1
|
Li J, Dan Y, Su W, Zhao M, Chen Z, Zhao Z. Immune-related biomarkers in the neuromyelitis optica spectrum disorder; pathogenesis and therapeutic approaches. Exp Eye Res 2025; 256:110395. [PMID: 40274184 DOI: 10.1016/j.exer.2025.110395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune inflammatory disease of the central nervous system that mostly affects the optic nerves and spinal cord. About eighty percent of patients have antibodies that are directed against the water channel aquaporin-4 (AQP4)-IgG, which is expressed on astrocytes. This protein was shown to be both a biomarker and a pathogenic cause of NMOSD. Researchers have discovered that antibodies against myelin oligodendrocyte glycoprotein (MOG) IgG can serve as a biomarker for a distinct condition known as MOG antibody-associated disease (MOGAD). This condition shares some similarities with AQP4-IgG-positive NMOSD, but it has distinct differences in terms of its underlying causes, clinical characteristics, response to treatment, and prognosis. Identifying AQP4 antibodies in the blood serum confirms the diagnosis of seropositive NMOSD. Nevertheless, it remains uncertain if there is a correlation between AQP4-IgG levels and disease activity, severity, responsiveness to medication, or long-term effects. Furthermore, there is still a need to establish and confirm biomarkers specifically for patients diagnosed with seronegative NMOSD. This study primarily examines the immunological aspects of NMOSD, which might have significant consequences for clinical practice. These implications include the possible use of new biomarkers to aid in the early and correct diagnosis of NMOSD, as well as the development of current treatment options to enhance the long-term prognosis of NMOSD patients.
Collapse
Affiliation(s)
- Jingyong Li
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Ya Dan
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Wei Su
- Department of Medical Imaging, The Second People's Hospital of Yaan, 625000, China
| | - Mingjun Zhao
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Zhiguo Chen
- People's Liberation Army Joint Logistic Support Force Rehabilitation Center, Da Lian, Yaan, 625000, China
| | - Zhuyang Zhao
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China.
| |
Collapse
|
2
|
Lobanovska M, Feng Y, Zhang J, Williams AH, Portnoy DA. Stress-dependent activation of the Listeria monocytogenes virulence program ensures bacterial resilience during infection. mBio 2025:e0071925. [PMID: 40304513 DOI: 10.1128/mbio.00719-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Listeria monocytogenes (Lm) is a Gram-positive, facultative intracellular pathogen that uses both a housekeeping (P1) and stress-activated (Sigma B-dependent) promoter (P2) to express the master virulence regulator PrfA. The Sigma B regulon contains over 300 genes known to respond to different stressors. However, the role of Sigma B in the regulation of prfA during the infection remains uncertain. To define pathways that lead to Sigma B-dependent prfA activation, we performed a genetic screen in L2 fibroblasts using ΔP1 Lm that only has the Sigma B-dependent promoter directly upstream of prfA. The screen identified transposon insertions in a large bacterial sensory organelle known as the stressosome. The absence of functional stressosome components resulted in heterogeneity within bacterial populations, with some bacteria behaving like wild type, while other members of the population exhibited defects in either vacuolar escape and/or cell-to-cell spread. We show that the heterogeneity of the stressosome mutants cannot be rescued by constitutive activation of PrfA. These data defined the importance of the stressosome in controlling bacterial homogeneity and characterized the function of the stressosome in robust virulence activation during infection. ΔP1 Lm model provides new opportunities to identify host-specific signals necessary for stressosome-dependent signaling during Listeria pathogenesis.IMPORTANCEMicrobial pathogens must adapt to varying levels of stress to survive. This study uncovered a link between stress sensing and activation of the virulence program in a facultative intracellular pathogen, Listeria monocytogenes. We show that host-imposed stress is sensed by the signaling machinery known as the stressosome to ensure robust and resilient virulence responses in vivo. Stressosome-dependent activation of the master virulence regulator PrfA was necessary to maintain L. monocytogenes homogeneity within the bacteria population during the transition between early and late stages of intracellular infection. This work also provides a model to further characterize how specific stress stimuli affect bacterial survival within the host, which is critical for our understanding of bacterial pathogenesis.
Collapse
Affiliation(s)
- Mariya Lobanovska
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Ying Feng
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Jonathan Zhang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Allison H Williams
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Daniel A Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
3
|
Pian Y, OuYang X. TRIM32 positively regulates c-di-GMP-Induced type I interferon signaling pathway in Listeria monocytogenes infection. Microbes Infect 2025:105499. [PMID: 40049511 DOI: 10.1016/j.micinf.2025.105499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Listeria monocytogenes (Lm) poses a significant threat to human health. TRIM32, an E3 ubiquitin ligase, plays a critical role in regulating immune responses to pathogen infections. Previous studies have shown that TRIM32 deficiency significantly impairs IFN-β production. In this study, we demonstrate that TRIM32 enhances IFN-β release upon activation by cyclic di-GMP (c-di-GMP). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed that TRIM32 deficiency upregulates genes associated with metabolic pathways while downregulating those involved in cytokine signaling and inflammatory responses. Western blot analysis further indicated a significant reduction in ERK and JNK phosphorylation in splenocytes and peritoneal macrophages, suggesting that TRIM32 modulates the MAPK signaling pathway. Additionally, the duration of p38, STAT, and TBK1 phosphorylation was shortened in bone marrow-derived macrophages. Collectively, these findings highlight the role of TRIM32 in enhancing the host immune response against Lm infection.
Collapse
Affiliation(s)
- Yaya Pian
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China.
| | - Xuan OuYang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Danielson M, Nicolai CJ, Vo TT, Wolf NK, Burke TP. Cytosolic bacterial pathogens activate TLR pathways in tumors that synergistically enhance STING agonist cancer therapies. iScience 2024; 27:111385. [PMID: 39669426 PMCID: PMC11635009 DOI: 10.1016/j.isci.2024.111385] [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: 05/20/2024] [Revised: 06/04/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
Intracellular bacterial pathogens are distinctive tools for fighting cancer, as they can proliferate in tumors and deliver therapeutic payloads to the eukaryotic cytosol. Cytosol-dwelling bacteria have undergone extensive preclinical and clinical testing, yet the mechanisms of activating innate immunity in tumors are unclear. We report that phylogenetically distinct cytosolic pathogens, including Listeria, Rickettsia, and Burkholderia species, elicited anti-tumor responses in poorly immunogenic melanoma and lymphoma in mice. Although the bacteria required cytosolic access, anti-tumor responses were largely independent of the cytosolic sensors cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING), but instead required Toll-like receptor (TLR) signaling. Combining pathogens with STING agonists elicited profound, synergistic anti-tumor effects with complete responses in >80% of mice. Small molecule TLR agonists also synergistically enhanced STING agonists. The responses required RAG2 but not interferons, and cured mice developed immunity to cancer rechallenge requiring CD8+ T cells. These studies provide a framework for enhancing microbial and small molecule innate agonists for cancer, via co-activating STING and TLRs.
