1
|
Yu MC, Li XL, Ning ML, Yan ZZ, Yu WT. USP22 inhibits microglial M1 polarization by regulating the PU.1/NLRP3 inflammasome pathway. Brain Res Bull 2024; 220:111157. [PMID: 39631712 DOI: 10.1016/j.brainresbull.2024.111157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/10/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE This study aimed to investigate the effect of Ubiquitin-Specific Peptidase 22 (USP22) on the inflammatory response mediated by BV-2 mouse microglia and explore the role of the PU box binding protein 1 (PU.1)/NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome in the USP22-induced polarization of BV-2 cells. METHODS The BV-2 mouse microglia line was cultured in vitro, and plasmid and siRNA transfection was performed to overexpress or knockdown USP22. Subsequently, BV-2 cells were treated with lipopolysaccharide (LPS) and interferon-gamma (IFN-γ) and interleukin (IL)-4 to induce M1 and M2 polarization, respectively. Western blot was used to detect the expression levels of USP22, PU.1, M1 polarization markers [inducible nitric oxide synthase (iNOS), and cluster of differentiation (CD) 86], M2 polarization markers [arginase 1 (Arg1), and CD206], in BV-2 cells from different treatment groups. Additionally, measurement was performed on the inflammasome NLRP3, and its activation-related proteins [NIMA-related kinase7 (NEK7), cleaved-caspase 1, apoptosis-associated speck-like protein containing a CARD (ASC)]. Enzyme-linked immunosorbent (ELISA) assay was employed to determine the levels of inflammatory cytokines tumor necrosis factor-alpha (TNF-α), IL-1 β, and IL-10 in the cells. Furthermore, immunofluorescence was utilized to analyze the levels of iNOS and Arg1-positive BV-2 cells in different treatment groups. Moreover, the ubiquitination level of PU.1 was detected using immunoprecipitation. RESULTS The protein expression level of USP22 was significantly down-regulated in BV-2 cells treated with M1 polarization. Overexpression of USP22 remarkably reduced the protein levels of iNOS and CD86, but markedly increased the protein levels of Arg1 and CD206 in cells. Besides, there was a notable reduction in the levels of TNF-α and IL-1 β in the cell culture medium, while a remarkable increase was observed in the level of IL-10. Additionally, the level of iNOS-positive cells was significantly decreased, while the level of Arg1-positive cells was considerably increased. However, up-regulation of PU.1 expression could reverse the above results and promoted the expression of NLRP3 and its activation-related proteins. Notably, overexpression of USP22 significantly down-regulated the protein expression and ubiquitination level of PU.1. CONCLUSION USP22 inhibits the M1 polarization of BV-2 mouse microglia. The PU.1/NLRP3 inflammasome pathway may be a critical regulatory pathway of USP22 in BV-2 cell polarization.
Collapse
Affiliation(s)
- Ming-Chen Yu
- Department of Orthopedics, Changzhou Geriatric hospital affiliated to Soochow University, Changzhou 213000, China; Department of Orthopedics, Changzhou NO. 7 People's Hospital, Changzhou 213000, China
| | - Xiao-Lin Li
- Department of Orthopedics, Changzhou Geriatric hospital affiliated to Soochow University, Changzhou 213000, China; Department of Orthopedics, Changzhou NO. 7 People's Hospital, Changzhou 213000, China
| | - Ming-Liang Ning
- Department of Orthopedics, Changzhou Geriatric hospital affiliated to Soochow University, Changzhou 213000, China; Department of Orthopedics, Changzhou NO. 7 People's Hospital, Changzhou 213000, China
| | - Zhen-Zhong Yan
- Department of Orthopedics, Changzhou Geriatric hospital affiliated to Soochow University, Changzhou 213000, China; Department of Orthopedics, Changzhou NO. 7 People's Hospital, Changzhou 213000, China
| | - Wan-Tao Yu
- Department Of Orthopdics, The First People's Hospital of Changzhou, Changzhou 213000, China.
| |
Collapse
|
2
|
Rodríguez-Ubreva J, Calafell-Segura J, Calvillo CL, Keller B, Ciudad L, Handfield LF, de la Calle-Fabregat C, Godoy-Tena G, Andrés-León E, Hoo R, Porter T, Prigmore E, Hofmann M, Decker A, Martín J, Vento-Tormo R, Warnatz K, Ballestar E. COVID-19 progression and convalescence in common variable immunodeficiency patients show dysregulated adaptive immune responses and persistent type I interferon and inflammasome activation. Nat Commun 2024; 15:10344. [PMID: 39609471 PMCID: PMC11605083 DOI: 10.1038/s41467-024-54732-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/20/2024] [Indexed: 11/30/2024] Open
Abstract
Common variable immunodeficiency (CVID) is the most prevalent primary immunodeficiency, marked by hypogammaglobulinemia, poor antibody responses, and increased infection susceptibility. The COVID-19 pandemic provided a unique opportunity to study the effects of prolonged viral infections on the immune responses of CVID patients. Here we use single-cell RNA-seq and spectral flow cytometry of peripheral blood samples before, during, and after SARS-CoV-2 infection showing that COVID-19 CVID patients display a persistent type I interferon signature at convalescence across immune compartments. Alterations in adaptive immunity include sustained activation of naïve B cells, increased CD21low B cells, impaired Th1 polarization, CD4+ T central memory exhaustion, and increased CD8+ T cell cytotoxicity. NK cell differentiation is defective, although cytotoxicity remains intact. Monocytes show persistent activation of inflammasome-related genes. These findings suggest the involvement of intact humoral immunity in regulating these processes and might indicate the need for early intervention to manage viral infections in CVID patients.
