1
|
Blutt SE, Miller AD, Conner ME. Dendritic cell expression of MyD88 is required for rotavirus-induced B cell activation. J Virol 2025; 99:e0065325. [PMID: 40304491 PMCID: PMC12090804 DOI: 10.1128/jvi.00653-25] [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: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
Intestinal IgA, produced by local intestinal B cells, is thought to play a major role in protection against intestinal infections. Rotavirus, a well-characterized intestinal virus, induces a rapid viral-specific intestinal IgA response that occurs in the absence of T cells. Previous work has indicated that dendritic cells facilitate the early IgA response to rotavirus. To determine whether the early Peyer's patch B cell activation associated with rotavirus infection in mice requires dendritic cells, we depleted dendritic cells and assessed B cell activation. Depletion of CD11c+ cells in vivo prior to infection resulted in a complete abrogation of Peyer's patch B cell activation. With the use of in vitro cell-based assays, CD11c+, but not T or CD11b+ cells, was shown to be essential for rotavirus-induced activation of B cells. Investigation of several pathways of B cell activation revealed that dendritic cell expression of MyD88 and signaling through the type I interferon receptor were critical for the ability of the virus to induce B cell activation. These findings indicate that CD11c+ dendritic cells can modulate B cell responses to viruses through toll-like receptor and type I interferon signaling pathways.IMPORTANCEDendritic cells are key mediators of immune responses in the intestine. They can capture and process rotavirus antigens and present these antigens to B cells, which produce critical IgA antibody that is essential for clearance of rotavirus infection and protection from reinfection. In the work presented here, we demonstrate that dendritic cell expression of MyD88, a key component of pattern recognition pathways, and not classical IgA pathway molecules such as BAFF and APRIL, is critical for the ability of the dendritic cell to induce the activation of B cells. Our findings emphasize the important role that dendritic cells play in initiating and regulating immune responses including T cell-independent B cell activation. A consideration of the role of dendritic cells in B cell activation and antibody production is an important feature in the development of therapeutic and preventive modalities to combat intestinal viral infections.
Collapse
Affiliation(s)
- Sarah E. Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - Amber D. Miller
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Margaret E. Conner
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
- Huffington Department of Education, Innovation, and Technology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
2
|
Schultz TE, Mathmann CD, Domínguez Cadena LC, Muusse TW, Kim H, Wells JW, Ulett GC, Hamerman JA, Brooks AJ, Kobe B, Sweet MJ, Stacey KJ, Blumenthal A. TLR4 endocytosis and endosomal TLR4 signaling are distinct and independent outcomes of TLR4 activation. EMBO Rep 2025; 26:2740-2766. [PMID: 40204912 PMCID: PMC12116916 DOI: 10.1038/s44319-025-00444-2] [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: 08/10/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Toll-like receptor 4 (TLR4) signaling at the plasma membrane and in endosomes results in distinct contributions to inflammation and host defence. Current understanding indicates that endocytosis of cell surface-activated TLR4 is required to enable subsequent signaling from endosomes. Contrary to this prevailing model, our data show that endosomal TLR4 signaling is not reliant on cell surface-expressed TLR4 or ligand-induced TLR4 endocytosis. Moreover, previously recognized requirements for the accessory molecule CD14 in TLR4 endocytosis and endosomal signaling are likely attributable to CD14 binding as well as trafficking and transferring lipopolysaccharide (LPS) to TLR4 at different subcellular localizations. TLR4 endocytosis requires the TLR4 intracellular signaling domain, contributions by phospholipase C gamma 2, spleen tyrosine kinase, E1/E2 ubiquitination enzymes, but not canonical TLR signaling adaptors and cascades. Thus, our study identifies independently operating TLR4 signaling modes that control TLR4 endocytosis, pro-inflammatory cell surface-derived, as well as endosomal TLR4 signaling. This revised understanding of how TLR4 functions within cells might be harnessed to selectively amplify or restrict TLR4 activation for the development of adjuvants, vaccines and therapeutics.
Collapse
Affiliation(s)
- Thomas E Schultz
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Carmen D Mathmann
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | | | - Timothy W Muusse
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Hyoyoung Kim
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - James W Wells
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Glen C Ulett
- School of Pharmacy and Medical Sciences and Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4215, Australia
| | - Jessica A Hamerman
- Immunology Program, Benaroya Research Institute, Seattle, WA, 98101, USA
| | - Andrew J Brooks
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Katryn J Stacey
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Antje Blumenthal
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia.
| |
Collapse
|
3
|
Yeap HW, Goh GR, Rosli SN, Pung HS, Giogha C, Eng VV, Pearson JS, Hartland EL, Chen KW. A bacterial network of T3SS effectors counteracts host pro-inflammatory responses and cell death to promote infection. EMBO J 2025; 44:2424-2445. [PMID: 40128366 PMCID: PMC12048508 DOI: 10.1038/s44318-025-00412-5] [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: 07/30/2024] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Innate immune signalling and cell death pathways are highly interconnected processes involving receptor-interacting protein kinases (RIPKs) as mediators of potent anti-microbial responses. However, these processes are often antagonised by bacterial type III secretion system (T3SS) effectors, and the cellular mechanisms by which the host retaliates are not completely understood. Here, we demonstrate that during Citrobacter rodentium infection, murine macrophages and colonic epithelial cells exhibit RIPK1 kinase-dependent caspase-8 activation to counteract NleE effector-mediated suppression of pro-inflammatory signalling. While C. rodentium injects into the host cells a second effector, NleB, to block caspase-8 signalling, macrophages respond by triggering RIPK3-mediated necroptosis, whereupon a third T3SS effector, EspL, acts to inactivate necroptosis. We further show that NleB and EspL collaborate to suppress caspase-8 and NLRP3 inflammasome activation in macrophages. Our findings suggest that C. rodentium has evolved to express a complex network of effectors as an adaptation to the importance of cell death for anti-bacterial defence in the host-pathogen arms race.
Collapse
Affiliation(s)
- Hui Wen Yeap
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Ghin Ray Goh
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Safwah Nasuha Rosli
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Hai Shin Pung
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Cristina Giogha
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Vik Ven Eng
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Jaclyn S Pearson
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
- School of Medicine, University of St Andrews, St Andrews, KY16 9TF, Fife, UK
| | - Elizabeth L Hartland
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Kaiwen W Chen
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
4
|
Salauddin M, Bhattacharyya D, Samanta I, Saha S, Xue M, Hossain MG, Zheng C. Role of TLRs as signaling cascades to combat infectious diseases: a review. Cell Mol Life Sci 2025; 82:122. [PMID: 40105962 PMCID: PMC11923325 DOI: 10.1007/s00018-025-05631-x] [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: 06/05/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
Investigating innate immunity and its signaling transduction is essential to understand inflammation and host defence mechanisms. Toll-like receptors (TLRs), an evolutionarily ancient group of pattern recognition receptors, are crucial for detecting microbial components and initiating immune responses. This review summarizes the mechanisms and outcomes of TLR-mediated signaling, focusing on motifs shared with other immunological pathways, which enhances our understanding of the innate immune system. TLRs recognize molecular patterns in microbial invaders, activate innate immunity and promote antigen-specific adaptive immunity, and each of them triggers unique downstream signaling patterns. Recent advances have highlighted the importance of supramolecular organizing centers (SMOCs) in TLR signaling, ensuring precise cellular responses and pathogen detection. Furthermore, this review illuminates how TLR pathways coordinate metabolism and gene regulation, contributing to adaptive immunity and providing novel insights for next-generation therapeutic strategies. Ongoing studies hold promise for novel treatments against infectious diseases, autoimmune conditions, and cancers.
Collapse
Affiliation(s)
- Md Salauddin
- Department of Microbiology and Public Health, Faculty of Veterinary, Animal and Biomedical Sciences, Khulna Agricultural University, Khulna, 9202, Bangladesh
| | - Debaraj Bhattacharyya
- Department of Veterinary Biochemistry, West Bengal University of Animal and Fishery Sciences, 37, K.B. Sarani, Kolkata, West Bengal, 700037, India
| | - Indranil Samanta
- Department of Veterinary Microbiology, West Bengal University of Animal and Fishery Sciences, 37, K.B. Sarani, Kolkata, West Bengal, 700037, India
| | - Sukumar Saha
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, 450001, Henan, China.
| | - Md Golzar Hossain
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
5
|
Chen Z, Behrendt R, Wild L, Schlee M, Bode C. Cytosolic nucleic acid sensing as driver of critical illness: mechanisms and advances in therapy. Signal Transduct Target Ther 2025; 10:90. [PMID: 40102400 PMCID: PMC11920230 DOI: 10.1038/s41392-025-02174-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Nucleic acids from both self- and non-self-sources act as vital danger signals that trigger immune responses. Critical illnesses such as acute respiratory distress syndrome, sepsis, trauma and ischemia lead to the aberrant cytosolic accumulation and massive release of nucleic acids that are detected by antiviral innate immune receptors in the endosome or cytosol. Activation of receptors for deoxyribonucleic acids and ribonucleic acids triggers inflammation, a major contributor to morbidity and mortality in critically ill patients. In the past decade, there has been growing recognition of the therapeutic potential of targeting nucleic acid sensing in critical care. This review summarizes current knowledge of nucleic acid sensing in acute respiratory distress syndrome, sepsis, trauma and ischemia. Given the extensive research on nucleic acid sensing in common pathological conditions like cancer, autoimmune disorders, metabolic disorders and aging, we provide a comprehensive summary of nucleic acid sensing beyond critical illness to offer insights that may inform its role in critical conditions. Additionally, we discuss potential therapeutic strategies that specifically target nucleic acid sensing. By examining nucleic acid sources, sensor activation and function, as well as the impact of regulating these pathways across various acute diseases, we highlight the driving role of nucleic acid sensing in critical illness.
Collapse
Affiliation(s)
- Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Rayk Behrendt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Lennart Wild
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
6
|
Hsieh ML, Nishizaki D, Adashek JJ, Kato S, Kurzrock R. Toll-like receptor 3: a double-edged sword. Biomark Res 2025; 13:32. [PMID: 39988665 PMCID: PMC11849352 DOI: 10.1186/s40364-025-00739-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/02/2025] [Indexed: 02/25/2025] Open
Abstract
The discovery of Toll-like receptors (TLRs) and their role in dendritic cells earned the Nobel Prize for 2011 because TLRs profoundly enhanced our understanding of the immune system. Specifically, TLR3 is located within the endosomal compartments of dendritic cells and plays a crucial role in the immune response by acting as a pattern recognition receptor that detects both exogenous (viral) and endogenous (mammalian) double-stranded RNA. However, TLR3 activation is a double-edged sword in various immune-mediated diseases. On one hand, it can enhance anti-viral defenses and promote pathogen clearance, contributing to host protection. On the other hand, excessive or dysregulated TLR3 signaling can lead to chronic inflammation and tissue damage, exacerbating conditions such as autoimmune diseases, chronic viral infections, and cancer. In cancer, TLR3 expression has been linked to both favorable and poor prognoses, though the underlying mechanisms remain unclear. Recent clinical and preclinical advances have explored the use of TLR3 agonists in cancer immunotherapy, attempting to capitalize on their potential to enhance anti-tumor responses. The dual role of TLR3 highlights its complexity as a therapeutic target, necessitating careful modulation to maximize its protective effects while minimizing potential pathological consequences. In this review, we explore the intricate roles of TLR3 in immune responses across different disease contexts, including cancer, infections, autoimmune disorders, and allergies, highlighting both its protective and detrimental effects in these disorders, as well as progress in developing TLR3 agonists as part of the immunotherapy landscape.
Collapse
Affiliation(s)
| | - Daisuke Nishizaki
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Shumei Kato
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Razelle Kurzrock
- Medical College of Wisconsin, Milwaukee, WI, USA.