Collapse
Affiliation(s)
- Meggie Danielson
- Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA 92617, USA
| | | | - Thaomy T. Vo
- Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA 92617, USA
| | - Natalie K. Wolf
- Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Thomas P. Burke
- Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA 92617, USA
| |
Collapse
|
5
|
Zhou G, Xie RF, Li SN, Chen SX, Feng YM, Xiang N, Tan ZY, Zhou X. Synergic effects and possible mechanism of emodin and stilbene glycosides on colorectal cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155821. [PMID: 39004030 DOI: 10.1016/j.phymed.2024.155821] [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: 03/08/2024] [Revised: 05/25/2024] [Accepted: 06/11/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Polygonum multiflorum (PM) is a core herb that enhances immunity. It can also detoxify, reduce swelling, and intercept malaria. Its main components, emodin (EMD) and 2,3,5,4'-Tetrahydroxy stilbene-2-O-β-D-glucoside (stilbene glycoside, TSG), have good anti-cancer potential. PURPOSE The study aims to investigate synergic effects of EMD and TSG on CRC and its possible mechanism. METHODS Network pharmacology and bioinformatics were used to identify targets. HPLC was used to analyze the effective ingredients in PM and to determine the content of the main ingredients. HT-29 cells were used for in vitro experiments. Cell Counting Kit-8 (CCK8) and scratch test were used to detect the effects of various chemical components of PM on the proliferation and migration of HT-29 cells, and Western Bolt (WB) test was used to evaluate the effects of EMD and TSG on P53 pathway. In vivo experiments, the effects of EMD and TSG were evaluated by measuring tumor weight and tumor volume in CRC mice model and histological analysis were carried out with HE staining. The expressions of HSP90, P53, COX2, and ROS were detected by quantitative reverse transcription polymerase chain reaction (PCR), and IL-1β, IL-4, IL-6, IL-10, TGF-β and IFN-γ were detected by enzyme linked immunosorbent assay (ELISA). WB and Immunohistochemistry (IHC) were used to detect the expression of P53 related proteins. RESULTS Network pharmacology showed PM closely related to colorectal cancer pathway and the core targets included STAT3 and P53; bioinformatics indicated P53 played an important role in the development and prognosis of CRC; chemical analysis showed identified and quantified gallic acid (GA), cis-TSG, trans-TSG, Emodin glucoside(EMDG), physcion glucoside (PHYG), EMD in PM; EMD induced apoptosis and TSG inhibited migration of HT-29 cells; EMD and TSG could coordinately shrink tumor size of CRC mice, elevate expressions of F4/80, decrease the content of IL-6 and TGF-β, promote tumor oxidized and reduce expression of P53 and STAT3 in the tumor. CONCLUSIONS In vitro experiments showed that TSG inhibited cancer cell migration and EMD induced apoptosis. EMD and TSG had synergic effects on CRC, whose possible mechanism might be to regulate the expression of cytokines and inhibit P53 pathway.
Collapse
Affiliation(s)
- Gui Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Rui-Fang Xie
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shan-Ni Li
- Shanghai Nanyang Model Private High School, Shanghai, China
| | - Shi-Xiu Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yi-Ming Feng
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Nan Xiang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Ze-Ye Tan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xin Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
6
|
Balan I, Boero G, Chéry SL, McFarland MH, Lopez AG, Morrow AL. Neuroactive Steroids, Toll-like Receptors, and Neuroimmune Regulation: Insights into Their Impact on Neuropsychiatric Disorders. Life (Basel) 2024; 14:582. [PMID: 38792602 PMCID: PMC11122352 DOI: 10.3390/life14050582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/18/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Pregnane neuroactive steroids, notably allopregnanolone and pregnenolone, exhibit efficacy in mitigating inflammatory signals triggered by toll-like receptor (TLR) activation, thus attenuating the production of inflammatory factors. Clinical studies highlight their therapeutic potential, particularly in conditions like postpartum depression (PPD), where the FDA-approved compound brexanolone, an intravenous formulation of allopregnanolone, effectively suppresses TLR-mediated inflammatory pathways, predicting symptom improvement. Additionally, pregnane neurosteroids exhibit trophic and anti-inflammatory properties, stimulating the production of vital trophic proteins and anti-inflammatory factors. Androstane neuroactive steroids, including estrogens and androgens, along with dehydroepiandrosterone (DHEA), display diverse effects on TLR expression and activation. Notably, androstenediol (ADIOL), an androstane neurosteroid, emerges as a potent anti-inflammatory agent, promising for therapeutic interventions. The dysregulation of immune responses via TLR signaling alongside reduced levels of endogenous neurosteroids significantly contributes to symptom severity across various neuropsychiatric disorders. Neuroactive steroids, such as allopregnanolone, demonstrate efficacy in alleviating symptoms of various neuropsychiatric disorders and modulating neuroimmune responses, offering potential intervention avenues. This review emphasizes the significant therapeutic potential of neuroactive steroids in modulating TLR signaling pathways, particularly in addressing inflammatory processes associated with neuropsychiatric disorders. It advances our understanding of the complex interplay between neuroactive steroids and immune responses, paving the way for personalized treatment strategies tailored to individual needs and providing insights for future research aimed at unraveling the intricacies of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Irina Balan
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Giorgia Boero
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA;
| | - Samantha Lucenell Chéry
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Minna H. McFarland
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alejandro G. Lopez
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
7
|
Padovani CM, Wilson RM, Rodriguez A, Spur BW, Yin K. Resolvin D2 attenuates LPS-induced macrophage exhaustion. FASEB J 2024; 38:e23569. [PMID: 38551610 DOI: 10.1096/fj.202302521r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024]
Abstract
Early in sepsis, a hyperinflammatory response is dominant, but later, an immunosuppressive phase dominates, and the host is susceptible to opportunistic infections. Anti-inflammatory agents may accelerate the host into immunosuppression, and few agents can reverse immunosuppression without causing inflammation. Specialized pro-resolving mediators (SPMs) such as resolvin D2 (RvD2) have been reported to resolve inflammation without being immunosuppressive, but little work has been conducted to examine their effects on immunosuppression. To assess the effects of RvD2 on immunosuppression, we established a model of macrophage exhaustion using two lipopolysaccharide (LPS) treatments or hits. THP-1 monocyte-derived macrophages were first treated with RvD2 or vehicle for 1 h. One LPS hit increased NF-κB activity 11-fold and TNF-α release 60-fold compared to unstimulated macrophages. RvD2 decreased LPS-induced NF-κB activity and TNF-α production but increased bacterial clearance. Two LPS hits reduced macrophage bacterial clearance and decreased macrophage NF-κB activity (45%) and TNF-α release (75%) compared to one LPS hit, demonstrating exhaustion. RvD2 increased NF-κB activity, TNF-α release, and bacterial clearance following two LPS hits compared to controls. TLR2 inhibition abolished RvD2-mediated changes. In a mouse sepsis model, splenic macrophage response to exogenous LPS was reduced compared to controls and was restored by in vivo administration of RvD2, supporting the in vitro results. If RvD2 was added to monocytes before differentiation into macrophages, however, RvD2 reduced LPS responses and increased bacterial clearance following both one and two LPS hits. The results show that RvD2 attenuated macrophage suppression in vitro and in vivo and that this effect was macrophage-specific.