Collapse
Affiliation(s)
- Javier Rodríguez-Ubreva
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain.
| | - Josep Calafell-Segura
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain
| | - Celia L Calvillo
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain
| | - Baerbel Keller
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laura Ciudad
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain
| | | | - Carlos de la Calle-Fabregat
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain
| | - Gerard Godoy-Tena
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain
| | - Eduardo Andrés-León
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Granada, Spain
| | - Regina Hoo
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Tarryn Porter
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Elena Prigmore
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Maike Hofmann
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Annegrit Decker
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Javier Martín
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Granada, Spain
| | | | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain.
- Epigenetics in Inflammatory and Metabolic Diseases Laboratory, Health Science Center (HSC), East China Normal University (ECNU), Shanghai, China.
| |
Collapse
|
3
|
Martinez JC, Morandini F, Fitzgibbons L, Sieczkiewicz N, Bae SJ, Meadow ME, Hillpot E, Cutting J, Paige V, Biashad SA, Simon M, Sedivy J, Seluanov A, Gorbunova V. cGAS deficient mice display premature aging associated with de-repression of LINE1 elements and inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617645. [PMID: 39416083 PMCID: PMC11482887 DOI: 10.1101/2024.10.10.617645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Aging-associated inflammation, or 'inflammaging" is a driver of multiple age-associated diseases. Cyclic GMP-AMP Synthase (cGAS) is a cytosolic DNA sensor that functions to activate interferon response upon detecting viral DNA in the cytoplasm. cGAS contributes to inflammaging by responding to endogenous signals such as damaged DNA or LINE1 (L1) cDNA which forms in aged cells. While cGAS knockout mice are viable their aging has not been examined. Unexpectedly, we found that cGAS knockout mice exhibit accelerated aging phenotype associated with induction of inflammation. Transcription of L1 elements was increased in both cGAS knockout mice and in cGAS siRNA knockdown cells associated with high levels of cytoplasmic L1 DNA and expression of ORF1 protein. Cells from cGAS knockout mice showed increased chromatin accessibility and decreased DNA methylation on L1 transposons. Stimulated emission depletion microscopy (STED) showed that cGAS forms nuclear condensates that co-localize with H3K9me3 heterochromatin marks, and H3K9me3 pattern is disrupted in cGAS knockout cells. Taken together these results suggest a previously undescribed role for cGAS in maintaining heterochromatin on transposable elements. We propose that loss of cGAS leads to loss of chromatin organization, de-repression of transposable elements and induction of inflammation resulting in accelerated aging.
Collapse
Affiliation(s)
- John C Martinez
- Translational Biomedical Sciences Program, University of Rochester, NY, 14627, USA
- Department of Biology, University of Rochester, NY, 14627, USA
| | | | | | | | - Sung Jae Bae
- Department of Biology, University of Rochester, NY, 14627, USA
| | | | - Eric Hillpot
- Department of Biology, University of Rochester, NY, 14627, USA
| | - Joseph Cutting
- Department of Biology, University of Rochester, NY, 14627, USA
| | - Victoria Paige
- Department of Biology, University of Rochester, NY, 14627, USA
| | | | - Matthew Simon
- Department of Biology, University of Rochester, NY, 14627, USA
| | - John Sedivy
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, RI, 02912, USA
| | - Andrei Seluanov
- Department of Biology, University of Rochester, NY, 14627, USA
- Department of Medicine, University of Rochester, NY, 14627, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, NY, 14627, USA
- Department of Medicine, University of Rochester, NY, 14627, USA
| |
Collapse
|
4
|
Kulakova K, Lawal TR, Mccarthy E, Floudas A. The Contribution of Macrophage Plasticity to Inflammatory Arthritis and Their Potential as Therapeutic Targets. Cells 2024; 13:1586. [PMID: 39329767 PMCID: PMC11430612 DOI: 10.3390/cells13181586] [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: 07/19/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Inflammatory arthritis are common chronic inflammatory autoimmune diseases characterised by progressive, destructive inflammation of the joints leading to a loss of function and significant comorbidities; importantly, there are no cures and only 20% of patients achieve drug-free remission for over 2 years. Macrophages play a vital role in maintaining homeostasis, however, under the wrong environmental cues, become drivers of chronic synovial inflammation. Based on the current "dogma", M1 macrophages secrete pro-inflammatory cytokines and chemokines, promoting tissue degradation and joint and bone erosion which over time lead to accelerated disease progression. On the other hand, M2 macrophages secrete anti-inflammatory mediators associated with wound healing, tissue remodelling and the resolution of inflammation. Currently, four subtypes of M2 macrophages have been identified, namely M2a, M2b, M2c and M2d. However, more subtypes may exist due to macrophage plasticity and the ability for repolarisation. Macrophages are highly plastic, and polarisation exists as a continuum with diverse intermediate phenotypes. This plasticity is achieved by a highly amenable epigenome in response to environmental stimuli and shifts in metabolism. Initiating treatment during the early stages of disease is important for improved prognosis and patient outcomes. Currently, no treatment targeting macrophages specifically is available. Such therapeutics are being investigated in ongoing clinical trials. The repolarisation of pro-inflammatory macrophages towards the anti-inflammatory phenotype has been proposed as an effective approach in targeting the M1/M2 imbalance, and in turn is a potential therapeutic strategy for IA diseases. Therefore, elucidating the mechanisms that govern macrophage plasticity is fundamental for the success of novel macrophage targeting therapeutics.