- MCW Cancer Center and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
7
|
Chan FHM, Yeap HW, Liu Z, Rosli SN, Low KE, Bonne I, Wu Y, Chong SZ, Chen KW. Plasticity of cell death pathways ensures GSDMD activation during Yersinia pseudotuberculosis infection. Cell Rep 2025; 44:115216. [PMID: 39823227 DOI: 10.1016/j.celrep.2024.115216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/30/2024] [Accepted: 12/26/2024] [Indexed: 01/19/2025] Open
Abstract
Macrophages express pattern recognition and cytokine receptors that mediate proinflammatory signal transduction pathways to combat microbial infection. To retaliate against such responses, pathogenic microorganisms have evolved multiple strategies to impede innate immune signaling. Recent studies demonstrated that YopJ suppression of TAK1 signaling during Yersinia pseudotuberculosis infection promotes the assembly of a RIPK1-dependent death-inducing complex that enables caspase-8 to directly cleave and activate gasdermin D (GSDMD). However, whether and how macrophages respond to Yersinia infection in the absence of YopJ or caspase-8 activity remains unclear. Here, we demonstrate that loss of YopJ or its catalytic activity triggers non-canonical inflammasome activation in macrophages and that caspase-11 is required to restrict the bacterial burden in vivo. Under conditions of low caspase-8 activity, wild-type Y. pseudotuberculosis invades macrophages and accesses the cytosol, leading to non-canonical inflammasome activation. Thus, our study highlights the plasticity of death pathways to ensure GSDMD activation during Yersinia infection.
Collapse
Affiliation(s)
- Felicia Hui Min Chan
- Immunology Translational Research Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| | - Hui Wen Yeap
- Immunology Translational Research Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| | - Zonghan Liu
- Immunology Translational Research Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| | - Safwah Nasuha Rosli
- Immunology Translational Research Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| | - Kay En Low
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117549, Singapore
| | - Isabelle Bonne
- Immunology Translational Research Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore; Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117549, Singapore
| | - Yixuan Wu
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Republic of Singapore
| | - Shu Zhen Chong
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore; Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Republic of Singapore
| | - Kaiwen W Chen
- Immunology Translational Research Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.
| |
Collapse
|
8
|
Shaw JR, Caprio N, Truong N, Weldemariam M, Tran A, Pilli N, Pandey S, Jones JW, Kane MA, Pearson RM. Inflammatory disease progression shapes nanoparticle biomolecular corona-mediated immune activation profiles. Nat Commun 2025; 16:924. [PMID: 39843415 PMCID: PMC11754911 DOI: 10.1038/s41467-025-56210-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
Polymeric nanoparticles (NPs) are promising tools used for immunomodulation and drug delivery in various disease contexts. The interaction between NP surfaces and plasma-resident biomolecules results in the formation of a biomolecular corona, which varies patient-to-patient and as a function of disease state. This study investigates how the progression of acute systemic inflammatory disease influences NP corona compositions and the corresponding effects on innate immune cell interactions, phenotypes, and cytokine responses. NP coronas alter cell associations in a disease-dependent manner, induce differential co-stimulatory and co-inhibitory molecule expression, and influence cytokine release. Integrated multi-omics analysis of proteomics, lipidomics, metabolomics, and cytokine datasets highlight a set of differentially enriched TLR4 ligands that correlate with dynamic NP corona-mediated immune activation. Pharmacological inhibition and genetic knockout studies validate that NP coronas mediate this response through TLR4/MyD88/NF-κB signaling. Our findings illuminate the personalized nature of corona formation under a dynamic inflammatory condition and its impact on NP-mediated immune activation profiles and inflammation, suggesting that disease progression-related alterations in plasma composition can manifest in the corona to cause unintended toxicity and altered therapeutic efficacy.
Collapse
Affiliation(s)
- Jacob R Shaw
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Nicholas Caprio
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
| | - Nhu Truong
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
| | - Mehari Weldemariam
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
| | - Anh Tran
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
| | - Nageswara Pilli
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
| | - Swarnima Pandey
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
| | - Jace W Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
| | - Ryan M Pearson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD, 21201, USA.
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD, 21201, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
9
|
Manik MK, Pan M, Xiao L, Gu W, Kim H, Pospich S, Hedger A, Vajjhala PR, Lee MYL, Qian X, Landsberg MJ, Ve T, Nanson JD, Raunser S, Stacey KJ, Wu H, Kobe B. Structural basis for TIR domain-mediated innate immune signaling by Toll-like receptor adaptors TRIF and TRAM. Proc Natl Acad Sci U S A 2025; 122:e2418988122. [PMID: 39786929 PMCID: PMC11745336 DOI: 10.1073/pnas.2418988122] [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: 09/17/2024] [Accepted: 12/10/2024] [Indexed: 01/30/2025] Open
Abstract
Innate immunity relies on Toll-like receptors (TLRs) to detect pathogen-associated molecular patterns. The TIR (Toll/interleukin-1 receptor) domain-containing TLR adaptors TRIF (TIR domain-containing adaptor-inducing interferon-β) and TRAM (TRIF-related adaptor molecule) are essential for MyD88-independent TLR signaling. However, the structural basis of TRIF and TRAM TIR domain-based signaling remains unclear. Here, we present cryo-EM structures of filaments formed by TRIF and TRAM TIR domains at resolutions of 3.3 Å and 5.6 Å, respectively. Both structures reveal two-stranded parallel helical arrangements. Functional studies underscore the importance of intrastrand interactions, mediated by the BB-loop, and interstrand interactions in TLR4-mediated signaling. We also report the crystal structure of the monomeric TRAM TIR domain bearing the BB loop mutation C117H, which reveals conformational differences consistent with its inactivity. Our findings suggest a unified signaling mechanism by the TIR domains of the four signaling TLR adaptors MyD88, MAL, TRIF, and TRAM and reveal potential therapeutic targets for immunity-related disorders.
Collapse
MESH Headings
- Adaptor Proteins, Vesicular Transport/metabolism
- Adaptor Proteins, Vesicular Transport/chemistry
- Adaptor Proteins, Vesicular Transport/genetics
- Adaptor Proteins, Vesicular Transport/immunology
- Signal Transduction/immunology
- Immunity, Innate
- Humans
- Protein Domains
- Cryoelectron Microscopy
- Receptors, Interleukin-1/metabolism
- Receptors, Interleukin-1/chemistry
- Receptors, Interleukin-1/genetics
- Receptors, Interleukin-1/immunology
- Toll-Like Receptor 4/metabolism
- Toll-Like Receptor 4/chemistry
- Toll-Like Receptor 4/immunology
- Myeloid Differentiation Factor 88/metabolism
- Myeloid Differentiation Factor 88/chemistry
- Myeloid Differentiation Factor 88/genetics
- Toll-Like Receptors/metabolism
- Toll-Like Receptors/immunology
- HEK293 Cells
- Crystallography, X-Ray
- Models, Molecular
- Adaptor Proteins, Signal Transducing
Collapse
Affiliation(s)
- Mohammad K. Manik
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Mengqi Pan
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD4072, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Le Xiao
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Weixi Gu
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD4072, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Hyoyoung Kim
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD4072, Australia
| | - Sabrina Pospich
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund44227, Germany
| | - Andrew Hedger
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD4072, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Parimala R. Vajjhala
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD4072, Australia
| | - Morris Y. L. Lee
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD4072, Australia
| | - Xiaoqi Qian
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD4072, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Michael J. Landsberg
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD4072, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Thomas Ve
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, QLD4215, Australia
| | - Jeffrey D. Nanson
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD4072, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
- Gulbali Institute, Charles Sturt University, Wagga Wagga, NSW2678, Australia
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund44227, Germany
| | - Katryn J. Stacey
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD4072, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD4072, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| |
Collapse
|
10
|
Holze J, Lauber F, Soler S, Kostenis E, Weindl G. Label-free biosensor assay decodes the dynamics of Toll-like receptor signaling. Nat Commun 2024; 15:9554. [PMID: 39532846 PMCID: PMC11558003 DOI: 10.1038/s41467-024-53770-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The discovery of Toll-like receptors (TLRs) represented a significant breakthrough that paved the way for the study of host-pathogen interactions in innate immunity. However, there are still major gaps in understanding TLR function, especially regarding the early dynamics of downstream TLR pathways. Here, we present a label-free optical biosensor-based assay as a method for detecting TLR activation in a native and label-free environment and defining the dynamics of TLR pathway activation. This technology is sufficiently sensitive to detect TLR signaling and readily discriminates between different TLR signaling pathways. We define pharmacological modulators of cell surface and endosomal TLRs and downstream signaling molecules and uncover TLR signaling signatures, including potential biased receptor signaling. These findings highlight that optical biosensor assays complement traditional assays that use a single endpoint and have the potential to facilitate the future design of selective drugs targeting TLRs and their downstream effector cascades.
Collapse
Affiliation(s)
- Janine Holze
- Pharmaceutical Institute, Section Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Felicitas Lauber
- Pharmaceutical Institute, Section Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Sofía Soler
- Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Evi Kostenis
- Institute for Pharmaceutical Biology, Molecular, Cellular and Pharmacobiology Section, University of Bonn, Bonn, Germany
| | - Günther Weindl
- Pharmaceutical Institute, Section Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
| |
Collapse
|
11
|
Wu J, Ai T, He P, Shi Q, Li Y, Zhang Z, Chen M, Huang Z, Wu S, Chen W, Han J. Cecal necroptosis triggers lethal cardiac dysfunction in TNF-induced severe SIRS. Cell Rep 2024; 43:114778. [PMID: 39325617 DOI: 10.1016/j.celrep.2024.114778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Tumor necrosis factor (TNF) induces systemic inflammatory response syndrome (SIRS), and severe SIRS can serve as a model for studying animal death caused by organ failure. Through strategic cecectomy, we demonstrate that necroptosis in the cecum initiates the death process in TNF-treated mice, but it is not the direct cause of death. Instead, we show that it is the cardiac dysfunction downstream of cecum damage that ultimately leads to the death of TNF-treated mice. By in vivo and ex vivo physiological analyses, we reveal that TNF and the damage-associated molecular patterns (DAMPs) released from necroptotic cecal cells jointly target cardiac endothelial cells, triggering caspase-8 activation and subsequent cardiac endothelial damage. Cardiac endothelial damage is a primary cause of the deterioration of diastolic function in the heart of TNF-treated mice. Our research provides insights into the pathophysiological process of TNF-induced lethality.
Collapse
Affiliation(s)
- Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Tingting Ai
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Peng He
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Qilin Shi
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yangxin Li
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Ziguan Zhang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Minwei Chen
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Zhengrong Huang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Suqin Wu
- Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Wanze Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518000, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
12
|
Zhang C, Singla RK, Tang M, Shen B. Natural products act as game-changer potentially in treatment and management of sepsis-mediated inflammation: A clinical perspective. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155710. [PMID: 38759311 DOI: 10.1016/j.phymed.2024.155710] [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: 11/21/2023] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Sepsis, a life-threatening condition resulting from uncontrolled host responses to infection, poses a global health challenge with limited therapeutic options. Due to high heterogeneity, sepsis lacks specific therapeutic drugs. Additionally, there remains a significant gap in the clinical management of sepsis regarding personalized and precise medicine. PURPOSE This review critically examines the scientific landscape surrounding natural products in sepsis and sepsis-mediated inflammation, highlighting their clinical potential. METHODS Following the PRISMA guidelines, we retrieved articles from PubMed to explore potential natural products with therapeutic effects in sepsis-mediated inflammation. RESULTS 434 relevant in vitro and in vivo studies were identified and screened. Ultimately, 55 studies were obtained as the supporting resources for the present review. We divided the 55 natural products into three categories: those influencing the synthesis of inflammatory factors, those affecting surface receptors and modulatory factors, and those influencing signaling pathways and the inflammatory cascade. CONCLUSION Natural products' potential as game-changers in sepsis-mediated inflammation management lies in their ability to modulate hallmarks in sepsis, including inflammation, immunity, and coagulopathy, which provides new therapeutic avenues that are readily accessible and capable of undergoing rapid clinical validation and deployment, offering a gift from nature to humanity. Innovative techniques like bioinformatics, metabolomics, and systems biology offer promising solutions to overcome these obstacles and facilitate the development of natural product-based therapeutics, holding promise for personalized and precise sepsis management and improving patient outcomes. However, standardization, bioavailability, and safety challenges arise during experimental validation and clinical trials of natural products.