Collapse
Affiliation(s)
- Cristina M Padovani
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Rachael M Wilson
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Ana Rodriguez
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Bernd W Spur
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Kingsley Yin
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| |
Collapse
|
8
|
Tavares LS, Oliveira-Silva RL, Moura MT, da Silva JB, Benko-Iseppon AM, Lima-Filho JV. Reference genes for gene expression profiling in mouse models of Listeria monocytogenes infection. Biotechniques 2024; 76:104-113. [PMID: 38112054 DOI: 10.2144/btn-2023-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023] Open
Abstract
RT-qPCR dissects transcription-based processes but requires reference genes (RGs) for data normalization. This study prospected RGs for mouse macrophages (pMØ) and spleen infected with Listeria monocytogenes. The pMØ were infected in vitro with L. monocytogenes or vehicle for 4 h. Mice were injected with L. monocytogenes (or vehicle) and euthanized 24 h post-injection. The RGs came from a multispecies primer set, from the literature or designed here. The RG ranking relied on GeNorm, NormFinder, BestKeeper, Delta-CT and RefFinder. B2m-H3f3a-Ppia were the most stable RGs for pMØ, albeit RG indexes fine-tuned estimations of cytokine relative expression. Actβ-Ubc-Ppia were the best RGs for spleen but modestly impacted the cytokine relative expression. Hence, mouse models of L. monocytogenes require context-specific RGs for RT-qPCR, thus reinforcing its paramount contribution to accurate gene expression profiling.
Collapse
Affiliation(s)
| | | | - Marcelo Tigre Moura
- Departamento de Biologia Celular e Molecular, Centro de Biotecnologia, Campus I, Universidade Federal da Paraíba, João Pessoa, PB, Brasil
| | | | | | - José Vitor Lima-Filho
- Departamento de Biologia, Universidade Federal Rural de Pernambuco, Recife, PE, Brasil
| |
Collapse
|
9
|
Danielson M, Nicolai CJ, Vo TT, Wolf N, Burke TP. Cytosolic bacterial pathogens activate TLR pathways in tumors that synergistically enhance STING agonist cancer therapies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.578087. [PMID: 38352567 PMCID: PMC10862861 DOI: 10.1101/2024.01.30.578087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Bacterial pathogens that invade the eukaryotic cytosol are distinctive tools for fighting cancer, as they preferentially target tumors and can deliver cancer antigens to MHC-I. Cytosolic bacterial pathogens have undergone extensive preclinical development and human clinical trials, yet the molecular mechanisms by which they are detected by innate immunity in tumors is unclear. We report that intratumoral delivery of phylogenetically distinct cytosolic pathogens, including Listeria, Rickettsia, and Burkholderia species, elicited anti-tumor responses in established, poorly immunogenic melanoma and lymphoma in mice. We were surprised to observe that although the bacteria required entry to the cytosol, the anti-tumor responses were largely independent of the cytosolic sensors cGAS/STING and instead required TLR signaling. Combining pathogens with TLR agonists did not enhance anti-tumor efficacy, while combinations with STING agonists elicited profound, synergistic anti-tumor effects with complete responses in >80% of mice after a single dose. Small molecule TLR agonists also synergistically enhanced the anti-tumor activity of STING agonists. The anti-tumor effects were diminished in Rag2-deficient mice and upon CD8 T cell depletion. Mice cured from combination therapy developed immunity to cancer rechallenge that was superior to STING agonist monotherapy. Together, these data provide a framework for enhancing the efficacy of microbial cancer therapies and small molecule innate immune agonists, via the co-activation of STING and TLRs.
Collapse
Affiliation(s)
- Meggie Danielson
- Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA USA
| | - Chris J. Nicolai
- Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Thaomy T. Vo
- Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA USA
| | - Natalie Wolf
- Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Thomas P. Burke
- Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA USA
| |
Collapse
|
10
|
Feng Y, Chang SK, Portnoy DA. The major role of Listeria monocytogenes folic acid metabolism during infection is the generation of N-formylmethionine. mBio 2023; 14:e0107423. [PMID: 37695058 PMCID: PMC10653936 DOI: 10.1128/mbio.01074-23] [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: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 09/12/2023] Open
Abstract
IMPORTANCE Folic acid is an essential vitamin for bacteria, plants, and animals. The lack of folic acid leads to various consequences such as a shortage of amino acids and nucleotides that are fundamental building blocks for life. Though antifolate drugs are widely used for antimicrobial treatments, the underlying mechanism of bacterial folate deficiency during infection is unclear. This study compares the requirements of different folic acid end-products during the infection of Listeria monocytogenes, a facultative intracellular pathogen of animals and humans. The results reveal the critical importance of N-formylmethionine, the amino acid used by bacteria to initiate protein synthesis. This work extends the current understanding of folic acid metabolism in pathogens and potentially provides new insights into antifolate drug development in the future.
Collapse
Affiliation(s)
- Ying Feng
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - Shannon K. Chang
- Department of Plant and Microbial Biology, University of California, Berkeley, California, USA
| | - Daniel A. Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, California, USA
| |
Collapse
|
11
|
Colleselli K, Stierschneider A, Wiesner C. An Update on Toll-like Receptor 2, Its Function and Dimerization in Pro- and Anti-Inflammatory Processes. Int J Mol Sci 2023; 24:12464. [PMID: 37569837 PMCID: PMC10419760 DOI: 10.3390/ijms241512464] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
While a certain level of inflammation is critical for humans to survive infection and injury, a prolonged inflammatory response can have fatal consequences. Pattern recognition Toll-like receptors (TLRs) are key players in the initiation of an inflammatory process. TLR2 is one of the most studied pattern recognition receptors (PRRs) and is known to form heterodimers with either TLR1, TLR4, TLR6, and TLR10, allowing it to recognize a wide range of pathogens. Although a large number of studies have been conducted over the past decades, there are still many unanswered questions regarding TLR2 mechanisms in health and disease. In this review, we provide an up-to-date overview of TLR2, including its homo- and heterodimers. Furthermore, we will discuss the pro- and anti-inflammatory properties of TLR2 and recent findings in prominent TLR2-associated infectious and neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | - Christoph Wiesner
- Department of Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences, 3500 Krems, Austria
| |
Collapse
|
12
|
Zou J, Chu S, Bao Q, Zhang Y. Telomere maintenance genes-derived prognosis signature characterizes immune landscape and predicts prognosis of head and neck squamous cell carcinoma. Medicine (Baltimore) 2023; 102:e34586. [PMID: 37543795 PMCID: PMC10402981 DOI: 10.1097/md.0000000000034586] [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: 04/26/2023] [Revised: 07/05/2023] [Accepted: 07/13/2023] [Indexed: 08/07/2023] Open
Abstract
Telomere dysfunction has been identified as a biological marker of cancer progression in several types of cancer, including Head and Neck Squamous Cell Carcinoma (HNSCC). This study aimed to characterize the telomere maintenance genes (TMG)-related signature in prognosis and treatment response in HNSCC. The transcriptome and clinical data of HNSCC were obtained from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus databases, respectively. Non-negative matrix factorization (NMF) was used to identify molecular subtypes derived from TMG. Gene set enrichment analysis (GSEA) was performed to analyze the differentially expressed pathways between subtypes, and a risk score model derived from TMG was established. Kaplan-Meier survival analysis was used to evaluate inter-group prognostic features, and the correlation between TMG-derived molecular subtypes and risk score model with immune infiltration, immunotherapy, and chemosensitivity was assessed. Two HNSCC subtypes were identified based on 59 TMG-related genes, which exhibit significant heterogeneity in prognosis, immune cell infiltration, and treatment response. Additionally, a TMG-derived risk signature containing 9 genes was developed to assess the prognosis of HNSCC patients. The signature had significant predictive ability for HNSCC prognosis and was significantly correlated with immune cell infiltration and immunotherapy response. A nomogram integrating the risk signature, N stage and radiotherapy was constructed to predict 1-, 3-, and 5-year overall survival (OS) of HNSCC patients, which had better performance than other prognostic models and included TMG-derived risk score, radiotherapy, and N stage. This study identified TMG-derived molecular subtypes in HNSCC and developed a novel prognostic score model, highlighting the potential value of TMG in HNSCC prognosis and immunotherapy.