Collapse
Affiliation(s)
- Karina Kulakova
- School of Biotechnology, Dublin City University, D09 V209 Dublin, Ireland; (K.K.)
- Life Sciences Institute, Dublin City University, D09 V209 Dublin, Ireland
| | - Tope Remilekun Lawal
- School of Biotechnology, Dublin City University, D09 V209 Dublin, Ireland; (K.K.)
| | - Eoghan Mccarthy
- Department of Rheumatology, Beaumont Hospital, D09 V2N0 Dublin, Ireland
- Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Achilleas Floudas
- School of Biotechnology, Dublin City University, D09 V209 Dublin, Ireland; (K.K.)
- Life Sciences Institute, Dublin City University, D09 V209 Dublin, Ireland
- Medical School, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
5
|
Li N, Steiger S, Zhong M, Lu M, Lei Y, Tang C, Chen J, Guo Y, Li J, Zhang D, Li J, Zhu E, Zheng Z, Lichtnekert J, Chen Y, Wang X. IRF8 maintains mononuclear phagocyte and neutrophil function in acute kidney injury. Heliyon 2024; 10:e31818. [PMID: 38845872 PMCID: PMC11153194 DOI: 10.1016/j.heliyon.2024.e31818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
Immune cells are key players in acute tissue injury and inflammation, including acute kidney injury (AKI). Their development, differentiation, activation status, and functions are mediated by a variety of transcription factors, such as interferon regulatory factor 8 (IRF8) and IRF4. We speculated that IRF8 has a pathophysiologic impact on renal immune cells in AKI and found that IRF8 is highly expressed in blood type 1 conventional dendritic cells (cDC1s), monocytes, monocyte-derived dendritic cells (moDCs) and kidney biopsies from patients with AKI. In a mouse model of ischemia‒reperfusion injury (IRI)-induced AKI, Irf8 -/- mice displayed increased tubular cell necrosis and worsened kidney dysfunction associated with the recruitment of a substantial amount of monocytes and neutrophils but defective renal infiltration of cDC1s and moDCs. Mechanistically, global Irf8 deficiency impaired moDC and cDC1 maturation and activation, as well as cDC1 proliferation, antigen uptake, and trafficking to lymphoid organs for T-cell priming in ischemic AKI. Moreover, compared with Irf8 +/+ mice, Irf8 -/- mice exhibited increased neutrophil recruitment and neutrophil extracellular trap (NET) formation following AKI. IRF8 primarily regulates cDC1 and indirectly neutrophil functions, and thereby protects mice from kidney injury and inflammation following IRI. Our results demonstrate that IRF8 plays a predominant immunoregulatory role in cDC1 function and therefore represents a potential therapeutic target in AKI.
