1
|
Li S, Liu J, Wu J, Zheng X. Immunological Mechanisms and Effects of Bacterial Infections in Acute-on-Chronic Liver Failure. Cells 2025; 14:718. [PMID: 40422221 DOI: 10.3390/cells14100718] [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: 04/09/2025] [Revised: 05/09/2025] [Accepted: 05/10/2025] [Indexed: 05/28/2025] Open
Abstract
Acute-on-chronic liver failure (ACLF) is a severe clinical syndrome characterized by high morbidity and mortality rates. Bacterial infection is a frequent precipitating factor and complication in ACLF patients, significantly worsening patient outcomes. Elucidating the mechanisms underlying bacterial infections and their impact on ACLF pathophysiology is crucial for developing effective therapies to reduce infection rates and mortality. Current research highlights that immune suppression in ACLF increases susceptibility to bacterial infections, which in turn exacerbate immune dysfunction. However, a comprehensive review summarizing the emerging mechanisms underlying this immunosuppression is currently lacking. This review aims to provide an overview of the latest research, focusing on alterations in the immune responses of innate immune cells-including monocytes, macrophages, and neutrophils-as well as adaptive immune cells such as T and B lymphocytes during the onset and progression of bacterial infections in ACLF. In addition, recent advances in immunomodulatory therapies, including stem cell-based interventions, will also be discussed.
Collapse
Affiliation(s)
- Sumeng Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Wu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xin Zheng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
2
|
Martelossi Cebinelli GC, de Oliveira Leandro M, Rocha Oliveira AE, Alves de Lima K, Donate PB, da Cruz Oliveira Barros C, Ramos ADS, Costa V, Bernardo Nascimento DC, Alves Damasceno LE, Tavares AC, Aquime Gonçalves AN, Imoto Nakaya HT, Cunha TM, Alves-Filho JC, Cunha FQ. CXCR4 + PD-L1 + neutrophils are increased in non-survived septic mice. iScience 2025; 28:112083. [PMID: 40241761 PMCID: PMC12003019 DOI: 10.1016/j.isci.2025.112083] [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: 09/05/2024] [Revised: 12/21/2024] [Accepted: 02/18/2025] [Indexed: 04/18/2025] Open
Abstract
The dysregulated host response to infections can lead to sepsis, a complex disease characterized by a spectrum of clinical phenotypes. Using scRNA-seq, we analyzed the immune cell of survived and non-survived CLP-septic mice to gain insights into the immunological mechanisms by which neutrophils contribute to the hyperinflammatory phenotype. Our findings reveal that non-survived mice exhibit increased frequencies of immature CXCR4+ PD-L1+ neutrophils in the bloodstream, accompanied by an accumulation of trafficking-specific CXCR4+ PD-L1+ neutrophils into the lungs. The IFN-gamma and LPS promote the PD-L1 expression on neutrophils and an activation profile associated with inflammation and organ damage. Notably, abrogating the IFN-gamma reduced susceptibility to CLP-sepsis and diminished CXCR4+ PD-L1+ neutrophils frequency. This study provides insights into the immune cell activation profiles associated with the worsening of the CLP-sepsis, and the CXCR4+ PD-L1+ neutrophils population highlighted here represents a promising target for therapeutic modulation in clinical sepsis hyperinflammatory phenotype.
Collapse
Affiliation(s)
- Guilherme Cesar Martelossi Cebinelli
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Maísa de Oliveira Leandro
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | | | - Kalil Alves de Lima
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Paula Barbim Donate
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Cleyson da Cruz Oliveira Barros
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Núcleo de Biologia Experimental, Universidade de Fortaleza (UNIFOR), Fortaleza, CE, Brazil
| | - Anderson dos Santos Ramos
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Victor Costa
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Daniele Carvalho Bernardo Nascimento
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Luis Eduardo Alves Damasceno
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Amanda Curto Tavares
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - André Nicolau Aquime Gonçalves
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Helder Takashi Imoto Nakaya
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Thiago Mattar Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - José Carlos Alves-Filho
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Fernando Queiroz Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| |
Collapse
|
3
|
Hu M, Oliveira APBN, Fang Z, Feng Y, Miranda M, Kowli S, Arunachalam PS, Vasudevan G, Hui HSY, Grifoni A, Sette A, Litvack M, Rouphael N, Suthar MS, Ji X, Maecker HT, Hagan T, Dhillon G, Nicolls MR, Pulendran B. Altered baseline immunological state and impaired immune response to SARS-CoV-2 mRNA vaccination in lung transplant recipients. Cell Rep Med 2025; 6:102050. [PMID: 40187358 PMCID: PMC12047491 DOI: 10.1016/j.xcrm.2025.102050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/08/2024] [Accepted: 03/07/2025] [Indexed: 04/07/2025]
Abstract
The effectiveness of COVID-19 mRNA vaccines is diminished in organ transplant patients. Using a multi-omics approach, we investigate the immunological state of lung transplant (LTX) recipients at baseline and after SARS-CoV-2 mRNA vaccination compared to healthy controls (HCs). LTX patients exhibit a baseline immune profile resembling severe COVID-19 and sepsis, characterized by elevated pro-inflammatory cytokines (e.g., EN-RAGE [also known as S100A12], interleukin [IL]-6), reduced human leukocyte antigen (HLA)-DR expression on monocytes and dendritic cells, impaired cytokine production, and increased plasma microbial products. Single-cell RNA sequencing identifies an enriched monocyte cluster in LTX patients marked by high S100A family expression and reduced cytokine and antigen presentation genes. Post vaccination, LTX patients show diminished antibody, B cell, and T cell responses, along with blunted innate immune signatures. Integrative analysis links these altered baseline immunological features to impaired vaccine responses. These findings provide critical insights into the immunosuppressed condition of LTX recipients and their reduced vaccine-induced adaptive and innate immune responses.
Collapse
Affiliation(s)
- Mengyun Hu
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Ana Paula B N Oliveira
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zhuoqing Fang
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Yupeng Feng
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA; Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Molly Miranda
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Sangeeta Kowli
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA; Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Gowri Vasudevan
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Harold Sai-Yin Hui
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Matthew Litvack
- Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA, USA
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA, USA
| | - Mehul S Suthar
- Department of Pediatrics, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - Xuhuai Ji
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Thomas Hagan
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gundeep Dhillon
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Mark R Nicolls
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
4
|
Terrisse R, Stephan F, Walter M, Lemey C. Predicting the evolution from first-episode psychosis to mood or psychotic disorder: A systematic review of biological markers. J Affect Disord 2025; 374:26-38. [PMID: 39793620 DOI: 10.1016/j.jad.2025.01.015] [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: 09/19/2024] [Revised: 11/27/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND AND HYPOTHESIS The development of paraclinical tools to assist clinical assessment is already widespread in nearly all other medical specialties. In psychiatry, many efforts are being made to improve management strategies using these new techniques. The first episode psychosis (FEP) is a clinical entity whose evolution after onset is difficult to predict in the current state of our practices. Our main objective was to identify from the literature the most promising biological markers for early prediction of thymic or psychotic trajectories following FEP. STUDY DESIGN We performed a systematic literature review on 4 databases: PubMed, Scopus, PsycINFO, and Web of Science following PRISMA guidelines and using search terms related to FEP and biomarkers. STUDY RESULTS Eight studies were included in our final analysis. Several biomarkers showed promising discriminatory capacities for predicting post-FEP evolution: the interleukins IL-6, IL-12p40, IL-1β, and the mRNA expression levels of the DICER-1 and AKT-1 genes. Other studies that opted for broad-spectrum strategies also highlighted new leads for the discovery of additional biomarkers. CONCLUSIONS Overall, our results indicate the value of replicating studies targeting the analysis of the predictive capacities of several biological markers. It also appears important to homogenize methodologies and favor the construction of predictive models on several of these markers to reinforce their statistical significance.
Collapse
Affiliation(s)
- Raphaël Terrisse
- Service hospitalo-universitaire de psychiatrie générale et de réhabilitation psychosociale 29G01 et 29G02, ER 7479 SPURBO, CHRU de Brest, hôpital de Bohars, Brest, France.
| | - Florian Stephan
- Service hospitalo-universitaire de psychiatrie générale et de réhabilitation psychosociale 29G01 et 29G02, ER 7479 SPURBO, CHRU de Brest, hôpital de Bohars, Brest, France
| | - Michel Walter
- Service hospitalo-universitaire de psychiatrie générale et de réhabilitation psychosociale 29G01 et 29G02, ER 7479 SPURBO, CHRU de Brest, hôpital de Bohars, Brest, France
| | - Christophe Lemey
- Service hospitalo-universitaire de psychiatrie générale et de réhabilitation psychosociale 29G01 et 29G02, ER 7479 SPURBO, CHRU de Brest, hôpital de Bohars, Brest, France
| |
Collapse
|
5
|
Kathuria S, Gupta A, Tracy AR, Luna Ramirez RI, Thulasingam SK, Zaghloul N, Ahmed M, Limesand SW. Systemic inflammatory responses to repeated and increasing endotoxin challenges in fetal sheep. Physiol Rep 2025; 13:e70316. [PMID: 40268878 PMCID: PMC12018166 DOI: 10.14814/phy2.70316] [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: 02/27/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
Repeated low-dose administration of lipopolysaccharide (LPS) attenuates subsequent fetal responses, which makes it challenging to investigate interventions to prolonged exposure. Our aim was to develop a fetal inflammatory response syndrome (FIRS) model that consistently and effectively elicits a marked physiological response to increasing LPS doses. Four intravenous LPS boluses (0.3, 1.5, 3, and 15 μg) were administered to fetal sheep over 5 days. Physiological responses were measured via blood gases, pH, lactate, and cortisol concentrations. Fetal peripheral blood mononuclear cells (PBMCs) were analyzed for transcriptomic changes and tissue cytokine expression postmortem. All LPS challenges increased lactate, cortisol, and pCO2 concentrations and decreased pH and pO2 levels at 3 and 5 hours. No interaction was found between day (increasing LPS doses) and hour (LPS response to each dose). PBMC numbers increase with LPS challenges. Transcriptional analysis on PBMCs identified several enriched gene clusters indicating upregulation of inflammatory gene signatures along with complement activation and NFκB signaling pathways. Expression of pro-inflammatory cytokines (TNFα, IL-6, or IL-1β) was measured in lung, heart, liver, placenta, and spleen. Physiological indices show both respiratory and metabolic acidosis with successive and increasing LPS challenges that demonstrate a robust systemic response despite tachyphylaxis to LPS in fetal sheep.
Collapse
Affiliation(s)
- Sanya Kathuria
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| | - Akash Gupta
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Ayna R. Tracy
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| | - Rosa I. Luna Ramirez
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| | - Senthil Kumar Thulasingam
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Nahla Zaghloul
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Mohamed Ahmed
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Division of Neonatology, Department of Pediatrics, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Sean W. Limesand
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
6
|
Wang J, Liu C, Wu T, Tan L, Wang J, Zhang W, Liu Y, Wei X, Zhang D. Unveiling the role of PPIF and macrophage subtypes in LSCC progression via single-cell and exosome RNA sequencing. Sci Rep 2025; 15:10141. [PMID: 40128571 PMCID: PMC11933383 DOI: 10.1038/s41598-025-93584-3] [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: 10/12/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a highly aggressive malignancy with a rising incidence over time. The tumor microenvironment (TME) plays a crucial role in LSCC development, yet the precise cellular characteristics of laryngeal cancer and its TME remain unclear. Here, we employed single-cell RNA sequencing analysis to uncover the heterogeneous populations of tumor and immune cells and investigate the role of the TME in LSCC. This analysis revealed significant heterogeneity among malignant cells, T cells, and macrophages. Notably, regulatory T cells were markedly increased at tumor sites, and macrophage analysis identified an increased presence of the Macrophage-C1-C1QC subset with up-regulated PPIF expression. Bulk RNA-seq further confirmed PPIF up-regulation in exosomes derived from LSCC tissues. Consistently, survival analysis indicated that high PPIF expression was associated with poor prognosis in LSCC. Further analyses suggested that PPIF up-regulation in Macrophage-C1-C1QC cells was associated with the enhancement of their anti-inflammatory phenotype and the promotion of F11R-F11R signaling with malignant cells, allowing LSCC cells to evade macrophage-mediated cytotoxicity. Our study provides new insights into the cellular dynamics of LSCC and highlights the critical role of Macrophage-C1-C1QC and PPIF in LSCC progression, offering potential therapeutic targets for treatment.