Collapse
Affiliation(s)
- Jianjun Zou
- Department of Otolaryngology, Hangzhou Red Cross Hospital (Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine), Hangzhou, Zhejiang, China
| | - Shidong Chu
- Department of Otolaryngology, Hangzhou Red Cross Hospital (Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine), Hangzhou, Zhejiang, China
| | - Qingfu Bao
- Department of Otolaryngology, Hangzhou Red Cross Hospital (Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine), Hangzhou, Zhejiang, China
| | - Yiyun Zhang
- Department of Otolaryngology, Hangzhou Red Cross Hospital (Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
13
|
OuYang X, Liu P, Zheng Y, Jiang H, Lv Q, Huang W, Hao H, Pian Y, Kong D, Jiang Y. TRIM32 reduced the recruitment of innate immune cells and the killing capacity of Listeria monocytogenes by inhibiting secretion of chemokines. Gut Pathog 2023; 15:32. [PMID: 37415157 DOI: 10.1186/s13099-023-00558-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/10/2023] [Indexed: 07/08/2023] Open
Abstract
Listeria monocytogenes (Lm) is a facultative, intracellular Gram-positive pathogenic bacterium that causes sepsis, a condition characterized by persistent excessive inflammation and organ dysfunction. However, the pathogenesis of Lm-induced sepsis is unknown. In this research, we discovered that TRIM32 is required for innate immune regulation during Lm infection. Trim32 deficiency remarkably reduced bacteremia and proinflammatory cytokine secretion in mice with severe Lm infection, preventing sepsis. Trim32-/- mice had a lower bacterial burden after Lm infection and survived significantly longer than wild-type (WT) mice, as well as lower serum levels of inflammatory cytokines TNF-α, IL-6, IL-18, IL-12p70, IFN-β, and IFN-γ at 1 day post infection (dpi) compared to WT mice. On the other hand, the chemokines CXCL1, CCL2, CCL7, and CCL5 were enhanced at 3 dpi in Trim32-/- mice than WT mice, reflecting increased recruitment of neutrophils and macrophages. Furthermore, Trim32-/- mice had higher levels of macrophage-associated iNOS to kill Lm. Collectively, our findings suggest that TRIM32 reduces innate immune cells recruitment and Lm killing capabilities via iNOS production.
Collapse
Affiliation(s)
- Xuan OuYang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Peng Liu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Yuling Zheng
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Hua Jiang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Qingyu Lv
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Wenhua Huang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Huaijie Hao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Yaya Pian
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China.
| | - Decong Kong
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China.
| | - Yongqiang Jiang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China.
| |
Collapse
|
14
|
Rahman S, Das AK. A subtractive proteomics and immunoinformatics approach towards designing a potential multi-epitope vaccine against pathogenic Listeriamonocytogenes. Microb Pathog 2022; 172:105782. [PMID: 36150556 DOI: 10.1016/j.micpath.2022.105782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/04/2022] [Accepted: 09/11/2022] [Indexed: 11/29/2022]
Abstract
Listeria monocytogenes is the causative agent of listeriosis, which is dangerous for pregnant women, the elderly or individuals with a weakened immune system. Individuals with leukaemia, cancer, HIV/AIDS, kidney transplant and steroid therapy suffer from immunological damage are menaced. World Health Organization (WHO) reports that human listeriosis has a high mortality rate of 20-30% every year. To date, no vaccine is available to treat listeriosis. Thereby, it is high time to design novel vaccines against L. monocytogenes. Here, we present computational approaches to design an antigenic, stable and safe vaccine against the L. monocytogenes that could help to control the infections associated with the pathogen. Three vital pathogenic proteins of L. monocytogenes, such as Listeriolysin O (LLO), Phosphatidylinositol-specific phospholipase C (PI-PLC), and Actin polymerization protein (ActA), were selected using a subtractive proteomics approach to design the multi-epitope vaccine (MEV). A total of 5 Cytotoxic T-lymphocyte (CTL) and 9 Helper T-lymphocyte (HTL) epitopes were predicted from these selected proteins. To design the multi-epitope vaccine (MEV) from the selected proteins, CTL epitopes were joined with the AAY linker, and HTL epitopes were joined with the GPGPG linker. Additionally, a human β-defensin-3 (hBD-3) adjuvant was added to the N-terminal side of the final MEV construct to increase the immune response to the vaccine. The final MEV was predicted to be antigenic, non-allergen and non-toxic in nature. Physicochemical property analysis suggested that the MEV construct is stable and could be easily purified through the E. coli expression system. This in-silico study showed that MEV has a robust binding interaction with Toll-like receptor 2 (TLR2), a key player in the innate immune system. Current subtractive proteomics and immunoinformatics study provides a background for designing a suitable, safe and effective vaccine against pathogenic L. monocytogenes.