Collapse
Affiliation(s)
- Na Li
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Stefanie Steiger
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Ludwig-Maximilian-University Munich, 80336, Munich, Bavaria, Germany
| | - Ming Zhong
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Meihua Lu
- Department of Geriatrics, People's Hospital of Banan District, 401320, Chongqing, China
| | - Yan Lei
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Chun Tang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Jiasi Chen
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Yao Guo
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Jinhong Li
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Dengyang Zhang
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Jingyi Li
- Department of Pulmonary & Critical Care Medicine, Shenzhen Hospital of Southern Medical University, 518107, Shenzhen, China
| | - Enyi Zhu
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Zhihua Zheng
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Julia Lichtnekert
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Ludwig-Maximilian-University Munich, 80336, Munich, Bavaria, Germany
| | - Yun Chen
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Xiaohua Wang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| |
Collapse
|
6
|
Koller BH, Nguyen M, Snouwaert JN, Gabel CA, Ting JPY. Species-specific NLRP3 regulation and its role in CNS autoinflammatory diseases. Cell Rep 2024; 43:113852. [PMID: 38427558 DOI: 10.1016/j.celrep.2024.113852] [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: 09/27/2023] [Revised: 01/16/2024] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
The NLRP3 inflammasome is essential for caspase-1 activation and the release of interleukin (IL)-1β, IL-18, and gasdermin-D in myeloid cells. However, research on species-specific NLRP3's physiological impact is limited. We engineer mice with the human NLRP3 gene, driven by either the human or mouse promoter, via syntenic replacement at the mouse Nlrp3 locus. Both promoters facilitate hNLRP3 expression in myeloid cells, but the mouse promoter responds more robustly to LPS. Investigating the disease impact of differential NLRP3 regulation, we introduce the D305N gain-of-function mutation into both humanized lines. Chronic inflammation is evident with both promoters; however, CNS outcomes vary significantly. Despite poor response to LPS, the human promoter results in D305N-associated aseptic meningitis, mirroring human pathology. The mouse promoter, although leading to increased CNS expression post-LPS, does not induce meningitis in D305N mutants. Therefore, human-like NLRP3 expression may be crucial for accurate modeling of its role in disease pathogenesis.
Collapse
Affiliation(s)
- Beverly H Koller
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - MyTrang Nguyen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John N Snouwaert
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Jenny P-Y Ting
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
7
|
Liu W, Tian X, Gu L, Yu B, Wang Z, Chi M, Lin J, Wang Q, Liu G, Zhao G, Cui Li. Oxymatrine mitigates Aspergillus fumigatus keratitis by suppressing fungal activity and restricting pyroptosis. Exp Eye Res 2024; 240:109830. [PMID: 38364932 DOI: 10.1016/j.exer.2024.109830] [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: 01/09/2024] [Revised: 02/04/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
Fungal keratitis (FK) is a refractory keratitis caused by excessive inflammation and fungal damage. Excessive inflammation can lead to tissue damage and corneal opacity, resulting in a poor prognosis for FK. Oxymatrine (OMT) is a natural alkaloid, which has rich pharmacological effects, such as antioxidant and anti-inflammation. However, its antifungal activity and the mechanism of action in FK have not been elucidated. This study confirmed that OMT suppressed Aspergillus fumigatus growth, biofilm formation, the integrity of fungal cell and conidial adherence. OMT not only effectively reduced corneal fungal load but also inflammation responses. OMT lessened the recruitment of neutrophils and macrophages in FK. In addition, OMT up-regulated the expression of Nrf2 and down-regulated the expression of IL-18, IL-1β, caspase-1, NLRP3 and GSDMD. Pre-treatment with Nrf2 inhibitor up-regulated the expression of IL-1β, IL-18, caspase-1, NLRP3 and GSDMD supressed by OMT. In conclusion, OMT has efficient anti-inflammatory and antifungal effects by suppressing fungal activity and restricting pyroptosis via Nrf2 pathway. OMT is considered as a potential option for the treatment of FK.
Collapse
Affiliation(s)
- Weichen Liu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xue Tian
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Lingwen Gu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Bing Yu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Ziyi Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Menghui Chi
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Guibo Liu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China.
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China.
| |
Collapse
|
8
|
Liu Y, He Y, Cao J, Lu H, Zou R, Zuo Z, Li R, Zhang Y, Sun J. Correlative analysis of transcriptome and proteome in Penaeus vannamei reveals key signaling pathways are involved in IFN-like antiviral regulation mediated by interferon regulatory factor (PvIRF). Int J Biol Macromol 2023; 253:127138. [PMID: 37776923 DOI: 10.1016/j.ijbiomac.2023.127138] [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: 07/01/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Interferon regulatory factors (IRFs) are crucial transcription factors that regulate interferon (IFN) induction in response to pathogen invasion. The regulatory mechanism of IRF has been well studied in vertebrates, but little has been known in arthropods. Therefore, in order to obtain new insights into the potential molecular mechanism of Peneaus vannamei IRF (PvIRF) in response to viral infection, comprehensive comparative analysis of the transcriptome and proteome profiles in shrimp infected with WSSV after knocking down PvIRF was conducted by using RNA sequencing (RNA-seq) and isobaric tags for relative and absolute quantification (iTRAQ). The sequence characterization, molecular functional evolution and 3D spatial structure of PvIRF were analyzed by using bioinformatics methods. PvIRF share the higher homology with different species in N-terminal end (containing DNA binding domain (DBD) including DNA sequence recognition sites and metal binding site) than that in C-terminal end. Within 4 IRF subfamilies of vertebrates, PvIRF had closer relationship with IRF1 subfamily. The DBD of PvIRF and C. gigas IRF1a were composed of