the regulatory role of histones in innate immune response, which provides a new strategy for the development of antibiotics and the use of histones as therapeutic targets for inflammatory diseases, sepsis, autoimmune diseases, and COVID-19.
Collapse
Affiliation(s)
- Xia Li
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Youyuan Ye
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Kailan Peng
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Zhuo Zeng
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Li Chen
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Yanhua Zeng
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China,Department of Dermatology and Venereology, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China,*Correspondence: Yanhua Zeng, ;
| |
Collapse
|