Collapse
Affiliation(s)
- Shakilur Rahman
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, West Bengal, India
| | - Amit Kumar Das
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, West Bengal, India.
| |
Collapse
|
15
|
Tavares LS, Mancebo BD, Santana LN, Adelson do Nascimento Silva A, Silva RLDO, Benko-Iseppon AM, Ramos MV, Monteiro do Nascimento CT, Grangeiro TB, Sousa JS, Mota RA, Júnior VADS, Lima-Filho JV. Recombinant osmotin inclusion bodies from Calotropis procera produced in E. coli BL21(DE3) prevent acute inflammation in a mouse model of listeriosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 102:154186. [PMID: 35617890 DOI: 10.1016/j.phymed.2022.154186] [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: 02/28/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The osmotin from the medicinal plant Calotropis procera (CpOsm) has characteristics similar to adiponectin, a human protein with immunoregulatory actions. PURPOSE This study aimed to investigate whether recombinant osmotin inclusion bodies from C. procera (IB/rCpOsm) produced in E. coli BL21(DE3) can prevent infection-induced inflammation. A virulent strain of Listeria monocytogenes was used as an infection model. METHODS Cells of E. coli BL21(DE3) carrying the plasmid pET303-CpOsm were used to express the recombinant osmotin, which accumulated at reasonable levels as inclusion bodies (IB/rCpOsm). IB/rCpOsm were purified from induced cells and SDS-polyacrylamide gel electrophoresis followed by mass spectrometry analyses confirmed the identity of the major protein band (23 kDa apparent molecular mass) as CpOsm. Peritoneal macrophages (pMØ) from Swiss mice were cultured with IB/rCpOsm (1 or 10 µg/ml) in 96-well plates and then infected with L. monocytogenes. IB/rCpOsm (0.1, 1 or 10 mg/kg) was also administered intravenously to Swiss mice, which were then infected intraperitoneally with L. monocytogenes. RESULTS Pretreatment of the pMØ with IB/rCpOsm significantly increased cell viability after infection and reduced the intracellular bacterial load. The infiltration of neutrophils into the peritoneal cavity of mice pretreated with IB/rCpOsm at 10 mg/kg (but not 0.1 and 1 mg/kg) was reduced after infection. In these mice, the bacterial load was high in the peritoneal fluid and the liver, but histological damage was discrete. The treatments with IB/rCpOsm at 10 mg/kg significantly increased the expression of the anti-inflammatory cytokine IL-10. CONCLUSION This study shows that recombinant osmotin inclusion bodies from C. procera were bioactive and prompted anti-inflammatory actions at therapeutic dosages in the L. monocytogenes infection model.
Collapse
|
16
|
Wang L, Ma Y, Shi J, Zhang Y, Tong J, Han Q. Corynebacterium pyruviciproducens-peptidoglycan: A novel bacterial peptidoglycan inhibiting overexpression of MyD88 in macrophages. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221095378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objectives: Bacterial peptidoglycan (PGN) is an essential ligand of TLR2 inducing inflammatory damage by boosting MyD88 overexpression in pathogen invasion, such as Methicillin-resistant Staphylococcus aureus (MRSA) infection. CP-PGN is a novel PGN from an adjuvant bacterium, displaying anti-infection immune regulation. This study aimed to clarify the unique moderation of MyD88 expression by CP-PGN. Methods: Compared with other ligands of TLR2, high expression of MyD88 in macrophages was established by MRSA and virus to investigate the immunomodulation of CP-PGN. Results: Compared with PGN derived from MRSA (M-PGN) and chemosynthetic Pam3CSK4 of model agonists of TLR2, CP-PGN could inhibit overexpression of MyD88 in a time- and dose-dependent way in infected macrophages by MRSA or Abelson leukemia virus. CP-PGN also promoted more anti-inflammatory IL-10 and less pro-inflammatory TNF-α in immature primary macrophages. Furthermore, IL-10 secretion induced by CP-PGN was reduced most significantly by blocking the dimer formation of MyD88 with ST2825 and lowering down expression by si-MyD88. Conclusion: CP-PGN could inhibit MyD88 overexpression by infection to moderate inflammatory cytokines. Therefore, CP-PGN is a novel potential ligand of TLR2 to induce inflammatory balance in the process of host defense against invading pathogens.
Collapse
Affiliation(s)
- Lin Wang
- Center of Clinical Laboratory, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, People’s Republic of China
| | - Yuan Ma
- Center of Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Jinfang Shi
- Center of Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Yang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Jia Tong
- Department of Immunology, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Qingzhen Han
- Center of Clinical Laboratory, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, People’s Republic of China
| |
Collapse
|
17
|
Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front Immunol 2022. [PMID: 35309296 DOI: 10.3389/fimmu.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Innate immunity is the first defense system against invading pathogens. Toll-like receptors (TLRs) are well-defined pattern recognition receptors responsible for pathogen recognition and induction of innate immune responses. Since their discovery, TLRs have revolutionized the field of immunology by filling the gap between the initial recognition of pathogens by innate immune cells and the activation of the adaptive immune response. TLRs critically link innate immunity to adaptive immunity by regulating the activation of antigen-presenting cells and key cytokines. Furthermore, recent studies also have shown that TLR signaling can directly regulate the T cell activation, growth, differentiation, development, and function under diverse physiological conditions. This review provides an overview of TLR signaling pathways and their regulators and discusses how TLR signaling, directly and indirectly, regulates cell-mediated immunity. In addition, we also discuss how TLR signaling is critically important in the host's defense against infectious diseases, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yang Du
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Changsheng Xing
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Helen Y Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
18
|
Rivera-Lugo R, Deng D, Anaya-Sanchez A, Tejedor-Sanz S, Tang E, Reyes Ruiz VM, Smith HB, Titov DV, Sauer JD, Skaar EP, Ajo-Franklin CM, Portnoy DA, Light SH. Listeria monocytogenes requires cellular respiration for NAD + regeneration and pathogenesis. eLife 2022; 11:e75424. [PMID: 35380108 PMCID: PMC9094743 DOI: 10.7554/elife.75424] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/04/2022] [Indexed: 11/20/2022] Open
Abstract
Cellular respiration is essential for multiple bacterial pathogens and a validated antibiotic target. In addition to driving oxidative phosphorylation, bacterial respiration has a variety of ancillary functions that obscure its contribution to pathogenesis. We find here that the intracellular pathogen Listeria monocytogenes encodes two respiratory pathways which are partially functionally redundant and indispensable for pathogenesis. Loss of respiration decreased NAD+ regeneration, but this could be specifically reversed by heterologous expression of a water-forming NADH oxidase (NOX). NOX expression fully rescued intracellular growth defects and increased L. monocytogenes loads >1000-fold in a mouse infection model. Consistent with NAD+ regeneration maintaining L. monocytogenes viability and enabling immune evasion, a respiration-deficient strain exhibited elevated bacteriolysis within the host cytosol and NOX expression rescued this phenotype. These studies show that NAD+ regeneration represents a major role of L. monocytogenes respiration and highlight the nuanced relationship between bacterial metabolism, physiology, and pathogenesis.