α-helices and β-folds which was similar with the DBD structure of M. musculus IRF2. Interestingly, different from the five Tryptophan repeats highly homologous in the DBD of vertebrate IRF, the first and fifth tryptophans of PvIRF mutate to Phenylalanine and Leucine respectively, while the mutations were conserved among shrimp IRFs. RNAi knockdown of PvIRF gene by double-strand RNA could obviously promote the in vivo propagation of WSSV in shrimp and increase the mortality of WSSV-infected shrimp. It suggested that PvIRF was involved in inhibiting the replication of WSSV in shrimp. A total of 8787 transcripts and 2846 proteins were identified with significantly differential abundances in WSSV-infected shrimp after PvIRF knockdown, among which several immune-related members were identified and categorized into 10 groups according to their possible functions. Furthermore, the variation of expression profile from members of key signaling pathways involving JAK/STAT and Toll signaling pathway implied that they might participate IRF-mediated IFN-like regulation in shrimp. Correlative analyses indicated that 722 differentially expressed proteins (DEPs) shared the same expression profiles with their corresponding transcripts, including recognition-related proteins (CTLs and ITGs), chitin-binding proteins (peritrophin), and effectors (ALFs and SWD), while 401 DEPs with the opposite expression profiles across the two levels emphasized the critical role of post-transcriptional and post-translational modification. The results provide candidate signaling pathway including pivotal genes and proteins involved in the regulatory mechanism of interferon mediated by IRF on shrimp antiviral response. This is the first report in crustacean to explore the IFN-like antiviral regulation pathway mediated by IRF on the basis of transcriptome and proteomics correlative analysis, and will provide new ideas for further research on innate immune and defense mechanisms of crustacean.
Collapse
Affiliation(s)
- Yichen Liu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Yuxin He
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Jinlai Cao
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Hangjia Lu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Ruifeng Zou
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Zhihan Zuo
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Ran Li
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Yichen Zhang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Jinsheng Sun
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China.
| |
Collapse
|
9
|
Pouyabahar D, Chung SW, Pezzutti OI, Perciani CT, Wang X, Ma XZ, Jiang C, Camat D, Chung T, Sekhon M, Manuel J, Chen XC, McGilvray ID, MacParland SA, Bader GD. A rat liver cell atlas reveals intrahepatic myeloid heterogeneity. iScience 2023; 26:108213. [PMID: 38026201 PMCID: PMC10651689 DOI: 10.1016/j.isci.2023.108213] [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: 12/06/2022] [Revised: 08/20/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
The large size and vascular accessibility of the laboratory rat (Rattus norvegicus) make it an ideal hepatic animal model for diseases that require surgical manipulation. Often, the disease susceptibility and outcomes of inflammatory pathologies vary significantly between strains. This study uses single-cell transcriptomics to better understand the complex cellular network of the rat liver, as well as to unravel the cellular and molecular sources of inter-strain hepatic variation. We generated single-cell and single-nucleus transcriptomic maps of the livers of healthy Dark Agouti and Lewis rat strains and developed a factor analysis-based bioinformatics analysis pipeline to study data covariates, such as strain and batch. Using this approach, we discovered transcriptomic variation within the hepatocyte and myeloid populations that underlie distinct cell states between rat strains. This finding will help provide a reference for future investigations on strain-dependent outcomes of surgical experiment models.
Collapse
Affiliation(s)
- Delaram Pouyabahar
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Sai W. Chung
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Olivia I. Pezzutti
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Catia T. Perciani
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Xinle Wang
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Xue-Zhong Ma
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Chao Jiang
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Damra Camat
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Trevor Chung
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Manmeet Sekhon
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Justin Manuel
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Xu-Chun Chen
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Ian D. McGilvray
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Sonya A. MacParland
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gary D. Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
- Princess Margaret Research Institute, University Health Network, Toronto, ON, Canada
- The Multiscale Human Program, Canadian Institute for Advanced Research, Toronto, ON, Canada
| |
Collapse
|
10
|
Vancolen S, Sébire G, Robaire B. Influence of androgens on the innate immune system. Andrology 2023; 11:1237-1244. [PMID: 36840517 DOI: 10.1111/andr.13416] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/15/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023]
Abstract
BACKGROUND Sexual dimorphism is observed in the occurrence, course, and severity of human disease. The difference in immune response between males and females can in part be attributed to sexual genotype. However, immunological differences can also be explained by endocrine-immune interactions. Specifically, androgens possess the ability of directly modulating the development and function of immune cells. Although androgens generally contribute to immunosuppressive effects, this is not necessarily always the case. AIM The aim of the review is to uncover the role of androgens in shaping the innate immune response. MATERIAL & METHODS Authors included papers in this review which discussed the impact of androgens on specific innate immune cells. RESULTS Androgens modulate the innate immune response through various mechanisms. However, there is conflicting evidence in the literature regarding the interplay betwen androgens and the innate immune system. DISCUSSION Conflicting evidence presented in this review could in part be explained by the limitations present in interpreting results. CONCLUSION This review is of great importance for our understanding of occurence and mechanism of human inflammatory disease.
