1
|
Raavi, Koehler AN, Vegas AJ. At The Interface: Small-Molecule Inhibitors of Soluble Cytokines. Chem Rev 2025; 125:4528-4568. [PMID: 40233276 DOI: 10.1021/acs.chemrev.4c00469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cytokines are crucial regulators of the immune system that orchestrate interactions between cells and, when dysregulated, contribute to the progression of chronic inflammation, cancer, and autoimmunity. Numerous biologic-based clinical agents, mostly monoclonal antibodies, have validated cytokines as important clinical targets and are now part of the standard of care for a number of diseases. These agents, while impactful, still suffer from limitations including a lack of oral bioavailability, high cost of production, and immunogenicity. Small-molecule cytokine inhibitors are attractive alternatives that can address these limitations. Although targeting cytokine-cytokine receptor complexes with small molecules has been a challenging research endeavor, multiple small-molecule inhibitors have now been identified, with a number of them undergoing clinical evaluation. In this review, we highlight the recent advancements in the discovery and development of small-molecule inhibitors targeting soluble cytokines. The strategies for identifying these novel ligands as well as the structural and mechanistic insights into their activity represent important milestones in tackling these challenging and clinically important protein-protein interactions.
Collapse
Affiliation(s)
- Raavi
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Angela N Koehler
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Arturo J Vegas
- Department of Chemistry, Boston University, Boston, Massachusetts 02115, United States
| |
Collapse
|
2
|
Kalailingam P, Ngan SC, Iyappan R, Nehchiri A, Mohd‐Kahliab K, Lee BST, Sharma B, Machan R, Bo ST, Chambers ES, Fajardo VA, Macpherson REK, Liu J, Klentrou P, Tsiani EL, Lim KL, Su IH, Gao Y, Richar AM, Kalaria RN, Chen CP, Balion C, de Kleijn D, McCarthy NE, Sze SK. Immunotherapeutic targeting of aging-associated isoDGR motif in chronic lung inflammation. Aging Cell 2025; 24:e14425. [PMID: 39757428 PMCID: PMC11984686 DOI: 10.1111/acel.14425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 08/18/2024] [Accepted: 11/04/2024] [Indexed: 01/07/2025] Open
Abstract
Accumulation of damaged biomolecules in body tissues is the primary cause of aging and age-related chronic diseases. Since this damage often occurs spontaneously, it has traditionally been regarded as untreatable, with typical therapeutic strategies targeting genes or enzymes being ineffective in this domain. In this report, we demonstrate that an antibody targeting the isoDGR damage motif in lung tissue can guide immune clearance of harmful damaged proteins in vivo, effectively reducing age-linked lung inflammation. We observed age-dependent accumulation of the isoDGR motif in human lung tissues, as well as an 8-fold increase in isoDGR-damaged proteins in lung fibrotic tissues compared with healthy tissue. This increase was accompanied by marked infiltration of CD68+/CD11b + macrophages, consistent with a role for isoDGR in promoting chronic inflammation. We therefore assessed isoDGR function in mice that were either naturally aged or lacked the isoDGR repair enzyme. IsoDGR-protein accumulation in mouse lung tissue was strongly correlated with chronic inflammation, pulmonary edema, and hypoxemia. This accumulation also induced mitochondrial and ribosomal dysfunction, in addition to features of cellular senescence, thereby contributing to progressive lung damage over time. Importantly, treatment with anti-isoDGR antibody was able to reduce these molecular features of disease and significantly reduced lung pathology in vivo.
Collapse
Affiliation(s)
- Pazhanichamy Kalailingam
- Center for Genomic MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - SoFong Cam Ngan
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Ranjith Iyappan
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Afra Nehchiri
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | | | | | - Bhargy Sharma
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - Radek Machan
- SCELSENanyang Technological UniversitySingaporeSingapore
| | - Sint Thida Bo
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - Emma S. Chambers
- Centre for Immunobiology, the Blizard Institute, Bart's and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Val A. Fajardo
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | | | - Jian Liu
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Panagiota Klentrou
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | | | - Kah Leong Lim
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - I. Hsin Su
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - Yong‐Gui Gao
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - A. Mark Richar
- Cardiovascular Research InstituteNational University Health SystemSingaporeSingapore
| | - Raj N. Kalaria
- Institute of Neuroscience, Campus for Ageing and VitalityNewcastle UniversityNewcastle upon TyneUK
| | - Christopher P. Chen
- Memory, Aging and Cognition CentreNational University Health SystemSingaporeSingapore
| | - Cynthia Balion
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonOntarioCanada
| | | | - Neil E. McCarthy
- Centre for Immunobiology, the Blizard Institute, Bart's and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Siu Kwan Sze
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| |
Collapse
|
3
|
Boonhok R, Senghoi W, Sangkanu S, Lim CL, Pudla M, Pereira MDL, Wilairatana P, Mahboob T, Rahman MA, Utaisincharoen P, Hiransai P, Nissapatorn V. Acanthamoeba castellanii-Mediated Reduction of Interleukin-1β Secretion and Its Association With Macrophage Autophagy. SCIENTIFICA 2025; 2025:3430892. [PMID: 40109888 PMCID: PMC11922611 DOI: 10.1155/sci5/3430892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Noncanonical autophagy including unconventional protein secretion has been extensively studied. Our work focused on a leaderless IL-1β protein secretion from human macrophage in response to Acanthamoeba castellanii components, Acanthamoeba culture supernatant (CS) and cell lysate (CL), as well as its association with macrophage autophagy. Phorbol 12-myristate 13-acetate (PMA)-induced THP-1 macrophages were treated with Acanthamoeba components of pathogenic (ATCC50739) and nonpathogenic (ATCC30010) strains in vitro. The data showed that Acanthamoeba treatment resulted in low IL-1β secretion from macrophages. In addition, Acanthamoeba CL of both strains was able to upregulate autophagy-related (Atg) protein 8, an autophagy marker, whereas Acanthamoeba CS downregulated Atg8 expression. We further manipulated autophagy and found that autophagy induction by starvation diminished IL-1β secretion while autophagy inhibition by 3-methyladenine (3MA) increased IL-1β secretion. Interestingly, in the presence of Acanthamoeba components either under starvation or 3MA treatment, IL-1β secretion was significantly reduced. Transcriptional expression of other ATG genes, i.e., ATG6, ATG7, and ATG5, were investigated and showed that their mRNA expression was maintained at the basal level under A. castellanii CS or CL treatment. Inflammasome-related genes, NLRP3 and CASPASE1, were upregulated following A. castellanii 50739 CS treatment but not in A. castellanii 50739 CL-treated condition. However, both conditions were able to increase IL-1β mRNA expression. TEM micrographs revealed that 3MA treatment induced the formation of large vacuoles and accumulation of autophagosome at the edge of THP-1 macrophages. However, the number and size of their structures were declined in the presence of A. castellanii 50739 CS with 3MA. Furthermore, immunofluorescence staining demonstrated the association between Atg8/LC3 and IL-1β expression, where downregulation of Atg8 by A. castellanii 50739 CS led to the upregulation of IL-1β. Altogether, the data indicate that Acanthamoeba can manipulate macrophage autophagy, thereby controlling low IL-1β secretion. The expression of autophagy- and inflammasome-related genes also indicates multiple mechanisms in IL-1β secretion in response to Acanthamoeba components. However, further characterization of Atg proteins and investigations into other intracellular pathways or defense mechanisms are needed to fully understand the unconventional secretion of IL-1β in macrophages. This knowledge could eventually lead to the development of innovative therapeutic strategies against Acanthamoeba infection by modulating autophagy or macrophage responses.
Collapse
Affiliation(s)
- Rachasak Boonhok
- Department of Medical Technology, School of Allied Health Sciences, and Research Excellence Center for Innovation and Health Products (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Wilaiwan Senghoi
- Department of Medical Technology, School of Allied Health Sciences, and Center of Excellence Research for Melioidosis and Microorganisms (CERMM), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Suthinee Sangkanu
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla 90112, Thailand
| | - Chooi Ling Lim
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Matsayapan Pudla
- Department of Oral Microbiology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials and Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Tooba Mahboob
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Md Atiar Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Pongsak Utaisincharoen
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Poonsit Hiransai
- Department of Medical Technology, School of Allied Health Sciences, and Center of Excellence in Marijuana, Hemp, and Kratom, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences, Southeast Asia Water Team (SEA Water Team) and World Union for Herbal Drug Discovery (WUHeDD), Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
4
|
Zhu J, Zhou T, Chen G, Wu Y, Chen X, Song Y, Tuohetali A, Gao H, Pang D, Wen H, Aimulajiang K. Inhibition of the MyD88 signaling pathway could upregulates Ghrelin expression to synergistically regulate hepatic Echinococcus multilocularis-infected progression. Front Immunol 2024; 15:1512180. [PMID: 39749332 PMCID: PMC11693510 DOI: 10.3389/fimmu.2024.1512180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction AE and whether the inhibition of the MyD88 inflammatory pathway can enhance Ghrelin expression to collaboratively modulate AE progression remains unclear. Methods In this study, we evaluated Ghrelin serum levels and changes in TLR4/MyD88/NF-κB pathway proteins and inflammatory factors in AE patients and E. multilocularis mouse models at different stages of infection (-4, -8, and -12 weeks). Additionally, we administered the MyD88 inhibitor TJ-M2010-5 intraperitoneally to infected mice to evaluate alterations in inflammation and Ghrelin levels, as well as disease progression. Results A decrease in serum Ghrelin levels in AE patients, whereas both Ghrelin and GHSR, along with TLR4/MyD88/NF-κB pathway proteins and markers of M1/M2 macrophage polarization, exhibited increased expression in the inflammatory cell zones surrounding hepatic lesions. Similar findings were observed in E. multilocularis-infected mice. M1-type inflammatory expression predominated throughout the infection's progression, with sustained high levels of Ghrelin counteracting inflammation. The TLR4/ MyD88/NF-κB pathway remained suppressed during the first 8 weeks, becoming activated only at 12 weeks. Inhibition of the MyD88 pathway resulted in reduced inflammation levels and upregulated Ghrelin expression, thereby collaboratively regulating the progression of hepatic infection. Conclusion These findings suggest an interactive regulation between the MyD88 inflammatory signaling pathway and Ghrelin, indicating that MyD88 inhibition could enhance Ghrelin expression to modulate the progression of E. multilocularis infection.