Collapse
Affiliation(s)
- Chi Zhang
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China
| | - Rajeev K Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India
| | - Min Tang
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China; West China School of Nursing, Sichuan University, Chengdu, PR China
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China.
| |
Collapse
|
13
|
An W, Lakhina S, Leong J, Rawat K, Husain M. Host Innate Antiviral Response to Influenza A Virus Infection: From Viral Sensing to Antagonism and Escape. Pathogens 2024; 13:561. [PMID: 39057788 PMCID: PMC11280125 DOI: 10.3390/pathogens13070561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Influenza virus possesses an RNA genome of single-stranded, negative-sensed, and segmented configuration. Influenza virus causes an acute respiratory disease, commonly known as the "flu" in humans. In some individuals, flu can lead to pneumonia and acute respiratory distress syndrome. Influenza A virus (IAV) is the most significant because it causes recurring seasonal epidemics, occasional pandemics, and zoonotic outbreaks in human populations, globally. The host innate immune response to IAV infection plays a critical role in sensing, preventing, and clearing the infection as well as in flu disease pathology. Host cells sense IAV infection through multiple receptors and mechanisms, which culminate in the induction of a concerted innate antiviral response and the creation of an antiviral state, which inhibits and clears the infection from host cells. However, IAV antagonizes and escapes many steps of the innate antiviral response by different mechanisms. Herein, we review those host and viral mechanisms. This review covers most aspects of the host innate immune response, i.e., (1) the sensing of incoming virus particles, (2) the activation of downstream innate antiviral signaling pathways, (3) the expression of interferon-stimulated genes, (4) and viral antagonism and escape.
Collapse
Affiliation(s)
| | | | | | | | - Matloob Husain
- Department of Microbiology and Immunology, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand; (W.A.); (S.L.); (J.L.); (K.R.)
| |
Collapse
|
14
|
Ota N, Endo S, Honma K, Iwayama K, Yamashita H, Tatsunami R, Sato K. Chloroquine regulates the lipopolysaccharide-induced inflammatory response in RAW264.7 cells. Allergol Immunopathol (Madr) 2024; 52:97-103. [PMID: 38970272 DOI: 10.15586/aei.v52i4.1083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/29/2024] [Indexed: 07/08/2024]
Abstract
INTRODUCTION AND OBJECTIVES Macrophage-induced inflammation plays a key role in defense against injury and harmful pathogens. Autophagy and the inflammatory response are associated; however, the relationship between the autophagy pathway and lipopolysaccharide (LPS)- induced inflammatory responses remains unknown. We aimed to determine the effect of autophagy on the LPS-induced myeloid differentiation factor 88 (MyD88)/nuclear transcription factor kB (NF-kB) pathway-mediated inflammatory response in RAW264.7 cells. MATERIALS AND METHODS To determine the effect of autophagy on the LPS-induced inflammatory response, using various in vitro assays, we determined the effect of autophagy inhibitors and inducers on the inflammatory response in RAW264.7 cells. RESULTS Chloroquine (CQ), an autophagy inhibitor, suppressed pro-inflammatory cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor α (TNFα) in LPS-stimulated RAW264.7 cells. CQ also affected inflammatory mediators such as myeloid differentiation factor 88 and NF-kB in LPS-stimulated RAW264.7 cells. CONCLUSION This study demonstrated that CQ regulates the LPS-induced inflammatory response in RAW264.7 cells. We propose that targeting the regulation of pro-inflammatory cytokine levels and inflammatory mediators using CQ is a promising therapeutic approach for preventing inflammatory injury. CQ serves as a potential therapeutic target for treating various inflammatory diseases.
Collapse
Affiliation(s)
- Natsuki Ota
- Department of Pharmacy, Hokkaido University of Science, 0068585 Sapporo, Japan
| | - Shoya Endo
- Department of Pharmacy, Hokkaido University of Science, 0068585 Sapporo, Japan
| | - Kouki Honma
- Department of Pharmacy, Hokkaido University of Science, 0068585 Sapporo, Japan
| | - Kuninori Iwayama
- Department of Pharmacy, Hokkaido University of Science, 0068585 Sapporo, Japan
| | - Hiroshi Yamashita
- Department of Pharmacy, Hokkaido University of Science, 0068585 Sapporo, Japan
| | - Ryosuke Tatsunami
- Department of Pharmacy, Hokkaido University of Science, 0068585 Sapporo, Japan
| | - Keisuke Sato
- Department of Pharmacy, Hokkaido University of Science, 0068585 Sapporo, Japan;
| |
Collapse
|
15
|
Monteiro CEDS, de Cerqueira Fiorio B, Silva FGO, de Fathima Felipe de Souza M, Franco ÁX, Lima MADS, Sales TMAL, Mendes TS, Havt A, Barbosa ALR, Resende ÂC, de Moura RS, de Souza MHLP, Soares PMG. A polyphenol-rich açaí seed extract protects against 5-fluorouracil-induced intestinal mucositis in mice through the TLR-4/MyD88/PI3K/mTOR/NF-κBp65 signaling pathway. Nutr Res 2024; 125:1-15. [PMID: 38428258 DOI: 10.1016/j.nutres.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 03/03/2024]
Abstract
Açaí seed extract (ASE) is obtained from Euterpe oleracea Mart. (açaí) plant (Amazon region) has high nutritional and functional value. ASE is rich in polyphenolic compounds, mainly proanthocyanidins. Proanthocyanidins can modulate the immune system and oxidative stress by inhibiting the toll-like receptor-4 (TLR-4)/myeloid differentiation primary response 88 (MyD88)/nuclear factor-κB (NF-κB) pathway. A great deal of evidence suggests that inflammatory cytokines and oxidative stress contribute to the pathogenesis of intestinal mucositis, and these events can lead to intestinal dysmotility. We hypothesized that ASE acts as an anti-inflammatory and antioxidant compound in intestinal mucositis induced by 5-fluorouracil (5-FU) through modulation of the TLR-4/MyD88/phosphatidylinositol-3-kinase α/mechanistic target of rapamycin/NF-κBp65 pathway. The animals were divided into linear 5-FU (450 mg/kg) and 5-FU + ASE (10, 30, and 100 mg/kg) groups. The weight loss of the animals was evaluated daily. Samples from duodenum, jejunum, and ileum were obtained for histopathological, biochemical, and functional analyses. ASE reduced weight loss, inflammatory parameters (interleukin-1β; tumor necrosis factor-α; myeloperoxidase activity) and the gene expression of mediators involved in the TLR-2/MyD88/NF-κB pathway. ASE prevented histopathological changes with beneficial effects on gastrointestinal transit delay, gastric emptying, and intestinal absorption/permeability. In conclusion, ASE protects the integrity of the intestinal epithelial barrier by inhibiting the TLR/MyD88/PI3K/mechanistic target of rapamycin/NF-κBp65 pathway.
Collapse
Affiliation(s)
- Carlos Eduardo da Silva Monteiro
- LEFFAG- Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Bárbara de Cerqueira Fiorio
- LEFFAG- Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Francisca Géssica Oliveira Silva
- LEFFAG- Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Maria de Fathima Felipe de Souza
- LEFFAG- Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Álvaro Xavier Franco
- LEFFAG- Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Marcos Aurélio de Sousa Lima
- LEFFAG- Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Thiago Meneses Araujo Leite Sales
- LEFFAG- Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Tiago Santos Mendes
- LEFFAG- Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Alexandre Havt
- Laboratory of Molecular Toxinology, LTM, Federal University of Ceará, Fortaleza, CE, Brazil
| | - André Luiz Reis Barbosa
- LAFFEX- Laboratory of Experimental Physiopharmacology, Parnaiba Delta Federal University (UFDPAR), Parnaíba, PI, Brazil
| | - Ângela Castro Resende
- Department of Pharmacology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Roberto Soares de Moura
- Department of Pharmacology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Pedro Marcos Gomes Soares
- LEFFAG- Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
16
|
Bali P, Lozano-Pope I, Hernandez J, Estrada MV, Corr M, Turner MA, Bouvet M, Benner C, Obonyo M. TRIF-IFN-I pathway in Helicobacter-induced gastric cancer in an accelerated murine disease model and patient biopsies. iScience 2024; 27:109457. [PMID: 38558931 PMCID: PMC10981133 DOI: 10.1016/j.isci.2024.109457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/20/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Helicobacter pylori (H. pylori) infection is a known cause of many digestive diseases, including gastritis, peptic ulcers, and gastric cancer. However, the underlying mechanisms by which H. pylori infection triggers these disorders are still not clearly understood. Gastric cancer is a slow progressing disease, which makes it difficult to study. We have developed an accelerated disease progression mouse model, which leverages mice deficient in the myeloid differentiation primary response 88 gene (Myd88-/-) infected with Helicobacter felis (H. felis). Using this model and gastric biopsy samples from patients, we report that activation of the Toll/interleukin-1 receptor (TIR)-domain-containing adaptor inducing interferon-β (TRIF)-type I interferon (IFN-I) signaling pathway promotes Helicobacter-induced disease progression toward severe gastric pathology and gastric cancer development. Further, results implicated downstream targets of this pathway in disease pathogenesis. These findings may facilitate stratification of Helicobacter-infected patients and thus enable treatment prioritization of patients.
Collapse
Affiliation(s)
- Prerna Bali
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ivonne Lozano-Pope
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jonathan Hernandez
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Monica V. Estrada
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Maripat Corr
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Michael A. Turner
- Department of Surgery, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Michael Bouvet
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
- Department of Surgery, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Christopher Benner
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Marygorret Obonyo
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
17
|
Korangath P, Jin L, Yang CT, Healy S, Guo X, Ke S, Grüttner C, Hu C, Gabrielson K, Foote J, Clarke R, Ivkov R. Iron Oxide Nanoparticles Inhibit Tumor Progression and Suppress Lung Metastases in Mouse Models of Breast Cancer. ACS NANO 2024; 18:10509-10526. [PMID: 38564478 PMCID: PMC11025112 DOI: 10.1021/acsnano.3c12064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/07/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024]
Abstract
Systemic exposure to starch-coated iron oxide nanoparticles (IONPs) can stimulate antitumor T cell responses, even when little IONP is retained within the tumor. Here, we demonstrate in mouse models of metastatic breast cancer that IONPs can alter the host immune landscape, leading to systemic immune-mediated disease suppression. We report that a single intravenous injection of IONPs can inhibit primary tumor growth, suppress metastases, and extend survival. Gene expression analysis revealed the activation of Toll-like receptor (TLR) pathways involving signaling via Toll/Interleukin-1 receptor domain-containing adaptor-inducing IFN-β (TRIF), a TLR pathway adaptor protein. Requisite participation of TRIF in suppressing tumor progression was demonstrated with histopathologic evidence of upregulated IFN-regulatory factor 3 (IRF3), a downstream protein, and confirmed in a TRIF knockout syngeneic mouse model of metastatic breast cancer. Neither starch-coated polystyrene nanoparticles lacking iron, nor iron-containing dextran-coated parenteral iron replacement agent, induced significant antitumor effects, suggesting a dependence on the type of IONP formulation. Analysis of multiple independent clinical databases supports a hypothesis that upregulation of TLR3 and IRF3 correlates with increased overall survival among breast cancer patients. Taken together, these data support a compelling rationale to re-examine IONP formulations as harboring anticancer immune (nano)adjuvant properties to generate a therapeutic benefit without requiring uptake by cancer cells.
Collapse
Affiliation(s)
- Preethi Korangath
- Department
of Radiation Oncology and Molecular Radiation Sciences, School of
Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
| | - Lu Jin
- The
Hormel Institute, University of Minnesota, Austin, Minnesota 55912, United States
| | - Chun-Ting Yang
- Department
of Radiation Oncology and Molecular Radiation Sciences, School of
Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
| | - Sean Healy
- Department
of Radiation Oncology and Molecular Radiation Sciences, School of
Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
| | - Xin Guo
- Department
of Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Suqi Ke
- Department
of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer
Centre, School of Medicine, Johns Hopkins
University, Baltimore, Maryland 21231, United States
| | | | - Chen Hu
- Department
of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer
Centre, School of Medicine, Johns Hopkins
University, Baltimore, Maryland 21231, United States
| | - Kathleen Gabrielson
- Department
of Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Jeremy Foote
- Department
of Microbiology, School of Medicine, University
of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Robert Clarke
- The
Hormel Institute, University of Minnesota, Austin, Minnesota 55912, United States
| | - Robert Ivkov
- Department
of Radiation Oncology and Molecular Radiation Sciences, School of
Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
- Department
of Oncology, Sidney Kimmel Comprehensive Cancer Centre, School of
Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
- Department
of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department
of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
18
|
Liu J, Kang R, Tang D. Lipopolysaccharide delivery systems in innate immunity. Trends Immunol 2024; 45:274-287. [PMID: 38494365 DOI: 10.1016/j.it.2024.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 03/19/2024]
Abstract
Lipopolysaccharide (LPS), a key component of the outer membrane in Gram-negative bacteria (GNB), is widely recognized for its crucial role in mammalian innate immunity and its link to mortality in intensive care units. While its recognition via the Toll-like receptor (TLR)-4 receptor on cell membranes is well established, the activation of the cytosolic receptor caspase-11 by LPS is now known to lead to inflammasome activation and subsequent induction of pyroptosis. Nevertheless, a fundamental question persists regarding the mechanism by which LPS enters host cells. Recent investigations have identified at least four primary pathways that can facilitate this process: bacterial outer membrane vesicles (OMVs); the spike (S) protein of SARS-CoV-2; host-secreted proteins; and host extracellular vesicles (EVs). These delivery systems provide new avenues for therapeutic interventions against sepsis and infectious diseases.