Collapse
Affiliation(s)
- Seline Vancolen
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Pediatrics and Department of Neurology, McGill University, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Guillaume Sébire
- Department of Pediatrics and Department of Neurology, McGill University, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Bernard Robaire
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Zong Q, Qu H, Zheng X, Wang H, Wu S, Yin Z, Bao W. Hypomethylated interferon regulatory factor 8 recruits activating protein-2α to attenuate porcine epidemic diarrhea virus infection in porcine jejunum. Front Immunol 2023; 14:1187144. [PMID: 37593742 PMCID: PMC10427914 DOI: 10.3389/fimmu.2023.1187144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/12/2023] [Indexed: 08/19/2023] Open
Abstract
Interferon regulatory factor 8 (IRF8) is a key regulator of innate immune receptor signaling that resists pathogen invasion by regulating cell growth and differentiation. Porcine epidemic diarrhea virus (PEDV) targets the intestine and damages the mucosal barrier. However, whether IRF8 regulates PEDV replication remains unclear. We revealed that PEDV infection activated IRF8 expression. Moreover, IRF8 deletion drastically promoted PEDV replication and invasion, increasing the virus copies and titers. Hypomethylation enrichment of activating protein (AP)-2α was significantly negatively correlated with high IRF8 expression, and AP-2α directly targeted the IRF8 promoter to regulate PEDV replication. Furthermore, IRF8 overexpression decreased the cellular reactive oxygen species levels and mitochondrial membrane potential and increased the antioxidant enzyme activities to alleviate PEDV-induced oxidative damage. IRF8 overexpression suppressed apoptotic gene expression, thereby inhibiting apoptosis in response to PEDV stimulation. Taken together, this study demonstrates that AP-2α is involved in PEDV-induced epigenetic modification of IRF8 to reduce cell apoptosis and oxidative stress and facilitate host resistance to PEDV in the intestinal epithelium.
Collapse
Affiliation(s)
- Qiufang Zong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Huan Qu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xianrui Zheng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Haifei Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Shenglong Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
12
|
Tan RZ, Zhong X, Han RY, Xie KH, Jia J, Yang Y, Cheng M, Yang CY, Lan HY, Wang L. Macrophages mediate psoriasis via Mincle-dependent mechanism in mice. Cell Death Discov 2023; 9:140. [PMID: 37117184 PMCID: PMC10147944 DOI: 10.1038/s41420-023-01444-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023] Open
Abstract
Psoriasis is currently considered to be an immune and inflammatory disease characterized by massive immune cells infiltration including macrophages. It has been reported that macrophage-inducible C-type lectin (Mincle) is essential to maintain the pro-inflammatory phenotype of M1 macrophages, however, its role and mechanisms in psoriasis remain largely unknown. A model of psoriasis was induced in mice by a daily topical application of imiquimod for 7 days. Role and mechanisms of Mincle in macrophage-mediated psoriasis were investigated in clodronate liposomes induced macrophage depletion mice followed by adoptively transferring with Mincle-expressing or -knockout (KO) macrophages, and in macrophage specific Mincle knockout mice (Mincleloxp/loxp/Lyz2-cre+/+). Finally, a Mincle neutralizing antibody was employed to the psoriasis mice to reveal the therapeutic potential for psoriasis by targeting Mincle. Mincle was highly expressed by M1 macrophages in the skin lesions of patients and mice with psoriasis. Clodronate liposomes-induced macrophage depletion inhibited psoriasis in mice, which was restored by adoptive transfer with Mincle-expressing macrophages but not by Mincle-KO macrophages. This was further confirmed in macrophage-specific Mincle-KO mice. Mechanistically, macrophages mediated psoriasis via the Mincle-Syk-NF-κB pathway as blocking macrophage Mincle inhibited Syk/NF-κB-driven skin lesions and epidermal injury in vivo and in vitro. We also found that LPS induced Mincle expression by M1 macrophages via the PU.1-dependent mechanism. Most importantly, we revealed that targeting Mincle with a neutralizing antibody significantly improved psoriasis in mice. In summary, our findings demonstrated that macrophages mediate psoriasis in mice via the Mincle-dependent mechanism, targeting Mincle may represent as a novel therapy for psoriasis. A simplified pathway model of Mincle in macrophage-mediated psoriasis.
Collapse
Affiliation(s)
- Rui-Zhi Tan
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Xia Zhong
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Rang-Yue Han
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Ke-Huan Xie
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jian Jia
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Ye Yang
- Department of Orthopaedics, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Mei Cheng
- Dermatological Department, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Chun-Yan Yang
- Dermatological Department, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Li Wang
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China.