Collapse
Affiliation(s)
- Jiang Zhu
- Department of Abdominal Surgery, The Third People Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Tanfang Zhou
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Guangfeng Chen
- Department of Abdominal Surgery, The Third People Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Yuhui Wu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xia Chen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Ya Song
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Ayinula Tuohetali
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Huijing Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Dongming Pang
- Department of Abdominal Surgery, The Third People Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Hao Wen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Kalibixiati Aimulajiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
5
|
Hedegger K, Hommel T, Schaubeck M, Gimpfl M, Dahlhoff M. Different infant formulas can activate toll-like receptor 9 in vitro and inhibit interleukin 6 in human primary intestinal epithelial cells. Eur J Nutr 2024; 64:16. [PMID: 39567370 PMCID: PMC11579199 DOI: 10.1007/s00394-024-03507-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 10/12/2024] [Indexed: 11/22/2024]
Abstract
PURPOSE Necrotizing enterocolitis (NEC) is the most severe gastrointestinal disease in preterm infants caused by an exaggerated intestinal epithelial immune activation. Several studies show that modulation of toll-like receptor 9 (TLR9) activity may have positive effects on preventing intestinal inflammatory mechanisms ultimately resulting in NEC development. In this study, the effect of various infant formulas (IF) and the probiotic strain Limosilactobacillus fermentum CECT5716 on TLR9 activation were analyzed in vitro. METHODS First, TLR4 and TLR9 expression was analyzed on human primary intestinal epithelial cells (P-IECs) by qPCR and Western blot analysis. Then genetically designed HEK-Dual™ hTLR9 (NF/IL8) reporter cells (HEK-Dual) were treated with different IFs, L. fermentum CECT5716, and different functional components to measure TLR9 activation via luminescence. Finally, the IFs were investigated in P-IECs to analyze TLR downstream signaling by Western blot analysis. RESULTS IFs containing intact protein and L. fermentum CECT5716 activated TLR9 in HEK-Dual cells, but the functional components lactoferrin, L-5-methyltetrahydrofolate, and hydrolyzed whey proteins failed to activate TLR9. In P-IECs, the IFs induced increased phosphorylation of MAPK8/9 of the TLR signaling pathway and significantly reduced IL6 levels. Consistently, IL6 levels were increased in P-IECs when TLR9-signaling was inhibited. Interestingly, L. fermentum CECT5716 enhanced TLR9-signaling and increased NF-kappa-B inhibitor alpha-phosphorylation. CONCLUSION We found out that the used control formula, prebiotic formula, prebiotic formula with hydrolyzed-protein, and L. fermentum CECT5716 reduce IL6 levels in human P-IECs through TLR9 activation. L. fermentum CECT5716 and the here tested IFs could be a promising approach for modulation of gut health in infants.
Collapse
Affiliation(s)
- Kathrin Hedegger
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU München, Feodor-Lynen-Straße 25, 81377, Munich, Germany
| | - Theresa Hommel
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria
| | - Monika Schaubeck
- HiPP GmbH & Co. Vertrieb KG, Georg-Hipp-Straße 7, 85276, Pfaffenhofen, Germany
| | - Martina Gimpfl
- HiPP GmbH & Co. Vertrieb KG, Georg-Hipp-Straße 7, 85276, Pfaffenhofen, Germany
| | - Maik Dahlhoff
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria.
| |
Collapse
|
6
|
Liu S, Yue Y, Wu Q, Zhang LM. Interleukin-33: A Double-Edged Sword in Sepsis. TRANSLATIONAL PERIOPERATIVE AND PAIN MEDICINE 2024; 11:612-619. [PMID: 39991638 PMCID: PMC11845250 DOI: 10.31480/2330-4871/188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Despite some advances that have been made in the clinical management of sepsis, its morbidity and mortality rates remain high. The pathogenesis of systemic inflammation and organ damage leading to septic mortality is not fully understood. Interleukin-33 (IL-33), a new member of the IL-1 superfamily, can be a traditional cytokine and a nuclear factor regulating gene transcription. Due to its biological characteristics, IL-33 can act as a double-edged sword in sepsis: It not only contributes to clear bacteria and improves survival but also causes inflammation, immunosuppression, and organ damage via modulating immune response. In this review, we will summarize updated information on the dual functions of IL-33 in the host immune response to sepsis.
Collapse
Affiliation(s)
- Shuai Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yinyan Yue
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qiuge Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li-Ming Zhang
- Department of Anesthesiology & Perioperative Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Goumenaki P, Günther S, Kikhi K, Looso M, Marín-Juez R, Stainier DYR. The innate immune regulator MyD88 dampens fibrosis during zebrafish heart regeneration. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1158-1176. [PMID: 39271818 PMCID: PMC11399109 DOI: 10.1038/s44161-024-00538-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/06/2024] [Indexed: 09/15/2024]
Abstract
The innate immune response is triggered rapidly after injury and its spatiotemporal dynamics are critical for regeneration; however, many questions remain about its exact role. Here we show that MyD88, a key component of the innate immune response, controls not only the inflammatory but also the fibrotic response during zebrafish cardiac regeneration. We find in cryoinjured myd88-/- ventricles a significant reduction in neutrophil and macrophage numbers and the expansion of a collagen-rich endocardial population. Further analyses reveal compromised PI3K/AKT pathway activation in the myd88-/- endocardium and increased myofibroblasts and scarring. Notably, endothelial-specific overexpression of myd88 reverses these neutrophil, fibrotic and scarring phenotypes. Mechanistically, we identify the endocardial-derived chemokine gene cxcl18b as a target of the MyD88 signaling pathway, and using loss-of-function and gain-of-function tools, we show that it controls neutrophil recruitment. Altogether, these findings shed light on the pivotal role of MyD88 in modulating inflammation and fibrosis during tissue regeneration.
Collapse
Affiliation(s)
- Pinelopi Goumenaki
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Stefan Günther
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Khrievono Kikhi
- Flow Cytometry Service Group, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mario Looso
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rubén Marín-Juez
- Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montreal, Quebec, Canada
- Department of Pathology and Cell Biology, University of Montreal, Montreal, Quebec, Canada
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany.
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.
| |
Collapse
|
8
|
Liao HX, Mao X, Wang L, Wang N, Ocansey DKW, Wang B, Mao F. The role of mesenchymal stem cells in attenuating inflammatory bowel disease through ubiquitination. Front Immunol 2024; 15:1423069. [PMID: 39185411 PMCID: PMC11341407 DOI: 10.3389/fimmu.2024.1423069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Inflammatory bowel disease (IBD), a condition of the digestive tract and one of the autoimmune diseases, is becoming a disease of significant global public health concern and substantial clinical burden. Various signaling pathways have been documented to modulate IBD, but the exact activation and regulatory mechanisms have not been fully clarified; thus, a need for constant exploration of the molecules and pathways that play key roles in the development of IBD. In recent years, several protein post-translational modification pathways, such as ubiquitination, phosphorylation, methylation, acetylation, and glycolysis, have been implicated in IBD. An aberrant ubiquitination in IBD is often associated with dysregulated immune responses and inflammation. Mesenchymal stem cells (MSCs) play a crucial role in regulating ubiquitination modifications through the ubiquitin-proteasome system, a cellular machinery responsible for protein degradation. Specifically, MSCs have been shown to influence the ubiquitination of key signaling molecules involved in inflammatory pathways. This paper reviews the recent research progress in MSC-regulated ubiquitination in IBD, highlighting their therapeutic potential in treating IBD and offering a promising avenue for developing targeted interventions to modulate the immune system and alleviate inflammatory conditions.
Collapse
Affiliation(s)
- Hong Xi Liao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| | - Xiaojun Mao
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu, China
| | - Lan Wang
- Department of Laboratory Medicine, Danyang Blood Station, Zhenjiang, Jiangsu, China
| | - Naijian Wang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bo Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| |
Collapse
|
9
|
Poulis N, Martin M, Hoerstrup SP, Emmert MY, Fioretta ES. Development of an iPSC-derived tissue-resident macrophage-based platform for the in vitro immunocompatibility assessment of human tissue engineered matrices. Sci Rep 2024; 14:12171. [PMID: 38806547 PMCID: PMC11133401 DOI: 10.1038/s41598-024-62745-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
Upon implanting tissue-engineered heart valves (TEHVs), blood-derived macrophages are believed to orchestrate the remodeling process. They initiate the immune response and mediate the remodeling of the TEHV, essential for the valve's functionality. The exact role of another macrophage type, the tissue-resident macrophages (TRMs), has not been yet elucidated even though they maintain the homeostasis of native tissues. Here, we characterized the response of hTRM-like cells in contact with a human tissue engineered matrix (hTEM). HTEMs comprised intracellular peptides with potentially immunogenic properties in their ECM proteome. Human iPSC-derived macrophages (iMφs) could represent hTRM-like cells in vitro and circumvent the scarcity of human donor material. iMφs were derived and after stimulation they demonstrated polarization towards non-/inflammatory states. Next, they responded with increased IL-6/IL-1β secretion in separate 3/7-day cultures with longer production-time-hTEMs. We demonstrated that iMφs are a potential model for TRM-like cells for the assessment of hTEM immunocompatibility. They adopt distinct pro- and anti-inflammatory phenotypes, and both IL-6 and IL-1β secretion depends on hTEM composition. IL-6 provided the highest sensitivity to measure iMφs pro-inflammatory response. This platform could facilitate the in vitro immunocompatibility assessment of hTEMs and thereby showcase a potential way to achieve safer clinical translation of TEHVs.
Collapse
Affiliation(s)
- Nikolaos Poulis
- Institute for Regenerative Medicine (IREM), University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Marcy Martin
- Institute for Regenerative Medicine (IREM), University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Simon P Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Maximilian Y Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland.
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland.
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany.