Collapse
Affiliation(s)
- Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
19
|
Feng AC, Thomas BJ, Purbey PK, de Melo FM, Liu X, Daly AE, Sun F, Lo JHH, Cheng L, Carey MF, Scumpia PO, Smale ST. The transcription factor NF-κB orchestrates nucleosome remodeling during the primary response to Toll-like receptor 4 signaling. Immunity 2024; 57:462-477.e9. [PMID: 38430908 PMCID: PMC10984581 DOI: 10.1016/j.immuni.2024.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 11/26/2023] [Accepted: 02/07/2024] [Indexed: 03/05/2024]
Abstract
Inducible nucleosome remodeling at hundreds of latent enhancers and several promoters shapes the transcriptional response to Toll-like receptor 4 (TLR4) signaling in macrophages. We aimed to define the identities of the transcription factors that promote TLR-induced remodeling. An analysis strategy based on ATAC-seq and single-cell ATAC-seq that enriched for genomic regions most likely to undergo remodeling revealed that the transcription factor nuclear factor κB (NF-κB) bound to all high-confidence peaks marking remodeling during the primary response to the TLR4 ligand, lipid A. Deletion of NF-κB subunits RelA and c-Rel resulted in the loss of remodeling at high-confidence ATAC-seq peaks, and CRISPR-Cas9 mutagenesis of NF-κB-binding motifs impaired remodeling. Remodeling selectivity at defined regions was conferred by collaboration with other inducible factors, including IRF3- and MAP-kinase-induced factors. Thus, NF-κB is unique among TLR4-activated transcription factors in its broad contribution to inducible nucleosome remodeling, alongside its ability to activate poised enhancers and promoters assembled into open chromatin.
Collapse
Affiliation(s)
- An-Chieh Feng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Brandon J Thomas
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA 98195, USA
| | - Prabhat K Purbey
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Filipe Menegatti de Melo
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xin Liu
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Allison E Daly
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Fei Sun
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jerry Hung-Hao Lo
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lijing Cheng
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael F Carey
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Philip O Scumpia
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephen T Smale
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
20
|
Schaaf KR, Landstreet SR, Pugazenthi S, Qian EY, Putz ND, Siderova T, Owen AM, Bohannon JK, Ware LB, Bastarache JA, Shaver CM. Cell-free hemoglobin triggers macrophage cytokine production via TLR4 and MyD88. Am J Physiol Lung Cell Mol Physiol 2024; 326:L29-L38. [PMID: 37991487 PMCID: PMC11279742 DOI: 10.1152/ajplung.00123.2023] [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: 04/14/2023] [Revised: 11/10/2023] [Accepted: 11/18/2023] [Indexed: 11/23/2023] Open
Abstract
Cell-free hemoglobin (CFH) is elevated in the airspace of patients with acute respiratory distress syndrome (ARDS) and is sufficient to cause acute lung injury in a murine model. However, the pathways through which CFH causes lung injury are not well understood. Toll-like receptor 4 (TLR4) is a mediator of inflammation after detection of damage- and pathogen-associated molecular patterns. We hypothesized that TLR4 signaling mediates the proinflammatory effects of CFH in the airspace. After intratracheal CFH, BALBc mice deficient in TLR4 had reduced inflammatory cell influx into the airspace [bronchoalveolar lavage (BAL) cell counts, median TLR4 knockout (KO): 0.8 × 104/mL [IQR 0.4-1.2 × 104/mL], wild-type (WT): 3.0 × 104/mL [2.2-4.0 × 104/mL], P < 0.001] and attenuated lung permeability (BAL protein, TLR4KO: 289 µg/mL [236-320], WT: 488 µg/mL [422-536], P < 0.001). These mice also had attenuated production of interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α in the airspace. C57Bl/6 mice lacking TLR4 on myeloid cells only (LysM.Cre+/-TLR4fl/fl) had reduced cytokine production in the airspace after CFH, without attenuation of lung permeability. In vitro studies confirm that WT primary murine alveolar macrophages exposed to CFH (0.01-1 mg/mL) had dose-dependent increases in IL-6, IL-1 β, CXC motif chemokine ligand 1 (CXCL-1), TNF-α, and IL-10 (P < 0.001). Murine MH-S alveolar-like macrophages show TLR4-dependent expression of IL-1β, IL-6, and CXCL-1 in response to CFH. Primary alveolar macrophages from mice lacking TLR4 adaptor proteins myeloid differentiation primary response 88 (MyD88) or TIR-domain-containing adapter-inducing interferon-β (TRIF) revealed that MyD88KO macrophages had 71-96% reduction in CFH-dependent proinflammatory cytokine production (P < 0.001), whereas macrophages from TRIFKO mice had variable changes in cytokine responses. These data demonstrate that myeloid TLR4 signaling through MyD88 is a key regulator of airspace inflammation in response to CFH.NEW & NOTEWORTHY Cell-free hemoglobin (CFH) is elevated in the airspace of most patients with acute respiratory distress syndrome and causes severe inflammation. Here, we identify that CFH contributes to macrophage-induced cytokine production via Toll-like receptor 4 (TLR4) and myeloid differentiation primary response 88 (MyD88) signaling. These data increase our knowledge of the mechanisms through which CFH contributes to lung injury and may inform development of targeted therapeutics to attenuate inflammation.
Collapse
Affiliation(s)
- Kaitlyn R Schaaf
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Stuart R Landstreet
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Sangami Pugazenthi
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Emily Y Qian
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Nathan D Putz
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Tatiana Siderova
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Allison M Owen
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Julia K Bohannon
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Julie A Bastarache
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Cell and Molecular Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Ciara M Shaver
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| |
Collapse
|
21
|
Jacob TV, Doshi GM. New Promising Routes in Peptic Ulcers: Toll-like Receptors and Semaphorins. Endocr Metab Immune Disord Drug Targets 2024; 24:865-878. [PMID: 37605412 DOI: 10.2174/1871530323666230821102718] [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: 12/03/2022] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 08/23/2023]
Abstract
Peptic ulcers (PU) are one of the commonest yet problematic diseases found to be existing in the majority of the population. Today, drugs from a wide range of therapeutic classes are available for the management of the disease. Still, the complications of the condition are difficult to tackle and the side effect profile is quite a concern. The literature indicates that Toll-like receptors (TLRs) and Semaphorins (SEMAs) have been under study for their various pharmacological actions over the past few decades. Both these signalling pathways are found to regulate immunological and inflammatory responses. Moreover, receptors and signalling molecules from the family of TLRs and SEMAs are found to have bacterial recognition and antibacterial properties which are essential in eradicating Helicobacter pylori (H. pylori), one of the major causative agents of PU. Our understanding of SEMAs, a class of proteins involved in cell signalling, is relatively less developed compared to TLRs, another class of proteins involved in the immune response. SEMAs and TLRs play different roles in biological processes, with SEMAs primarily involved in guiding cell migration and axon guidance during development, while TLRs are responsible for recognizing pathogens and initiating an immune response. Here, in this review, we will discuss in detail the signalling cascade of TLRs and SEMAs and thereby understand its association with PU for future therapeutic targeting. The review also aims at providing an overview of the study that has been into exploring the role of these signalling pathways in the management of PU.
Collapse
Affiliation(s)
- Teresa V Jacob
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| | - Gaurav M Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| |
Collapse
|
22
|
Mahapatra S, Ganguly B, Pani S, Saha A, Samanta M. A comprehensive review on the dynamic role of toll-like receptors (TLRs) in frontier aquaculture research and as a promising avenue for fish disease management. Int J Biol Macromol 2023; 253:126541. [PMID: 37648127 DOI: 10.1016/j.ijbiomac.2023.126541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023]
Abstract
Toll-like receptors (TLRs) represent a conserved group of germline-encoded pattern recognition receptors (PRRs) that recognize pathogen-associated molecular patterns (PAMPs) and play a crucial role in inducing the broadly acting innate immune response against pathogens. In recent years, the detection of 21 different TLR types in various fish species has sparked interest in exploring the potential of TLRs as targets for boosting immunity and disease resistance in fish. This comprehensive review offers the latest insights into the diverse facets of fish TLRs, highlighting their history, classification, architectural insights through 3D modelling, ligands recognition, signalling pathways, crosstalk, and expression patterns at various developmental stages. It provides an exhaustive account of the distinct TLRs induced during the invasion of specific pathogens in various fish species and delves into the disparities between fish TLRs and their mammalian counterparts, highlighting the specific contribution of TLRs to the immune response in fish. Although various facets of TLRs in some fish, shellfish, and molluscs have been described, the role of TLRs in several other aquatic organisms still remained as potential gaps. Overall, this article outlines frontier aquaculture research in advancing the knowledge of fish immune systems for the proper management of piscine maladies.
Collapse
Affiliation(s)
- Smruti Mahapatra
- Immunology Laboratory, Fish Health Management Division, ICAR-Central Institute of Freshwater Aquaculture (ICAR-CIFA), Kausalyaganga, Bhubaneswar 751002, Odisha, India
| | - Bristy Ganguly
- Immunology Laboratory, Fish Health Management Division, ICAR-Central Institute of Freshwater Aquaculture (ICAR-CIFA), Kausalyaganga, Bhubaneswar 751002, Odisha, India
| | - Saswati Pani
- Immunology Laboratory, Fish Health Management Division, ICAR-Central Institute of Freshwater Aquaculture (ICAR-CIFA), Kausalyaganga, Bhubaneswar 751002, Odisha, India
| | - Ashis Saha
- Reproductive Biology and Endocrinology Laboratory, Fish Nutrition and Physiology Division, ICAR-Central Institute of Freshwater Aquaculture (ICAR-CIFA), Kausalyaganga, Bhubaneswar 751002, Odisha, India
| | - Mrinal Samanta
- Immunology Laboratory, Fish Health Management Division, ICAR-Central Institute of Freshwater Aquaculture (ICAR-CIFA), Kausalyaganga, Bhubaneswar 751002, Odisha, India.
| |
Collapse
|
23
|
Kotov DI, Lee OV, Fattinger SA, Langner CA, Guillen JV, Peters JM, Moon A, Burd EM, Witt KC, Stetson DB, Jaye DL, Bryson BD, Vance RE. Early cellular mechanisms of type I interferon-driven susceptibility to tuberculosis. Cell 2023; 186:5536-5553.e22. [PMID: 38029747 PMCID: PMC10757650 DOI: 10.1016/j.cell.2023.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 06/16/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023]
Abstract
Mycobacterium tuberculosis (Mtb) causes 1.6 million deaths annually. Active tuberculosis correlates with a neutrophil-driven type I interferon (IFN) signature, but the cellular mechanisms underlying tuberculosis pathogenesis remain poorly understood. We found that interstitial macrophages (IMs) and plasmacytoid dendritic cells (pDCs) are dominant producers of type I IFN during Mtb infection in mice and non-human primates, and pDCs localize near human Mtb granulomas. Depletion of pDCs reduces Mtb burdens, implicating pDCs in tuberculosis pathogenesis. During IFN-driven disease, we observe abundant DNA-containing neutrophil extracellular traps (NETs) described to activate pDCs. Cell-type-specific disruption of the type I IFN receptor suggests that IFNs act on IMs to inhibit Mtb control. Single-cell RNA sequencing (scRNA-seq) indicates that type I IFN-responsive cells are defective in their response to IFNγ, a cytokine critical for Mtb control. We propose that pDC-derived type I IFNs act on IMs to permit bacterial replication, driving further neutrophil recruitment and active tuberculosis disease.