Collapse
Affiliation(s)
- Jianchao Wang
- Department of Otolaryngology, The Third People's Hospital of Shenzhen (The Second Affiliated Hospital of Southern University of Science and Technology), No.29 Bulan Rd, Shenzhen, 518112, Guangdong, China
| | - Chunhan Liu
- Department of Otolaryngology, The Third People's Hospital of Shenzhen (The Second Affiliated Hospital of Southern University of Science and Technology), No.29 Bulan Rd, Shenzhen, 518112, Guangdong, China
- Vertigo Clinic, The Third People's Hospital of Shenzhen (The Second Affiliated Hospital of Southern University of Science and Technology), No.29 Bulan Rd, Shenzhen, 518112, Guangdong, China
| | - Tailin Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Lei Tan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jiangyan Wang
- Department of Otolaryngology-Head and Neck Surgery, Gansu Provincial Hospital, No.204 Donggang West Rd, Lanzhou, 730000, Gansu, China
| | - Wenliang Zhang
- Department of Bioinformatics, Outstanding Biotechnology Co., Ltd.-Shenzhen, No. 5022 Binhe Avenue, Shenzhen, 518000, Guangdong, China
| | - Yang Liu
- Department of Bioinformatics, Outstanding Biotechnology Co., Ltd.-Shenzhen, No. 5022 Binhe Avenue, Shenzhen, 518000, Guangdong, China
| | - Xudong Wei
- Department of Otolaryngology-Head and Neck Surgery, Gansu Provincial Hospital, No.204 Donggang West Rd, Lanzhou, 730000, Gansu, China.
| | - Di Zhang
- Department of Otolaryngology, The Third People's Hospital of Shenzhen (The Second Affiliated Hospital of Southern University of Science and Technology), No.29 Bulan Rd, Shenzhen, 518112, Guangdong, China.
| |
Collapse
|
7
|
Khan R, Ji W, Guzman Rivera J, Madhvi A, Andrews T, Richlin B, Suarez C, Gaur S, Hasan UN, Cuddy W, Singh AR, Bukulmez H, Kaelber D, Kimura Y, Ganapathi U, Michailidis IE, Ukey R, Moroso-Fela S, Kuster JK, Casseus M, Roy J, Burns JC, Kleinman LC, Horton DB, Lakhani SA, Gennaro ML. A genetically modulated Toll-like receptor-tolerant phenotype in peripheral blood cells of children with multisystem inflammatory syndrome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkaf006. [PMID: 40101747 PMCID: PMC11952872 DOI: 10.1093/jimmun/vkaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/02/2025] [Indexed: 03/20/2025]
Abstract
Dysregulated innate immune responses contribute to multisystem inflammatory syndrome in children (MIS-C), characterized by gastrointestinal, mucocutaneous, and/or cardiovascular injury occurring weeks after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposure. To investigate innate immune functions, we stimulated ex vivo peripheral blood cells from MIS-C patients with agonists of Toll-like receptors (TLR), key innate immune response initiators. We found severely dampened cytokine responses and elevated gene expression of negative regulators of TLR signaling. Increased plasma levels of zonulin, a gut leakage marker, were also detected. These effects were also observed in fully convalescent children months after MIS-C recovery. When we investigated the genetic background of patients in relation to TLR responsiveness, we found that cells from MIS-C children carrying rare heterozygous variants of lysosomal trafficking regulator (LYST) were less refractory to TLR stimulation and exhibited lysosomal and mitochondrial abnormalities with altered energy metabolism. Moreover, these rare LYST variant heterozygous carriers tended to exhibit unfavorable clinical laboratory indicators of inflammation, including more profound lymphopenia. The results of our observational study have several implications. First, TLR hyporesponsiveness may be associated with hyperinflammation and/or excessive or prolonged stimulation with gut-originated TLR ligands. Second, TLR hyporesponsiveness during MIS-C may be protective, since LYST variant heterozygous carriers exhibited reduced TLR hyporesponsiveness and unfavorable clinical laboratory indicators of inflammation. Thus, links may exist between genetic background, ability to establish a refractory immune state, and MIS-C clinical spectrum. Third, the possibility exists that prolonged TLR hyporesponsiveness is one of the mechanisms driving long coronavirus disease (COVID), which highlights the need to monitor long-term consequences of MIS-C.
Collapse
Affiliation(s)
- Rehan Khan
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Weizhen Ji
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| | - Jeisac Guzman Rivera
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Abhilasha Madhvi
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Tracy Andrews
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, United States
| | - Benjamin Richlin
- Pediatric Clinical Research Center, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Christian Suarez
- Pediatric Clinical Research Center, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Sunanda Gaur
- Department of Pediatrics, Clinical Research Center, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Uzma N Hasan
- Department of Pediatrics, Cooperman Barnabas Medical Center, Livingston, NJ, United States
| | - William Cuddy
- Maria Fareri Children’s Hospital, Valhalla, NY, United States
| | - Aalok R Singh
- Maria Fareri Children’s Hospital, Valhalla, NY, United States
- New York Medical College, Touro University, Valhalla, NY, United States
| | - Hulya Bukulmez
- Department of Pediatrics, Division of Rheumatology, MetroHealth System, Cleveland, OH, United States
| | - David Kaelber
- Department of Pediatrics, Division of Rheumatology, MetroHealth System, Cleveland, OH, United States
- Center for Clinical Informatics Research and Education, MetroHealth System, Cleveland, OH, United States
- Department of Internal Medicine, Pediatrics, and Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Yukiko Kimura
- Hackensack University Medical Center, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| | - Usha Ganapathi
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Ioannis E Michailidis
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Rahul Ukey
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Sandra Moroso-Fela
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - John K Kuster
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| | - Myriam Casseus
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Jason Roy
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, United States
| | - Jane C Burns
- Department of Pediatrics, University of California, San Diego, CA, United States
- Rady Children’s Hospital-San Diego, University of California, San Diego School of Medicine, San Diego, CA, United States
| | - Lawrence C Kleinman
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
- Department of Global Urban Health, Rutgers School of Public Health, Piscataway, NJ, United States
| | - Daniel B Horton
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, United States
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
- Rutgers Center for Pharmacoepidemiology and Treatment Science, Institute for Health, Health Care Policy and Aging Research, New Brunswick, NJ, United States
| | - Saquib A Lakhani
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| | - Maria Laura Gennaro
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
- Department of Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| |
Collapse
|
8
|
Zhang Z, Liu Y, Yu T, Liu Z. Unraveling the Complex Nexus of Macrophage Metabolism, Periodontitis, and Associated Comorbidities. J Innate Immun 2025; 17:211-225. [PMID: 40058341 PMCID: PMC11968099 DOI: 10.1159/000542531] [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: 03/17/2024] [Accepted: 11/07/2024] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Periodontitis is recognized as one of the most prevalent oral dysbiotic inflammatory diseases, ultimately leading to the irreversible destruction of periodontal tissues. Macrophages play a pivotal role in the development and progression of periodontitis, and the feasibility of targeting them therapeutically has been established. Since metabolic switching significantly contributes to macrophage regulation, conducting an in-depth review of macrophage metabolism in periodontitis may serve as the foundation for developing innovative treatments. SUMMARY This paper has been carefully reviewed to provide a comprehensive overview of the roles played by macrophages in periodontitis and associated comorbidities. Initially, detailed presentations on the metabolic reprogramming of macrophages, including glucose, lipid, and amino acid metabolism, were provided. Subsequently, dominating macrophage phenotype and metabolism under lipopolysaccharide (LPS) stimulation or during periodontitis were presented with emphasize on critical molecules involved. Furthermore, in recognition of the close association between periodontitis and several comorbidities, the interaction among macrophage metabolism, periodontitis, and related metabolic diseases, was thoroughly discussed. KEY MESSAGES Through the examination of current research on macrophage metabolic reprogramming induced by periodontitis, this review provides potential immunometabolic therapeutic targets for the future and raises many important, yet unstudied, subjects for follow-up. BACKGROUND Periodontitis is recognized as one of the most prevalent oral dysbiotic inflammatory diseases, ultimately leading to the irreversible destruction of periodontal tissues. Macrophages play a pivotal role in the development and progression of periodontitis, and the feasibility of targeting them therapeutically has been established. Since metabolic switching significantly contributes to macrophage regulation, conducting an in-depth review of macrophage metabolism in periodontitis may serve as the foundation for developing innovative treatments. SUMMARY This paper has been carefully reviewed to provide a comprehensive overview of the roles played by macrophages in periodontitis and associated comorbidities. Initially, detailed presentations on the metabolic reprogramming of macrophages, including glucose, lipid, and amino acid metabolism, were provided. Subsequently, dominating macrophage phenotype and metabolism under lipopolysaccharide (LPS) stimulation or during periodontitis were presented with emphasize on critical molecules involved. Furthermore, in recognition of the close association between periodontitis and several comorbidities, the interaction among macrophage metabolism, periodontitis, and related metabolic diseases, was thoroughly discussed. KEY MESSAGES Through the examination of current research on macrophage metabolic reprogramming induced by periodontitis, this review provides potential immunometabolic therapeutic targets for the future and raises many important, yet unstudied, subjects for follow-up.
Collapse
Affiliation(s)
- Zihan Zhang
- The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Liu
- The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China,
| | - Tian Yu
- Department of Stomatology, Nanbu Country People's Hospital, Nanchong, China
| | - Zhen Liu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
9
|
Moerings BGJ, Govers C, van Bergenhenegouwen J, Mes JJ, van Dijk M, Witkamp RF, van Norren K, Abbring S. Induction of endotoxin tolerance in murine monocyte and macrophage cell populations - optimal LPS dose and compartment-specific reversal by β-glucan. Food Funct 2025; 16:1576-1587. [PMID: 39917820 PMCID: PMC11803501 DOI: 10.1039/d4fo05223d] [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: 10/25/2024] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
Beta-glucans, naturally present in foods like wheat, mushrooms, and yeast, have shown potential in reversing immunosuppression. However, the existing evidence solely relies on ex vivo studies assessing direct effects of β-glucans on macrophages. To investigate whether such effects also occur after their oral administration, this study first systematically examined the immunosuppressive effects of LPS in mice. Subsequently, we assessed the ability of yeast-derived whole β-glucan particles (yWGP), administered through the diet, to counteract LPS-induced immunological tolerance. Immunosuppression following intraperitoneal administration of 20, 200, or 2000 μg kg-1 LPS was demonstrated by reduced TNF-α and IL-6 release upon ex vivo LPS stimulation of immune cells harvested from the peritoneal fluid, spleen, and bone marrow. Immunosuppression in blood was detected only after 200 and 2000 μg kg-1 LPS. LPS tolerance extended to heterologous stimuli (PAM3Cys, heat-killed Pseudomonas aeruginosa), indicating cross-tolerance. Due to animal discomfort at 2000 μg kg-1 LPS, as evidenced by a significantly enhanced clinical severity score, a dose of 200 μg kg-1 LPS was selected for the follow-up trial. In this experiment, mice fed a yWGP-supplemented diet for two weeks prior to LPS administration showed effective reversal of LPS tolerance, reflected by restored TNF-α levels in peritoneal cells but not in other monocyte- and macrophage-containing cell populations. Together, these studies demonstrate that peritoneal administration of 200 μg kg-1 LPS induced ex vivo LPS tolerance in all immunological organs studied, without significantly compromising animal welfare. The selective efficacy of dietary β-glucans to counteract immunosuppression, which is often observed in vulnerable and immunocompromised patient populations, warrants further clinical evaluation.
Collapse
Affiliation(s)
- Bart G J Moerings
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands.
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Coen Govers
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | | | - Jurriaan J Mes
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | | | - Renger F Witkamp
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands.
| | - Klaske van Norren
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands.
| | - Suzanne Abbring
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands.
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
10
|
Shrestha D, Pant BD, Roychowdhury S, Gandhirajan A, Cross E, Chhabria M, Bauer SR, Jeng M, Mitchell M, Mehkri O, Zaidi F, Ahuja A, Wang X, Wang Y, McDonald C, Longworth MS, Stappenbeck TS, Stark GR, Scheraga RG, Vachharajani V. Immunometabolic chaos in septic shock. J Leukoc Biol 2025; 117:qiae211. [PMID: 39340428 PMCID: PMC11879763 DOI: 10.1093/jleuko/qiae211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/09/2024] [Accepted: 09/26/2024] [Indexed: 09/30/2024] Open
Abstract
Septic shock is associated with over 40% mortality. The immune response in septic shock is tightly regulated by cellular metabolism and transitions from early hyper-inflammation to later hypo-inflammation. Patients are susceptible to secondary infections during hypo-inflammation. The magnitude of the metabolic dysregulation and the effect of plasma metabolites on the circulating immune cells in septic shock are not reported. We hypothesized that the accumulated plasma metabolites affect the immune response in septic shock during hypo-inflammation. Our study took a unique approach. Using peripheral blood from adult septic shock patients and healthy controls, we studied: (i) Whole blood stimulation ± E. Coli lipopolysaccharide (LPS: endotoxin) to analyze plasma TNF protein, and (ii). Plasma metabolomic profile by Metabolon. Inc. (iii) We exposed peripheral blood mononuclear cells (PBMCs) from healthy controls to commercially available carbohydrate, amino acid, and fatty acid metabolites and studied the response to LPS. We report that: (i) The whole blood stimulation of the healthy control group showed a significantly upregulated TNF protein, while the septic shock group remained endotoxin tolerant, a biomarker for hypo-inflammation. (ii) A significant accumulation of carbohydrate, amino acid, fatty acid, ceramide, sphingomyelin, and TCA cycle pathway metabolites in septic shock plasma. (iii) In vitro exposure to 5 metabolites repressed while 2 metabolites upregulated the inflammatory response of PBMCs to LPS. We conclude that the endotoxin-tolerant phenotype of septic shock is associated with a simultaneous accumulation of plasma metabolites from multiple metabolic pathways, and these metabolites fundamentally influence the immune response profile of circulating cells.
Collapse
Affiliation(s)
- Deepmala Shrestha
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Bishnu D. Pant
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Sanjoy Roychowdhury
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Anugraha Gandhirajan
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Emily Cross
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Mamta Chhabria
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Seth R. Bauer
- Pharmacy Department, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Margaret Jeng
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Megan Mitchell
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Omar Mehkri
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Fatima Zaidi
- Discovery and Translational Science, Metabolon, 617 Davis Drive, Suite 100, Morrisville, NC 27560, United States
| | - Akash Ahuja
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Xiaofeng Wang
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Yuxin Wang
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Christine McDonald
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Michelle S. Longworth
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Thaddeus S. Stappenbeck
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - George R. Stark
- Cancer Biology, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Rachel G. Scheraga
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Vidula Vachharajani
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| |
Collapse
|
11
|
Kim NH, Sim SJ, Han HG, Yoon JH, Han YH. Immunosenescence and age-related immune cells: causes of age-related diseases. Arch Pharm Res 2025; 48:132-149. [PMID: 39725853 DOI: 10.1007/s12272-024-01529-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Immunosenescence is a weakening of the immune system due to aging, characterized by changes in immune cells and dysregulated immune function. Age-related immune cells are increasing with aging. They are associated with chronic prolonged inflammation, causing tissue dysfunction and age-related diseases. Here, we discuss increased pro-inflammatory activity of aged macrophages, accumulation of lymphocytes with an age-associated phenotype, and specific alterations in both functions and characteristics of these immune cells. These cellular changes are associated with development of age-related diseases. Additionally, we reviewed various therapeutic strategies targeting age-related immunosenescence, providing pathways to mitigate effects of age-related diseases.
Collapse
Affiliation(s)
- Nam-Hee Kim
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - So-Jin Sim
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Hong-Gyu Han
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Jeong-Hyuk Yoon
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Yong-Hyun Han
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea.