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Institut für Regenerative Medizin (IREM), University of Zurich, Moussonstrasse 13, 8044, Zurich, Switzerland.
| | - Emanuela S Fioretta
- Institute for Regenerative Medicine (IREM), University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| |
Collapse
|
10
|
Li Y, Huang L, Li H, Zhu Y, Yu Z, Zheng X, Weng C, Feng WH. ASFV pA151R negatively regulates type I IFN production via degrading E3 ligase TRAF6. Front Immunol 2024; 15:1339510. [PMID: 38449860 PMCID: PMC10914938 DOI: 10.3389/fimmu.2024.1339510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
African swine fever (ASF) caused by African swine fever virus (ASFV) is a highly mortal and hemorrhagic infectious disease in pigs. Previous studies have indicated that ASFV modulates interferon (IFN) production. In this study, we demonstrated that ASFV pA151R negatively regulated type I IFN production. Ectopic expression of pA151R dramatically inhibited K63-linked polyubiquitination and Ser172 phosphorylation of TANK-binding kinase 1 (TBK1). Mechanically, we demonstrated that E3 ligase TNF receptor-associated factor 6 (TRAF6) participated in the ubiquitination of TBK1 in cGAS-STING signaling pathway. We showed that pA151R interacted with TRAF6 and degraded it through apoptosis pathway, leading to the disruption of TBK1 and TRAF6 interaction. Moreover, we clarified that the amino acids H102, C109, C132, and C135 in pA151R were crucial for pA151R to inhibit type I interferon production. In addition, we verified that overexpression of pA151R facilitated DNA virus Herpes simplex virus 1 (HSV-1) replication by inhibiting IFN-β production. Importantly, knockdown of pA151R inhibited ASFV replication and enhanced IFN-β production in porcine alveolar macrophages (PAMs). Our findings will help understand how ASFV escapes host antiviral immune responses and develop effective ASFV vaccines.
Collapse
Affiliation(s)
- You Li
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Li Huang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hui Li
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yingqi Zhu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zilong Yu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiaojie Zheng
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Changjiang Weng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Wen-hai Feng
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
11
|
Bayer AL, Smolgovsky S, Ngwenyama N, Hernández-Martínez A, Kaur K, Sulka K, Amrute J, Aronovitz M, Lavine K, Sharma S, Alcaide P. T-Cell MyD88 Is a Novel Regulator of Cardiac Fibrosis Through Modulation of T-Cell Activation. Circ Res 2023; 133:412-429. [PMID: 37492941 PMCID: PMC10529989 DOI: 10.1161/circresaha.123.323030] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/17/2023] [Indexed: 07/27/2023]
Abstract
BACKGROUND Cardiac inflammation in heart failure is characterized by the presence of damage-associated molecular patterns, myeloid cells, and T cells. Cardiac damage-associated molecular patterns provide continuous proinflammatory signals to myeloid cells through TLRs (toll-like receptors) that converge onto the adaptor protein MyD88 (myeloid differentiation response 88). These induce activation into efficient antigen-presenting cells that activate T cells through their TCR (T-cell receptor). T-cell activation results in cardiotropism, cardiac fibroblast transformation, and maladaptive cardiac remodeling. T cells rely on TCR signaling for effector function and survival, and while they express MyD88 and damage-associated molecular pattern receptors, their role in T-cell activation and cardiac inflammation is unknown. METHODS We performed transverse aortic constriction in mice lacking MyD88 in T cells and analyzed remodeling, systolic function, survival, and T-cell activation. We profiled wild type versus Myd88-/- mouse T cells at the transcript and protein level and performed several functional assays. RESULTS Analysis of single-cell RNA-sequencing data sets revealed that MyD88 is expressed in mouse and human cardiac T cells. MyD88 deletion in T cells resulted in increased levels of cardiac T-cell infiltration and fibrosis in response to transverse aortic constriction. We discovered that TCR-activated Myd88-/- T cells had increased proinflammatory signaling at the transcript and protein level compared with wild type, resulting in increased T-cell effector functions such as adhesion, migration across endothelial cells, and activation of cardiac fibroblast. Mechanistically, we found that MyD88 modulates T-cell activation and survival through TCR-dependent rather than TLR-dependent signaling. CONCLUSIONS Our results outline a novel intrinsic role for MyD88 in limiting T-cell activation that is central to tune down cardiac inflammation during cardiac adaptation to stress.
Collapse
Affiliation(s)
| | | | | | | | - Kuljeet Kaur
- Department of Immunology, Tufts University, Boston MA
| | | | - Junedh Amrute
- Department of Medicine, Washington University School of Medicine, Saint Louis MO
| | | | - Kory Lavine
- Department of Medicine, Washington University School of Medicine, Saint Louis MO
| | - Shruti Sharma
- Department of Immunology, Tufts University, Boston MA
| | - Pilar Alcaide
- Department of Immunology, Tufts University, Boston MA
| |
Collapse
|
12
|
Han X, Sun Y. PROTACs: A novel strategy for cancer drug discovery and development. MedComm (Beijing) 2023; 4:e290. [PMID: 37261210 PMCID: PMC10227178 DOI: 10.1002/mco2.290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023] Open
Abstract
Proteolysis targeting chimera (PROTAC) technology has become a powerful strategy in drug discovery, especially for undruggable targets/proteins. A typical PROTAC degrader consists of three components: a small molecule that binds to a target protein, an E3 ligase ligand (consisting of an E3 ligase and its small molecule recruiter), and a chemical linker that hooks first two components together. In the past 20 years, we have witnessed advancement of multiple PROTAC degraders into the clinical trials for anticancer therapies. However, one of the major challenges of PROTAC technology is that only very limited number of E3 ligase recruiters are currently available as E3 ligand for targeted protein degradation (TPD), although human genome encodes more than 600 E3 ligases. Thus, there is an urgent need to identify additional effective E3 ligase recruiters for TPD applications. In this review, we summarized the existing RING-type E3 ubiquitin ligase and their small molecule recruiters that act as effective E3 ligands of PROTAC degraders and their application in anticancer drug discovery. We believe that this review could serve as a reference in future development of efficient E3 ligands of PROTAC technology for cancer drug discovery and development.
Collapse
Affiliation(s)
- Xin Han
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionChina National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational MedicineZhejiang University School of MedicineHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERZhejiang ProvinceChina
- Key Laboratory of Molecular Biology in Medical SciencesZhejiang ProvinceChina
| | - Yi Sun
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionChina National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational MedicineZhejiang University School of MedicineHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERZhejiang ProvinceChina
- Key Laboratory of Molecular Biology in Medical SciencesZhejiang ProvinceChina
- Research Center for Life Science and Human HealthBinjiang Institute of Zhejiang UniversityHangzhouChina
| |
Collapse
|
13
|
Elsayed S, Elsaid KA. Protein phosphatase 2A regulates xanthine oxidase-derived ROS production in macrophages and influx of inflammatory monocytes in a murine gout model. Front Pharmacol 2022; 13:1033520. [PMID: 36467056 PMCID: PMC9712728 DOI: 10.3389/fphar.2022.1033520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/07/2022] [Indexed: 08/08/2023] Open
Abstract
Background: Gout is a common arthritis, due to deposition of monosodium urate (MSU) crystals which results in IL-1β secretion by tissue-resident macrophages. Xanthine oxidase (XO) catalyzes uric acid (UA) production and in the process, reactive oxygen species (ROS) are generated which contributes to NLRP3 inflammasome activation. Protein phosphatase 2A (PP2A) may be involved in regulating inflammatory pathways in macrophages. The objective of this study was to investigate whether PP2A regulates gout inflammation, mediated by XO activity modulation. We studied UA and ROS generations in MSU stimulated murine bone marrow derived macrophages (BMDMs) in response to fingolimod phosphate, a PP2A activator, and compared its anti-inflammatory efficacy to that of an XO inhibitor, febuxostat. Methods: BMDMs were stimulated with MSU, GM-CSF/IL-1β or nigericin ± fingolimod (2.5 μM) or febuxostat (200 μM) and UA levels, ROS, XO, and PP2A activities, Xdh (XO) expression and secreted IL-1β levels were determined. PP2A activity and IL-1β in MSU stimulated BMDMs ± N-acetylcysteine (NAC) (10 μM) ± okadaic acid (a PP2A inhibitor) were also determined. M1 polarization of BMDMs in response to MSU ± fingolimod treatment was assessed by a combination of iNOS expression and multiplex cytokine assay. The in vivo efficacy of fingolimod was assessed in a murine peritoneal model of acute gout where peritoneal lavages were studied for pro-inflammatory classical monocytes (CMs), anti-inflammatory nonclassical monocytes (NCMs) and neutrophils by flow cytometry and IL-1β by ELISA. Results: Fingolimod reduced intracellular and secreted UA levels (p < 0.05), Xdh expression (p < 0.001), XO activity (p < 0.001), ROS generation (p < 0.0001) and IL-1β secretion (p < 0.0001), whereas febuxostat enhanced PP2A activity (p < 0.05). NAC treatment enhanced PP2A activity and reduced XO activity and PP2A restoration mediated NAC's efficacy as co-treatment with okadaic acid increased IL-1β secretion (p < 0.05). Nigericin activated caspase-1 and reduced PP2A activity (p < 0.001) and fingolimod reduced caspase-1 activity in BMDMs (p < 0.001). Fingolimod reduced iNOS expression (p < 0.0001) and secretion of IL-6 and TNF-α (p < 0.05). Fingolimod reduced CMs (p < 0.0001), neutrophil (p < 0.001) and IL-1β (p < 0.05) lavage levels while increasing NCMs (p < 0.001). Conclusion: Macrophage PP2A is inactivated in acute gout by ROS and a PP2A activator exhibited a broad anti-inflammatory effect in acute gout in vitro and in vivo.