Collapse
Affiliation(s)
- Dmitri I Kotov
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Ophelia V Lee
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Stefan A Fattinger
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Charlotte A Langner
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jaresley V Guillen
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Joshua M Peters
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Andres Moon
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | - Eileen M Burd
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | - Kristen C Witt
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Daniel B Stetson
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - David L Jaye
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Russell E Vance
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
24
|
Almazmomi MA, Esmat A, Naeem A. Acute Kidney Injury: Definition, Management, and Promising Therapeutic Target. Cureus 2023; 15:e51228. [PMID: 38283512 PMCID: PMC10821757 DOI: 10.7759/cureus.51228] [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] [Accepted: 12/28/2023] [Indexed: 01/30/2024] Open
Abstract
Acute kidney injury (AKI) is caused by a sudden loss of renal function, resulting in the build-up of waste products and a significant increase in mortality and morbidity. It is commonly diagnosed in critically ill patients, with its occurrence estimated at up to 50% in patients hospitalized in the intensive critical unit. Despite ongoing efforts, the death rate associated with AKI has remained high over the past half-century. Thus, it is critical to investigate novel therapy options for preventing the epidemic. Many studies have found that inflammation and Toll-like receptor-4 (TLR-4) activation have a significant role in the pathogenesis of AKI. Noteworthy, challenges in the search for efficient pharmacological therapy for AKI have arisen due to the multifaceted origin and complexity of the clinical history of people with the disease. This article focuses on kidney injury's epidemiology, risk factors, and pathophysiological processes. Specifically, it focuses on the role of TLRs especially type 4 in disease development.
Collapse
Affiliation(s)
- Meaad A Almazmomi
- Pharmaceutical Care Department, Ministry of National Guard - Health Affairs, Jeddah, SAU
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Ahmed Esmat
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Anjum Naeem
- Pharmaceutical Care Department, Ministry of National Guard - Health Affairs, Jeddah, SAU
| |
Collapse
|
25
|
Zhou H, Shi X, Yu Y, Yang L, OuYang J, Bian Y, Liu Y, Li G. Puerarin Alleviates Oxidized Oil-Induced Oxidative Injury and Inflammation via Inhibition of the Nrf2/Keap1 and HMGB1/TLR4/MAPK Signaling Pathways: An Investigation in a Chicken Model. Mol Nutr Food Res 2023; 67:e2200663. [PMID: 37776050 DOI: 10.1002/mnfr.202200663] [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: 09/29/2022] [Revised: 05/29/2023] [Indexed: 10/01/2023]
Abstract
SCOPE Puerarin has possessed a wide range of pharmacological activities. However, little is known about the protective effects of puerarin on the oxidized oil-induced injury. Here, the antioxidant and anti-inflammatory effects of puerarin are described using a chicken model. METHODS AND RESULTS A total of 360 broilers are arranged in four treatments. Diets include two types of soybean oil (fresh or oxidized) and two levels of puerarin (0 or 750 mg kg-1 ). Results show that puerarin alleviates oxidized soybean oil-induced hepatic and thymic oxidative injury. This effect is observed by increasing the SOD activity and the expressions of Nrf2 signaling pathway-related genes and reducing the MDA content in the liver and thymus. Moreover, puerarin supplementation decreases the concentrations and mRNA levels of pro-inflammatory factors in the liver and thymus. The potential mechanism responsible for this is the decrease in the mRNA or protein levels of HMGB1, TLR4, MyD88, and p65 in the liver or thymus. Western blotting results indicate that puerarin also decreases the phosphorylation of JNK1/2, ERK1/2, and p38 in the liver and thymus. CONCLUSION This study demonstrates puerarin may be a potential nutrient supplement in the treatment of oxidized oil-induced damage, and the Nrf2/Keap1 and HMGB1/TLR4/MAPK signaling pathways might be its important target.
Collapse
Affiliation(s)
- Hua Zhou
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, P. R. China
- Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Nanchang, 330045, P. R. China
| | - Xuan Shi
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, P. R. China
- Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Nanchang, 330045, P. R. China
| | - Yingmei Yu
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, P. R. China
- Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Nanchang, 330045, P. R. China
| | - Lei Yang
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, P. R. China
- Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Nanchang, 330045, P. R. China
| | - Jingxin OuYang
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, P. R. China
- Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Nanchang, 330045, P. R. China
| | - Yinhao Bian
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, P. R. China
- Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Nanchang, 330045, P. R. China
| | - Yichun Liu
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, P. R. China
- Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Nanchang, 330045, P. R. China
| | - Guanhong Li
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, P. R. China
- Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Nanchang, 330045, P. R. China
| |
Collapse
|
26
|
Chakraborty S, Ye J, Wang H, Sun M, Zhang Y, Sang X, Zhuang Z. Application of toll-like receptors (TLRs) and their agonists in cancer vaccines and immunotherapy. Front Immunol 2023; 14:1227833. [PMID: 37936697 PMCID: PMC10626551 DOI: 10.3389/fimmu.2023.1227833] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/10/2023] [Indexed: 11/09/2023] Open
Abstract
Toll-like receptors (TLRs) are pattern recognition receptors (PRRs) expressed in various immune cell types and perform multiple purposes and duties involved in the induction of innate and adaptive immunity. Their capability to propagate immunity makes them attractive targets for the expansion of numerous immunotherapeutic approaches targeting cancer. These immunotherapeutic strategies include using TLR ligands/agonists as monotherapy or combined therapeutic strategies. Several TLR agonists have demonstrated significant efficacy in advanced clinical trials. In recent years, multiple reports established the applicability of TLR agonists as adjuvants to chemotherapeutic drugs, radiation, and immunotherapies, including cancer vaccines. Cancer vaccines are a relatively novel approach in the field of cancer immunotherapy and are currently under extensive evaluation for treating different cancers. In the present review, we tried to deliver an inclusive discussion of the significant TLR agonists and discussed their application and challenges to their incorporation into cancer immunotherapy approaches, particularly highlighting the usage of TLR agonists as functional adjuvants to cancer vaccines. Finally, we present the translational potential of rWTC-MBTA vaccination [irradiated whole tumor cells (rWTC) pulsed with phagocytic agonists Mannan-BAM, TLR ligands, and anti-CD40 agonisticAntibody], an autologous cancer vaccine leveraging membrane-bound Mannan-BAM, and the immune-inducing prowess of TLR agonists as a probable immunotherapy in multiple cancer types.
Collapse
Affiliation(s)
- Samik Chakraborty
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- NE1 Inc., New York, NY, United States
| | - Juan Ye
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Herui Wang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mitchell Sun
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yaping Zhang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xueyu Sang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
27
|
Low R, Ha SD, Sleapnicov N, Maneesh P, Kim SO. Prolonged Inhibition of the MEK1/2-ERK Signaling Axis Primes Interleukin-1 Beta Expression through Histone 3 Lysine 9 Demethylation in Murine Macrophages. Int J Mol Sci 2023; 24:14428. [PMID: 37833877 PMCID: PMC10572145 DOI: 10.3390/ijms241914428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Macrophages undergo different cellular states upon activation that can be hyporesponsive (tolerated) or hyperresponsive (primed or trained) to subsequent stimuli. Epigenetic modifications are known to play key roles in determining these cellular states. However, little is known about the role of signaling pathways that lead to these epigenetic modifications. Here, we examined the effects of various inhibitors targeting key signaling pathways induced by lipopolysaccharide (LPS) on tolerance and priming in murine macrophages. We found that a prolonged inhibition (>18 h) of the mitogen-activated protein kinase (MEK)1/2-extracellular signal-regulated kinase (ERK)1/2 signaling axis reversed tolerance and primed cells in expressing interleukin (IL)-1β and other inflammatory cytokines such as IL-6, tumor necrosis factor (TNF)α, and CXCL10. The ectopic expression of catalytically active and inactive MEK1 mutants suppressed and enhanced IL-1β expression, respectively. A transcriptomic analysis showed that cells primed by the MEK1/2 inhibitor U0126 expressed higher levels of gene sets associated with immune responses and cytokine/chemokine production, but expressed lower levels of genes with cell cycle progression, chromosome organization, and heterochromatin formation than non-primed cells. Of interest, the mRNA expressions of the histone 3 lysine 9 (H3K9) methyltransferase Suv39h1 and the H3K9 methylation reader Cbx5 were substantially suppressed, whereas the H3K9 demethylase Kdm7a was enhanced, suggesting a role of the MEK1/2-ERK signaling axis in H3K9 demethylation. The H3K9 trimethylation levels in the genomic regions of IL-1β, TNFα, and CXCL10 were decreased by U0126. Also, the H3K9 methyltransferase inhibitor BIX01294 mimicked the U0126 training effects and the overexpression of chromobox homolog (CBX)5 prevented the U0126 training effects in both RAW264.7 cells and bone-marrow-derived macrophages. Collectively, these data suggest that the prolonged inhibition of the MEK1/2-ERK signaling axis reverses tolerance and primed macrophages likely through decreasing the H3K9 methylation levels.
Collapse
Affiliation(s)
| | | | | | | | - Sung Ouk Kim
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6G 2V4, Canada; (R.L.); (S.-D.H.); (N.S.); (P.M.)
| |
Collapse
|
28
|
Holloway KN, Douglas JC, Rafferty TM, Kane CJM, Drew PD. Ethanol Induces Neuroinflammation in a Chronic Plus Binge Mouse Model of Alcohol Use Disorder via TLR4 and MyD88-Dependent Signaling. Cells 2023; 12:2109. [PMID: 37626919 PMCID: PMC10453365 DOI: 10.3390/cells12162109] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/14/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Ethanol induces neuroinflammation, which is believed to contribute to the pathogenesis of alcohol use disorder (AUD). Toll-like receptors (TLRs) are a group of pattern recognition receptors (PRRs) expressed on both immune cells, including microglia and astrocytes, and non-immune cells in the central nervous system (CNS). Studies have shown that alcohol activates TLR4 signaling, resulting in the induction of pro-inflammatory cytokines and chemokines in the CNS. However, the effect of alcohol on signaling pathways downstream of TLR4, such as MyD88 and TRIF (TICAM) signaling, has not been evaluated extensively. In the current study, we treated male wild-type, TLR4-, MyD88-, and TRIF-deficient mice using a chronic plus binge mouse model of AUD. Evaluation of mRNA expression by qRT-PCR revealed that ethanol increased IL-1β, TNF-α, CCL2, COX2, FosB, and JunB in the cerebellum in wild-type and TRIF-deficient mice, while ethanol generally did not increase the expression of these molecules in TLR4- and MyD88-deficient mice. Furthermore, IRF3, IRF7, and IFN-β1, which are associated with the TRIF-dependent signaling cascade, were largely unaffected by alcohol. Collectively, these results suggest that the TLR4 and downstream MyD88-dependent signaling pathways are essential in ethanol-induced neuroinflammation in this mouse model of AUD.
Collapse
Affiliation(s)
- Kalee N. Holloway
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - James C. Douglas
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Tonya M. Rafferty
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Cynthia J. M. Kane
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Paul D. Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
29
|
Bali P, Lozano-Pope I, Hernandez J, Estrada MV, Corr M, Turner MA, Bouvet M, Benner C, Obonyo M. Activation of the TRIF pathway and downstream targets results in the development of precancerous lesions during infection with Helicobacter. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.04.543598. [PMID: 37333238 PMCID: PMC10274671 DOI: 10.1101/2023.06.04.543598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Helicobacter pylori ( H. pylori) infection is an established cause of many digestive diseases, including gastritis, peptic ulcers, and gastric cancer. However, the mechanism by which infection with H. pylori causes these disorders is still not clearly understood. This is due to insufficient knowledge of pathways that promote H. pylori -induced disease progression. We have established a Helicobacter -induced accelerated disease progression mouse model, which involves infecting mice deficient in the myeloid differentiation primary response 88 gene ( Myd88 -/- ) with H. felis . Using this model, we report here that that progression of H. felis -induced inflammation to high-grade dysplasia was associated with activation of type I interferon (IFN-I) signaling pathway and upregulation of related downstream target genes, IFN-stimulated genes (ISGs). These observations were further corroborated by the enrichment of ISRE motifs in the promoters of upregulated genes. Further we showed that H. felis -induced inflammation in mice deficient in Toll/interleukin-1 receptor (TIR)-domain-containing adaptor inducing interferon-β (TRIF, Trif Lps 2 ) did not progress to severe gastric pathology, indicating a role of the TRIF signaling pathway in disease pathogenesis and progression. Indeed, survival analysis in gastric biopsy samples from gastric cancer patients illustrated that high expression of Trif was significantly associated with poor survival in gastric cancer.