- Multidimentional Genomics Research Center, Kangwon National University, Chuncheon, 24341, South Korea.
| |
Collapse
|
12
|
Dou MH, Huang JY, Li PY, Chen WL, Wang XR, Yang TZ, Fan XY, Zhang XY, Lu Y, Bai J, Du SY. How can traditional Chinese medicine enhance the efficacy of antibiotics in the treatment of MRSA-infected pneumonia: An experimental study on the combination of Reyanning mixture (RYN) and linezolid. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119221. [PMID: 39653103 DOI: 10.1016/j.jep.2024.119221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Reyanning Mixture (RYN) is a Chinese patent medicine widely used in the treatment of respiratory inflammatory diseases in China and has potential in the treatment of bacteria-infected pneumonia. AIM OF THE STUDY The present study aimed to demonstrate the therapeutic potential of RYN in combination with linezolid for the treatment of MRSA-infected pneumonia and to explore the mechanisms of action and active components. MATERIALS AND METHODS The pharmacodynamics of RYN alone and in combination with linezolid was investigated in a rat model of MRSA-induced pneumonia. Transcriptomics, ELISA, Western blot and qRT-PCR were used to explore and verify the pharmacological mechanism of the anti-inflammatory effect of RYN. UPLC-MS and molecular docking were used to explore the anti-inflammatory components of RYN for the treatment of MRSA-infected pneumonia. RESULTS In vivo, RYN reduced lung injury and inflammation in rats with pneumonia. In particular, the combination of RYN and linezolid enhanced the therapeutic effect compared to that of either treatment alone. Further research suggests that the synergistic therapeutic effect of the combination may be related to the inhibition of the inflammatory response by RYN and the enhancement of linezolid inhibition and clearance of MRSA in lung tissues by RYN. RYN plays an anti-inflammatory role in MRSA-infected pneumonia by inhibiting the TLR2/NF-κB/NLRP3 pathway, with 7 active components that may play a dominant role. CONCLUSIONS These results indicate that RYN may serve as an adjuvant drug to antibiotics for the treatment of MRSA-associated pneumonia. Exploration of its mechanisms and active components is conducive to the clinical application and quality improvement of RYN. More importantly, this study showed that the synergistic therapeutic effect of the combination of traditional Chinese medicine and antibiotics may be a valuable therapeutic strategy.
Collapse
MESH Headings
- Animals
- Linezolid/pharmacology
- Linezolid/administration & dosage
- Linezolid/therapeutic use
- Anti-Bacterial Agents/pharmacology
- Anti-Bacterial Agents/administration & dosage
- Anti-Bacterial Agents/therapeutic use
- Methicillin-Resistant Staphylococcus aureus/drug effects
- Male
- Rats, Sprague-Dawley
- Rats
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/administration & dosage
- Drugs, Chinese Herbal/therapeutic use
- Drug Therapy, Combination
- Medicine, Chinese Traditional/methods
- Drug Synergism
- Staphylococcal Infections/drug therapy
- Pneumonia, Staphylococcal/drug therapy
- Pneumonia, Staphylococcal/microbiology
- Lung/drug effects
- Lung/pathology
- Lung/microbiology
- Anti-Inflammatory Agents/pharmacology
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Disease Models, Animal
- Molecular Docking Simulation
Collapse
Affiliation(s)
- Min-Hang Dou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jia-Yi Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Peng-Yue Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wan-Ling Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin-Ran Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tian-Zi Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiao-Yu Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin-Yu Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Jie Bai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shou-Ying Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
13
|
Yoon SH, Kim CY, Lee E, Lee C, Lee KS, Lee J, Park H, Choi B, Hwang I, Kim J, Kim TG, Son J, Hyun YM, Hong S, Yu JW. Microglial NLRP3-gasdermin D activation impairs blood-brain barrier integrity through interleukin-1β-independent neutrophil chemotaxis upon peripheral inflammation in mice. Nat Commun 2025; 16:699. [PMID: 39814731 PMCID: PMC11735931 DOI: 10.1038/s41467-025-56097-1] [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: 04/09/2024] [Accepted: 01/09/2025] [Indexed: 01/30/2025] Open
Abstract
Blood-brain barrier (BBB) disintegration is a key contributor to neuroinflammation; however, the biological processes governing BBB permeability under physiological conditions remain unclear. Here, we investigate the role of NLRP3 inflammasome in BBB disruption following peripheral inflammatory challenges. Repeated intraperitoneal lipopolysaccharide administration causes NLRP3-dependent BBB permeabilization and myeloid cell infiltration into the brain. Using a mouse model with cell-specific hyperactivation of NLRP3, we identify microglial NLRP3 activation as essential for peripheral inflammation-induced BBB disruption. Conversely, NLRP3 and microglial gasdermin D (GSDMD) deficiency markedly attenuates lipopolysaccharide-induced BBB breakdown. Notably, IL-1β is not required for NLRP3-GSDMD-mediated BBB disruption. Instead, microglial NLRP3-GSDMD axis upregulates CXCL chemokines and matrix metalloproteinases around BBB via producing GDF-15, promoting the recruitment of CXCR2-containing neutrophils. Inhibition of neutrophil infiltration and matrix metalloproteinase activity significantly reduces NLRP3-mediated BBB impairment. Collectively, these findings reveal the important role of NLRP3-driven chemokine production in BBB disintegration, suggesting potential therapeutic targets to mitigate neuroinflammation.
Collapse
Affiliation(s)
- Sung-Hyun Yoon
- Department of Microbiology and Immunology, Brain Korea 21 Project for Medical Science, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chae Youn Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Eunju Lee
- Department of Microbiology and Immunology, Brain Korea 21 Project for Medical Science, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| | - Changjun Lee
- Department of Microbiology and Immunology, Brain Korea 21 Project for Medical Science, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Seo Lee
- Department of Microbiology and Immunology, Brain Korea 21 Project for Medical Science, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jaeho Lee
- Department of Anatomy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hana Park
- Doping Control Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Bokeum Choi
- Department of Microbiology and Immunology, Brain Korea 21 Project for Medical Science, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Inhwa Hwang
- Department of Microbiology and Immunology, Brain Korea 21 Project for Medical Science, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Junhan Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Tae-Gyun Kim
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Junghyun Son
- Doping Control Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Young-Min Hyun
- Department of Anatomy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seunghee Hong
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | - Je-Wook Yu
- Department of Microbiology and Immunology, Brain Korea 21 Project for Medical Science, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Chavez C, Lin K, Malveaux A, Gorin A, Brizuela S, Cheng QJ, Hoffmann A. IRF1 cooperates with ISGF3 or GAF to form innate immune de novo enhancers in macrophages. Sci Signal 2025; 18:eado8860. [PMID: 39772531 PMCID: PMC12052582 DOI: 10.1126/scisignal.ado8860] [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] [Accepted: 12/04/2024] [Indexed: 01/11/2025]
Abstract
Macrophages exposed to immune stimuli reprogram their epigenomes to alter their subsequent functions. Exposure to bacterial lipopolysaccharide (LPS) causes widespread nucleosome remodeling and the formation of thousands of de novo enhancers. We dissected the regulatory logic by which the network of interferon regulatory factors (IRFs) induces the opening of chromatin and the formation of de novo enhancers. We found that LPS-activated IRF3 mediated de novo enhancer formation indirectly by activating the type I interferon (IFN)-induced ISGF3. However, ISGF3 was generally needed to collaborate with IRF1, particularly where chromatin was less accessible. At these locations, IRF1 was required for the initial opening of chromatin, with ISGF3 extending accessibility and promoting the deposition of H3K4me1, marking poised enhancers. Because IRF1 expression depends on the transcription factor NF-κB, which is activated in infected but not bystander cells, IRF-regulated enhancers required activation of both the IRF3 and NF-κB branches of the innate immune signaling network. However, type II IFN (IFN-γ), which is typically produced by T cells, may also induce IRF1 expression through the STAT1 homodimer GAF. We showed that, upon IFN-γ stimulation, IRF1 was also responsible for opening inaccessible chromatin sites that could then be exploited by GAF to form de novo enhancers. Together, our results reveal how combinatorial logic gates of IRF1-ISGF3 or IRF1-GAF restrict immune epigenomic memory formation to macrophages exposed to pathogens or IFN-γ-secreting T cells but not bystander macrophages exposed transiently to type I IFN.
Collapse
Affiliation(s)
- Carolina Chavez
- Molecular and Medical Pharmacology PhD Program, UCLA
- Department of Microbiology, Immunology and Molecular Genetics, UCLA
| | - Kelly Lin
- Department of Microbiology, Immunology and Molecular Genetics, UCLA
| | - Alexis Malveaux
- Department of Microbiology, Immunology and Molecular Genetics, UCLA
| | - Aleksandr Gorin
- Department of Medicine, David Geffen School of Medicine, UCLA
| | | | - Quen J. Cheng
- Department of Medicine, David Geffen School of Medicine, UCLA
| | | |
Collapse
|
15
|
McCain KM, Mansilla G, Sheldon EL, Zimmer C, Schrey AW, Rowe M, Dor R, Kohl KD, Søraker JS, Jensen H, Mathot KJ, Vu T, Phuong HT, Jimeno B, Buchanan KL, Thiam M, Briskie J, Martin LB. Microbial surveillance versus cytokine responsiveness in native and non-native house sparrows. Biol Lett 2025; 21:20240431. [PMID: 39878139 PMCID: PMC11776021 DOI: 10.1098/rsbl.2024.0431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/24/2024] [Accepted: 11/12/2024] [Indexed: 01/31/2025] Open
Abstract
The success of introduced species often relies on flexible traits, including immune system traits. While theories predict non-natives will have weak defences due to decreased parasite pressure, effective parasite surveillance remains crucial, as infection risk is rarely zero and the evolutionary novelty of infection is elevated in non-native areas. This study examines the relationship between parasite surveillance and cytokine responsiveness in native and non-native house sparrows, hypothesizing that non-natives maintain high pathogen surveillance while avoiding costly inflammation. We made this specific prediction, as this pattern could enable invaders to effectively mitigate pathogen risk in a manner commensurate with the life-history priorities of a colonizing organism (i.e. rapid maturation and high reproductive effort). To test this hypothesis, we measured TLR-2 and TLR-4 expression, markers of pathogen surveillance and cytokine responses (changes in IL-1β and IL-10), regulators of inflammation, to a simulated bacterial infection. In non-native sparrows, we found that as TLR-4 expression increased, IL-1β and IL-10 responses decreased, a relationship not observed in native sparrows. Additionally, higher body condition predicted larger IL-1β and IL-10 responses in all birds. These findings suggest that high TLR-4 surveillance may mitigate strong inflammatory responses in non-native sparrows, with pathological and resource-based costs driving immune variation among and within populations.
Collapse
Affiliation(s)
- Kailey M. McCain
- Global Enviornmental and Genomic Health Sciences, University of South Florida, Tampa, FL33612, USA
| | - Gabby Mansilla
- Global Enviornmental and Genomic Health Sciences, University of South Florida, Tampa, FL33612, USA
| | - Elizabeth L. Sheldon
- Sorbonne Université, Villefranche Oceanography Laboratory, 181 Chem. du Lazaret, Villefranche-sur-Mer06230, France
| | - Cedric Zimmer
- Laboratoire d’Ethologie Expérimentale et Comparée, LEEC, Université Sorbonne Paris Nord, UR 4443, Villetaneuse93430, France
| | - Aaron W. Schrey
- Department of Biology, Georgia Southern University Armstrong Campus, Science Center 1505,16 11935 Abercorn Street, Savannah, GA31419, USA
| | - Melissah Rowe
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO‐KNAW), Wageningen6700 AB, The Netherlands
| | - Roi Dor
- Department of Natural Sciences, The Open University of Israel, Ra’anana, Israel
| | - Kevin D. Kohl
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue Pittsburgh, Pittsburgh, PA15260, USA
| | - Jørgen S. Søraker
- Department of Biology, Centre for Conservation Biology, Norwegian University of Science and Technology, N-7491 Trondheim, Trondheim, Norway
| | - Henrik Jensen
- Department of Biology, Centre for Conservation Biology, Norwegian University of Science and Technology, N-7491 Trondheim, Trondheim, Norway
| | - Kimberley J. Mathot
- Canada Research Chair in Integrative Ecology, Department of Biological Sciences,CW405 Biological Sciences Building, University of Alberta, EdmontonAB T6G 2E9, Canada
| | - Thinh Vu
- Department of Wildlife, Faculty of Forest Resource and Environmental Management, Vietnam National University of Forestry, Chương Mỹ, Vietnam
| | - Ho Thu Phuong
- Department of Wildlife, Faculty of Forest Resource and Environmental Management, Vietnam National University of Forestry, Chương Mỹ, Vietnam
| | - Blanca Jimeno
- Department of Biological Conservation and Ecosystem Restoration, Pyrenean Institute of Ecology (IPE-CSIC). Av. Nuestra Señora de la Victoria, 22700, Jaca, Spain
| | - Katherine L. Buchanan
- School of Life and Environmental Sciences, Deakin University Geelong, Geelong, Vic3216, Australia
| | - Massamba Thiam
- Laboratory of Zoology of Terrestrial Vertebrates, Fundamental Institute of Black Africa,Cheikh Anta Diop University of Dakar Senegal, Dakar, Senegal
| | - James Briskie
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Lynn B. Martin
- Global Enviornmental and Genomic Health Sciences, University of South Florida, Tampa, FL33612, USA
| |
Collapse
|
16
|
Jiang W, Chen Y, Yu CY, Zou B, Lu Y, Yang Q, Tang Z, Mao W, Li J, Han H, Shao L, Zeng J, Chu Y, Tang J, Lu M. Alveolar epithelial cells shape lipopolysaccharide-induced inflammatory responses and reprogramming of alveolar macrophages. Eur J Immunol 2025; 55:e2350378. [PMID: 39498697 DOI: 10.1002/eji.202350378] [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: 02/19/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 11/07/2024]
Abstract
Alveolar macrophages (AMs) are sentinels in the airways, where they sense and respond to invading microbes and other stimuli. Unlike macrophages in other locations, AMs can remain responsive to Gram-negative lipopolysaccharides (LPS) after they have responded to LPS in vivo (they do not develop "endotoxin tolerance"), suggesting that the alveolar microenvironment may influence their responses. Although alveolar epithelial cells (AECs) normally limit AMs' innate responses, preventing inflammation induced by harmless antigens in the lung, how AECs influence the innate responses of AMs to infectious agents has been uncertain. Here we report that (1) after exposure to aspirated (intranasal instillation) LPS, AMs increase their responses to TLR agonists and elevate their phagocytic and bactericidal activities in mice; (2) Aspirated LPS pre-exposure increases host resistance to pulmonary infection caused by Gram-negative bacteria and the protection effect lasts for at least 35 days; (3) LPS stimulation of AECs both increases AMs' innate immune responses and prevents AMs from developing tolerance in vitro; (4) Upon LPS stimulation, AMs secreted TNF-α induces AECs to release GM-CSF, which potentiates AMs' response. These experiments have revealed a previously unappreciated role that AECs may play in boosting the innate responses of AMs and promoting resistance to pulmonary infections.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- Department of Rheumatology and Immunology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yeying Chen
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Cheng-Yun Yu
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Benkun Zou
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Yimeng Lu
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Qian Yang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Zihui Tang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Weiying Mao
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Li
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Han Han
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Lingyun Shao
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiashun Zeng
- Department of Rheumatology and Immunology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Mingfang Lu
- Department of Immunology, School of Basic Medical Sciences, Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
| |
Collapse
|
17
|
Jaisue J, Suzuki N, Nii T, Isobe N. Mammary leukocytes function of endotoxin tolerant goat induced by intrauterine infusion of lipopolysaccharide. Innate Immun 2025; 31:17534259251341659. [PMID: 40405625 PMCID: PMC12102568 DOI: 10.1177/17534259251341659] [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: 12/06/2024] [Revised: 03/17/2025] [Accepted: 04/27/2025] [Indexed: 05/24/2025] Open
Abstract
A previous study found that repeated intrauterine infusions of lipopolysaccharide (LPS) followed by an LPS infusion into the mammary glands attenuated the mammary inflammatory response. This suggests that repeated LPS infusion into the uterus promotes endotoxin tolerance (ET) in the mammary gland. However, the specific changes in mammary glands under ET conditions remain unclear. We hypothesized that ET affects leukocyte function in milk. This study aimed to investigate leukocyte function in milk under ET conditions induced through repeated LPS infusions into the uterus for three days followed by LPS infusion into the mammary glands of goats. Goats in the IU group (n = 17) received an infusion of 100 μg LPS in 5 ml saline into the uterus for three consecutive days (d -3, -2, and -1), whereas the goats in the control group did not receive this infusion (n = 19). On d 0, 1 μg LPS in 5 ml saline was infused into the mammary glands of both groups. Milk was collected 0, 4, 8, 12, 24, 48, 72, and 120 h after LPS intramammary infusion. The IU group decreased cytokine production (interleukin (IL)-1β, tumor necrosis factor (TNF)-α, and IL-1Ra) in milk following intramammary LPS infusion. Moreover, leukocyte activation, measured by phagocytic activity and CD11b expression, was higher in the IU group than in the control group. These findings suggest that goats exhibit enhanced leukocyte function in mammary glands under ET conditions, induced by repeated intrauterine infusion of LPS.