Collapse
Affiliation(s)
- Rafael Rivera-Lugo
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - David Deng
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Andrea Anaya-Sanchez
- Graduate Group in Microbiology, University of California, BerkeleyBerkeleyUnited States
| | - Sara Tejedor-Sanz
- Department of Biosciences, Rice UniversityHoustonUnited States
- The Molecular Foundry, Lawrence Berkeley National LaboratoryBerkeleyUnited States
| | - Eugene Tang
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Valeria M Reyes Ruiz
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical CenterNashvilleUnited States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical CenterNashvilleUnited States
| | - Hans B Smith
- Department of Medical Microbiology and Immunology, University of Wisconsin-MadisonMadisonUnited States
| | - Denis V Titov
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
- Department of Nutritional Sciences and Toxicology, University of California, BerkeleyBerkeleyUnited States
- Center for Computational Biology, University of California, BerkeleyBerkeleyUnited States
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-MadisonMadisonUnited States
| | - Eric P Skaar
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical CenterNashvilleUnited States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical CenterNashvilleUnited States
| | - Caroline M Ajo-Franklin
- Department of Biosciences, Rice UniversityHoustonUnited States
- The Molecular Foundry, Lawrence Berkeley National LaboratoryBerkeleyUnited States
| | - Daniel A Portnoy
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
- Department of Plant and Microbial Biology, University of California, BerkeleyBerkeleyUnited States
| | - Samuel H Light
- Department of Microbiology, University of ChicagoChicagoUnited States
- Duchossois Family Institute, University of ChicagoChicagoUnited States
| |
Collapse
|
19
|
Rosenberg G, Riquelme S, Prince A, Avraham R. Immunometabolic crosstalk during bacterial infection. Nat Microbiol 2022; 7:497-507. [PMID: 35365784 DOI: 10.1038/s41564-022-01080-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 02/03/2022] [Indexed: 01/22/2023]
Abstract
Following detection of bacteria, macrophages switch their metabolism from oxidative respiration through the tricarboxylic acid cycle to high-rate aerobic glycolysis. This immunometabolic shift enables pro-inflammatory and antimicrobial responses and is facilitated by the accumulation of fatty acids, tricarboxylic acid-derived metabolites and catabolism of amino acids. Recent studies have shown that these immunometabolites are co-opted by pathogens as environmental cues for expression of virulence genes. We review mechanisms by which host immunometabolites regulate bacterial pathogenicity and discuss opportunities for the development of therapeutics targeting metabolic host-pathogen crosstalk.
Collapse
Affiliation(s)
- Gili Rosenberg
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | - Alice Prince
- Columbia University Medical Center, New York, NY, USA.
| | - Roi Avraham
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
20
|
Rivera-Lugo R, Light SH, Garelis NE, Portnoy DA. RibU is an essential determinant of Listeria pathogenesis that mediates acquisition of FMN and FAD during intracellular growth. Proc Natl Acad Sci U S A 2022; 119:e2122173119. [PMID: 35316134 PMCID: PMC9060500 DOI: 10.1073/pnas.2122173119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/15/2022] [Indexed: 11/18/2022] Open
Abstract
Flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD) are essential riboflavin-derived cofactors involved in a myriad of redox reactions across all forms of life. Nevertheless, the basis of flavin acquisition strategies by riboflavin auxotrophic pathogens remains poorly defined. In this study, we examined how the facultative intracellular pathogen Listeria monocytogenes, a riboflavin auxotroph, acquires flavins during infection. A L. monocytogenes mutant lacking the putative riboflavin transporter (RibU) was completely avirulent in mice but had no detectable growth defect in nutrient-rich media. However, unlike wild type, the RibU mutant was unable to grow in defined media supplemented with FMN or FAD or to replicate in macrophages starved for riboflavin. Consistent with RibU functioning to scavenge FMN and FAD inside host cells, a mutant unable to convert riboflavin to FMN or FAD retained virulence and grew in cultured macrophages and in spleens and livers of infected mice. However, this FMN- and FAD-requiring strain was unable to grow in the gallbladder or intestines, where L. monocytogenes normally grows extracellularly, suggesting that these sites do not contain sufficient flavin cofactors to promote replication. Thus, by deleting genes required to synthesize FMN and FAD, we converted L. monocytogenes from a facultative to an obligate intracellular pathogen. Collectively, these data indicate that L. monocytogenes requires riboflavin to grow extracellularly in vivo but scavenges FMN and FAD to grow in host cells.
Collapse
Affiliation(s)
- Rafael Rivera-Lugo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Samuel H. Light
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Nicholas E. Garelis
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Daniel A. Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720
| |
Collapse
|
21
|
Hu J, Deng F, Zhao B, Lin Z, Sun Q, Yang X, Wu M, Qiu S, Chen Y, Yan Z, Luo S, Zhao J, Liu W, Li C, Liu KX. Lactobacillus murinus alleviate intestinal ischemia/reperfusion injury through promoting the release of interleukin-10 from M2 macrophages via Toll-like receptor 2 signaling. MICROBIOME 2022; 10:38. [PMID: 35241180 PMCID: PMC8896269 DOI: 10.1186/s40168-022-01227-w] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/07/2022] [Indexed: 05/05/2023]
Abstract
BACKGROUND Intestinal ischemia/reperfusion (I/R) injury has high morbidity and mortality rates. Gut microbiota is a potential key factor affecting intestinal I/R injury. Populations exhibit different sensitivities to intestinal I/R injury; however, whether this interpopulation difference is related to variation in gut microbiota is unclear. Here, to elucidate the interaction between the gut microbiome and intestinal I/R injury, we performed 16S DNA sequencing on the preoperative feces of C57BL/6 mice and fecal microbiota transplantation (FMT) experiments in germ-free mice. The transwell co-culture system of small intestinal organoids extracted from control mice and macrophages extracted from control mice or Toll-like receptor 2 (TLR2)-deficient mice or interleukin-10 (IL-10)-deficient mice were established separately to explore the potential mechanism of reducing intestinal I/R injury. RESULTS Intestinal I/R-sensitive (Sen) and intestinal I/R-resistant (Res) mice were first defined according to different survival outcomes of mice suffering from intestinal I/R. Fecal microbiota composition and diversity prior to intestinal ischemia differed between Sen and Res mice. The relative abundance of Lactobacillus murinus (L. murinus) at the species level was drastically higher in Res than that in Sen mice. Clinically, the abundance of L. murinus in preoperative feces of patients undergoing cardiopulmonary bypass surgery was closely related to the degree of intestinal I/R injury after surgery. Treatment with L. murinus significantly prevented intestinal I/R-induced intestinal injury and improved mouse survival, which depended on macrophages involvement. Further, in vitro experiments indicated that promoting the release of IL-10 from macrophages through TLR2 may be a potential mechanism for L. murinus to reduce intestinal I/R injury. CONCLUSION The gut microbiome is involved in the postoperative outcome of intestinal I/R. Lactobacillus murinus alleviates mice intestinal I/R injury through macrophages, and promoting the release of IL-10 from macrophages through TLR2 may be a potential mechanism for L. murinus to reduce intestinal I/R injury. This study revealed a novel mechanism of intestinal I/R injury and a new therapeutic strategy for clinical practice. Video Abstract.