Collapse
|
30
|
Seya T, Shingai M, Kawakita T, Matsumoto M. Two Modes of Th1 Polarization Induced by Dendritic-Cell-Priming Adjuvant in Vaccination. Cells 2023; 12:1504. [PMID: 37296625 PMCID: PMC10252737 DOI: 10.3390/cells12111504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Viral infections are usually accompanied by systemic cytokinemia. Vaccines need not necessarily mimic infection by inducing cytokinemia, but must induce antiviral-acquired immunity. Virus-derived nucleic acids are potential immune-enhancers and particularly good candidates as adjuvants in vaccines in mouse models. The most important nucleic-acid-sensing process involves the dendritic cell (DC) Toll-like receptor (TLR), which participates in the pattern recognition of foreign DNA/RNA structures. Human CD141+ DCs preferentially express TLR3 in endosomes and recognize double-stranded RNA. Antigen cross-presentation occurs preferentially in this subset of DCs (cDCs) via the TLR3-TICAM-1-IRF3 axis. Another subset, plasmacytoid DCs (pDCs), specifically expresses TLR7/9 in endosomes. They then recruit the MyD88 adaptor, and potently induce type I interferon (IFN-I) and proinflammatory cytokines to eliminate the virus. Notably, this inflammation leads to the secondary activation of antigen-presenting cDCs. Hence, the activation of cDCs via nucleic acids involves two modes: (i) with bystander effect of inflammation and (ii) without inflammation. In either case, the acquired immune response finally occurs with Th1 polarity. The level of inflammation and adverse events depend on the TLR repertoire and the mode of response to their agonists in the relevant DC subsets, and could be predicted by assessing the levels of cytokines/chemokines and T cell proliferation in vaccinated subjects. The main differences in the mode of vaccine sought in infectious diseases and cancer are defined by whether it is prophylactic or therapeutic, whether it can deliver sufficient antigens to cDCs, and how it behaves in the microenvironment of the lesion. Adjuvant can be selected on a case-to-case basis.
Collapse
Affiliation(s)
- Tsukasa Seya
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori 030-0943, Japan;
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
| | - Masashi Shingai
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
- Division of Biologics Development, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan
- International Collaboration Unit, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo 001-0021, Japan
| | - Tomomi Kawakita
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo 001-0021, Japan
| | - Misako Matsumoto
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori 030-0943, Japan;
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
| |
Collapse
|
31
|
Reynoso M, Hobbs S, Kolb AL, Matheny RW, Roberts BM. MyD88 and not TRIF knockout is sufficient to abolish LPS-induced inflammatory responses in bone-derived macrophages. FEBS Lett 2023; 597:1225-1232. [PMID: 36971014 DOI: 10.1002/1873-3468.14616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/29/2023]
Abstract
Macrophages play an important role in the response to infection and/or repair of injury in tissues. To examine the NF-κB pathway in response to an inflammatory stimulus, we used wild-type bone-marrow-derived macrophages (BMDMs) or BMDMs with knockout (KO) of myeloid differentiation primary response 88 (MyD88) and/or Toll/interleukin-1 receptor domain-containing adapter-inducing interferon-β (TRIF) via CRISPR/Cas9. Following treatment of BMDMs with lipopolysaccharide (LPS) to induce an inflammatory response, translational signalling of NF-κB was quantified via immunoblot and cytokines were measured. Our findings reveal that MyD88 KO, but not TRIF KO, decreased LPS-induced NF-κB signalling, and 10% expression of basal MyD88 expression was sufficient to partially rescue the abolished inflammatory cytokine secretion observed upon MyD88 KO.
Collapse
Affiliation(s)
- Marinaliz Reynoso
- U.S. Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Stuart Hobbs
- U.S. Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Alexander L Kolb
- U.S. Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Ronald W Matheny
- Military Operational Medicine Research Program, Fort Detrick, MA, USA
| | - Brandon M Roberts
- U.S. Army Research Institute of Environmental Medicine, Natick, MA, USA
| |
Collapse
|
32
|
Baran M, Feriotti C, McGinley A, Carlile SR, Jiang Z, Calderon-Gonzalez R, Dumigan A, Sá-Pessoa J, Sutton CE, Kearney J, McLoughlin RM, Mills KHG, Fitzgerald KA, Bengeochea JA, Bowie AG. PYHIN protein IFI207 regulates cytokine transcription and IRF7 and contributes to the establishment of K. pneumoniae infection. Cell Rep 2023; 42:112341. [PMID: 37018072 DOI: 10.1016/j.celrep.2023.112341] [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: 03/24/2022] [Revised: 02/02/2023] [Accepted: 03/20/2023] [Indexed: 04/06/2023] Open
Abstract
PYHIN proteins AIM2 and IFI204 sense pathogen DNA, while other PYHINs have been shown to regulate host gene expression through as-yet unclear mechanisms. We characterize mouse PYHIN IFI207, which we find is not involved in DNA sensing but rather is required for cytokine promoter induction in macrophages. IFI207 co-localizes with both active RNA polymerase II (RNA Pol II) and IRF7 in the nucleus and enhances IRF7-dependent gene promoter induction. Generation of Ifi207-/- mice shows no role for IFI207 in autoimmunity. Rather, IFI207 is required for the establishment of a Klebsiella pneumoniae lung infection and for Klebsiella macrophage phagocytosis. These insights into IFI207 function illustrate that PYHINs can have distinct roles in innate immunity independent of DNA sensing and highlight the need to better characterize the whole mouse locus, one gene at a time.
Collapse
Affiliation(s)
- Marcin Baran
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 Dublin, Ireland
| | - Claudia Feriotti
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, 97 Lisburn Road, Belfast, UK
| | - Aoife McGinley
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 Dublin, Ireland
| | - Simon R Carlile
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 Dublin, Ireland
| | - Zhaozhao Jiang
- Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ricardo Calderon-Gonzalez
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, 97 Lisburn Road, Belfast, UK
| | - Amy Dumigan
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, 97 Lisburn Road, Belfast, UK
| | - Joana Sá-Pessoa
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, 97 Lisburn Road, Belfast, UK
| | - Caroline E Sutton
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 Dublin, Ireland
| | - Jay Kearney
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 Dublin, Ireland
| | - Rachel M McLoughlin
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 Dublin, Ireland
| | - Kingston H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 Dublin, Ireland
| | - Katherine A Fitzgerald
- Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jose A Bengeochea
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, 97 Lisburn Road, Belfast, UK
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 Dublin, Ireland.
| |
Collapse
|
33
|
Bhagwani AR, Ali M, Piper B, Liu M, Hudson J, Kelly N, Bogamuwa S, Yang H, Londino JD, Bednash JS, Farkas D, Mallampalli RK, Nicolls MR, Ryan JJ, Thompson AR, Chan SY, Gomez D, Goncharova EA, Farkas L. A p53-TLR3 axis ameliorates pulmonary hypertension by inducing BMPR2 via IRF3. iScience 2023; 26:105935. [PMID: 36685041 PMCID: PMC9852960 DOI: 10.1016/j.isci.2023.105935] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/17/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) features pathogenic and abnormal endothelial cells (ECs), and one potential origin is clonal selection. We studied the role of p53 and toll-like receptor 3 (TLR3) in clonal expansion and pulmonary hypertension (PH) via regulation of bone morphogenetic protein (BMPR2) signaling. ECs of PAH patients had reduced p53 expression. EC-specific p53 knockout exaggerated PH, and clonal expansion reduced p53 and TLR3 expression in rat lung CD117+ ECs. Reduced p53 degradation (Nutlin 3a) abolished clonal EC expansion, induced TLR3 and BMPR2, and ameliorated PH. Polyinosinic/polycytidylic acid [Poly(I:C)] increased BMPR2 signaling in ECs via enhanced binding of interferon regulatory factor-3 (IRF3) to the BMPR2 promoter and reduced PH in p53-/- mice but not in mice with impaired TLR3 downstream signaling. Our data show that a p53/TLR3/IRF3 axis regulates BMPR2 expression and signaling in ECs. This link can be exploited for therapy of PH.
Collapse
Affiliation(s)
- Aneel R. Bhagwani
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Mehboob Ali
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Bryce Piper
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Mingjun Liu
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Jaylen Hudson
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Neil Kelly
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Srimathi Bogamuwa
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Hu Yang
- Chemical & Biochemical Engineering, Missouri S&T, Rolla, MO 65409, USA
| | - James D. Londino
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Joseph S. Bednash
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Daniela Farkas
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Rama K. Mallampalli
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Mark R. Nicolls
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John J. Ryan
- College of Humanities & Sciences, Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - A.A. Roger Thompson
- Department of Infection, Immunity & Cardiovascular Disease, Faculty of Medicine, Dentistry & Health, University of Sheffield, Sheffield S10 2RX, UK
| | - Stephen Y. Chan
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Delphine Gomez
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Elena A. Goncharova
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA 95616, USA
| | - Laszlo Farkas
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
34
|
TLR3 forms a laterally aligned multimeric complex along double-stranded RNA for efficient signal transduction. Nat Commun 2023; 14:164. [PMID: 36631495 PMCID: PMC9834221 DOI: 10.1038/s41467-023-35844-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Toll-like receptor 3 (TLR3) is a member of the TLR family, which plays an important role in the innate immune system and is responsible for recognizing viral double-stranded RNA (dsRNA). Previous biochemical and structural studies have revealed that a minimum length of approximately 40-50 base pairs of dsRNA is necessary for TLR3 binding and dimerization. However, efficient TLR3 activation requires longer dsRNA and the molecular mechanism underlying its dsRNA length-dependent activation remains unknown. Here, we report cryo-electron microscopy analyses of TLR3 complexed with longer dsRNA. TLR3 dimers laterally form a higher multimeric complex along dsRNA, providing the basis for cooperative binding and efficient signal transduction.
Collapse
|
35
|
León B. Understanding the development of Th2 cell-driven allergic airway disease in early life. FRONTIERS IN ALLERGY 2023; 3:1080153. [PMID: 36704753 PMCID: PMC9872036 DOI: 10.3389/falgy.2022.1080153] [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: 10/25/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
Allergic diseases, including atopic dermatitis, allergic rhinitis, asthma, and food allergy, are caused by abnormal responses to relatively harmless foreign proteins called allergens found in pollen, fungal spores, house dust mites (HDM), animal dander, or certain foods. In particular, the activation of allergen-specific helper T cells towards a type 2 (Th2) phenotype during the first encounters with the allergen, also known as the sensitization phase, is the leading cause of the subsequent development of allergic disease. Infants and children are especially prone to developing Th2 cell responses after initial contact with allergens. But in addition, the rates of allergic sensitization and the development of allergic diseases among children are increasing in the industrialized world and have been associated with living in urban settings. Particularly for respiratory allergies, greater susceptibility to developing allergic Th2 cell responses has been shown in children living in urban environments containing low levels of microbial contaminants, principally bacterial endotoxins [lipopolysaccharide (LPS)], in the causative aeroallergens. This review highlights the current understanding of the factors that balance Th2 cell immunity to environmental allergens, with a particular focus on the determinants that program conventional dendritic cells (cDCs) toward or away from a Th2 stimulatory function. In this context, it discusses transcription factor-guided functional specialization of type-2 cDCs (cDC2s) and how the integration of signals derived from the environment drives this process. In addition, it analyzes observational and mechanistic studies supporting an essential role for innate sensing of microbial-derived products contained in aeroallergens in modulating allergic Th2 cell immune responses. Finally, this review examines whether hyporesponsiveness to microbial stimulation, particularly to LPS, is a risk factor for the induction of Th2 cell responses and allergic sensitization during infancy and early childhood and the potential factors that may affect early-age response to LPS and other environmental microbial components.
Collapse
Affiliation(s)
- Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
36
|
Huang S, Cheng A, Wang M, Yin Z, Huang J, Jia R. Viruses utilize ubiquitination systems to escape TLR/RLR-mediated innate immunity. Front Immunol 2022; 13:1065211. [PMID: 36505476 PMCID: PMC9732732 DOI: 10.3389/fimmu.2022.1065211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/10/2022] [Indexed: 11/26/2022] Open
Abstract
When the viruses invade the body, they will be recognized by the host pattern recognition receptors (PRRs) such as Toll like receptor (TLR) or retinoic acid-induced gene-I like receptor (RLR), thus causing the activation of downstream antiviral signals to resist the virus invasion. The cross action between ubiquitination and proteins in these signal cascades enhances the antiviral signal. On the contrary, more and more viruses have also been found to use the ubiquitination system to inhibit TLR/RLR mediated innate immunity. Therefore, this review summarizes how the ubiquitination system plays a regulatory role in TLR/RLR mediated innate immunity, and how viruses use the ubiquitination system to complete immune escape.