Collapse
Affiliation(s)
- Jirapat Jaisue
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Naoki Suzuki
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Takahiro Nii
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Naoki Isobe
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| |
Collapse
|
18
|
Piccioni A, Spagnuolo F, Baroni S, Savioli G, Valletta F, Bungaro MC, Tullo G, Candelli M, Gasbarrini A, Franceschi F. The Need for Standardized Guidelines for the Use of Monocyte Distribution Width (MDW) in the Early Diagnosis of Sepsis. J Pers Med 2024; 15:5. [PMID: 39852198 PMCID: PMC11766780 DOI: 10.3390/jpm15010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/06/2024] [Accepted: 12/25/2024] [Indexed: 01/26/2025] Open
Abstract
Sepsis is a complex and potentially life-threatening syndrome characterized by an abnormal immune response to an infection, which can lead to organ dysfunction, septic shock, and death. Early diagnosis is crucial to improving prognosis and reducing hospital management costs. This narrative review aims to summarize and evaluate the current literature on the role of monocyte distribution width (MDW) as a diagnostic biomarker for sepsis, highlighting its advantages, limitations, and potential clinical applications. MDW measures the volumetric distribution width of monocytes, reflecting monocytic anisocytosis, and is detected using advanced hematological analyzers. In 2019, it was approved by the FDA as a biomarker for sepsis due to its ability to identify systemic inflammatory response at an early stage. Thirty-one studies analyzed by us have shown that an increased MDW value is associated with a higher risk of sepsis and that its combination with clinical parameters (such as qSOFA) and other biomarkers (CRP, PCT) can enhance diagnostic sensitivity and risk stratification capacity. Despite its high sensitivity, MDW has lower specificity compared to more established biomarkers such as procalcitonin, thus requiring a multimodal integration for an accurate diagnosis. The use of MDW in emergency and intensive care settings represents an opportunity to improve early sepsis diagnosis and critical patient management, particularly when combined with other markers and clinical tools. However, further studies are needed to define a universal cut-off and confirm its validity in different clinical contexts and pathological scenarios.
Collapse
Affiliation(s)
- Andrea Piccioni
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (F.V.); (M.C.B.); (G.T.); (M.C.); (F.F.)
| | - Fabio Spagnuolo
- Facutly of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (A.G.)
| | - Silvia Baroni
- Facutly of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (A.G.)
- Unit of Chemistry, Biochemistry and Clinical Molecular Biology, Department of Laboratory and Hematological Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gabriele Savioli
- Department of Emergency Medicine and Surgery, IRCCS Fondanzione Policlinico San Matteo, 27100 Pavia, Italy;
| | - Federico Valletta
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (F.V.); (M.C.B.); (G.T.); (M.C.); (F.F.)
| | - Maria Chiara Bungaro
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (F.V.); (M.C.B.); (G.T.); (M.C.); (F.F.)
| | - Gianluca Tullo
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (F.V.); (M.C.B.); (G.T.); (M.C.); (F.F.)
| | - Marcello Candelli
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (F.V.); (M.C.B.); (G.T.); (M.C.); (F.F.)
- Facutly of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (A.G.)
| | - Antonio Gasbarrini
- Facutly of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (A.G.)
- Medical and Surgical Science Department, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (F.V.); (M.C.B.); (G.T.); (M.C.); (F.F.)
- Facutly of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (A.G.)
| |
Collapse
|
19
|
Klaimi C, Kong W, Blériot C, Haas JT. The immunological interface: dendritic cells as key regulators in metabolic dysfunction-associated steatotic liver disease. FEBS Lett 2024. [PMID: 39668616 DOI: 10.1002/1873-3468.15072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/29/2024] [Accepted: 11/12/2024] [Indexed: 12/14/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) refers to a broad spectrum of conditions associating fat accumulation in the liver (steatosis) with varying degrees of inflammation (hepatitis) and fibrosis, which can progress to cirrhosis and potentially cancer (hepatocellular carcinoma). The first stages of these diseases are reversible and the immune system, together with metabolic factors (obesity, insulin resistance, Western diet, etc.), can influence the disease trajectory leading to progression or regression. Dendritic cells are professional antigen-presenting cells that constantly sense environmental stimuli and orchestrate immune responses. Herein, we discuss the existing literature on the heterogeneity of dendritic cell lineages, states, and functions, to provide a comprehensive overview of how liver dendritic cells influence the onset and evolution of MASLD.
Collapse
Affiliation(s)
- Camilla Klaimi
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | | | - Camille Blériot
- Gustave Roussy, CNRS UMR9018, Metabolic and Systemic Aspects of Oncogenesis for New Therapeutic Approaches, Université Paris-Saclay, Villejuif, France
- Institut Necker Enfants Malades, CNRS, INSERM, Université Paris Cité, France
| | - Joel T Haas
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| |
Collapse
|
20
|
Islam MM, Watanabe E, Salma U, Ozaki M, Irahara T, Tanabe S, Katsuki R, Oishi D, Takeyama N. Immunoadjuvant therapy in the regulation of cell death in sepsis: recent advances and future directions. Front Immunol 2024; 15:1493214. [PMID: 39720718 PMCID: PMC11666431 DOI: 10.3389/fimmu.2024.1493214] [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: 09/08/2024] [Accepted: 11/18/2024] [Indexed: 12/26/2024] Open
Abstract
Sepsis is characterized by a concomitant early pro-inflammatory response by immune cells to an infection, and an opposing anti-inflammatory response that results in protracted immunosuppression. The primary pathological event in sepsis is widespread programmed cell death, or cellular self-sacrifice, of innate and adaptive immune cells, leading to profound immunological suppression. This severe immune dysfunction hampers effective primary pathogen clearance, thereby increasing the risk of secondary opportunistic infections, latent viral reactivation, multiple organ dysfunction, and elevated mortality. The types of cell death include apoptosis (type I programmed cell death), autophagy (type II programmed cell death), NETosis (a program for formation of neutrophil extracellular traps (NETs)) and other programmed cell deaths like pyroptosis, ferroptosis, necroptosis, each contributing to immunosuppression in distinct ways during the later phases of sepsis. Extensive apoptosis of lymphocytes, such as CD4+, CD8+ T cells, and B cells, is strongly associated with immunosuppression. Apoptosis of dendritic cells further compromises T and B cell survival and can induce T cell anergy or promote regulatory Treg cell proliferation. Moreover, delayed apoptosis and impaired neutrophil function contribute to nosocomial infections and immune dysfunction in sepsis. Interestingly, aberrant NETosis and the subsequent depletion of mature neutrophils also trigger immunosuppression, and neutrophil pyroptosis can positively regulate NETosis. The interaction between programmed cell death 1 (PD-1) or programmed cell death 1 ligand (PD-L1) plays a key role in T cell modulation and neutrophil apoptosis in sepsis. The dendritic cell growth factor, Fms-like tyrosine kinase (FLTEL), increases DC numbers, enhances CD 28 expression, attenuates PD-L1, and improves survival in sepsis. Recently, immunoadjuvant therapies have attracted attention for their potential to restore host physiological immunity and homeostasis in patients with sepsis. This review focuses on several potential immunotherapeutic agents designed to bolster suppressed innate and adaptive immune responses in the management of sepsis.
Collapse
Affiliation(s)
- Md. Monirul Islam
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong (USTC), Chattogram, Bangladesh
| | - Eizo Watanabe
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Umme Salma
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Masayuki Ozaki
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Takayuki Irahara
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Subaru Tanabe
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Ryusuke Katsuki
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Dai Oishi
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Naoshi Takeyama
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
21
|
Medina CK, Aykut B. Gut Microbial Dysbiosis and Implications in Solid Organ Transplantation. Biomedicines 2024; 12:2792. [PMID: 39767699 PMCID: PMC11673786 DOI: 10.3390/biomedicines12122792] [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: 08/15/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The gut microbiome has been shown to play a significant role in solid organ transplantation, potentially influencing graft function and patient outcomes. Dysbiosis, characterized by reduced microbial diversity and an increase in pathogenic taxa, has been linked to higher incidences of allograft rejection, graft dysfunction, and post-transplant mortality. Several studies suggest that the gut microbiome might be able to serve as both a biomarker and a therapeutic target, potentially guiding personalized immunosuppressive therapies and other interventions to improve outcomes after solid organ transplantation. As summarized in this review, clinical studies have shown that specific microbial shifts correlate with adverse outcomes, including acute rejection and chronic allograft dysfunction. As research surrounding the relationship between the gut microbiome and solid organ transplant progresses, the integration of microbial analysis into clinical practice has the potential to revolutionize post-transplant care, offering new avenues to improve graft survival and patient quality of life. This review aims to provide a comprehensive overview of the relationship between gut microbial dysbiosis and transplantation outcomes, emphasizing the impact on kidney, liver, lung, and heart transplant recipients.
Collapse
Affiliation(s)
| | - Berk Aykut
- Department of Surgery, Duke University, Durham, NC 27710, USA
| |
Collapse
|
22
|
Nagashima M, Takeda Y, Saitoh S, Sabrina S, Araki A, Nagase S, Asao H. A loss of tuning of both pro-coagulant and inflammatory responses in monocytes in patients with preeclampsia. J Reprod Immunol 2024; 166:104334. [PMID: 39332076 DOI: 10.1016/j.jri.2024.104334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/30/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024]
Abstract
An imbalance between pro- and anti-angiogenesis is one of the leading causes of preeclampsia (PE). Monocytes, known as regulators of angiogenesis during immune responses, cooperate with platelets, but the specifics of these responses during pregnancy remain unclear. In this study, we investigated the relationship between pro-coagulant responses on monocytes [platelet activation marker CD61 as a monocyte-platelet aggregate (MPA), tissue factor (CD142), etc.], inflammatory responses [soluble CD40 ligand (sCD40L), soluble suppression of tumorigenesis-2 (sST2), etc.], and the balance of angiogenesis [soluble Fms-related receptor tyrosine kinase 1/placental growth factor (sFlt-1/PLGF) ratio]. In PE, markers of pro-coagulant and inflammatory responses were higher than those in normal pregnancy (NP). Interestingly, in NP, these markers harmonized with the sFlt-1/PLGF ratio, but not in PE. Furthermore, ex vivo examinations showed that upregulation of CD142 induced by additional platelet activation with adenosine diphosphate was diminished in PE. Conversely, low-dose aspirin, which is used as a preventive treatment for PE, could inhibit the increase of CD61 and sST2 under inflammatory stimuli and platelet activation in NP but not in PE. These results indicate that monocytes in PE upregulate basal activity and lose responsiveness to stimulation. The elevation of pro-coagulant and inflammatory responses may be mitigated by prophylaxis with low-dose aspirin. Therefore, the findings suggesting a loss of tuning of both pro-coagulant and inflammatory responses on monocytes help in understanding the pathology of PE. The harmonization between pro-coagulant responses, inflammatory responses, and angiogenesis may serve as useful indicators for the prediction and preventive treatment of PE.
Collapse
Affiliation(s)
- Mikako Nagashima
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan; Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yuji Takeda
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan.
| | - Shinichi Saitoh
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Saima Sabrina
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Akemi Araki
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Satoru Nagase
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Hironobu Asao
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan
| |
Collapse
|
23
|
Liao Y, Zhang W, Zhou M, Zhu C, Zou Z. Ubiquitination in pyroptosis pathway: A potential therapeutic target for sepsis. Cytokine Growth Factor Rev 2024; 80:72-86. [PMID: 39294049 DOI: 10.1016/j.cytogfr.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Sepsis remains a significant clinical challenge, causing numerous deaths annually and representing a major global health burden. Pyroptosis, a unique form of programmed cell death characterized by cell lysis and the release of inflammatory mediators, is a crucial factor in the pathogenesis and progression of sepsis, septic shock, and organ dysfunction. Ubiquitination, a key post-translational modification influencing protein fate, has emerged as a promising target for managing various inflammatory conditions, including sepsis. This review integrates the current knowledge on sepsis, pyroptosis, and the ubiquitin system, focusing on the molecular mechanisms of ubiquitination within pyroptotic pathways activated during sepsis. By exploring how modulating ubiquitination can regulate pyroptosis and its associated inflammatory signaling pathways, this review provides insights into potential therapeutic strategies for sepsis, highlighting the need for further research into these complex molecular networks.