Collapse
Affiliation(s)
- Jingjuan Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Fan Deng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Bingcheng Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Zebin Lin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Qishun Sun
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Xiao Yang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Mei Wu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Shida Qiu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Yu Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Zhengzheng Yan
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Sidan Luo
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Jin Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Weifeng Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Cai Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| | - Ke Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| |
Collapse
|
22
|
Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front Immunol 2022; 13:812774. [PMID: 35309296 PMCID: PMC8927970 DOI: 10.3389/fimmu.2022.812774] [Citation(s) in RCA: 419] [Impact Index Per Article: 139.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
Innate immunity is the first defense system against invading pathogens. Toll-like receptors (TLRs) are well-defined pattern recognition receptors responsible for pathogen recognition and induction of innate immune responses. Since their discovery, TLRs have revolutionized the field of immunology by filling the gap between the initial recognition of pathogens by innate immune cells and the activation of the adaptive immune response. TLRs critically link innate immunity to adaptive immunity by regulating the activation of antigen-presenting cells and key cytokines. Furthermore, recent studies also have shown that TLR signaling can directly regulate the T cell activation, growth, differentiation, development, and function under diverse physiological conditions. This review provides an overview of TLR signaling pathways and their regulators and discusses how TLR signaling, directly and indirectly, regulates cell-mediated immunity. In addition, we also discuss how TLR signaling is critically important in the host's defense against infectious diseases, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yang Du
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Changsheng Xing
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Helen Y. Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
23
|
Santana LN, Tavares LS, Dorvigny BM, Souza FDAL, Paiva BHDA, Evêncio-Neto J, Hounkonnou SGC, Silva AFB, Ramos MV, Lima-Filho JV. Anti-infective activity of Cratylia argentea lectin (CFL) against experimental infection with virulent Listeria monocytogenes in Swiss mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 94:153839. [PMID: 34781231 DOI: 10.1016/j.phymed.2021.153839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/06/2021] [Accepted: 10/27/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND The lectin from Cratylia argentea (CFL) is able to modulate the immune system response and is thus a potential phytotherapeutic substance. HYPOTHESIS/PURPOSE In this study, we investigated the role of CFL on control of bacterial infection caused by Listeria monocytogenes, the causative agent of human listeriosis. STUDY DESIGN Swiss mice were infected with L. monocytogenes and then treated with CFL. METHODS Adult Swiss mice weighing with 30-40 g were infected intraperitoneally with a bacterial suspension (0.2 ml; 1 × 107 CFU/ml). After 30 min, the mice were treated with CFL intravenously at concentrations of 0.1 or 10 mg/kg. Control mice received phosphate-buffered saline (PBS). The animals were euthanized 24 h after infection. RESULTS We observed that i.v. administration of CFL to Swiss mice did not cause acute toxicity, and reduced the leukocyte counts in the bloodstream 24 h after infection with virulent L. monocytogenes. There was a reduction in the bacterial burden within peritoneal macrophages after infection in CFL-treated mice. Accordingly, the bacterial counts in the bloodstream, spleen and liver also decreased in comparison with the PBS group. Histological damage in the spleen and liver was lower in mice that received CFL treatment. In vitro antimicrobial assays demonstrated that CFL does not inhibit the growth of L. monocytogenes. The mRNA expression of the anti-inflammatory cytokine IL-10 was enhanced with CFL treatment after infection. CONCLUSION The lectin from C. argentea (CFL) has immunomodulatory and anti-infective properties of pharmacological interest for control of infectious diseases.
Collapse
Affiliation(s)
- Lucas Nunes Santana
- Department of Biology, Microbiology and Immunology Laboratory, Federal Rural University of Pernambuco, Dom Manoel de Medeiros, s/n, B. Dois Irmãos, Recife, PE CEP 52171-900, Brazil
| | - Lethicia Souza Tavares
- Department of Biology, Microbiology and Immunology Laboratory, Federal Rural University of Pernambuco, Dom Manoel de Medeiros, s/n, B. Dois Irmãos, Recife, PE CEP 52171-900, Brazil
| | - Betty Mancebo Dorvigny
- Department of Biology, Microbiology and Immunology Laboratory, Federal Rural University of Pernambuco, Dom Manoel de Medeiros, s/n, B. Dois Irmãos, Recife, PE CEP 52171-900, Brazil
| | | | | | - Joaquim Evêncio-Neto
- Department of Morphology and Animal Physiology, Federal Rural University of Pernambuco, Recife, PE, Brazil
| | | | | | - Márcio Viana Ramos
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Jose Vitor Lima-Filho
- Department of Biology, Microbiology and Immunology Laboratory, Federal Rural University of Pernambuco, Dom Manoel de Medeiros, s/n, B. Dois Irmãos, Recife, PE CEP 52171-900, Brazil.
| |
Collapse
|
24
|
Li X, Cao G, Yang H, Zhi D, Li L, Wang D, Liu M, Su H. S100A8 expression in oviduct mucosal epithelial cells is regulated by estrogen and affects mucosal immune homeostasis. PLoS One 2021; 16:e0260188. [PMID: 34793556 PMCID: PMC8601440 DOI: 10.1371/journal.pone.0260188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/03/2021] [Indexed: 12/05/2022] Open
Abstract
Chronic inflammation can cause oviduct mucosal damage and immune dysfunction, leading to infertility, early pregnancy loss, ectopic pregnancy, tumors, and a decrease in reproductive capacities in female animals. Estrogen can suppress immune responses in different tissues and oviducts, and regulate the oviduct immune balance; however, the underlying mechanisms remain unclear. The objective of this study was to explore the mechanism of estrogen-regulated oviduct mucosal immunity and discover new estrogen targets for regulating oviduct mucosal immune homeostasis. Sheep oviduct epithelial cells (SOECs) were treated with 17-β estradiol (E2). Transcriptome sequencing and analysis showed differentially expressed S100 calcium-binding protein A (S100A) genes that may participate in the oviduct mucosa immunoregulation of estrogen. Quantitative polymerase chain reaction and immunocytochemistry analysis showed that S100A8 expression changed dynamically in E2-treated SOECs and peaked after 7 h of treatment. Estrogen nuclear receptors and G protein-coupled membrane receptors promoted E2-dependent S100A8 upregulation. The S100A8 gene was disrupted using the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 method. Levels of inflammatory factors interleukin (IL)-1β and IL-4 were significantly upregulated in S100A8-knockdown SOECs, whereas those of the anti-inflammatory factor IL-10 was downregulated. Following S100A8 knockdown in SOECs treated with E2 for 7 h, IL-10 levels increased significantly. Estrogen affected oviduct mucosa immune function and dynamically regulated S100A8 in SOECs. S100A8 knockdown caused an excessive immune response, indicating that S100A8 is beneficial for maintaining immune homeostasis in the oviduct mucosa. Moreover, estrogen can compensate for the effect of S100A8 knockdown by upregulating IL-10.
Collapse
Affiliation(s)
- Xiaodan Li
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
- Department of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Guifang Cao
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
- * E-mail:
| | - Hongxin Yang
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
- Department of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Dafu Zhi
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lei Li
- Maternal and Child Health Hospital of Hohhot, Hohhot, China
| | - Daqing Wang
- Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot, China
| | - Moning Liu
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Hong Su
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
25
|
Li H, Liu S, Han J, Li S, Gao X, Wang M, Zhu J, Jin T. Role of Toll-Like Receptors in Neuroimmune Diseases: Therapeutic Targets and Problems. Front Immunol 2021; 12:777606. [PMID: 34790205 PMCID: PMC8591135 DOI: 10.3389/fimmu.2021.777606] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/15/2021] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptors (TLRs) are a class of proteins playing a key role in innate and adaptive immune responses. TLRs are involved in the development and progression of neuroimmune diseases via initiating inflammatory responses. Thus, targeting TLRs signaling pathway may be considered as a potential therapy for neuroimmune diseases. However, the role of TLRs is elusive and complex in neuroimmune diseases. In addition to the inadequate immune response of TLRs inhibitors in the experiments, the recent studies also demonstrated that partial activation of TLRs is conducive to the production of anti-inflammatory factors and nervous system repair. Exploring the mechanism of TLRs in neuroimmune diseases and combining with developing the emerging drug may conquer neuroimmune diseases in the future. Herein, we provide an overview of the role of TLRs in several neuroimmune diseases, including multiple sclerosis, neuromyelitis optica spectrum disorder, Guillain-Barré syndrome and myasthenia gravis. Emerging difficulties and potential solutions in clinical application of TLRs inhibitors will also be discussed.