Collapse
Affiliation(s)
- Shanzhi Huang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Anchun Cheng
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mingshu Wang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Juan Huang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China,*Correspondence: Renyong Jia,
| |
Collapse
|
37
|
Hou W, Xia X, Li Y, Lv H, Liu J, Li X. Recent progress and perspectives on the relationship between hyperuricemia and periodontitis. Front Immunol 2022; 13:995582. [PMID: 36466813 PMCID: PMC9708725 DOI: 10.3389/fimmu.2022.995582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 11/01/2022] [Indexed: 08/31/2023] Open
Abstract
Periodontitis is one of the most prevalent diseases in oral cavity, which could not merely lead to the destruction of supporting or surrounding tooth structures but also affect the whole-body health such as the digestive and nervous systems. Epidemiological investigations suggested that in some developed countries, more than 45% or even 50% population were suffering from periodontitis. However, the prevalence increases with age remarkably and it is investigated that a high prevalence (>50%) is affecting the elderly who is over 65 years old. There is an increasing interest in the direct and indirect relationships between periodontitis and hyperuricemia. Currently, hyperuricemia has become the second major metabolic disease in modern society and the prevalence of hyperuricemia among adult males and females was 21.7% and 14.4% respectively. As an inflammatory disease associated with various systemic diseases, periodontitis may have certain connections with hyperuricemia. Partial existing research announced that hyperuricemia could act as an inhibitory factor for periodontitis, while other scholars presented that a high uric acid (UA) level was more likely to aggravate inflammatory immune response and lead to more serious tissue destruction. This article provides a detailed and comprehensive overview of the relationship underlying hyperuricemia and periodontitis in the molecular mechanisms. Given the impact of hyperuricemia, this review could provide insight into its comorbidities.
Collapse
Affiliation(s)
- Wenxue Hou
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| | - Xiaomin Xia
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| | - Ying Li
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| | - Hanlin Lv
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| | - Jie Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| | - Xue Li
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| |
Collapse
|
38
|
Heinz R, Schneider UC. TLR4-Pathway-Associated Biomarkers in Subarachnoid Hemorrhage (SAH): Potential Targets for Future Anti-Inflammatory Therapies. Int J Mol Sci 2022; 23:ijms232012618. [PMID: 36293468 PMCID: PMC9603851 DOI: 10.3390/ijms232012618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/20/2022] Open
Abstract
Subarachnoid hemorrhage is associated with severe neurological deficits for survivors. Among survivors of the initial bleeding, secondary brain injury leads to additional brain damage. Apart from cerebral vasospasm, secondary brain injury mainly results from cerebral inflammation taking place in the brain parenchyma after bleeding. The brain’s innate immune system is activated, which leads to disturbances in brain homeostasis, cleavage of inflammatory cytokines and, subsequently, neuronal cell death. The toll-like receptor (TLR)4 signaling pathway has been found to play an essential role in the pathophysiology of acute brain injuries such as subarachnoid hemorrhage (SAH). TLR4 is expressed on the cell surface of microglia, which are key players in the cellular immune responses of the brain. The participants in the signaling pathway, such as TLR4-pathway-like ligands, the receptor itself, and inflammatory cytokines, can act as biomarkers, serving as clues regarding the inflammatory status after SAH. Moreover, protein complexes such as the NLRP3 inflammasome or receptors such as TREM1 frame the TLR4 pathway and are indicative of inflammation. In this review, we focus on the activity of the TLR4 pathway and its contributors, which can act as biomarkers of neuroinflammation or even offer potential new treatment targets for secondary neuronal cell death after SAH.
Collapse
Affiliation(s)
- Rebecca Heinz
- Experimental Neurosurgery, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany
| | - Ulf C. Schneider
- Experimental Neurosurgery, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany
- Department of Neurosurgery, Cantonal Hospital of Lucerne, 6000 Lucerne, Switzerland
- Correspondence:
| |
Collapse
|
39
|
Weiss HJ, O’Neill LAJ. Of Flies and Men—The Discovery of TLRs. Cells 2022; 11:cells11193127. [PMID: 36231089 PMCID: PMC9563146 DOI: 10.3390/cells11193127] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/29/2022] [Accepted: 10/02/2022] [Indexed: 11/30/2022] Open
Abstract
In 2011, the Nobel Prize in Physiology or Medicine was awarded to three immunologists: Bruce A. Beutler, Jules A. Hoffmann, and Ralph M. Steinman. While Steinman was honored for his work on dendritic cells and adaptive immunity, Beutler and Hoffman received the prize for their contributions to discoveries in innate immunity. In 1996, Hoffmann found the toll gene to be crucial for mounting antimicrobial responses in fruit flies, first implicating this developmental gene in immune signaling. Two years later, Beutler built on this observation by describing a Toll-like gene, tlr4, as the receptor for the bacterial product LPS, representing a crucial step in innate immune activation and protection from bacterial infections in mammals. These publications spearheaded research in innate immune sensing and sparked a huge interest regarding innate defense mechanisms in the following years and decades. Today, Beutler and Hoffmann’s research has not only resulted in the discovery of the role of multiple TLRs in innate immunity but also in a much broader understanding of the molecular components of the innate immune system. In this review, we aim to collect the discoveries leading up to the publications of Beutler and Hoffmann, taking a close look at how early advances in both developmental biology and immunology converged into the research awarded with the Nobel Prize. We will also discuss how these discoveries influenced future research and highlight the importance they hold today.
Collapse
|
40
|
Kusiak A, Brady G. Bifurcation of signalling in human innate immune pathways to NF-kB and IRF family activation. Biochem Pharmacol 2022; 205:115246. [PMID: 36088989 DOI: 10.1016/j.bcp.2022.115246] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/28/2022]
Abstract
The human innate immune response can be activated through a wide range of stimuli. This multi-faceted system can be triggered by a range of immunostimulants including pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). These stimuli drive intracellular signalling pathways that branch off downstream to activate several distinct transcription factors. The two most impactful of which in innate immune outcomes are the NF-κB and the IRF family members. Both transcription factor families play defining roles in driving inflammation as well as the antiviral response. Pathways leading to their simultaneous activation share common upstream components but eventually distinct regulators which directly facilitate their activation. This review will discuss the current state of knowledge about what is known about how these pathways bifurcate to activate NF-κB and IRF family members.
Collapse
Affiliation(s)
- Aleksandra Kusiak
- Trinity Translational Medicine Institute, St James' Campus, Trinity College Dublin, D08 W9RT Dublin, Ireland.
| | - Gareth Brady
- Trinity Translational Medicine Institute, St James' Campus, Trinity College Dublin, D08 W9RT Dublin, Ireland.
| |
Collapse
|
41
|
Jin R, Cao X, Lu M, Gao Q, Ma T. The intersection molecule MDA5 in Cancer and COVID-19. Front Immunol 2022; 13:963051. [PMID: 36119095 PMCID: PMC9471860 DOI: 10.3389/fimmu.2022.963051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
The connections between pattern recognition receptors (PRRs) and pathogen-associated molecular patterns (PAMPs) constitutes the crucial signaling pathways in the innate immune system. Cytoplasmic nucleic acid sensor melanoma differentiation-associated gene 5 (MDA5) serves as an important pattern recognition receptor in the innate immune system by recognizing viral RNA. MDA5 also plays a role in identifying the cytoplasmic RNA from damaged, dead cancer cells or autoimmune diseases. MDA5’s recognition of RNA triggers innate immune responses, induces interferon (IFN) response and a series of subsequent signaling pathways to produce immunomodulatory factors and inflammatory cytokines. Here we review the latest progress of MDA5 functions in triggering anti-tumor immunity by sensing cytoplasmic dsRNA, and recognizing SARS-CoV-2 virus infection for antiviral response, in which the virus utilizes multiple ways to evade the host defense mechanism.
Collapse
Affiliation(s)
- Renjing Jin
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiaoqing Cao
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Mingjun Lu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Qing Gao
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Teng Ma
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- *Correspondence: Teng Ma,
| |
Collapse
|
42
|
Lopatina T, Sarcinella A, Brizzi MF. Tumour Derived Extracellular Vesicles: Challenging Target to Blunt Tumour Immune Evasion. Cancers (Basel) 2022; 14:cancers14164020. [PMID: 36011012 PMCID: PMC9406972 DOI: 10.3390/cancers14164020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Tumour onset and development occur because of specific immune support. The immune system, which is originally able to perceive and eliminate incipient cancer cells, becomes suppressed and hijacked by cancer. For these purposes, tumour cells use extracellular vesicles (TEVs). Specific molecular composition allows TEVs to reprogram immune cells towards tumour tolerance. Circulating TEVs move from their site of origin to other organs, preparing “a fertile soil” for metastasis formation. This implies that TEV molecular content can provide a valuable tool for cancer biomarker discovery and potential targets to reshape the immune system into tumour recognition and eradication. Abstract Control of the immune response is crucial for tumour onset and progression. Tumour cells handle the immune reaction by means of secreted factors and extracellular vesicles (EV). Tumour-derived extracellular vesicles (TEV) play key roles in immune reprogramming by delivering their cargo to different immune cells. Tumour-surrounding tissues also contribute to tumour immune editing and evasion, tumour progression, and drug resistance via locally released TEV. Moreover, the increase in circulating TEV has suggested their underpinning role in tumour dissemination. This review brings together data referring to TEV-driven immune regulation and antitumour immune suppression. Attention was also dedicated to TEV-mediated drug resistance.
Collapse
|
43
|
Ataide MA, Knöpper K, Cruz de Casas P, Ugur M, Eickhoff S, Zou M, Shaikh H, Trivedi A, Grafen A, Yang T, Prinz I, Ohlsen K, Gomez de Agüero M, Beilhack A, Huehn J, Gaya M, Saliba AE, Gasteiger G, Kastenmüller W. Lymphatic migration of unconventional T cells promotes site-specific immunity in distinct lymph nodes. Immunity 2022; 55:1813-1828.e9. [PMID: 36002023 DOI: 10.1016/j.immuni.2022.07.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/06/2022] [Accepted: 07/27/2022] [Indexed: 12/31/2022]
Abstract
Lymphatic transport of molecules and migration of myeloid cells to lymph nodes (LNs) continuously inform lymphocytes on changes in drained tissues. Here, using LN transplantation, single-cell RNA-seq, spectral flow cytometry, and a transgenic mouse model for photolabeling, we showed that tissue-derived unconventional T cells (UTCs) migrate via the lymphatic route to locally draining LNs. As each tissue harbored a distinct spectrum of UTCs with locally adapted differentiation states and distinct T cell receptor repertoires, every draining LN was thus populated by a distinctive tissue-determined mix of these lymphocytes. By making use of single UTC lineage-deficient mouse models, we found that UTCs functionally cooperated in interconnected units and generated and shaped characteristic innate and adaptive immune responses that differed between LNs that drained distinct tissues. Lymphatic migration of UTCs is, therefore, a key determinant of site-specific immunity initiated in distinct LNs with potential implications for vaccination strategies and immunotherapeutic approaches.