Collapse
|
14
|
Montaño DE, Hartung S, Wich M, Ali R, Jungnickel B, von Lilienfeld-Toal M, Voigt K. The TLR-NF-kB axis contributes to the monocytic inflammatory response against a virulent strain of Lichtheimia corymbifera, a causative agent of invasive mucormycosis. Front Immunol 2022; 13:882921. [PMID: 36311802 PMCID: PMC9608459 DOI: 10.3389/fimmu.2022.882921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022] Open
Abstract
Invasive mucormycosis (IM) is a life-threatening infection caused by the fungal order Mucorales, its diagnosis is often delayed, and mortality rates range from 40-80% due to its rapid progression. Individuals suffering from hematological malignancies, diabetes mellitus, organ transplantations, and most recently COVID-19 are particularly susceptible to infection by Mucorales. Given the increase in the occurrence of these diseases, mucormycosis has emerged as one of the most common fungal infections in the last years. However, little is known about the host immune response to Mucorales. Therefore, we characterized the interaction among L. corymbifera—one of the most common causative agents of IM—and human monocytes, which are specialized phagocytes that play an instrumental role in the modulation of the inflammatory response against several pathogenic fungi. This study covered four relevant aspects of the host-pathogen interaction: i) The recognition of L. corymbifera by human monocytes. ii) The intracellular fate of L. corymbifera. iii) The inflammatory response by human monocytes against the most common causative agents of mucormycosis. iv) The main activated Pattern-Recognition Receptors (PRRs) inflammatory signaling cascades in response to L. corymbifera. Here, we demonstrate that L. corymbifera exhibits resistance to intracellular killing over 24 hours, does not germinate, and inflicts minimal damage to the host cell. Nonetheless, viable fungal spores of L. corymbifera induced early production of the pro-inflammatory cytokine IL-1β, and late release of TNF-α and IL-6 by human monocytes. Moreover, we revealed that IL-1β production predominantly depends on Toll-like receptors (TLRs) priming, especially via TLR4, while TNF-α is secreted via C-type lectin receptors (CTLs), and IL-6 is produced by synergistic activation of TLRs and CTLs. All these signaling pathways lead to the activation of NF-kB, a transcription factor that not only regulates the inflammatory response but also the apoptotic fate of monocytes during infection with L. corymbifera. Collectively, our findings provide new insights into the host-pathogen interactions, which may serve for future therapies to enhance the host inflammatory response to L. corymbifera.
Collapse
Affiliation(s)
- Dolly E. Montaño
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany
- Jena Microbial Resource Collection, Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Susann Hartung
- Infections in Hematology and Oncology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany
| | - Melissa Wich
- Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany
| | - Rida Ali
- Jena Microbial Resource Collection, Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Berit Jungnickel
- Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany
| | - Marie von Lilienfeld-Toal
- Infections in Hematology and Oncology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany
- Department of Hematology and Medical Oncology, Jena University Hospital, Jena, Germany
| | - Kerstin Voigt
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany
- Jena Microbial Resource Collection, Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
- *Correspondence: Kerstin Voigt,
| |
Collapse
|
15
|
Kelsall IR. Non-lysine ubiquitylation: Doing things differently. Front Mol Biosci 2022; 9:1008175. [PMID: 36200073 PMCID: PMC9527308 DOI: 10.3389/fmolb.2022.1008175] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/01/2022] [Indexed: 11/23/2022] Open
Abstract
The post-translational modification of proteins with ubiquitin plays a central role in nearly all aspects of eukaryotic biology. Historically, studies have focused on the conjugation of ubiquitin to lysine residues in substrates, but it is now clear that ubiquitylation can also occur on cysteine, serine, and threonine residues, as well as on the N-terminal amino group of proteins. Paradigm-shifting reports of non-proteinaceous substrates have further extended the reach of ubiquitylation beyond the proteome to include intracellular lipids and sugars. Additionally, results from bacteria have revealed novel ways to ubiquitylate (and deubiquitylate) substrates without the need for any of the enzymatic components of the canonical ubiquitylation cascade. Focusing mainly upon recent findings, this review aims to outline the current understanding of non-lysine ubiquitylation and speculate upon the molecular mechanisms and physiological importance of this non-canonical modification.
Collapse
|
16
|
Cucurbitacin B Down-Regulates TNF Receptor 1 Expression and Inhibits the TNF-α-Dependent Nuclear Factor κB Signaling Pathway in Human Lung Adenocarcinoma A549 Cells. Int J Mol Sci 2022; 23:ijms23137130. [PMID: 35806134 PMCID: PMC9267118 DOI: 10.3390/ijms23137130] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 12/30/2022] Open
Abstract
Pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), induce the expression of intracellular adhesion molecule-1 (ICAM-1) by activating the nuclear factor κB (NF-κB) signaling pathway. In the present study, we found that cucurbitacin B decreased the expression of ICAM-1 in human lung adenocarcinoma A549 cells stimulated with TNF-α or interleukin-1α. We further investigated the mechanisms by which cucurbitacin B down-regulates TNF-α-induced ICAM-1 expression. Cucurbitacin B inhibited the nuclear translocation of the NF-κB subunit RelA and the phosphorylation of IκBα in A549 cells stimulated with TNF-α. Cucurbitacin B selectively down-regulated the expression of TNF receptor 1 (TNF-R1) without affecting three adaptor proteins (i.e., TRADD, RIPK1, and TRAF2). The TNF-α-converting enzyme inhibitor suppressed the down-regulation of TNF-R1 expression by cucurbitacin B. Glutathione, N-acetyl-L-cysteine, and, to a lesser extent, L-cysteine attenuated the inhibitory effects of cucurbitacin B on the TNF-α-induced expression of ICAM-1, suggesting that an α,β-unsaturated carbonyl moiety is essential for anti-inflammatory activity. The present results revealed that cucurbitacin B down-regulated the expression of TNF-R1 at the initial step in the TNF-α-dependent NF-κB signaling pathway.
Collapse
|
17
|
Butler D, Ambite I, Wan MLY, Tran TH, Wullt B, Svanborg C. Immunomodulation therapy offers new molecular strategies to treat UTI. Nat Rev Urol 2022; 19:419-437. [PMID: 35732832 PMCID: PMC9214477 DOI: 10.1038/s41585-022-00602-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 12/13/2022]
Abstract
Innovative solutions are needed for the treatment of bacterial infections, and a range of antibacterial molecules have been explored as alternatives to antibiotics. A different approach is to investigate the immune system of the host for new ways of making the antibacterial defence more efficient. However, the immune system has a dual role as protector and cause of disease: in addition to being protective, increasing evidence shows that innate immune responses can become excessive and cause acute symptoms and tissue pathology during infection. This role of innate immunity in disease suggests that the immune system should be targeted therapeutically, to inhibit over-reactivity. The ultimate goal is to develop therapies that selectively attenuate destructive immune response cascades, while augmenting the protective antimicrobial defence but such treatment options have remained underexplored, owing to the molecular proximity of the protective and destructive effects of the immune response. The concept of innate immunomodulation therapy has been developed successfully in urinary tract infections, based on detailed studies of innate immune activation and disease pathogenesis. Effective, disease-specific, immunomodulatory strategies have been developed by targeting specific immune response regulators including key transcription factors. In acute pyelonephritis, targeting interferon regulatory factor 7 using small interfering RNA or treatment with antimicrobial peptide cathelicidin was protective and, in acute cystitis, targeting overactive effector molecules such as IL-1β, MMP7, COX2, cAMP and the pain-sensing receptor NK1R has been successful in vivo. Furthermore, other UTI treatment strategies, such as inhibiting bacterial adhesion and vaccination, have also shown promise. Hyperactivation of innate immunity is a disease determinant in urinary tract infections (UTIs). Modulation of innate immunity has promise as a therapy for UTIs. In this Review, the authors discuss potential mechanisms and immunomodulatory therapeutic strategies in UTIs. Excessive innate immune responses to infection cause symptoms and pathology in acute pyelonephritis and acute cystitis. Innate immunomodulation therapy is, therefore, a realistic option for treating these conditions. Targeting excessive innate immune responses at the level of transcription has been successful in animal models. Innate immunomodulation therapy reduces excessive inflammation and tissue pathology and accelerates bacterial clearance from infected kidneys and bladders in mice. Innate immunomodulation therapy also accelerates the clearance of antibiotic-resistant bacterial strains.
Collapse
Affiliation(s)
- Daniel Butler
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ines Ambite
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Murphy Lam Yim Wan
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thi Hien Tran
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Björn Wullt
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
18
|
Alcoceba M, García-Álvarez M, Medina A, Maldonado R, González-Calle V, Chillón MC, Sarasquete ME, González M, García-Sanz R, Jiménez C. MYD88 Mutations: Transforming the Landscape of IgM Monoclonal Gammopathies. Int J Mol Sci 2022; 23:5570. [PMID: 35628381 PMCID: PMC9141891 DOI: 10.3390/ijms23105570] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/05/2023] Open
Abstract
The MYD88 gene has a physiological role in the innate immune system. Somatic mutations in MYD88, including the most common L265P, have been associated with the development of certain types of lymphoma. MYD88L265P is present in more than 90% of patients with Waldenström's macroglobulinemia (WM) and IgM monoclonal gammopathy of undetermined significance (IgM-MGUS). The absence of MYD88 mutations in WM patients has been associated with a higher risk of transformation into aggressive lymphoma, resistance to certain therapies (BTK inhibitors), and shorter overall survival. The MyD88 signaling pathway has also been used as a target for specific therapies. In this review, we summarize the clinical applications of MYD88 testing in the diagnosis, prognosis, follow-up, and treatment of patients. Although MYD88L265P is not specific to WM, few tumors present a single causative mutation in a recurrent position. The role of the oncogene in the pathogenesis of WM is still unclear, especially considering that the mutation can be found in normal B cells of patients, as recently reported. This may have important implications for early lymphoma detection in healthy elderly individuals and for the treatment response assessment based on a MYD88L265P analysis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ramón García-Sanz
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.A.); (M.G.-Á.); (A.M.); (R.M.); (V.G.-C.); (M.C.C.); (M.E.S.); (M.G.); (C.J.)
| | | |
Collapse
|
19
|
Zhu G, Cheng Z, Wang Q, Lin C, Lin P, He R, Chen H, Hoffman RM, Ye J. TRAF6 regulates the signaling pathway influencing colorectal cancer function by ubiquitination mechanisms. Cancer Sci 2022; 113:1393-1405. [PMID: 35179811 PMCID: PMC8990288 DOI: 10.1111/cas.15302] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 12/24/2022] Open
Abstract
Tumor necrosis factor receptor‐associated factor‐6 (TRAF6) is a ubiquitin E3 ligase. TRAF6 plays an important role in tumor invasion and metastasis. However, the specific mechanism by which TRAF6 promotes colorectal cancer (CRC) metastasis is incompletely understood. This study aimed to determine whether TRAF6 affects the LPS‐NF‐κB‐VEGF‐C signaling pathway through ubiquitination, which plays a role in colorectal cancer metastasis. Here, our results showed that TRAF6 affected lymphangiogenesis through the LPS‐NF‐κB‐VEGF‐C signaling pathway. Using ubiquitination experiments, we found that TRAF6 was mainly ubiquitinated with the K63‐linked chains, and LPS promoted ubiquitination of TRAF6 and K63‐linked chains. More importantly, TRAF6 124mut is the main ubiquitination site of TRAF6 interacting with K63‐linked chains. TRAF6 affected the migration, invasion, and lymphatic metastasis of colorectal cancer through its ubiquitination. In subcutaneous xenograft models, TRAF6 124mut inhibited tumor growth. In conclusion, our results provide new insight for studying the mechanism of lymphangiogenesis in colorectal cancer to promote cancer metastasis, which may provide new ideas for tumor immunotherapy.