Collapse
Affiliation(s)
- Haixia Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Shan Liu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jinming Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
| | - Shengxian Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyan Gao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Meng Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital, Solna, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Kumar V. Going, Toll-like receptors in skin inflammation and inflammatory diseases. EXCLI JOURNAL 2021; 20:52-79. [PMID: 33510592 PMCID: PMC7838829 DOI: 10.17179/excli2020-3114] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023]
Abstract
The Indian Ayurvedic physicians knew the concept of inflammation dating back to 1500 BC. The continuous progress in the immunology of inflammation has explained its undiscovered mechanisms. For example, the discovery of Toll-like receptor 4 (TLR4) in humans (1997) has revolutionized the field of infection biology and innate immunity. The laboratory mice have shown twelve TLRs and express TLR10 (CD290) as a disrupted pseudogene, and humans have ten functional TLRs. Now, it is well established that TLRs play a significant role in different infectious and inflammatory diseases. Skin inflammation and other associated inflammatory diseases, including atopic dermatitis (AD), acne vulgaris, and psoriasis, along with many skin cancers are major health problems all over the world. The continuous development in the immunopathogenesis of inflammatory skin diseases has opened the window of opportunity for TLRs in studying their role. Hence, the manuscript explores the role of different TLRs in the pathogenesis of skin inflammation and associated inflammatory diseases. The article starts with the concept of inflammation, its origin, and the impact of TLRs discovery on infection and inflammation biology. The subsequent section describes the burden of skin-associated inflammatory diseases worldwide and the effect of the geographical habitat of people affecting it. The third section explains skin as an immune organ and explains the expression of different TLRs on different skin cells, including keratinocytes, Langerhans cells (LCs), skin fibroblasts, and melanocytes. The fourth section describes the impact of TLRs on these cells in different skin-inflammatory conditions, including acne vulgaris, AD, psoriasis, and skin cancers. The article also discusses the use of different TLR-based therapeutic approaches as specific to these inflammatory skin diseases.
Collapse
Affiliation(s)
- Vijay Kumar
- Children Health Clinical Unit, Faculty of Medicine and Biomedical Sciences, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia
| |
Collapse
|
27
|
Louie A, Bhandula V, Portnoy DA. Secretion of c-di-AMP by Listeria monocytogenes Leads to a STING-Dependent Antibacterial Response during Enterocolitis. Infect Immun 2020; 88:e00407-20. [PMID: 33020211 PMCID: PMC7671888 DOI: 10.1128/iai.00407-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/27/2020] [Indexed: 12/24/2022] Open
Abstract
Stimulator of interferon genes (STING) acts as a cytoplasmic signaling hub of innate immunity that is activated by host-derived or bacterially derived cyclic dinucleotides. Listeria monocytogenes is a foodborne, facultative intracellular pathogen that secretes c-di-AMP and activates STING, yet the in vivo role of the STING pathway during bacterial pathogenesis remains unclear. In this study, we found that STING-deficient mice had increased weight loss and roughly 10-fold-increased systemic bacterial burden during L. monocytogenes-induced enterocolitis. Infection with a L. monocytogenes mutant impaired in c-di-AMP secretion failed to elicit a protective response, whereas a mutant with increased c-di-AMP secretion triggered enhanced protection. Type I interferon (IFN) is a major output of STING signaling; however, disrupting IFN signaling during L. monocytogenes-induced enterocolitis did not recapitulate STING deficiency. In the absence of STING, the intestinal immune response was associated with a reduced influx of inflammatory monocytes. These studies suggest that in barrier sites such as the intestinal tract, where pathogen-associated molecular patterns are abundant, cytosolic surveillance systems such as STING are well positioned to detect pathogenic bacteria.
Collapse
Affiliation(s)
- Alexander Louie
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Varaang Bhandula
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Daniel A Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
28
|
Kumar V. Toll-like receptors in sepsis-associated cytokine storm and their endogenous negative regulators as future immunomodulatory targets. Int Immunopharmacol 2020; 89:107087. [PMID: 33075714 PMCID: PMC7550173 DOI: 10.1016/j.intimp.2020.107087] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/04/2020] [Accepted: 10/08/2020] [Indexed: 12/15/2022]
Abstract
Sepsis infects more than 48.9 million people world-wide, with 19.7 million deaths. Cytokine storm plays a significant role in sepsis, along with severe COVID-19. TLR signaling pathways plays a crucial role in generating the cytokine storm. Endogenous negative regulators of TLR signaling are crucial to regulate cytokine storm.
Cytokine storm generates during various systemic acute infections, including sepsis and current pandemic called COVID-19 (severe) causing devastating inflammatory conditions, which include multi-organ failure or multi-organ dysfunction syndrome (MODS) and death of the patient. Toll-like receptors (TLRs) are one of the major pattern recognition receptors (PRRs) expressed by immune cells as well as non-immune cells, including neurons, which play a crucial role in generating cytokine storm. They recognize microbial-associated molecular patterns (MAMPs, expressed by pathogens) and damage or death-associate molecular patterns (DAMPs; released and/expressed by damaged/killed host cells). Upon recognition of MAMPs and DAMPs, TLRs activate downstream signaling pathways releasing several pro-inflammatory mediators [cytokines, chemokines, interferons, and reactive oxygen and nitrogen species (ROS or RNS)], which cause acute inflammation meant to control the pathogen and repair the damage. Induction of an exaggerated response due to genetic makeup of the host and/or persistence of the pathogen due to its evasion mechanisms may lead to severe systemic inflammatory condition called sepsis in response to the generation of cytokine storm and organ dysfunction. The activation of TLR-induced inflammatory response is hardwired to the induction of several negative feedback mechanisms that come into play to conclude the response and maintain immune homeostasis. This state-of-the-art review describes the importance of TLR signaling in the onset of the sepsis-associated cytokine storm and discusses various host-derived endogenous negative regulators of TLR signaling pathways. The subject is very important as there is a vast array of genes and processes implicated in these negative feedback mechanisms. These molecules and mechanisms can be targeted for developing novel therapeutic drugs for cytokine storm-associated diseases, including sepsis, severe COVID-19, and other inflammatory diseases, where TLR-signaling plays a significant role.
Collapse
Affiliation(s)
- V Kumar
- Children Health Clinical Unit, Faculty of Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia.
| |
Collapse
|