Collapse
Affiliation(s)
- Marco A Ataide
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany.
| | - Konrad Knöpper
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Paulina Cruz de Casas
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Milas Ugur
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Sarah Eickhoff
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Mangge Zou
- Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Haroon Shaikh
- Department of Medicine II and Pediatrics, Würzburg University Hospital, ZEMM, 97078 Würzburg, Germany
| | - Apurwa Trivedi
- Centre d'Immunologie de Marseille-Luminy (CIML), Department of Immunology, 13288 Marseille, France
| | - Anika Grafen
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Tao Yang
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Immo Prinz
- Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Knut Ohlsen
- Institute for Molecular Infection Biology (IMIB), 97078 Würzburg, Germany
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Andreas Beilhack
- Department of Medicine II and Pediatrics, Würzburg University Hospital, ZEMM, 97078 Würzburg, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Mauro Gaya
- Centre d'Immunologie de Marseille-Luminy (CIML), Department of Immunology, 13288 Marseille, France
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), 97078 Würzburg, Germany
| | - Georg Gasteiger
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Wolfgang Kastenmüller
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany.
| |
Collapse
|
44
|
Wang J, Erlacher M, Fernandez-Orth J. The role of inflammation in hematopoiesis and bone marrow failure: What can we learn from mouse models? Front Immunol 2022; 13:951937. [PMID: 36032161 PMCID: PMC9403273 DOI: 10.3389/fimmu.2022.951937] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Hematopoiesis is a remarkable system that plays an important role in not only immune cell function, but also in nutrient transport, hemostasis and wound healing among other functions. Under inflammatory conditions, steady-state hematopoiesis switches to emergency myelopoiesis to give rise to the effector cell types necessary to fight the acute insult. Sustained or aberrant exposure to inflammatory signals has detrimental effects on the hematopoietic system, leading to increased proliferation, DNA damage, different forms of cell death (i.e., apoptosis, pyroptosis and necroptosis) and bone marrow microenvironment modifications. Together, all these changes can cause premature loss of hematopoiesis function. Especially in individuals with inherited bone marrow failure syndromes or immune-mediated aplastic anemia, chronic inflammatory signals may thus aggravate cytopenias and accelerate disease progression. However, the understanding of the inflammation roles in bone marrow failure remains limited. In this review, we summarize the different mechanisms found in mouse models regarding to inflammatory bone marrow failure and discuss implications for future research and clinical practice.
Collapse
Affiliation(s)
- Jun Wang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Miriam Erlacher
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Juncal Fernandez-Orth
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
- *Correspondence: Juncal Fernandez-Orth,
| |
Collapse
|
45
|
Garantziotis S, Savani RC. Proteoglycans in Toll-like receptor responses and innate immunity. Am J Physiol Cell Physiol 2022; 323:C202-C214. [PMID: 35675639 DOI: 10.1152/ajpcell.00088.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The extracellular matrix (ECM) is an active and dynamic feature of tissues that not only provides gross structure but also plays key roles in cellular responses. The ever-changing microenvironment responds dynamically to cellular and external signals, and in turn influences cell fate, tissue development, and response to environmental injury or microbial invasion. It is therefore paramount to understand how the ECM components interact with each other, the environment and cells, and how they mediate their effects. Among the ECM components that have recently garnered increased attention, proteoglycans (PGs) deserve special note. Recent evidence strongly suggests that they play a crucial role both in health maintenance and disease development. In particular, proteoglycans dictate whether homeostasis or cell death will result from a given injury, by triggering and modulating activation of the innate immune system, via a conserved array of receptors that recognize exogenous (infectious) or endogenous (tissue damage) molecular patterns. Innate immune activation by proteoglycans has important implications for the understanding of cell-matrix interactions in health and disease. In this review, we will summarize the current state of knowledge of innate immune signaling by proteoglycans, discuss the implications, and explore future directions to define progress in this area of extracellular matrix biology.
Collapse
Affiliation(s)
- Stavros Garantziotis
- Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Rashmin C Savani
- Division of Neonatal-Perinatal Medicine, Center for Pulmonary & Vascular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
46
|
Xie W, Xue Y, Zhang H, Wang Y, Meng M, Chang G, Shen X. A high-concentrate diet provokes inflammatory responses by downregulating Forkhead box protein A2 (FOXA2) through epigenetic modifications in the liver of dairy cows. Gene X 2022; 837:146703. [PMID: 35772653 DOI: 10.1016/j.gene.2022.146703] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/04/2022] [Accepted: 06/24/2022] [Indexed: 11/04/2022] Open
Abstract
A high-concentrate diet has been reported to promote an inflammatory response in dairy cows. The purpose of this study was to clarify the effect of the high-concentrate (HC) diet on hepatic Forkhead box protein A2 (FOXA2) expression and uncover the molecular mechanisms in inflammatory responses in the liver. The results showed that the HC diet reduced the ruminal fluid pH and elevated the secretion of SAA3, IL-1α, and IL-8 and reduced that of IL-10 in peripheral blood plasma. Compared with the low-concentrate (LC) group, the concentration of myeloperoxidase (MPO) was higher in the liver of dairy cows in the HC group. In addition, the relative mRNA expression of acute phase proteins (HP, SAA3, and LBP), proinflammatory cytokines (TNFα, IL-1α, IL-1β, IL-8), TLR4, MyD88, TRAF6, TRIF, IκBα, p65, p38 and JNK1 was upregulated and that of IL-10 was downregulated in the liver of the HC group. Consistently, the protein abundance of TLR4, TNFα and phosphorylation of proteins involved in NF-κB (IκBα and p65) and MAPK (p38 and JNK) pathways were significantly increased in the HC group compared with the LC group. And both the mRNA and protein abundance of FOXA2 were downregulated in the HC group. Further epigenetic analysis results demonstrated that chromatin compaction and DNA hypermethylation contributed to inhibiting FOXA2 expression, in which the demethylase ten-eleven translocation 1 (TET1) and histone deacetylase 3 (HDAC3) might participate. Overall, these findings demonstrated that the high-concentrate diet triggered inflammatory cascades and downregulated FOXA2 by epigenetic modifications in the liver of dairy cows.
Collapse
Affiliation(s)
- Wan Xie
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yang Xue
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Hongzhu Zhang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yan Wang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Meijuan Meng
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Guangjun Chang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xiangzhen Shen
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
47
|
Heidari A, Yazdanpanah N, Rezaei N. The role of Toll-like receptors and neuroinflammation in Parkinson's disease. J Neuroinflammation 2022; 19:135. [PMID: 35668422 PMCID: PMC9172200 DOI: 10.1186/s12974-022-02496-w] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 05/26/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder, characterized by motor and non-motor symptoms, significantly affecting patients' life. Pathologically, PD is associated with the extensive degeneration of dopaminergic neurons in various regions of the central nervous system (CNS), specifically the substantia nigra. This neuronal loss is accompanied by the aggregation of misfolded protein, named α-synuclein. MAIN TEXT Recent studies detected several clues of neuroinflammation in PD samples using postmortem human PD brains and various PD animal models. Some evidence of neuroinflammation in PD patients included higher levels of proinflammatory cytokines in serum and cerebrospinal fluid (CSF), presence of activated microglia in various brain regions such as substantia nigra, infiltration of peripheral inflammatory cells in affected brain regions, and altered function of cellular immunity like monocytes phagocytosis defects. On the other side, Toll-like receptors (TLRs) are innate immune receptors primarily located on microglia, as well as other immune and non-immune cells, expressing pivotal roles in recognizing exogenous and endogenous stimuli and triggering inflammatory responses. Most studies indicated an increased expression of TLRs in the brain and peripheral blood cells of PD samples. Besides, this upregulation was associated with excessive neuroinflammation followed by neurodegeneration in affected regions. Therefore, evidence proposed that TLR-mediated neuroinflammation might lead to a dopaminergic neural loss in PD patients. In this regard, TLR2, TLR4, and TLR9 have the most prominent roles. CONCLUSION Although the presence of inflammation in acute phases of PD might have protective effects concerning the clearance of α-synuclein and delaying the disease advancement, the chronic activation of TLRs and neuroinflammation might lead to neurodegeneration, resulting in the disease progression. Therefore, this study aimed to review additional evidence of the contribution of TLRs and neuroinflammation to PD pathogenesis, with the hope that TLRs could serve as novel disease-modifying therapeutic targets in PD patients in the future.
Collapse
Affiliation(s)
- Arash Heidari
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran. .,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Wang JC, Sun L. PD-1/PD-L1, MDSC Pathways, and Checkpoint Inhibitor Therapy in Ph(-) Myeloproliferative Neoplasm: A Review. Int J Mol Sci 2022; 23:5837. [PMID: 35628647 PMCID: PMC9143160 DOI: 10.3390/ijms23105837] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 11/23/2022] Open
Abstract
There has been significant progress in immune checkpoint inhibitor (CPI) therapy in many solid tumor types. However, only a single failed study has been published in treating Ph(-) myeloproliferative neoplasm (MPN). To make progress in CPI studies on this disease, herein, we review and summarize the mechanisms of activation of the PD-L1 promoter, which are as follows: (a) the extrinsic mechanism, which is activated by interferon gamma (IFN γ) by tumor infiltration lymphocytes (TIL) and NK cells; (b) the intrinsic mechanism of EGFR or PTEN loss resulting in the activation of the MAPK and AKT pathways and then stat 1 and 3 activation; and (c) 9p24 amplicon amplification, resulting in PD-L1 and Jak2 activation. We also review the literature and postulate that many of the failures of CPI therapy in MPN are likely due to excessive MDSC activities. We list all of the anti-MDSC agents, especially those with ruxolitinib, IMID compounds, and BTK inhibitors, which may be combined with CPI therapy in the future as part of clinical trials applying CPI therapy to Ph(-) MPN.
Collapse
Affiliation(s)
- Jen-Chin Wang
- Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA;
| | | |
Collapse
|
49
|
Wolińska-Nizioł L, Romaniuk K, Wojciechowska K, Surga K, Kamaszewski M, Szudrowicz H, Miączyńska M. Tollip-deficient zebrafish display no abnormalities in development, organ morphology or gene expression in response to lipopolysaccharide. FEBS Open Bio 2022; 12:1453-1464. [PMID: 35506194 PMCID: PMC9340867 DOI: 10.1002/2211-5463.13423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/13/2022] [Accepted: 05/03/2022] [Indexed: 11/11/2022] Open
Abstract
Tollip is a multifunctional adaptor protein implicated in innate immunity, lysosomal trafficking/autophagy of protein aggregates and various signaling processes in mammalian models. To verify evolutionary conservation of these functions, we used CRISPR/Cas9 editing to construct a zebrafish line bearing a stable tollip knockout. In contrast to previously reported tollip morphants, Tollip‐deficient fish display normal development until adulthood, are fertile, and have no apparent physiological defects. When challenged with lipopolysaccharide (LPS), inflammatory gene expression is unaffected. Moreover, Tollip deficiency does not aggravate swimming deficiency resulting from lysosomal dysfunction and proteotoxicity in a fish model of Gaucher disease. Thus, individual functions of Tollip may be organism‐specific or manifest only upon certain conditions/challenges or disease backgrounds.
Collapse
Affiliation(s)
- Lidia Wolińska-Nizioł
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Karolina Romaniuk
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Karolina Wojciechowska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Krzysztof Surga
- Zebrafish Core Facility, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Maciej Kamaszewski
- Department of Ichthyology and Biotechnology in Aquaculture, Institute of Animal Sciences, University of Life Sciences, Warsaw, Poland
| | - Hubert Szudrowicz
- Department of Ichthyology and Biotechnology in Aquaculture, Institute of Animal Sciences, University of Life Sciences, Warsaw, Poland
| | - Marta Miączyńska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| |
Collapse
|
50
|
Iberg CA, Bourque J, Fallahee I, Son S, Hawiger D. TNF-α sculpts a maturation process in vivo by pruning tolerogenic dendritic cells. Cell Rep 2022; 39:110657. [PMID: 35417681 PMCID: PMC9113652 DOI: 10.1016/j.celrep.2022.110657] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 02/16/2022] [Accepted: 03/18/2022] [Indexed: 12/12/2022] Open
Abstract
It remains unclear how the pro-immunogenic maturation of conventional dendritic cells (cDCs) abrogates their tolerogenic functions. Here, we report that the loss of tolerogenic functions depends on the rapid death of BTLAhi cDC1s, which, in the steady state, are present in systemic peripheral lymphoid organs and promote tolerance that limits subsequent immune responses. A canonical inducer of maturation, lipopolysaccharide (LPS), initiates a burst of tumor necrosis factor alpha (TNF-α) production and the resultant acute death of BTLAhi cDC1s mediated by tumor necrosis factor receptor 1. The ablation of these individual tolerogenic cDCs is amplified by TNF-α produced by neighboring cells. This loss of tolerogenic cDCs is transient, accentuating the restoration of homeostatic conditions through biological turnover of cDCs in vivo. Therefore, our results reveal that the abrogation of tolerogenic functions during an acute immunogenic maturation depends on an ablation of the tolerogenic cDC population, resulting in a dynamic remodeling of the cDC functional landscape.
Collapse
Affiliation(s)
- Courtney A Iberg
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Jessica Bourque
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Ian Fallahee
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Sungho Son
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|