Collapse
Affiliation(s)
- Guangwei Zhu
- Department of Gastrointestinal Surgery, Section, Institute of Abdominal Surgery, Key Laboratory of accurate diagnosis and treatment of cancer, The First Hospital Affiliated to Fujian Medical University, Fuzhou, 350005, China
| | - Zhibin Cheng
- Department of Gastrointestinal Surgery, Section, Institute of Abdominal Surgery, Key Laboratory of accurate diagnosis and treatment of cancer, The First Hospital Affiliated to Fujian Medical University, Fuzhou, 350005, China
| | - Qin Wang
- Department of Gastrointestinal Surgery, Section, Institute of Abdominal Surgery, Key Laboratory of accurate diagnosis and treatment of cancer, The First Hospital Affiliated to Fujian Medical University, Fuzhou, 350005, China
| | - Chunlin Lin
- Department of Gastrointestinal Surgery, Section, Institute of Abdominal Surgery, Key Laboratory of accurate diagnosis and treatment of cancer, The First Hospital Affiliated to Fujian Medical University, Fuzhou, 350005, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Penghang Lin
- Department of Gastrointestinal Surgery, Section, Institute of Abdominal Surgery, Key Laboratory of accurate diagnosis and treatment of cancer, The First Hospital Affiliated to Fujian Medical University, Fuzhou, 350005, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Ruofan He
- Department of Gastrointestinal Surgery, Section, Institute of Abdominal Surgery, Key Laboratory of accurate diagnosis and treatment of cancer, The First Hospital Affiliated to Fujian Medical University, Fuzhou, 350005, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Hui Chen
- Department of Gastrointestinal Surgery, Section, Institute of Abdominal Surgery, Key Laboratory of accurate diagnosis and treatment of cancer, The First Hospital Affiliated to Fujian Medical University, Fuzhou, 350005, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Robert M Hoffman
- AntiCancer, Inc, San Diego, CA, U.S.A.,Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Jianxin Ye
- Department of Gastrointestinal Surgery, Section, Institute of Abdominal Surgery, Key Laboratory of accurate diagnosis and treatment of cancer, The First Hospital Affiliated to Fujian Medical University, Fuzhou, 350005, China
| |
Collapse
|
20
|
The Therapeutic Prospects of Targeting IL-1R1 for the Modulation of Neuroinflammation in Central Nervous System Disorders. Int J Mol Sci 2022; 23:ijms23031731. [PMID: 35163653 PMCID: PMC8915186 DOI: 10.3390/ijms23031731] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/24/2022] [Accepted: 01/30/2022] [Indexed: 11/16/2022] Open
Abstract
The interleukin-1 receptor type 1 (IL-1R1) holds pivotal roles in the immune system, as it is positioned at the “epicenter” of the inflammatory signaling networks. Increased levels of the cytokine IL-1 are a recognized feature of the immune response in the central nervous system (CNS) during injury and disease, i.e., neuroinflammation. Despite IL-1/IL-1R1 signaling within the CNS having been the subject of several studies, the roles of IL-1R1 in the CNS cellular milieu still cause controversy. Without much doubt, however, the persistent activation of the IL-1/IL-1R1 signaling pathway is intimately linked with the pathogenesis of a plethora of CNS disease states, ranging from Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS), all the way to schizophrenia and prion diseases. Importantly, a growing body of evidence is showing that blocking IL-1R1 signaling via pharmacological or genetic means in different experimental models of said CNS diseases leads to reduced neuroinflammation and delayed disease progression. The aim of this paper is to review the recent progress in the study of the biological roles of IL-1R1, as well as to highlight key aspects that render IL-1R1 a promising target for the development of novel disease-modifying treatments for multiple CNS indications.
Collapse
|
21
|
Talreja J, Bauerfeld C, Wang X, Hafner M, Liu Y, Samavati L. MKP-1 modulates ubiquitination/phosphorylation of TLR signaling. Life Sci Alliance 2021; 4:e202101137. [PMID: 34580177 PMCID: PMC8500224 DOI: 10.26508/lsa.202101137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 11/24/2022] Open
Abstract
Ubiquitination and phosphorylation are reversible posttranslational protein modifications regulating physiological and pathological processes. MAPK phosphatase (MKP)-1 regulates innate and adaptive immunity. The multifaceted roles of MKP-1 were attributed to dephosphorylation of p38 and JNK MAPKs. We show that the lack of MKP-1 modulates the landscape of ubiquitin ligases and deubiquitinase enzymes (DUBs). MKP-1-/- showed an aberrant regulation of several DUBs and increased expression of proteins and genes involved in IL-1/TLR signaling upstream of MAPK, including IL-1R1, IRAK1, TRAF6, phosphorylated TAK1, and an increased K63 polyubiquitination on TRAF6. Increased K63 polyubiquitination on TRAF6 was associated with an enhanced phosphorylated form of A20. Among abundant DUBs, ubiquitin-specific protease-13 (USP13), which cleaves polyubiquitin-chains on client proteins, was substantially enhanced in murine MKP-1-deficient BMDMs. An inhibitor of USP13 decreased the K63 polyubiquitination on TRAF6, TAK1 phosphorylation, IL-1β, and TNF-α induction in response to LPS in BMDMs. Our data show for the first time that MKP-1 modulates the ligase activity of TRAF6 through modulation of specific DUBs.
Collapse
Affiliation(s)
- Jaya Talreja
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, MI, USA
| | - Christian Bauerfeld
- Department of Pediatrics, Division of Critical Care, Central Michigan University, Mount Pleasant, MI, USA
| | - Xiantao Wang
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Disease, National Institutes of Health, Bethesda, MD, USA
| | - Markus Hafner
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Disease, National Institutes of Health, Bethesda, MD, USA
| | - Yusen Liu
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Lobelia Samavati
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
22
|
Mahmoud MF, Abdelaal S, Mohammed HO, El-Shazly AM, Daoud R, Abdelfattah MAO, Sobeh M. Syzygium aqueum (Burm.f.) Alston Prevents Streptozotocin-Induced Pancreatic Beta Cells Damage via the TLR-4 Signaling Pathway. Front Pharmacol 2021; 12:769244. [PMID: 34912223 PMCID: PMC8667316 DOI: 10.3389/fphar.2021.769244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/25/2021] [Indexed: 11/23/2022] Open
Abstract
Although several treatments are available for the treatment of type 2 diabetes mellitus, adverse effects and cost burden impose the search for safe, efficient, and cost-effective alternative herbal remedies. Syzygium aqueum (Burm.f.) Alston, a natural anti-inflammatory, antioxidant herb, may suppress diabetes-associated inflammation and pancreatic beta-cell death. Here, we tested the ability of the bioactive leaf extract (SA) to prevent streptozotocin (STZ)-induced oxidative stress and inflammation in pancreatic beta cells in rats and the involvement of the TLR-4 signaling pathway. Non-fasted rats pretreated with 100 or 200 mg kg-1 SA 2 days prior to the STZ challenge and for 14 days later had up to 52 and 39% reduction in the glucose levels, respectively, while glibenclamide, the reference standard drug (0.5 mg kg-1), results in 70% reduction. Treatment with SA extract was accompanied by increased insulin secretion, restoration of Langerhans islets morphology, and decreased collagen deposition as demonstrated from ELISA measurement, H and E, and Mallory staining. Both glibenclamide and SA extract significantly decreased levels of TLR-4, MYD88, pro-inflammatory cytokines TNF-α, and TRAF-6 in pancreatic tissue homogenates, which correlated well with minimal pancreatic inflammatory cell infiltration. Pre-treatment with SA or glibenclamide decreased malondialdehyde, a sensitive biomarker of ROS-induced lipid peroxidation, and restored depleted reduced glutathione in the pancreas. Altogether, these data indicate that S. aqueum is effective in improving STZ-induced pancreatic damage, which could be beneficial in treating type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Mona F. Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Shimaa Abdelaal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Heba Osama Mohammed
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Assem M. El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Rachid Daoud
- African Genome Center, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | | | - Mansour Sobeh
- AgroBioSciences, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| |
Collapse
|
23
|
Wang W, Gao W, Zhu Q, Alasbahi A, Seki E, Yang L. TAK1: A Molecular Link Between Liver Inflammation, Fibrosis, Steatosis, and Carcinogenesis. Front Cell Dev Biol 2021; 9:734749. [PMID: 34722513 PMCID: PMC8551703 DOI: 10.3389/fcell.2021.734749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/22/2021] [Indexed: 12/22/2022] Open
Abstract
Chronic insult and persistent injury can cause liver inflammation, fibrosis, and carcinogenesis; it can also be associated with metabolic disorders. Identification of critical molecules that link the process of inflammation and carcinogenesis will provide prospective therapeutic targets for liver diseases. Rapid advancements in gene engineering technology have allowed the elucidation of the underlying mechanism of transformation, from inflammation and metabolic disorders to carcinogenesis. Transforming growth factor-β-activated kinase 1 (TAK1) is an upstream intracellular protein kinase of nuclear factor kappa-B (NF-κB) and c-Jun N-terminal kinases, which are activated by numerous cytokines, growth factors, and microbial products. In this study, we highlighted the functional roles of TAK1 and its interaction with transforming growth factor-β, WNT, AMP-activated protein kinase, and NF-κB signaling pathways in liver inflammation, steatosis, fibrosis, and carcinogenesis based on previously published articles.