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China.
| |
Collapse
|
13
|
Kloc M, Kubiak JZ, Zdanowski R, Ghobrial RM. Memory Macrophages. Int J Mol Sci 2022; 24:ijms24010038. [PMID: 36613481 PMCID: PMC9819859 DOI: 10.3390/ijms24010038] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/12/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Immunological memory is a crucial part of the immune defense that allows organisms to respond against previously encountered pathogens or other harmful factors. Immunological memory is based on the establishment of epigenetic modifications of the genome. The ability to memorize encounters with pathogens and other harmful factors and mount enhanced defense upon subsequent encounters is an evolutionarily ancient mechanism operating in all animals and plants. However, the term immunological memory is usually restricted to the organisms (invertebrates and vertebrates) possessing the immune system. The mammalian immune system, with innate and adaptive branches, is the most sophisticated among vertebrates. The concept of innate memory and memory macrophages is relatively new and thus understudied. We introduce the concept of immunological memory and describe types of memory in different species and their evolutionary status. We discuss why the traditional view of innate immune cells as the first-line defenders is too restrictive and how the innate immune cells can accumulate and retain immunologic memory. We describe how the initial priming leads to chromatin remodeling and epigenetic changes, which allow memory macrophage formation. We also summarize what is currently known about the mechanisms underlying development of memory macrophages; their molecular and metabolic signature and surface markers; and how they may contribute to immune defense, diseases, and organ transplantation.
Collapse
Affiliation(s)
- Malgorzata Kloc
- The Houston Methodist Research Institute, Transplant Immunology, Houston, TX 77030, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Genetics, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
- Correspondence:
| | - Jacek Z. Kubiak
- Dynamics and Mechanics of Epithelia Group, Faculty of Medicine, Institute of Genetics and Development of Rennes, University of Rennes, CNRS, UMR 6290, 35043 Rennes, France
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine National Research Institute, Szaserow 128, 04-141 Warsaw, Poland
| | - Robert Zdanowski
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine National Research Institute, Szaserow 128, 04-141 Warsaw, Poland
| | - Rafik M. Ghobrial
- The Houston Methodist Research Institute, Transplant Immunology, Houston, TX 77030, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
14
|
Greenwood DL, Ramsey HE, Nguyen PTT, Patterson AR, Voss K, Bader JE, Sugiura A, Bacigalupa ZA, Schaefer S, Ye X, Dahunsi DO, Madden MZ, Wellen KE, Savona MR, Ferrell PB, Rathmell JC. Acly Deficiency Enhances Myelopoiesis through Acetyl Coenzyme A and Metabolic-Epigenetic Cross-Talk. Immunohorizons 2022; 6:837-850. [PMID: 36547387 PMCID: PMC9935084 DOI: 10.4049/immunohorizons.2200086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
Hematopoiesis integrates cytokine signaling, metabolism, and epigenetic modifications to regulate blood cell generation. These processes are linked, as metabolites provide essential substrates for epigenetic marks. In this study, we demonstrate that ATP citrate lyase (Acly), which metabolizes citrate to generate cytosolic acetyl-CoA and is of clinical interest, can regulate chromatin accessibility to limit myeloid differentiation. Acly was tested for a role in murine hematopoiesis by small-molecule inhibition or genetic deletion in lineage-depleted, c-Kit-enriched hematopoietic stem and progenitor cells from Mus musculus. Treatments increased the abundance of cell populations that expressed the myeloid integrin CD11b and other markers of myeloid differentiation. When single-cell RNA sequencing was performed, we found that Acly inhibitor-treated hematopoietic stem and progenitor cells exhibited greater gene expression signatures for macrophages and enrichment of these populations. Similarly, the single-cell assay for transposase-accessible chromatin sequencing showed increased chromatin accessibility at genes associated with myeloid differentiation, including CD11b, CD11c, and IRF8. Mechanistically, Acly deficiency altered chromatin accessibility and expression of multiple C/EBP family transcription factors known to regulate myeloid differentiation and cell metabolism, with increased Cebpe and decreased Cebpa and Cebpb. This effect of Acly deficiency was accompanied by altered mitochondrial metabolism with decreased mitochondrial polarization but increased mitochondrial content and production of reactive oxygen species. The bias to myeloid differentiation appeared due to insufficient generation of acetyl-CoA, as exogenous acetate to support alternate compensatory pathways to produce acetyl-CoA reversed this phenotype. Acly inhibition thus can promote myelopoiesis through deprivation of acetyl-CoA and altered histone acetylome to regulate C/EBP transcription factor family activity for myeloid differentiation.