Collapse
Affiliation(s)
- Yan Liao
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Wangzheqi Zhang
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Miao Zhou
- Department of Anesthesiology, the Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Chenglong Zhu
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China.
| | - Zui Zou
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
24
|
Yan W, Yan Y, Luo X, Dong Y, Liang G, Miao H, Huang Z, Jiang H. Lipopolysaccharide (LPS)-induced inflammation in RAW264.7 cells is inhibited by microRNA-494-3p via targeting lipoprotein-associated phospholipase A2. Eur J Trauma Emerg Surg 2024; 50:3289-3298. [PMID: 38955820 PMCID: PMC11666615 DOI: 10.1007/s00068-024-02588-7] [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: 09/26/2023] [Accepted: 06/14/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Gram-negative bacterial lipopolysaccharide (LPS) is a major component of inflammation and plays a key role in the pathogenesis of sepsis. According to our previous study, the expression of lipoprotein-associated phospholipase A2 (Lp-PLA2) is significantly upregulated in septic patients and is positively correlated with the severity of this disease. Herein, we investigated the potential roles of Lp-PLA2-targeting microRNAs (miRNAs) in LPS-induced inflammation in murine mononuclear macrophages (RAW264.7 cells). METHODS In LPS-stimulated RAW264.7 cells, Lp-PLA2 was confirmed to be expressed during the inflammatory response. The function of microRNA-494-3p (miR-494-3p) in the LPS-induced inflammatory response of RAW264.7 cells was determined by the transfection of a miR-494-3p mimic or inhibitor in vitro. RESULTS Compared to the control, LPS induced a significant increase in the Lp-PLA2 level, which was accompanied by the release of inflammatory mediators. The bioinformatics and qRT‒PCR results indicated that the miR-494-3p level was associated with Lp-PLA2 expression in the LPS-induced inflammatory response of RAW264.7 cells. Dual-luciferase reporter assay results confirmed that the 3'-UTR of Lp-PLA2 was a functional target of microRNA-494-3p. During the LPS-induced inflammatory response of RAW264.7 cells, targeting Lp-PLA2 and transfecting miR-494-3p mimics significantly upregulated the expression of miR-494-3p, leading to a reduction in the release of inflammatory factors and conferring a protective effect on LPS-stimulated RAW264.7 cells. CONCLUSION By targeting Lp-PLA2, miR-494-3p suppresses Lp-PLA2 secretion, thereby alleviating LPS-induced inflammation, which indicates that miR-494-3p may be a potential target for sepsis treatment.
Collapse
Affiliation(s)
- Wenxiao Yan
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Yan Yan
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong University, Nantong, China
- Department of Intensive Care Unit, Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xinye Luo
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Yansong Dong
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Guiwen Liang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Hua Miao
- Medical School of Nantong University, Nantong University, Nantong, China.
- Department of Emergency Medicine, Rudong County People's Hospital, Nantong, China.
| | - Zhongwei Huang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China.
- Medical School of Nantong University, Nantong University, Nantong, China.
| | - Haiyan Jiang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China.
- Medical School of Nantong University, Nantong University, Nantong, China.
| |
Collapse
|
25
|
Garaci E, Pariano M, Nunzi E, Costantini C, Bellet MM, Antognelli C, Russo MA, Romani L. Bacteria and fungi of the lung: allies or enemies? Front Pharmacol 2024; 15:1497173. [PMID: 39584143 PMCID: PMC11584946 DOI: 10.3389/fphar.2024.1497173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024] Open
Abstract
Moving from the earlier periods in which the lungs were believed to represent sterile environments, our knowledge on the lung microbiota has dramatically increased, from the first descriptions of the microbial communities inhabiting the healthy lungs and the definition of the ecological rules that regulate its composition, to the identification of the changes that occur in pathological conditions. Despite the limitations of lung as a microbiome reservoir due to the low microbial biomass and abundance, defining its microbial composition and function in the upper and lower airways may help understanding the impact on local homeostasis and its disruption in lung diseases. In particular, the understanding of the metabolic and immune significance of microbes, their presence or lack thereof, in health and disease states could be valuable in development of novel druggable targets in disease treatments. Next-generation sequencing has identified intricate inter-microbe association networks that comprise true mutualistic or antagonistic direct or indirect relationships in the respiratory tract. In this review, the tripartite interaction of bacteria, fungi and the mammalian host is addressed to provide an integrated view of the microbial-host cross-talk in lung health and diseases from an immune and metabolic perspective.
Collapse
Affiliation(s)
- Enrico Garaci
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Cinzia Antognelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Luigina Romani
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
26
|
Ming S, Li X, Xiao Q, Qu S, Wang Q, Fang Q, Liang P, Xu Y, Yang J, Yang Y, Huang X, Wu Y. TREM2 aggravates sepsis by inhibiting fatty acid oxidation via the SHP1/BTK axis. J Clin Invest 2024; 135:e159400. [PMID: 39405126 DOI: 10.1172/jci159400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 10/08/2024] [Indexed: 01/03/2025] Open
Abstract
Impaired fatty acid oxidation (FAO) and the therapeutic benefits of FAO restoration have been revealed in sepsis. However, the regulatory factors contributing to FAO dysfunction during sepsis remain inadequately clarified. In this study, we identified a subset of lipid-associated macrophages characterized by high expression of trigger receptor expressed on myeloid cells 2 (TREM2) and demonstrated that TREM2 acted as a suppressor of FAO to increase the susceptibility to sepsis. TREM2 expression was markedly upregulated in sepsis patients and correlated with the severity of sepsis. Knockout of TREM2 in macrophages improved the survival rate and reduced inflammation and organ injuries of sepsis mice. Notably, TREM2-deficient mice exhibited decreased triglyceride accumulation and an enhanced FAO rate. Further observations showed that the blockade of FAO substantially abolished the alleviated symptoms observed in TREM2-knockout mice. Mechanically, we demonstrated that TREM2 interacted with the phosphatase SHP1 to inhibit bruton tyrosine kinase-mediated (BTK-mediated) FAO in sepsis. Our findings expand the understanding of FAO dysfunction in sepsis and reveal TREM2 as a critical regulator of FAO that may provide a promising target for the clinical treatment of sepsis.
Collapse
Affiliation(s)
- Siqi Ming
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Zhuhai Hospital, Zhuhai, China
| | - Xingyu Li
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Key Research Laboratory of Traditional Chinese Medicine in the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, the Fifth Affiliated Hospital, SunYat-Sen University, Zhuhai, China
| | - Qiang Xiao
- Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Siying Qu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Qiaohua Wang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Qiongyan Fang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Pingping Liang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yating Xu
- National Clinical Research Center for Infectious Disease, Shenzhen Third People' s Hospital, the Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Jingwen Yang
- Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Yongqiang Yang
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Zhuhai Hospital, Zhuhai, China
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Key Research Laboratory of Traditional Chinese Medicine in the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, the Fifth Affiliated Hospital, SunYat-Sen University, Zhuhai, China
- National Clinical Research Center for Infectious Disease, Shenzhen Third People' s Hospital, the Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Key Research Laboratory of Traditional Chinese Medicine in the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, the Fifth Affiliated Hospital, SunYat-Sen University, Zhuhai, China
| |
Collapse
|
27
|
Zhao X, Wang M, Zhang Y, Zhang Y, Tang H, Yue H, Zhang L, Song D. Macrophages in the inflammatory response to endotoxic shock. Immun Inflamm Dis 2024; 12:e70027. [PMID: 39387442 PMCID: PMC11465138 DOI: 10.1002/iid3.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/24/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Endotoxic shock, particularly prevalent in intensive care units, represents a significant medical challenge. Endotoxin, upon invading the host, triggers intricate interactions with the innate immune system, particularly macrophages. This activation leads to the production of inflammatory mediators such as tumor necrosis factor-alpha, interleukin-6, and interleukin-1-beta, as well as aberrant activation of the nuclear factor-kappa-B and mitogen-activated protein kinase signaling pathways. OBJECTIVE This review delves into the intricate inflammatory cascades underpinning endotoxic shock, with a particular focus on the pivotal role of macrophages. It aims to elucidate the clinical implications of these processes and offer insights into potential therapeutic strategies. RESULTS Macrophages, central to immune regulation, manifest in two distinct subsets: M1 (classically activated subtype) macrophages and M2 (alternatively activated subtype) macrophages. The former exhibit an inflammatory phenotype, while the latter adopt an anti-inflammatory role. By modulating the inflammatory response in patients with endotoxic shock, these macrophages play a crucial role in restoring immune balance and facilitating recovery. CONCLUSION Macrophages undergo dynamic changes within the immune system, orchestrating essential processes for maintaining tissue homeostasis. A deeper comprehension of the mechanisms governing macrophage-mediated inflammation lays the groundwork for an anti-inflammatory, targeted approach to treating endotoxic shock. This understanding can significantly contribute to the development of more effective therapeutic interventions.
Collapse
Affiliation(s)
- Xinjie Zhao
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
- School of MedicineXizang Minzu UniversityXianyangChina
| | - Mengjie Wang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| | - Yanru Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| | - Yiyi Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| | - Haojie Tang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| | - Hongyi Yue
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| | - Li Zhang
- Affiliated Hospital of Xizang Minzu UniversityXianyangShaanxiChina
| | - Dan Song
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| |
Collapse
|
28
|
Apps R, Biancotto A, Candia J, Kotliarov Y, Perl S, Cheung F, Farmer R, Mulè MP, Rachmaninoff N, Chen J, Martins AJ, Shi R, Zhou H, Bansal N, Schum P, Olnes MJ, Milanez-Almeida P, Han KL, Sellers B, Cortese M, Hagan T, Rouphael N, Pulendran B, King L, Manischewitz J, Khurana S, Golding H, van der Most RG, Dickler HB, Germain RN, Schwartzberg PL, Tsang JS. Acute and persistent responses after H5N1 vaccination in humans. Cell Rep 2024; 43:114706. [PMID: 39235945 PMCID: PMC11949244 DOI: 10.1016/j.celrep.2024.114706] [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: 01/16/2024] [Revised: 04/14/2024] [Accepted: 08/16/2024] [Indexed: 09/07/2024] Open
Abstract
To gain insight into how an adjuvant impacts vaccination responses, we use systems immunology to study human H5N1 influenza vaccination with or without the adjuvant AS03, longitudinally assessing 14 time points including multiple time points within the first day after prime and boost. We develop an unsupervised computational framework to discover high-dimensional response patterns, which uncover adjuvant- and immunogenicity-associated early response dynamics, including some that differ post prime versus boost. With or without adjuvant, some vaccine-induced transcriptional patterns persist to at least 100 days after initial vaccination. Single-cell profiling of surface proteins, transcriptomes, and chromatin accessibility implicates transcription factors in the erythroblast-transformation-specific (ETS) family as shaping these long-lasting signatures, primarily in classical monocytes but also in CD8+ naive-like T cells. These cell-type-specific signatures are elevated at baseline in high-antibody responders in an independent vaccination cohort, suggesting that antigen-agnostic baseline immune states can be modulated by vaccine antigens alone to enhance future responses.
Collapse
Affiliation(s)
- Richard Apps
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | | | - Julián Candia
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | - Yuri Kotliarov
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA; Biometric Research Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Rockville, MD, USA
| | - Shira Perl
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | - Foo Cheung
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | - Rohit Farmer
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | - Matthew P Mulè
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA; NIH Oxford-Cambridge Scholars Program, Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge, UCB2 0QQ Cambridge, UK
| | - Nicholas Rachmaninoff
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Jinguo Chen
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | - Andrew J Martins
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Rongye Shi
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | - Huizhi Zhou
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | - Neha Bansal
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Paula Schum
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | - Matthew J Olnes
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | | | - Kyu Lee Han
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | - Brian Sellers
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA
| | - Mario Cortese
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Thomas Hagan
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Decatur, GA 30030, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA; Hope Clinic of the Emory Vaccine Center, Decatur, GA 30030, USA
| | - Lisa King
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20993 USA
| | - Jody Manischewitz
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20993 USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20993 USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20993 USA
| | | | | | - Ronald N Germain
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA; Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Pamela L Schwartzberg
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA; Cell Signaling and Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - John S Tsang
- NIH Center for Human Immunology, NIH, Bethesda, MD 20892, USA; Multiscale Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA; Center for Systems and Engineering Immunology, Departments of Immunobiology and Biomedical Engineering, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
29
|
Boyle MJ, Engwerda CR, Jagannathan P. The impact of Plasmodium-driven immunoregulatory networks on immunity to malaria. Nat Rev Immunol 2024; 24:637-653. [PMID: 38862638 PMCID: PMC11688169 DOI: 10.1038/s41577-024-01041-5] [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] [Accepted: 04/30/2024] [Indexed: 06/13/2024]
Abstract
Malaria, caused by infection with Plasmodium parasites, drives multiple regulatory responses across the immune landscape. These regulatory responses help to protect against inflammatory disease but may in some situations hamper the acquisition of adaptive immune responses that clear parasites. In addition, the regulatory responses that occur during Plasmodium infection may negatively affect malaria vaccine efficacy in the most at-risk populations. Here, we discuss the specific cellular mechanisms of immunoregulatory networks that develop during malaria, with a focus on knowledge gained from human studies and studies that involve the main malaria parasite to affect humans, Plasmodium falciparum. Leveraging this knowledge may lead to the development of new therapeutic approaches to increase protective immunity to malaria during infection or after vaccination.
Collapse
Affiliation(s)
- Michelle J Boyle
- Life Sciences Division, Burnet Institute, Melbourne, Victoria, Australia.
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| | | | - Prasanna Jagannathan
- Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
30
|
Silva AAB, Barbeiro DF, Ariga SKK, Barbeiro HV, Coelho AMM, Chaib E, Passarelli M, Soriano FG. SEPTIC SHOCK: LPS TOLERANCE PROTECTS MITOCHONDRIAL BIOGENESIS AND RESPIRATION. Shock 2024; 62:410-415. [PMID: 38888558 DOI: 10.1097/shk.0000000000002399] [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: 06/20/2024]
Abstract
ABSTRACT Mitochondrial dysfunction is a recognized feature of sepsis, characterized by ultrastructural damage, diminished oxidative phosphorylation, and depletion of mitochondrial antioxidant capacity observed in deceased septic patients. LPS tolerance induces a controlled response to sepsis. This study aimed to evaluate the function of tolerant mitochondria after cecal ligation and puncture (CLP)-induced sepsis. Mytochondrial oxygen consumption was determined using polarography. Extraction and quantification of RNA for the expression of Tfam, Nrf-1, and Ppargc-1α, and respiratory complex activity were measured. CLP-tolerant animals presented preserved respiratory rates of S3 and S4 and a ratio of respiratory control (RCR) compared to CLP-nontolerant animals with reduced oxidative phosphorylation and increased uncoupled respiration. Complex I Vmax was reduced in septic animals; however, CLP animals sustained normal Vmax. Mitochondrial biogenesis was preserved in CLP-tolerant animals compared to the CLP-nontolerant group, likely due to increased TFAM expression. LPS tolerance protected septic animals from mitochondrial dysfunction, favoring mitochondrial biogenesis and preserving mitochondrial respiration and respiratory complex I activity.