Collapse
Affiliation(s)
- Weijun Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenkang Gao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingjing Zhu
- Department of Liver Diseases, Wuhan Jinyintan Hospital, Wuhan, China
| | - Afnan Alasbahi
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ekihiro Seki
- Department of Medicine, Cedars-Sinai, Los Angeles, CA, United States
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
Lin H, Fan Y, Wieser A, Zhang J, Regel I, Nieß H, Mayerle J, Gerbes AL, Steib CJ. Albumin Might Attenuate Bacteria-Induced Damage on Kupffer Cells for Patients with Chronic Liver Disease. Cells 2021; 10:cells10092298. [PMID: 34571946 PMCID: PMC8469739 DOI: 10.3390/cells10092298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/20/2021] [Accepted: 09/01/2021] [Indexed: 12/29/2022] Open
Abstract
Chronic liver diseases (CLDs) are complex diseases that cause long-term inflammation and infection, which in turn accelerate their development. The usage of albumin in patients with CLDs has been debated for years. Human serum albumin (HSA) plays a key role in immunomodulation during the process of CLDs. The correlation between albumin and C-reactive protein (CRP) in CLD patients was analyzed by linear regression with the Pearson statistic. The damage of THP-1 and primary cells was evaluated by measuring the lactate dehydrogenase (LDH) in the supernatant. Immunofluorescence staining was performed to determine underlying pathways in Kupffer cells (KCs). Albumin negatively correlated with infection in patients with CLDs. In vitro experiments with THP-1 cells and KCs showed that albumin reduced LDH release after stimulation with bacterial products, while no differences in hepatic stellate cells (HSCs) and sinusoidal endothelial cells (SECs) were detected. Moreover, immunofluorescence staining revealed an increase of p-ERK and p-NF-kB p65 density after albumin treatment of KCs stimulated by bacterial products. In conclusion, albumin could assist CLD patients in alleviating inflammation caused by bacterial products and might be beneficial to patients with CLDs by securing KCs from bacteria-induced damage, providing a compelling rationale for albumin therapy in patients with CLDs.
Collapse
Affiliation(s)
- Hao Lin
- Liver Center Munich, Department of Medicine II, University Hospital, 81377 Munich, Germany; (H.L.); (Y.F.); (I.R.); (J.M.); (A.L.G.)
| | - Yuhui Fan
- Liver Center Munich, Department of Medicine II, University Hospital, 81377 Munich, Germany; (H.L.); (Y.F.); (I.R.); (J.M.); (A.L.G.)
| | - Andreas Wieser
- Max von Pettenkofer Institute, Faculty of Medicine, Medical Microbiology and Hospital Epidemiology, 80336 Munich, Germany;
- Division of Infectious Diseases and Tropical Medicine, University Hospital, 80802 Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, 80802 Munich, Germany
| | - Jiang Zhang
- Liver Transplantation Center, Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China;
| | - Ivonne Regel
- Liver Center Munich, Department of Medicine II, University Hospital, 81377 Munich, Germany; (H.L.); (Y.F.); (I.R.); (J.M.); (A.L.G.)
| | - Hanno Nieß
- Biobank of the Department of General, Visceral and Transplant Surgery, University Hospital, 80802 Munich, Germany;
| | - Julia Mayerle
- Liver Center Munich, Department of Medicine II, University Hospital, 81377 Munich, Germany; (H.L.); (Y.F.); (I.R.); (J.M.); (A.L.G.)
| | - Alexander L. Gerbes
- Liver Center Munich, Department of Medicine II, University Hospital, 81377 Munich, Germany; (H.L.); (Y.F.); (I.R.); (J.M.); (A.L.G.)
| | - Christian J. Steib
- Liver Center Munich, Department of Medicine II, University Hospital, 81377 Munich, Germany; (H.L.); (Y.F.); (I.R.); (J.M.); (A.L.G.)
- Correspondence: ; Tel.: +49-89-4400-72298; Fax: +49-89-4400-75299
| |
Collapse
|
25
|
Yuan S, Chen Y, Zhang M, Wang Z, Hu Z, Ruan Y, Ren Z, Shi F. Overexpression of miR-223 Promotes Tolerogenic Properties of Dendritic Cells Involved in Heart Transplantation Tolerance by Targeting Irak1. Front Immunol 2021; 12:676337. [PMID: 34421892 PMCID: PMC8374072 DOI: 10.3389/fimmu.2021.676337] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/13/2021] [Indexed: 02/05/2023] Open
Abstract
Dendritic cells (DCs) are key mediators of transplant rejection. Numerous factors have been identified that regulate transplant immunopathology by modulating the function of DCs. Among these, microRNAs (miRNAs), small non-coding RNA molecules, have received much attention. The miRNA miR-223 is very highly expressed and tightly regulated in hematopoietic cells. It plays an important role in modulating the immune response by regulating neutrophils and macrophages, and its dysregulation contributes to multiple types of immune diseases. However, the role of miR-223 in immune rejection is unclear. Here, we observed expression of miR-223 in patients and mice who had undergone heart transplantation and found that it increased in the serum of both, and also in DCs from the spleens of recipient mice, although it was unchanged in splenic T cells. We also found that miR-223 expression decreased in lipopolysaccharide-stimulated DCs. Increasing the level of miR-223 in DCs promoted polarization of DCs toward a tolerogenic phenotype, which indicates that miR-223 can attenuate activation and maturation of DCs. MiR-223 effectively induced regulatory T cells (Tregs) by inhibiting the function of antigen-presenting DCs. In addition, we identified Irak1 as a miR-223 target gene and an essential regulator of DC maturation. In mouse allogeneic heterotopic heart transplantation models, grafts survived longer and suffered less immune cell infiltration in mice with miR-223-overexpressing immature (im)DCs. In the miR-223-overexpressing imDC recipients, T cells from spleen differentiated into Tregs, and the level of IL-10 in heart grafts was markedly higher than that in the control group. In conclusion, miR-223 regulates the function of DCs via Irak1, differentiation of T cells into Tregs, and secretion of IL-10, thereby suppressing allogeneic heart graft rejection.
Collapse
Affiliation(s)
- Shun Yuan
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanyang Chen
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Min Zhang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhipeng Hu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yongle Ruan
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zongli Ren
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Shi
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
26
|
Di Ceglie I, van Lent PLEM, Geven EJW, Koenders MI, Blom AB, Vogl T, Roth J, van den Bosch MHJ. S100A8/A9 is not essential for the development of inflammation and joint pathology in interleukin-1 receptor antagonist knockout mice. Arthritis Res Ther 2021; 23:216. [PMID: 34412663 PMCID: PMC8375068 DOI: 10.1186/s13075-021-02602-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/06/2021] [Indexed: 11/24/2022] Open
Abstract
Background Excessive osteoclast activity, which is strongly stimulated by pro-inflammatory mediators, results in bone and cartilage degeneration as central features of many arthritides. Levels of the alarmin S100A8/A9 and interleukin (IL)-1β are both increased in arthritis patients and correlate with disease activity and progression of tissue erosion. We previously presented S100A8/A9 as a good biomarker for joint inflammation and arthritis pathology under circumstances of high IL-1 signaling in mice that lack the gene encoding IL-1 receptor antagonist (Il1rn−/− mice). Here, we investigated whether S100A8/A9 is also actively involved in the development of joint inflammation and both cartilage and bone pathology under these conditions by comparing Il1rn−/− mice with mice that have an additional deficiency for S100a9 (Il1rn−/−XS100a9−/−). Methods Il1rn−/−XS100a9−/− on a BALB/c background were obtained by crossing S100a9−/− mice and Il1rn−/− mice. Arthritis incidence and severity were macroscopically scored. Myeloid cell populations in the bone marrow and spleen were determined using flow cytometry. In vitro osteoclastogenesis of bone marrow cells was evaluated with TRAP staining. Microscopic joint inflammation, cartilage degeneration, and bone destruction were evaluated using histology of ankle joints of 12- and 20-week-old mice. Results Macroscopically scored arthritis severity was comparable between Il1rn−/− and Il1rn−/−XS100a9−/− mice. Inflammation, cartilage erosion, and bone erosion were clearly present in 12-week-old mice of both strains lacking Il1rn−/−, but not significantly different between Il1rn−/−XS100a9−/− and Il1rn−/−. Moreover, we observed that the numbers of neutrophils and monocytes were increased by the absence of Il1rn, which was affected by the absence of S100a9 only in the spleen but not in the bone marrow. In line with our other findings, the absence of S100a9 did not affect the osteoclastogenic potential of osteoclast precursors in the absence of Il1rn. Finally, in agreement with the findings in early arthritis development in 12-week-old mice, cartilage and bone erosion in 20-week-old mice was significantly higher in both Il1rn−/− strains, but the additional absence of S100a9 did not further affect tissue pathology. Conclusion S100A8/A9 deficiency does not significantly affect inflammation and joint destruction in mice with high IL1β signaling suggesting that S100A8/A9 is not essential for the development of arthritis under these conditions. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02602-y.
Collapse
Affiliation(s)
- Irene Di Ceglie
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 28, 6525, GA, Nijmegen, the Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 28, 6525, GA, Nijmegen, the Netherlands.
| | - Edwin J W Geven
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 28, 6525, GA, Nijmegen, the Netherlands
| | - Marije I Koenders
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 28, 6525, GA, Nijmegen, the Netherlands
| | - Arjen B Blom
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 28, 6525, GA, Nijmegen, the Netherlands
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Martijn H J van den Bosch
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 28, 6525, GA, Nijmegen, the Netherlands
| |
Collapse
|
27
|
Campbell GR, Spector SA. Induction of Autophagy to Achieve a Human Immunodeficiency Virus Type 1 Cure. Cells 2021; 10:cells10071798. [PMID: 34359967 PMCID: PMC8307643 DOI: 10.3390/cells10071798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Effective antiretroviral therapy has led to significant human immunodeficiency virus type 1 (HIV-1) suppression and improvement in immune function. However, the persistence of integrated proviral DNA in latently infected reservoir cells, which drive viral rebound post-interruption of antiretroviral therapy, remains the major roadblock to a cure. Therefore, the targeted elimination or permanent silencing of this latently infected reservoir is a major focus of HIV-1 research. The most studied approach in the development of a cure is the activation of HIV-1 expression to expose latently infected cells for immune clearance while inducing HIV-1 cytotoxicity—the “kick and kill” approach. However, the complex and highly heterogeneous nature of the latent reservoir, combined with the failure of clinical trials to reduce the reservoir size casts doubt on the feasibility of this approach. This concern that total elimination of HIV-1 from the body may not be possible has led to increased emphasis on a “functional cure” where the virus remains but is unable to reactivate which presents the challenge of permanently silencing transcription of HIV-1 for prolonged drug-free remission—a “block and lock” approach. In this review, we discuss the interaction of HIV-1 and autophagy, and the exploitation of autophagy to kill selectively HIV-1 latently infected cells as part of a cure strategy. The cure strategy proposed has the advantage of significantly decreasing the size of the HIV-1 reservoir that can contribute to a functional cure and when optimised has the potential to eradicate completely HIV-1.