Collapse
Affiliation(s)
- Dalton L. Greenwood
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Haley E. Ramsey
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Phuong T. T. Nguyen
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Andrew R. Patterson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Kelsey Voss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Jackie E. Bader
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Ayaka Sugiura
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | | | - Samuel Schaefer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Xiang Ye
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Debolanle O. Dahunsi
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Matthew Z. Madden
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Kathryn E. Wellen
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael R. Savona
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN
| | - P. Brent Ferrell
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
15
|
Li X, Luo S, Chen X, Li S, Hao L, Yang D. Adipose-derived stem cells attenuate acne-related inflammation via suppression of NLRP3 inflammasome. Stem Cell Res Ther 2022; 13:334. [PMID: 35871079 PMCID: PMC9308350 DOI: 10.1186/s13287-022-03007-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/02/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Acne is a chronic facial disease caused by Propionibacterium acnes, which proliferates within sebum-blocked skin follicles and increases inflammatory cytokine production. Several therapeutic drugs and products have been proposed to treat acne, yet no single treatment that ensures long-term treatment efficacy for all patients is available. Here, we explored the use of facial autologous fat transplant of adipose-derived stem cells (ADSCs) to dramatically reduce acne lesions. METHODS THP-1 cells were treated with active P. acnes for 24 h at different multiplicities of infection, and alterations in inflammatory factors were detected. To study the effect of THP-1 on inflammasome-related proteins, we first co-cultured ADSCs with THP-1 cells treated with P. acnes and evaluated the levels of these proteins in the supernatant. Further, an acne mouse model injected with ADSCs was used to assess inflammatory changes. RESULTS Propionibacterium acnes-mediated stimulation of THP-1 cells had a direct correlation with the expression of active caspase-1 and interleukin (IL)-1β in an infection-dependent manner. ADSCs significantly reduced the production of IL-1β induced by P. acnes stimulation through the reactive oxygen species (ROS)/Nod-like receptor family pyrin domain-containing 3 (NLRP3)/caspase-1 pathway. The results showed that ADSCs inhibit the skin inflammation induced by P. acnes by blocking the NLRP3 inflammasome via reducing the secretion of IL-1β in vivo. CONCLUSIONS Our findings suggest that ADSCs can alter IL-1β secretion by restricting the production of mitochondria ROS, thereby inhibiting the NLRP3/caspase-1 pathway in P. acnes-induced inflammatory responses. This study indicates that anti-acne therapy can potentially be developed by targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Xiaoxi Li
- The 1st Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Rd, NanGang Dist, Harbin, Heilongjiang, China
| | - Sai Luo
- The 1st Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Rd, NanGang Dist, Harbin, Heilongjiang, China
| | - Xinyao Chen
- The 1st Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Rd, NanGang Dist, Harbin, Heilongjiang, China
| | - Shasha Li
- The 1st Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Rd, NanGang Dist, Harbin, Heilongjiang, China
| | - Lijun Hao
- The 1st Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Rd, NanGang Dist, Harbin, Heilongjiang, China.
| | - Dan Yang
- Harbin Medical University, No. 157, BaoJian Rd, NanGang Dist, Harbin, Heilongjiang, China.
| |
Collapse
|
16
|
The Inflammasome NLRC4 Protects against Cryptococcus gattii by Inducing the Classic Caspase-1 to Activate the Pyroptosis Signal. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7355485. [PMID: 35340249 PMCID: PMC8942663 DOI: 10.1155/2022/7355485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022]
Abstract
Cryptococcus is one of the most pathogenic invasive fungi, and its interaction with the host's natural immunity, especially the role of the inflammasome family, has not been fully elucidated. As an important member of the inflammasome family, NOD-like receptor (NLR) family caspase recruitment domain (CARD) containing 4 (NLRC4) has been proven to protect lungs from damage from a variety of pathogens. In this study, we investigated the protective effect and mechanism of NLRC4 on cryptococcal pulmonary infection using NLRC4-/-mice in vivo and NLRC4-/-macrophages in vitro models stimulated by cryptococcal cells. We apply small animal fluorescence imaging to detect the fungal burden in the lungs and living body micro-CT scans of mice and in vitro tissue micro-CT scans to compare differences in infection foci nodules and histopathological lesions, and the activation of caspase-1 and downstream cytokines were detected by Western bolt and ELISA, etc. The results demonstrated that cryptococcal infection can activate the Nod-like receptors of caspase-1 activation and NLRC4 inflammasomes in macrophages and dendritic cells and affect downstream IL-1β and IL-18 release. After cryptococcal infection, the survival rate, lung fungal burden, and histopathological damage of NLRC4−/− mice were significantly impaired. NLRC4−/− macrophages showed a lower release of inflammatory factors, reactive oxygen species (ROS), and lactate dehydrogenase (LDH). Collectively, our results demonstrated that the activation of caspase-1 and downstream cytokines mediated by NLRC4 inflammasome in immune cells during Cryptococcus infection can enhance pyroptosis of macrophages, affect the phagocytic ability of macrophages, and inhibit the intracellular parasitism of cryptococcus, eventually reducing the burden of fungi.
Collapse
|