Collapse
Affiliation(s)
- Andre Augusto Botêga Silva
- Laboratório de Emergências Clínicas (LIM 51) do Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Denise Frediani Barbeiro
- Laboratório de Emergências Clínicas (LIM 51) do Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Suely Kunimi Kubo Ariga
- Laboratório de Emergências Clínicas (LIM 51) do Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Hermes Vieira Barbeiro
- Laboratório de Emergências Clínicas (LIM 51) do Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ana Maria Mendonça Coelho
- Laboratório de Gastrocirúrgia do Departamento de Gastroenterologia e Cirurgia Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Eleazar Chaib
- Laboratório de Gastrocirúrgia do Departamento de Gastroenterologia e Cirurgia Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Francisco Garcia Soriano
- Laboratório de Emergências Clínicas (LIM 51) do Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
31
|
Barcena ML, Christiansen-Mensch C, Aslam M, Haritonow N, Ladilov Y, Regitz-Zagrosek V. Upregulation of Mitochondrial Sirt3 and Alleviation of the Inflammatory Phenotype in Macrophages by Estrogen. Cells 2024; 13:1420. [PMID: 39272992 PMCID: PMC11393879 DOI: 10.3390/cells13171420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/16/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Aging and comorbidities like type 2 diabetes and obesity contribute to the development of chronic systemic inflammation, which impacts the development of heart failure and vascular disease. Increasing evidence suggests a role of pro-inflammatory M1 macrophages in chronic inflammation. A shift of metabolism from mitochondrial oxidation to glycolysis is essential for the activation of the pro-inflammatory M1 phenotype. Thus, reprogramming the macrophage metabolism may alleviate the pro-inflammatory phenotype and protect against cardiovascular diseases. In the present study, we hypothesized that the activation of estrogen receptors leads to the elevation of the mitochondrial deacetylase Sirt3, which supports mitochondrial function and mitigates the pro-inflammatory phenotype in macrophages. MATERIALS AND METHODS Experiments were performed using the mouse macrophage cell line RAW264.7, as well as primary male or female murine bone marrow macrophages (BMMs). Macrophages were treated for 24 h with estradiol (E2) or vehicle (dextrin). The effect of E2 on Sirt3 expression was investigated in pro-inflammatory M1, anti-inflammatory/immunoregulatory M2, and naïve M0 macrophages. Mitochondrial respiration was measured by Seahorse assay, and protein expression and acetylation were determined by western blotting. RESULTS E2 treatment upregulated mitochondrial Sirt3, reduced mitochondrial protein acetylation, and increased basal mitochondrial respiration in naïve RAW264.7 macrophages. Similar effects on Sirt3 expression and mitochondrial protein acetylation were observed in primary female but not in male murine BMMs. Although E2 upregulated Sirt3 in naïve M0, pro-inflammatory M1, and anti-inflammatory/immunoregulatory M2 macrophages, it reduced superoxide dismutase 2 acetylation and suppressed mitochondrial reactive oxygen species formation only in pro-inflammatory M1 macrophages. E2 alleviated the pro-inflammatory phenotype in M1 RAW264.7 cells. CONCLUSIONS The study suggests that E2 treatment upregulates Sirt3 expression in macrophages. In primary BMMs, female-specific Sirt3 upregulation was observed. The Sirt3 upregulation was accompanied by mitochondrial protein deacetylation and the alleviation of the oxidative and pro-inflammatory phenotype in M1 macrophages. Thus, the E2-Sirt3 axis might be used in a therapeutic strategy to fight chronic systemic inflammation and prevent the development of inflammation-linked diseases.
Collapse
Affiliation(s)
- Maria Luisa Barcena
- Department of Urology, Eberhard Karl University of Tuebingen, 72076 Tuebingen, Germany
- German Center for Cardiovascular Research (DZHK), Berlin Partner Site, 10115 Berlin, Germany
| | | | - Muhammad Aslam
- Experimental Cardiology, Department of Internal Medicine I, Justus Liebig University, 35392 Giessen, Germany;
- German Center for Cardiovascular Research (DZHK), RheinMain Partner Site, 61231 Bad Nauheim, Germany
| | - Natalie Haritonow
- Department of Geriatrics and Medical Gerontology, Charité–Universitätsmedizin Berlin, 12203 Berlin, Germany;
| | - Yury Ladilov
- Department of Cardiovascular Surgery, Heart Center Brandenburg, Brandenburg Medical School, Bernau bei Berlin, 16321 Brandenburg, Germany;
| | - Vera Regitz-Zagrosek
- Institute for Gender in Medicine, Charité–Universitätsmedizin Berlin, 10115 Berlin, Germany; (C.C.-M.)
- Department of Cardiology, University Hospital Zürich, University of Zürich, 8091 Zürich, Switzerland
| |
Collapse
|
32
|
Wang W, Ma L, Liu B, Ouyang L. The role of trained immunity in sepsis. Front Immunol 2024; 15:1449986. [PMID: 39221248 PMCID: PMC11363069 DOI: 10.3389/fimmu.2024.1449986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction syndrome caused by dysregulated host response to infection, characterized by a systemic inflammatory response to infection. The use of antibiotics, fluid resuscitation, and organ support therapy has limited prognostic benefit in patients with sepsis, and its incidence is not diminishing, which is attracting increased attention in medicine. Sepsis remains one of the most debilitating and expensive illnesses. One of the main reasons of septic mortality is now understood to be disruption of immune homeostasis. Immunotherapy is revolutionizing the treatment of illnesses in which dysregulated immune responses play a significant role. This "trained immunity", which is a potent defense against infection regardless of the type of bacteria, fungus, or virus, is attributed to the discovery that the innate immune cells possess immune memory via metabolic and epigenetic reprogramming. Here we reviewed the immunotherapy of innate immune cells in sepsis, the features of trained immunity, and the relationship between trained immunity and sepsis.
Collapse
Affiliation(s)
| | | | | | - Liangliang Ouyang
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
33
|
Joosten SC, Wiersinga WJ, Poll TVD. Dysregulation of Host-Pathogen Interactions in Sepsis: Host-Related Factors. Semin Respir Crit Care Med 2024; 45:469-478. [PMID: 38950605 PMCID: PMC11663080 DOI: 10.1055/s-0044-1787554] [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: 07/03/2024]
Abstract
Sepsis stands as a prominent contributor to sickness and death on a global scale. The most current consensus definition characterizes sepsis as a life-threatening organ dysfunction stemming from an imbalanced host response to infection. This definition does not capture the intricate array of immune processes at play in sepsis, marked by simultaneous states of heightened inflammation and immune suppression. This overview delves into the immune-related processes of sepsis, elaborating about mechanisms involved in hyperinflammation and immune suppression. Moreover, we discuss stratification of patients with sepsis based on their immune profiles and how this could impact future sepsis management.
Collapse
Affiliation(s)
- Sebastiaan C.M. Joosten
- Centre for Experimental and Molecular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Willem J. Wiersinga
- Centre for Experimental and Molecular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Tom van der Poll
- Centre for Experimental and Molecular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Opgenorth J, Mayorga EJ, Abeyta MA, Rodriguez-Jimenez S, Goetz BM, Freestone AD, Baumgard LH. Intravenous lipopolysaccharide challenge in early- versus mid-lactation dairy cattle. II: The production and metabolic responses. J Dairy Sci 2024; 107:6240-6251. [PMID: 38460878 DOI: 10.3168/jds.2023-24351] [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: 10/25/2023] [Accepted: 02/02/2024] [Indexed: 03/11/2024]
Abstract
Most immunometabolic research uses mid-lactation (ML) cows. Cows in early lactation (EL) are in a presumed state of immune suppression/dysregulation and less is known about how they respond to a pathogen. Study objectives were to compare the production and metabolic responses to i.v. LPS and to differentiate between the direct effects of immune activation and the indirect effects of illness-induced hypophagia in EL and ML cows. Cows in EL (n = 11; 20 ± 2 DIM) and ML (n = 12; 131 ± 31 DIM) were enrolled in a 2 × 2 factorial design containing 2 experimental periods (P). During P1 (3 d), cows were fed ad libitum and baseline data were collected. At the initiation of P2 (3 d), cows were randomly assigned to 1 of 2 treatments by lactation stage (LS): (1) EL (EL-LPS; n = 6) or ML (ML-LPS; n = 6) cows administered i.v. a single bolus of 0.09 µg LPS/kg of BW; Escherichia coli O55:B5 or (2) pair-fed (PF) EL (EL-PF; n = 5) or ML (ML-PF; n = 6) cows administered i.v. saline. Administering LPS decreased DMI and this was more severe in EL-LPS than ML-LPS cows (34% and 11% relative to baseline, respectively). By design, P2 DMI patterns were similar in the PF groups compared with their LPS counterparts. Milk yield decreased following LPS (42% on d 1 relative to P1) and despite an exacerbated decrease in EL-LPS cows on d 1 (25% relative to ML-LPS), remained similar between LS from d 2 to 3. The EL-LPS cows had increased milk fat content, but no difference in protein and lactose percentages compared with ML-LPS cows. Further, cumulative ECM yield was increased (21%) in EL-LPS compared with ML-LPS cows. During P2, EL-LPS cows had a more intense increase in MUN and BUN than ML-LPS and EL-PF cows. Administering LPS did not cause hypoglycemia in either EL-LPS or ML-LPS cows, but glucose was increased (33%) in EL-LPS compared with EL-PF. Hyperinsulinemia occurred after LPS, and insulin was further increased in ML-LPS than EL-LPS cows (2.2-fold at 12 h peak). During P2, circulating glucagon increased only in EL-LPS cows (64% relative to all other groups). Both EL groups had increased NEFA at 3 and 6 h after LPS from baseline (56%), but NEFA in EL-LPS cows gradually returned to baseline thereafter and were reduced relative to EL-PF until 36 h (50% from 12 to 24 h). Alterations in BHB did not differ between ML groups, but EL-LPS had reduced BHB compared with EL-PF from 24 to 72 h (51%). Results indicate that there are distinct LS differences in the anorexic and metabolic responses to immune activation. Collectively, EL cows are more sensitive to the catabolic effects of LPS than ML cows, but these exacerbated metabolic responses appear coordinated to fuel an augmented immune system while simultaneously supporting milk synthesis.
Collapse
Affiliation(s)
- J Opgenorth
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - E J Mayorga
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - M A Abeyta
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | | | - B M Goetz
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - A D Freestone
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - L H Baumgard
- Department of Animal Science, Iowa State University, Ames, IA 50011.
| |
Collapse
|
35
|
Nideffer J, Ty M, Donato M, John R, Kajubi R, Ji X, Nankya F, Musinguzi K, Press KD, Yang N, Camanag K, Greenhouse B, Kamya M, Feeney ME, Dorsey G, Utz PJ, Pulendran B, Khatri P, Jagannathan P. Clinical immunity to malaria involves epigenetic reprogramming of innate immune cells. PNAS NEXUS 2024; 3:pgae325. [PMID: 39161730 PMCID: PMC11331423 DOI: 10.1093/pnasnexus/pgae325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024]
Abstract
The regulation of inflammation is a critical aspect of disease tolerance and naturally acquired clinical immunity to malaria. Here, we demonstrate using RNA sequencing and epigenetic landscape profiling by cytometry by time-of-flight, that the regulation of inflammatory pathways during asymptomatic parasitemia occurs downstream of pathogen sensing-at the epigenetic level. The abundance of certain epigenetic markers (methylation of H3K27 and dimethylation of arginine residues) and decreased prevalence of histone variant H3.3 correlated with suppressed cytokine responses among monocytes of Ugandan children. Such an epigenetic signature was observed across diverse immune cell populations and not only characterized active asymptomatic parasitemia but also correlated with future long-term disease tolerance and clinical immunity when observed in uninfected children. Pseudotime analyses revealed a potential trajectory of epigenetic change that correlated with a child's age and recent parasite exposure and paralleled the acquisition of clinical immunity. Thus, our data support a model whereby exposure to Plasmodium falciparum induces epigenetic changes that regulate excessive inflammation and contribute to naturally acquire clinical immunity to malaria.
Collapse
Affiliation(s)
- Jason Nideffer
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Maureen Ty
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michele Donato
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Rek John
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Richard Kajubi
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Xuhuai Ji
- Institute for Immunity, Infection, and Transplantation, Stanford University, Stanford, CA 94305, USA
| | | | | | | | - Nora Yang
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Kylie Camanag
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Bryan Greenhouse
- Department of Medicine, University of California, San Francisco, CA 94142, USA
| | - Moses Kamya
- School of Medicine, Makerere University, Kampala, Uganda
| | - Margaret E Feeney
- Department of Pediatrics, University of California, San Francisco, CA 94142, USA
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, CA 94142, USA
| | - Paul J Utz
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Bali Pulendran
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Purvesh Khatri
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
36
|
Cheng X, Li Y, Wang H. Activation of Wnt/β-catenin signal induces DCs to differentiate into immune tolerant regDCs in septic mice. Mol Immunol 2024; 172:38-46. [PMID: 38870636 DOI: 10.1016/j.molimm.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/06/2024] [Accepted: 04/28/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Sepsis is a common complication among patients in intensive care units, and has a high mortality rate, with no effective therapies to date. As immunosuppression has become the research focus of sepsis, the regulatory role of dendritic cells (DCs) in the immune response to sepsis has received attention. OBJECTIVE To investigate the role of the Wnt/β-catenin signaling pathway in inducing the differentiation of splenic DCs in mice with sepsis caused by cecal ligation and puncture (CLP). METHODS C57bl/6 mice were randomly divided into three groups, namely the sham, 24 h post-CLP, and 72 h post-CLP groups. Levels of regulatory T cells (Tregs) among splenic mononuclear cells, suppressor T cells (TSs), and surface markers, such as major histocompatibility complex class II (MHC-II), co-stimulatory molecules (CD80 and CD86), negative co-stimulatory molecule death-ligand 1 (PD-L1), CC chemokine receptor-5 (CCR5), and CC chemokine receptor-7 (CCR7), were analyzed via flow cytometry for each group of mice post-surgery. CD11c+ DCs were purified from the splenic mononuclear cells of each group, and the expression of β-catenin, Wnt5a, and Wnt3a was detected using RT-PCR and western blotting.Each group of DCs was incubated with LPS-containing culture solution, and the supernatant of the culture solution was collected after 24 hours to detect the level of Tumor necrosis factor-α(TNF-α), interleukin (IL)-6, IL-12, and IL-10. RESULTS Compared with that in the sham group, the expression of β-catenin, Wnt5a, and Wnt3a in splenic DCs of the other two groups of mice increased with prolonged CLP exposure (P<0.05). Meanwhile, the proportion of Tregs and TSs increased in the mouse spleens after CLP, and levels of DC surface molecules, such as CCR5, CCR7, CD80, CD86, and MHC-II, decreased to different degrees, whereas those of PD-L1 increased. These results suggested that DCs differentiate towards regulatory DCs (regDCs) after CLP in mice. The results of ELISA showed that the longer the exposure time after CLP, the lower the ability of DCs to secrete TNF-α and IL-12, but the higher the level of IL-10 and IL-6. CONCLUSION The Wnt/β-catenin signaling pathway activates and induces regDCs differentiation in the splenic DCs of mice with sepsis and participates in the regulation of immune tolerance in the organism.