Collapse
Affiliation(s)
- Grant R. Campbell
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA;
- Correspondence: ; Tel.: +1-858-534-7477
| | - Stephen A. Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA;
- Rady Children’s Hospital, San Diego, CA 92123, USA
| |
Collapse
|
28
|
DOKUMACIOĞLU E, İSKENDER H, TERİM KAPAKİN KA, YENİCE G, MOKTHARE B, BOLAT İ, HAYIRLI A. Effect of betulinic acid administration on TLR-9/NF-κB /IL-18 levels in experimental liver injury. Turk J Med Sci 2021; 51:1544-1553. [PMID: 33773522 PMCID: PMC8283446 DOI: 10.3906/sag-2004-184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 03/27/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND/AIM Acetaminophen (APAP), used in the composition of thousands of preparations, is the most commonly used analgesic and antipyretic drug. The present study aimed to investigate the potential protective effects of the betulinic acid (BA) treatment through an APAP-induced hepatotoxicity rat model, using inflammatory, biochemical, and histopathological parameters. MATERIALS AND METHODS The study consisted of four groups: control group, APAP group, BA group, and APAP+BA group. Experimental studies continued for fifteen days. Serum samples were analysed for glucose, total cholesterol (TChol), triglyceride (TG), low density lipoprotein (LDL), high density lipoprotein (HDL), aspartate amino transferase (AST), malondialdehyde (MDA), toll-like receptor-9 (TLR-9), nuclear factor kappa B (NF-κB), and interleukin-18 (IL-18). RESULTS TLR9, IL-18, NF-κB, and MDA levels increased significantly in liver injury groups. These increases considerably decreased by the BA treatment. All groups showed immunopositivity for 8-hydroxy-2’–deoxyguanosine (8-OHdG) and interleukin (IL-1β) in the hepatocytes, inflammatory cells, and epithelial cells of bile ducts. CONCLUSION BA can be used as an effective agent in the prevention and treatment of acute liver diseases due to its inhibitory properties in multiple pathways and its potent antioxidant effects.
Collapse
Affiliation(s)
- Eda DOKUMACIOĞLU
- Department of Nutrition and Dietetics, Faculty of Healthy Sciences, Artvin Çoruh University, ArtvinTurkey
| | - Hatice İSKENDER
- Department of Nutrition and Dietetics, Faculty of Healthy Sciences, Artvin Çoruh University, ArtvinTurkey
| | | | - Güler YENİCE
- Department of Animal Nutrition and Nutritional Disorders, Faculty of Veterinary Medicine, Atatürk University, ErzurumTurkey
| | - Behzat MOKTHARE
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, ErzurumTurkey
| | - İsmail BOLAT
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, ErzurumTurkey
| | - Armağan HAYIRLI
- Department of Animal Nutrition and Nutritional Disorders, Faculty of Veterinary Medicine, Atatürk University, ErzurumTurkey
| |
Collapse
|
29
|
Cao W, Guo Y, Cheng Z, Xu G, Zuo Q, Nie L, Huang Y, Liu S, Zhu Y. Inducible ATP1B1 Upregulates Antiviral Innate Immune Responses by the Ubiquitination of TRAF3 and TRAF6. THE JOURNAL OF IMMUNOLOGY 2021; 206:2668-2681. [PMID: 34011520 DOI: 10.4049/jimmunol.2001262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/19/2021] [Indexed: 11/19/2022]
Abstract
The antiviral innate immune responses are crucial steps during host defense and must be strictly regulated, but the molecular mechanisms of control remain unclear. In this study, we report increased expression of human ATPase Na+/K+ transporting subunit β 1(ATP1B1) after DNA and RNA virus infections. We found that the expression of ATP1B1 can inhibit viral replication and increase the levels of IFNs, IFN-stimulated genes, and inflammatory cytokines. Knockdown of ATP1B1 by specific short hairpin RNA had the opposite effects. Upon viral infection, ATP1B1 was induced, interacted with TRAF3 and TRAF6, and potentiated the ubiquitination of these proteins, leading to increased phosphorylation of downstream molecules, including TGF-β-activated kinase 1 (TAK1) and TANK-binding kinase 1 (TBK1). These results reveal a previously unrecognized role of ATP1B1 in antiviral innate immunity and suggest a novel mechanism for the induction of IFNs and proinflammatory cytokines during viral infection.
Collapse
Affiliation(s)
- Wei Cao
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yifei Guo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhikui Cheng
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Gang Xu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Qi Zuo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yu Huang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
30
|
Umeoguaju FU, Ephraim-Emmanuel BC, Uba JO, Bekibele GE, Chigozie N, Orisakwe OE. Immunomodulatory and Mechanistic Considerations of Hibiscus sabdariffa (HS) in Dysfunctional Immune Responses: A Systematic Review. Front Immunol 2021; 12:550670. [PMID: 34040600 PMCID: PMC8141557 DOI: 10.3389/fimmu.2021.550670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 04/08/2021] [Indexed: 11/13/2022] Open
Abstract
Hibiscus sabdariffa calyx (HS) water decoction extract is a commonly consumed beverage with various pharmacological properties. This systematic review examines the possible effect of HS intake on immune mediators. The Scopus and PUBMED databases were searched for all human and animal studies that investigated the effect of HS administration on immune related biomarkers. For each of the immune biomarkers, the mean, standard deviation and number of subjects were extracted for both the HS treated and untreated group. These values were used in the computation of standardized mean difference (SMD). Statistical analysis and forest plot were done with R statistical software (version 3.6.1). Twenty seven (27) studies met the eligibility criteria. Twenty two (22) of the studies were used for the meta-analysis which included a total of 1211 subjects. The meta-analysis showed that HS administration significantly lowered the levels of TNF-α (n=10; pooled SMD: -1.55; 95% CI: -2.43, -0.67; P < 0.01), IL-6 (n=11; pooled SMD:-1.09; 95% CI: -1.77, -0.40; P < 0.01), IL-1β (n=7; pooled SMD:-0.62; 95% CI: -1.25, 0.00; P = 0.05), Edema formation (n=4; pooled SMD: -2.29; 95% CI: -4.47, -0.11; P = 0.04), Monocyte Chemoattractant Protein -1 (n=4; pooled SMD: -1.17; 95% CI: -1.78, -0.57; P < 0.01) and Angiotensin converting enzyme cascade (n=6; pooled SMD: -0.91; 95% CI: -1.57, -0.25; P < 0.01). The levels of IL-10 (n=4; pooled SMD: -0.38; 95% CI: -1.67, 0.91; P = 0.56), Interleukin 8 (n=2; pooled SMD:-0.12; 95% CI: -0.76, 0.51; P = 0.71), iNOS (n=2; pooled SMD:-0.69; 95% CI: -1.60, 0.23 P = 0.14) and C- Reactive Protein (n=4; pooled SMD: 0.05; 95% CI: -0.26, 0.36; P = 0.75), were not significantly changed by HS administration. Some of the results had high statistical heterogeneity. HS may be promising in the management of disorders involving hyperactive immune system or chronic inflammation.
Collapse
Affiliation(s)
- Francis U. Umeoguaju
- World Bank Africa Centre of Excellence in Public Health and Toxicological Research (PUTOR), University of Port Harcourt, Port Harcourt, Nigeria
| | - Benson C. Ephraim-Emmanuel
- World Bank Africa Centre of Excellence in Public Health and Toxicological Research (PUTOR), University of Port Harcourt, Port Harcourt, Nigeria
- Department of Dental Health Sciences, Ogbia, Bayelsa State College of Health Technology, Otakeme, Nigeria
| | - Joy O. Uba
- World Bank Africa Centre of Excellence in Public Health and Toxicological Research (PUTOR), University of Port Harcourt, Port Harcourt, Nigeria
| | - Grace E. Bekibele
- World Bank Africa Centre of Excellence in Public Health and Toxicological Research (PUTOR), University of Port Harcourt, Port Harcourt, Nigeria
| | - Nwondah Chigozie
- World Bank Africa Centre of Excellence in Public Health and Toxicological Research (PUTOR), University of Port Harcourt, Port Harcourt, Nigeria
| | - Orish Ebere Orisakwe
- World Bank Africa Centre of Excellence in Public Health and Toxicological Research (PUTOR), University of Port Harcourt, Port Harcourt, Nigeria
- Department of Experimental Pharmacology & Toxicology, Faculty of Pharmacy, University of Port Harcourtt, Port Harcourt, Nigeria
| |
Collapse
|
31
|
Loss of Mef2D function enhances TLR induced IL-10 production in macrophages. Biosci Rep 2021; 40:225925. [PMID: 32725155 PMCID: PMC7442974 DOI: 10.1042/bsr20201859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 12/24/2022] Open
Abstract
Mef2 transcription factors comprise a family of four different isoforms that regulate a number of processes including neuronal and muscle development. While roles for Mef2C and Mef2D have been described in B-cell development their role in immunity has not been extensively studied. In innate immune cells such as macrophages, TLRs drive the production of both pro- and anti-inflammatory cytokines. IL-10 is an important anti-inflammatory cytokine produced by macrophages and it establishes an autocrine feedback loop to inhibit pro-inflammatory cytokine production. We show here that macrophages from Mef2D knockout mice have elevated levels of IL-10 mRNA induction compared with wild-type cells following LPS stimulation. The secretion of IL-10 was also higher from Mef2D knockout macrophages and this correlated to a reduction in the secretion of TNF, IL-6 and IL-12p40. The use of an IL-10 neutralising antibody showed that this reduction in pro-inflammatory cytokine production in the Mef2D knockouts was IL-10 dependent. As the IL-10 promoter has previously been reported to contain a potential binding site for Mef2D, it is possible that the binding of other Mef2 isoforms in the absence of Mef2D may result in a higher activation of the IL-10 gene. Further studies with compound Mef2 isoforms would be required to address this. We also show that Mef2D is highly expressed in the thymus, but that loss of Mef2D does not affect thymic T-cell development or the production of IFNγ from CD8 T cells.