Collapse
Affiliation(s)
- Xia Cheng
- Graduate Training Base of Jinzhou Medical University (Department of Pathology, Fourth Medical Center, General Hospital of Chinese People's Liberation Army), Beijing 100048, China; Department of Pathology, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yazhuo Li
- Department of Pathology, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Hongwei Wang
- Department of Pathology, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China.
| |
Collapse
|
37
|
Charoensaensuk V, Yeh WL, Huang BR, Hsu TC, Xie SY, Chen CW, Wang YW, Yang LY, Tsai CF, Lu DY. Repetitive Administration of Low-Dose Lipopolysaccharide Improves Repeated Social Defeat Stress-Induced Behavioral Abnormalities and Aberrant Immune Response. J Neuroimmune Pharmacol 2024; 19:38. [PMID: 39066908 DOI: 10.1007/s11481-024-10141-x] [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: 01/30/2024] [Accepted: 07/14/2024] [Indexed: 07/30/2024]
Abstract
Repetitive exposure of innate immune cells to a subthreshold dosage of endotoxin components may modulate inflammatory responses. However, the regulatory mechanisms in the interactions between the central nervous system (CNS) and the immune system remain unclear. This study aimed to investigate the effects of lipopolysaccharide (LPS) preconditioning in repeated social defeat stress (RSDS)-induced abnormal immune responses and behavioral impairments. This study aimed to elucidate the mechanisms that underlie the protective effects of repeated administration of a subthreshold dose LPS on behavioral impairments using the RSDS paradigm. LPS preconditioning improved abnormal behaviors in RSDS-defeated mice, accompanied by decreased monoamine oxidases and increased glucocorticoid receptor expression in the hippocampus. In addition, pre-treated with LPS significantly decreased the recruited peripheral myeloid cells (CD11b+CD45hi), mainly circulating inflammatory monocytes (CD11b+CD45hiLy6ChiCCR2+) into the brain in response to RSDS challenge. Importantly, we found that LPS preconditioning exerts its protective properties by regulating lipocalin-2 (LCN2) expression in microglia, which subsequently induces expressions of chemokine CCL2 and pro-inflammatory cytokine. Subsequently, LPS-preconditioning lessened the resident microglia population (CD11b+CD45intCCL2+) in the brains of the RSDS-defeated mice. Moreover, RSDS-associated expressions of leukocytes (CD11b+CD45+CCR2+) and neutrophils (CD11b+CD45+Ly6G+) in the bone marrow, spleen, and blood were also attenuated by LPS-preconditioning. In particular, LPS preconditioning also promoted the expression of endogenous antioxidants and anti-inflammatory proteins in the hippocampus. Our results demonstrate that LPS preconditioning ameliorates lipocalin 2-associated microglial activation and aberrant immune response and promotes the expression of endogenous antioxidants and anti-inflammatory protein, thereby maintaining the homeostasis of pro-inflammation/anti-inflammation in both the brain and immune system, ultimately protecting the mice from RSDS-induced aberrant immune response and behavioral changes.
Collapse
Affiliation(s)
- Vichuda Charoensaensuk
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Lan Yeh
- Department of Biochemistry, School of Medicine, China Medical University, Taichung, 40402, Taiwan
- Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Bor-Ren Huang
- Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Tsung-Che Hsu
- School of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Sheng-Yun Xie
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chao-Wei Chen
- Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Yu-Wen Wang
- Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, China Medical University, Taichung, 404328, Taiwan
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
- Laboratory for Neural Repair, China Medical University Hospital, Taichung, 404327, Taiwan
| | - Cheng-Fang Tsai
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
38
|
Harvei S, Skogen V, Egelandsdal B, Birkeland S, Paulsen JE, Carlsen H. Chronic oral LPS administration does not increase inflammation or induce metabolic dysregulation in mice fed a western-style diet. Front Nutr 2024; 11:1376493. [PMID: 39077160 PMCID: PMC11284168 DOI: 10.3389/fnut.2024.1376493] [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: 01/25/2024] [Accepted: 06/25/2024] [Indexed: 07/31/2024] Open
Abstract
Introduction Lipopolysaccharides (LPS) present in the intestine are suggested to enter the bloodstream after consumption of high-fat diets and cause systemic inflammation and metabolic dysregulation through a process named "metabolic endotoxemia." This study aimed to determine the role of orally administered LPS to mice in the early stage of chronic low-grade inflammation induced by diet. Methods We supplemented the drinking water with E. coli derived LPS to mice fed either high-fat Western-style diet (WSD) or standard chow (SC) for 7 weeks (n = 16-17). Body weight was recorded weekly. Systemic inflammatory status was assessed by in vivo imaging of NF-κB activity at different time points, and glucose dysregulation was assessed by insulin sensitivity test and glucose tolerance test near the end of the study. Systemic LPS exposure was estimated indirectly via quantification of LPS-binding protein (LBP) and antibodies against LPS in plasma, and directly using an LPS-sensitive cell reporter assay. Results and discussion Our results demonstrate that weight development and glucose regulation are not affected by LPS. We observed a transient LPS dependent upregulation of NF-κB activity in the liver region in both diet groups, a response that disappeared within the first week of LPS administration and remained low during the rest of the study. However, WSD fed mice had overall a higher NF-κB activity compared to SC fed mice at all time points independent of LPS administration. Our findings indicate that orally administered LPS has limited to no impact on systemic inflammation and metabolic dysregulation in mice fed a high-fat western diet and we question the capability of intestinally derived LPS to initiate systemic inflammation through a healthy and uncompromised intestine, even when exposed to a high-fat diet.
Collapse
Affiliation(s)
- Silje Harvei
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Vemund Skogen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Bjørg Egelandsdal
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Signe Birkeland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Jan Erik Paulsen
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, As, Norway
| | - Harald Carlsen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| |
Collapse
|
39
|
Rentsch P, Ganesan K, Langdon A, Konen LM, Vissel B. Toward the development of a sporadic model of Alzheimer's disease: comparing pathologies between humanized APP and the familial J20 mouse models. Front Aging Neurosci 2024; 16:1421900. [PMID: 39040546 PMCID: PMC11260812 DOI: 10.3389/fnagi.2024.1421900] [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: 04/23/2024] [Accepted: 06/03/2024] [Indexed: 07/24/2024] Open
Abstract
Background Finding successful therapies for individuals with Alzheimer's disease (AD) remains an ongoing challenge. One contributing factor is that the mouse models commonly used in preclinical research primarily mimic the familial form of AD, whereas the vast majority of human cases are sporadic. Accordingly, for a sporadic mouse model of AD, incorporating the multifactorial aspects of the disease is of utmost importance. Methods In the current study, we exposed humanized Aβ knock-in mice (hAβ-KI) to weekly low-dose lipopolysaccharide (LPS) injections until 24 weeks of age and compared the development of AD pathologies to the familial AD mouse model known as the J20 mice. Results At the early time point of 24 weeks, hAβ-KI mice and J20 mice exhibited spatial memory impairments in the Barnes maze. Strikingly, both hAβ-KI mice and J20 mice showed significant loss of dendritic spines when compared to WT controls, despite the absence of Aβ plaques in hAβ-KI mice at 24 weeks of age. Glial cell numbers remained unchanged in hAβ-KI mice compared to WT, and LPS exposure in hAβ-KI mice did not result in memory deficits and failed to exacerbate any other examined AD pathology. Conclusion The study highlights the potential of hAβ-KI mice as a model for sporadic AD, demonstrating early cognitive deficits and synaptic alterations despite no evidence of Aβ plaque formation. These findings underscore the importance of considering multifactorial influences in sporadic AD pathogenesis and the need for innovative models to advance our understanding and treatment strategies for this complex disease.
Collapse
Affiliation(s)
- Peggy Rentsch
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
- UNSW St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Kiruthika Ganesan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Alexander Langdon
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Lyndsey M. Konen
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
| | - Bryce Vissel
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
- UNSW St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
40
|
Smith CT, Wang Z, Lewis JS. Engineering antigen-presenting cells for immunotherapy of autoimmunity. Adv Drug Deliv Rev 2024; 210:115329. [PMID: 38729265 DOI: 10.1016/j.addr.2024.115329] [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: 11/03/2023] [Revised: 03/05/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
Autoimmune diseases are burdensome conditions that affect a significant fraction of the global population. The hallmark of autoimmune disease is a host's immune system being licensed to attack its tissues based on specific antigens. There are no cures for autoimmune diseases. The current clinical standard for treating autoimmune diseases is the administration of immunosuppressants, which weaken the immune system and reduce auto-inflammatory responses. However, people living with autoimmune diseases are subject to toxicity, fail to mount a sufficient immune response to protect against pathogens, and are more likely to develop infections. Therefore, there is a concerted effort to develop more effective means of targeting immunomodulatory therapies to antigen-presenting cells, which are involved in modulating the immune responses to specific antigens. In this review, we highlight approaches that are currently in development to target antigen-presenting cells and improve therapeutic outcomes in autoimmune diseases.
Collapse
Affiliation(s)
- Clinton T Smith
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Zhenyu Wang
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Jamal S Lewis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| |
Collapse
|
41
|
He W, Yao C, Wang K, Duan Z, Wang S, Xie L. Single-cell landscape of immunological responses in elderly patients with sepsis. Immun Ageing 2024; 21:40. [PMID: 38909272 PMCID: PMC11193269 DOI: 10.1186/s12979-024-00446-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/11/2024] [Indexed: 06/24/2024]
Abstract
Sepsis is a dysregulated host response to severe infections, and immune dysfunction plays a crucial role in its pathogenesis. Elderly patients, a special population influenced by immunosenescence, are more susceptible to sepsis and have a worse prognosis. However, the immunopathogenic mechanisms underlying sepsis in elderly patients remain unclear. Here, we performed single-cell RNA sequencing of peripheral blood samples from young and old subjects and patients with sepsis. By exploring the transcriptional profiles of immune cells, we analyzed immune cell compositions, phenotype shifts, expression heterogeneities, and intercellular communication. In elderly patients with sepsis, innate immune cells (e.g., monocytes and DCs) exhibit decreased antigen presentation, presenting an overactive inflammatory and senescent phenotype. However, the immunophenotype of T cells shifted to characterize effector, memory, and exhaustion. Moreover, we identified strong interferon-γ responses of T cells in both aging and sepsis groups and a deranged inflammaging status in elderly sepsis patients. Tregs in elderly patients with sepsis showed increased abundance and enhanced immunosuppressive effects. In addition, metabolism-associated pathways were upregulated in T cells in elderly patients with sepsis, and the lysine metabolism pathway was enriched in Tregs. Cell-cell interaction analysis showed that the expression profile of ligand-receptor pairs was probably associated with aggravated immune dysfunction in elderly patients with sepsis. A novel HLA-KIR interaction was observed between Tregs and CD8 + T cells. These findings illustrate the immunological hallmarks of sepsis in elderly patients, and highlight that immunosuppressive and metabolic regulatory pathways may undergo important alterations in elderly patients with sepsis.
Collapse
Affiliation(s)
- Wanxue He
- Department of Pulmonary and Critical Care Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- College of Pulmonary and Critical Care Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chen Yao
- College of Pulmonary and Critical Care Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Kaifei Wang
- College of Pulmonary and Critical Care Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhimei Duan
- College of Pulmonary and Critical Care Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shuo Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - Lixin Xie
- College of Pulmonary and Critical Care Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
42
|
Sato Y, Noguchi H, Kubo S, Kaku K, Okabe Y, Onishi H, Nakamura M. Modulation of allograft immune responses by Porphyromonas gingivalis lipopolysaccharide administration in a rat model of kidney transplantation. Sci Rep 2024; 14:13969. [PMID: 38886503 PMCID: PMC11183068 DOI: 10.1038/s41598-024-64771-5] [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: 02/09/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease that affects the periodontal tissues. Although it is associated with various systemic diseases, the impact of periodontitis on kidney transplantation (KT) outcomes, particularly allograft rejection, remains unclear. This study investigated the effect of periodontitis on transplant immunity, specifically examining Porphyromonas gingivalis-derived lipopolysaccharide (LPS-PG). In vitro experiments revealed that LPS-PG increased regulatory T cells (Tregs) in Lewis rat spleen cells. In a mixed lymphocyte reaction assay, concentrations of interferon-γ, indicative of alloreactivity, were lower than in controls when LPS-PG was added to the culture and when LPS-PG-administered Lewis rat spleen cells were used as responders. In a rat KT model, LPS-PG administration to recipients promoted mild tubulitis and low serum creatinine and blood urea nitrogen levels 5 days post-KT compared with PBS-administered controls. Furthermore, LPS-PG-administered recipients had an elevated Treg proportion in their peripheral blood and spleen cells, and increased infiltrating Tregs in kidney allografts, compared with controls. The elevated Treg proportion in peripheral blood and spleen cells had a significant negative correlation with serum creatinine, suggesting elevated Tregs modulated allograft rejection. These findings suggest that periodontitis might modulate alloimmune reactivity through LPS-PG and Tregs, offering insights to refine immunosuppressive strategies for KT recipients.
Collapse
Affiliation(s)
- Yu Sato
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroshi Noguchi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Shinsuke Kubo
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Keizo Kaku
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yasuhiro Okabe
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hideya Onishi
- Department of Cancer Therapy and Research, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
43
|
Zhou M, Aziz M, Li J, Jha A, Ma G, Murao A, Wang P. BMAL2 promotes eCIRP-induced macrophage endotoxin tolerance. Front Immunol 2024; 15:1426682. [PMID: 38938563 PMCID: PMC11208452 DOI: 10.3389/fimmu.2024.1426682] [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: 05/01/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
Background The disruption of the circadian clock is associated with inflammatory and immunological disorders. BMAL2, a critical circadian protein, forms a dimer with CLOCK, activating transcription. Extracellular cold-inducible RNA-binding protein (eCIRP), released during sepsis, can induce macrophage endotoxin tolerance. We hypothesized that eCIRP induces BMAL2 expression and promotes macrophage endotoxin tolerance through triggering receptor expressed on myeloid cells-1 (TREM-1). Methods C57BL/6 wild-type (WT) male mice were subjected to sepsis by cecal ligation and puncture (CLP). Serum levels of eCIRP 20 h post-CLP were assessed by ELISA. Peritoneal macrophages (PerM) were treated with recombinant mouse (rm) CIRP (eCIRP) at various doses for 24 h. The cells were then stimulated with LPS for 5 h. The levels of TNF-α and IL-6 in the culture supernatants were assessed by ELISA. PerM were treated with eCIRP for 24 h, and the expression of PD-L1, IL-10, STAT3, TREM-1 and circadian genes such as BMAL2, CRY1, and PER2 was assessed by qPCR. Effect of TREM-1 on eCIRP-induced PerM endotoxin tolerance and PD-L1, IL-10, and STAT3 expression was determined by qPCR using PerM from TREM-1-/- mice. Circadian gene expression profiles in eCIRP-treated macrophages were determined by PCR array and confirmed by qPCR. Induction of BMAL2 activation in bone marrow-derived macrophages was performed by transfection of BMAL2 CRISPR activation plasmid. The interaction of BMAL2 in the PD-L1 promoter was determined by computational modeling and confirmed by the BIAcore assay. Results Serum levels of eCIRP were increased in septic mice compared to sham mice. Macrophages pre-treated with eCIRP exhibited reduced TNFα and IL-6 release upon LPS challenge, indicating macrophage endotoxin tolerance. Additionally, eCIRP increased the expression of PD-L1, IL-10, and STAT3, markers of immune tolerance. Interestingly, TREM-1 deficiency reversed eCIRP-induced macrophage endotoxin tolerance and significantly decreased PD-L1, IL-10, and STAT3 expression. PCR array screening of circadian clock genes in peritoneal macrophages treated with eCIRP revealed the elevated expression of BMAL2, CRY1, and PER2. In eCIRP-treated macrophages, TREM-1 deficiency prevented the upregulation of these circadian genes. In macrophages, inducible BMAL2 expression correlated with increased PD-L1 expression. In septic human patients, blood monocytes exhibited increased expression of BMAL2 and PD-L1 in comparison to healthy subjects. Computational modeling and BIAcore assay identified a putative binding region of BMAL2 in the PD-L1 promoter, suggesting BMAL2 positively regulates PD-L1 expression in macrophages. Conclusion eCIRP upregulates BMAL2 expression via TREM-1, leading to macrophage endotoxin tolerance in sepsis. Targeting eCIRP to maintain circadian rhythm may correct endotoxin tolerance and enhance host resistance to bacterial infection.
Collapse
Affiliation(s)
- Mian Zhou
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Jingsong Li
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Alok Jha
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Gaifeng Ma
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
44
|
Yaseen MM, Abuharfeil NM, Darmani H. MDSC expansion during HIV infection: regulators, ART and immune reconstitution. Genes Immun 2024; 25:242-253. [PMID: 38605259 DOI: 10.1038/s41435-024-00272-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) become expanded in different pathological conditions including human immunodeficiency virus (HIV) infection and this may worsen the disease status and accelerate disease progression. In HIV infection, MDSCs suppress anti-HIV immune responses and hamper immune reconstitution. Understanding the factors and mechanisms of MDSC expansion during HIV infection is central to understanding the pathophysiology of HIV infection. This may pave the way to developing new therapeutic targets or strategies. In this work we addressed (i) the mechanisms that regulate MDSC expansion, (ii) the impact of antiretroviral therapy (ART) on the frequency of MDSCs during HIV infection; (iii) the impact of MDSCs on immune reconstitution during successful ART; and (iv) the potential of MDSCs as a therapeutic target.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
| | - Nizar Mohammad Abuharfeil
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Homa Darmani
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| |
Collapse
|
45
|
Zhang S, Shan J, Jie Y, Zhang X, Zhu M, Shen J, Mao K, Chen W, Wang Y, Wen Y. Inhibition of PI3K p110δ rebalanced Th17/Treg and reduced macrophages pyroptosis in LPS-induced sepsis. Mol Immunol 2024; 170:110-118. [PMID: 38653076 DOI: 10.1016/j.molimm.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/13/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Sepsis is a systemic inflammatory response syndrome caused by trauma or infection, which can lead to multiple organ dysfunction. In severe cases, sepsis can also progress to septic shock and even death. Effective treatments for sepsis are still under development. This study aimed to determine if targeting the PI3K/Akt signaling with CAL-101, a PI3K p110δ inhibitor, could alleviate lipopolysaccharide (LPS)-induced sepsis and contribute to immune tolerance. Our findings indicated that CAL-101 treatment improved survival rates and alleviated the progression of LPS-induced sepsis. Compared to antibiotics, CAL-101 not only restored the Th17/regulatory T cells (Treg) balance but also enhanced Treg cell function. Additionally, CAL-101 promoted type 2 macrophage (M2) polarization, inhibited TNF-α secretion, and increased IL-10 secretion. Moreover, CAL-101 treatment reduced pyroptosis in peritoneal macrophages by inhibiting caspase-1/gasdermin D (GSDMD) activation. This study provides a mechanistic basis for future clinical exploration of targeted therapeutics and immunomodulatory strategies in the treatment of sepsis.
Collapse
Affiliation(s)
- Shiyun Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Jiajia Shan
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Yiyang Jie
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Xian Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Minyi Zhu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Jingwen Shen
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Kefan Mao
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Wenhao Chen
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Yong Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China.
| | - Yanting Wen
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, The Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China.
| |
Collapse
|
46
|
Roychowdhury S, Pant B, Cross E, Scheraga R, Vachharajani V. Effect of ethanol exposure on innate immune response in sepsis. J Leukoc Biol 2024; 115:1029-1041. [PMID: 38066660 PMCID: PMC11136611 DOI: 10.1093/jleuko/qiad156] [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: 05/10/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 01/06/2024] Open
Abstract
Alcohol use disorder, reported by 1 in 8 critically ill patients, is a risk factor for death in sepsis patients. Sepsis, the leading cause of death, kills over 270,000 patients in the United States alone and remains without targeted therapy. Immune response in sepsis transitions from an early hyperinflammation to persistent inflammation and immunosuppression and multiple organ dysfunction during late sepsis. Innate immunity is the first line of defense against pathogen invasion. Ethanol exposure is known to impair innate and adaptive immune response and bacterial clearance in sepsis patients. Specifically, ethanol exposure is known to modulate every aspect of innate immune response with and without sepsis. Multiple molecular mechanisms are implicated in causing dysregulated immune response in ethanol exposure with sepsis, but targeted treatments have remained elusive. In this article, we outline the effects of ethanol exposure on various innate immune cell types in general and during sepsis.
Collapse
Affiliation(s)
- Sanjoy Roychowdhury
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Bishnu Pant
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Emily Cross
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Rachel Scheraga
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Department of Pulmonary and Critical Care Medicine, Integrated Hospital-Care Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland OH 44195, United States
| | - Vidula Vachharajani
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Department of Pulmonary and Critical Care Medicine, Integrated Hospital-Care Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland OH 44195, United States
| |
Collapse
|
47
|
Wu Y, Wang L, Li Y, Cao Y, Wang M, Deng Z, Kang H. Immunotherapy in the context of sepsis-induced immunological dysregulation. Front Immunol 2024; 15:1391395. [PMID: 38835773 PMCID: PMC11148279 DOI: 10.3389/fimmu.2024.1391395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Sepsis is a clinical syndrome caused by uncontrollable immune dysregulation triggered by pathogen infection, characterized by high incidence, mortality rates, and disease burden. Current treatments primarily focus on symptomatic relief, lacking specific therapeutic interventions. The core mechanism of sepsis is believed to be an imbalance in the host's immune response, characterized by early excessive inflammation followed by late immune suppression, triggered by pathogen invasion. This suggests that we can develop immunotherapeutic treatment strategies by targeting and modulating the components and immunological functions of the host's innate and adaptive immune systems. Therefore, this paper reviews the mechanisms of immune dysregulation in sepsis and, based on this foundation, discusses the current state of immunotherapy applications in sepsis animal models and clinical trials.
Collapse
Affiliation(s)
- Yiqi Wu
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Lu Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yun Li
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yuan Cao
- Department of Emergency Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Min Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zihui Deng
- Department of Basic Medicine, Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Hongjun Kang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| |
Collapse
|
48
|
Amosu MM, Jankowski AM, McCright JC, Yang BE, de Oro Fernandez JG, Moore KA, Gadde HS, Donthi M, Kaluzienski ML, Sriram V, Maisel K. Plasmacytoid dendritic cells mediate CpG-ODN induced increase in survival in a mouse model of lymphangioleiomyomatosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.06.527331. [PMID: 36798234 PMCID: PMC9934559 DOI: 10.1101/2023.02.06.527331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a devastating disease primarily found in women of reproductive age that leads to cystic destruction of the lungs. Recent work has shown that LAM causes immunosuppression and that checkpoint inhibitors can be used as LAM treatment. Toll-like receptor (TLR) agonists can also re-activate immunity and the TLR9 agonist, CpG-ODN, has been effective in treating lung cancer in animal models. Here we investigate the use of TLR9 agonist CpG-ODN as LAM immunotherapy in combination with checkpoint inhibitor, anti-PD1, standard of care rapamycin and determine the immune mechanisms underlying therapeutic efficacy. We used survival studies, flow cytometry, ELISA, and histology to assess immune response and survival after intranasal treatment with CpG-ODN in combination with rapamycin or anti-PD1 therapy in a mouse model of metastatic LAM. We found that local administration of CpG-ODN enhances survival in a mouse model of LAM. We found that a lower dose led to longer survival likely due to fewer local side effects but increased LAM nodule count and size compared to the higher dose. CpG-ODN treatment also reduced regulatory T cells and increased the number of Th17 helper T cells as well as cytotoxic T cells. These effects appear to be mediated in part by plasmacytoid dendritic cells (pDCs), as depletion of pDCs reduces survival and abrogates Th17 T cell response. Finally, we found that CpG-ODN treatment is effective in early stage and progressive disease and is additive with anti-PD1 therapy and rapamycin. In summary, we have found that TLR9 agonist CpG-ODN can be used as LAM immunotherapy and effectively synergizes with rapamycin and anti-PD1 therapy in LAM.
Collapse
Affiliation(s)
- Mayowa M Amosu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Ashleigh M Jankowski
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Jacob C McCright
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Bennett E Yang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | | | - Kaitlyn A Moore
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Havish S Gadde
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Mehul Donthi
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Michele L Kaluzienski
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Vedanth Sriram
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| |
Collapse
|
49
|
Yurakova TR, Gorshkova EA, Nosenko MA, Drutskaya MS. Metabolic Adaptations and Functional Activity of Macrophages in Homeostasis and Inflammation. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:817-838. [PMID: 38880644 DOI: 10.1134/s0006297924050043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 06/18/2024]
Abstract
In recent years, the role of cellular metabolism in immunity has come into the focus of many studies. These processes form a basis for the maintenance of tissue integrity and homeostasis, as well as represent an integral part of the immune response, in particular, inflammation. Metabolic adaptations not only ensure energy supply for immune response, but also affect the functions of immune cells by controlling transcriptional and post-transcriptional programs. Studying the immune cell metabolism facilitates the search for new treatment approaches, especially for metabolic disorders. Macrophages, innate immune cells, are characterized by a high functional plasticity and play a key role in homeostasis and inflammation. Depending on the phenotype and origin, they can either perform various regulatory functions or promote inflammation state, thus exacerbating the pathological condition. Furthermore, their adaptations to the tissue-specific microenvironment influence the intensity and type of immune response. The review examines the effect of metabolic reprogramming in macrophages on the functional activity of these cells and their polarization. The role of immunometabolic adaptations of myeloid cells in tissue homeostasis and in various pathological processes in the context of inflammatory and metabolic diseases is specifically discussed. Finally, modulation of the macrophage metabolism-related mechanisms reviewed as a potential therapeutic approach.
Collapse
Affiliation(s)
- Taisiya R Yurakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Ekaterina A Gorshkova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Maxim A Nosenko
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, D02F306, Ireland
| | - Marina S Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, 354340, Russia
| |
Collapse
|
50
|
Dulfer EA, Joosten LAB, Netea MG. Enduring echoes: Post-infectious long-term changes in innate immunity. Eur J Intern Med 2024; 123:15-22. [PMID: 38135583 DOI: 10.1016/j.ejim.2023.12.020] [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: 11/11/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
Upon encountering pathogens, the immune system typically responds by initiating an acute and self-limiting reaction, with symptoms subsiding after the pathogen has been cleared. However, long-term post-infectious clinical symptoms can manifest months or even years after the initial infection. 'Trained immunity', the functional reprogramming of innate immune cells through epigenetic and metabolic rewiring, has been proposed as a key concept for understanding these long-term effects. Although trained immunity can result in enhanced protection against reinfection with heterologous pathogens, it can also contribute to detrimental outcomes. Persisting and excessive inflammation can cause tissue damage and aggravate immune-mediated conditions and cardiovascular complications. On the other hand, suppression of immune cell effector functions by long-lasting epigenetic changes can result in post-infectious immune paralysis. Distinct stimuli can evoke different trained immunity programs, potentially resulting in different consequences for the host. In this review, we provide an overview of both the adaptive and maladaptive consequences of infectious diseases. We discuss how long-term immune dysregulation in patients can be addressed by tailoring host-directed interventions and identify areas of scientific and therapeutic potential to advance further.
Collapse
Affiliation(s)
- Elisabeth A Dulfer
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud university medical center, Nijmegen, the Netherlands.
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud university medical center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud university medical center, Nijmegen, the Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Germany
| |
Collapse
|