Collapse
|
32
|
Griffiths JS, Camilli G, Kotowicz NK, Ho J, Richardson JP, Naglik JR. Role for IL-1 Family Cytokines in Fungal Infections. Front Microbiol 2021; 12:633047. [PMID: 33643264 PMCID: PMC7902786 DOI: 10.3389/fmicb.2021.633047] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Fungal pathogens kill approximately 1.5 million individuals per year and represent a severe disease burden worldwide. It is estimated over 150 million people have serious fungal disease such as recurrent mucosal infections or life-threatening systemic infections. Disease can ensue from commensal fungi or new infection and involves different fungal morphologies and the expression of virulence factors. Therefore, anti-fungal immunity is complex and requires coordination between multiple facets of the immune system. IL-1 family cytokines are associated with acute and chronic inflammation and are essential for the innate response to infection. Recent research indicates IL-1 cytokines play a key role mediating immunity against different fungal infections. During mucosal disease, IL-1R and IL-36R are required for neutrophil recruitment and protective Th17 responses, but function through different mechanisms. During systemic disease, IL-18 drives protective Th1 responses, while IL-33 promotes Th2 and suppresses Th1 immunity. The IL-1 family represents an attractive anti-fungal immunotherapy target. There is a need for novel anti-fungal therapeutics, as current therapies are ineffective, toxic and encounter resistance, and no anti-fungal vaccine exists. Furthering our understanding of the IL-1 family cytokines and their complex role during fungal infection may aid the development of novel therapies. As such, this review will discuss the role for IL-1 family cytokines in fungal infections.
Collapse
Affiliation(s)
- James S Griffiths
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Giorgio Camilli
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Natalia K Kotowicz
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jemima Ho
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jonathan P Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
33
|
Li X, Wei Z, Wang X, Duan F, Xiong L, Li J, Tian J, Jia L, Gao H. Premna microphylla Turcz leaf pectin exhibited antioxidant and anti-inflammatory activities in LPS-stimulated RAW 264.7 macrophages. Food Chem 2021; 349:129164. [PMID: 33550022 DOI: 10.1016/j.foodchem.2021.129164] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/18/2020] [Accepted: 01/19/2021] [Indexed: 02/08/2023]
Abstract
Premna microphylla turcz leaf juice with polysaccharides (PMPs) as its main component, are raw material of jelly-like Chinese traditional food "Guanyin tofu", which were also experiencedly used to relieve inflammation-related symptoms. Here three kinds of PMPs were extracted in alkaline (APMP), water (WPMP) and acidic (HPMP) conditions, being characteristic of RG I, high- and low-methoxyl HG pectin, respectively, in amorphous form with diverse surface microstructures, among which APMP predominantly composed of Glucose instead of galacturonic acid, showing wider molecular weight distribution and more branched chains. PMPs showed remarkable radical scavenging capability, and especially APMP at concentrations above 50 μg/mL effectively inhibited the reactive oxygen species and malondialdehyde production in LPS-stimulated RAW 264.7 macrophages, by enhancing enzymatic activities of endogenous superoxide dismutase, glutathione peroxidase and catalase, and accordingly alleviated inflammatory cytokines. Thus, PMPs could be promising non-toxic natural dietary supplement to improve chronic inflammation-induced diseases.
Collapse
Affiliation(s)
- Xiao Li
- Department of Food Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
| | - Zeliang Wei
- Laboratory of Ethnopharmacology, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Xingyue Wang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Feixia Duan
- Department of Food Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China.
| | - Lidan Xiong
- Cosmetics Safety and Efficacy Evaluation Center, West China Hospital, Sichuan University, No. 5, Gong Xing Road, Chengdu, Sichuan, Chengdu 610041, PR China
| | - Jingwen Li
- Department of Food Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
| | - Jing Tian
- Department of Food Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
| | - Lirong Jia
- Department of Food Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
| | - Hong Gao
- Department of Food Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
| |
Collapse
|
34
|
Weighted Gene Co-Expression Network Analysis Identifies Key Modules and Hub Genes Associated with Mycobacterial Infection of Human Macrophages. Antibiotics (Basel) 2021; 10:antibiotics10020097. [PMID: 33498280 PMCID: PMC7909288 DOI: 10.3390/antibiotics10020097] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/28/2020] [Accepted: 01/14/2021] [Indexed: 02/04/2023] Open
Abstract
Tuberculosis (TB) is still a leading cause of death worldwide. Treatments remain unsatisfactory due to an incomplete understanding of the underlying host–pathogen interactions during infection. In the present study, weighted gene co-expression network analysis (WGCNA) was conducted to identify key macrophage modules and hub genes associated with mycobacterial infection. WGCNA was performed combining our own transcriptomic results using Mycobacterium aurum-infected human monocytic macrophages (THP1) with publicly accessible datasets obtained from three types of macrophages infected with seven different mycobacterial strains in various one-to-one combinations. A hierarchical clustering tree of 11,533 genes was built from 198 samples, and 47 distinct modules were revealed. We identified a module, consisting of 226 genes, which represented the common response of host macrophages to different mycobacterial infections that showed significant enrichment in innate immune stimulation, bacterial pattern recognition, and leukocyte chemotaxis. Moreover, by network analysis applied to the 74 genes with the best correlation with mycobacteria infection, we identified the top 10 hub-connecting genes: NAMPT, IRAK2, SOCS3, PTGS2, CCL20, IL1B, ZC3H12A, ABTB2, GFPT2, and ELOVL7. Interestingly, apart from the well-known Toll-like receptor and inflammation-associated genes, other genes may serve as novel TB diagnosis markers and potential therapeutic targets.
Collapse
|
35
|
Chaichian Y, Strand V. Interferon-directed therapies for the treatment of systemic lupus erythematosus: a critical update. Clin Rheumatol 2021; 40:3027-3037. [PMID: 33411137 DOI: 10.1007/s10067-020-05526-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/05/2020] [Accepted: 11/25/2020] [Indexed: 11/28/2022]
Abstract
The interferon (IFN) pathway, especially type I IFN, plays a critical role in the immunopathogenesis of systemic lupus erythematosus (SLE). We have gained significant insights into this pathway over the past two decades, including a better understanding of the key mediators of inflammation upstream and downstream of type I IFN. This has led to the identification of multiple potential targets for the treatment of SLE, for which a significant unmet need remains due to the failure of many patients to adequately respond to standard-of-care medications. Unfortunately, most new therapies in SLE have disappointed in preclinical or clinical trials to date, including a number that target type I IFN. Nevertheless, several IFN-directed therapies aimed at specific steps within this immunologic pathway have recently shown promise, and additional agents are in the treatment pipeline. In this review, we focus on the results of key therapeutic studies targeting the type I IFN pathway and discuss the future state of IFN-blockade in SLE.
Collapse
Affiliation(s)
- Yashaar Chaichian
- Division of Immunology and Rheumatology, Stanford University, Palo Alto, CA, USA.
| | - Vibeke Strand
- Division of Immunology and Rheumatology, Stanford University, Palo Alto, CA, USA
| |
Collapse
|
36
|
Michalski JE, Schwartz DA. Genetic Risk Factors for Idiopathic Pulmonary Fibrosis: Insights into Immunopathogenesis. J Inflamm Res 2021; 13:1305-1318. [PMID: 33447070 PMCID: PMC7801923 DOI: 10.2147/jir.s280958] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis is an etiologically complex interstitial lung disease characterized by progressive scarring of the lungs with a subsequent decline in lung function. While much of the pathogenesis of IPF still remains unclear, it is now understood that genetic variation accounts for at least one-third of the risk of developing the disease. The single-most validated and most significant risk factor, genetic or otherwise, is a gain-of-function promoter variant in the MUC5B gene. While the functional impact of these IPF risk variants at the cellular and tissue levels are areas of active investigation, there is a growing body of evidence that these genetic variants may influence disease pathogenesis through modulation of innate immune processes.
Collapse
Affiliation(s)
- Jacob E Michalski
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - David A Schwartz
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
37
|
Dagvadorj J, Mikulska-Ruminska K, Tumurkhuu G, Ratsimandresy RA, Carriere J, Andres AM, Marek-Iannucci S, Song Y, Chen S, Lane M, Dorfleutner A, Gottlieb RA, Stehlik C, Cassel S, Sutterwala FS, Bahar I, Crother TR, Arditi M. Recruitment of pro-IL-1α to mitochondrial cardiolipin, via shared LC3 binding domain, inhibits mitophagy and drives maximal NLRP3 activation. Proc Natl Acad Sci U S A 2021; 118:e2015632118. [PMID: 33361152 PMCID: PMC7817159 DOI: 10.1073/pnas.2015632118] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The balance between NLRP3 inflammasome activation and mitophagy is essential for homeostasis and cellular health, but this relationship remains poorly understood. Here we found that interleukin-1α (IL-1α)-deficient macrophages have reduced caspase-1 activity and diminished IL-1β release, concurrent with reduced mitochondrial damage, suggesting a role for IL-1α in regulating this balance. LPS priming of macrophages induced pro-IL-1α translocation to mitochondria, where it directly interacted with mitochondrial cardiolipin (CL). Computational modeling revealed a likely CL binding motif in pro-IL-1α, similar to that found in LC3b. Thus, binding of pro-IL-1α to CL in activated macrophages may interrupt CL-LC3b-dependent mitophagy, leading to enhanced Nlrp3 inflammasome activation and more robust IL-1β production. Mutation of pro-IL-1α residues predicted to be involved in CL binding resulted in reduced pro-IL-1α-CL interaction, a reduction in NLRP3 inflammasome activity, and increased mitophagy. These data identify a function for pro-IL-1α in regulating mitophagy and the potency of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Jargalsaikhan Dagvadorj
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Karolina Mikulska-Ruminska
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213
- Institute of Physics, Faculty of Physics Astronomy and Informatics, Nicolaus Copernicus University in Toruń, 87-100 Torun, Poland
| | - Gantsetseg Tumurkhuu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | | | - Jessica Carriere
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Allen M Andres
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Stefanie Marek-Iannucci
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Yang Song
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Shuang Chen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics. David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Malcolm Lane
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Andrea Dorfleutner
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Roberta A Gottlieb
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Christian Stehlik
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Suzanne Cassel
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Fayyaz S Sutterwala
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Ivet Bahar
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213;
| | - Timothy R Crother
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048;
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics. David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Moshe Arditi
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048;
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pediatrics. David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| |
Collapse
|
38
|
Mechanism by which TRAF6 Participates in the Immune Regulation of Autoimmune Diseases and Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4607197. [PMID: 33294443 PMCID: PMC7714562 DOI: 10.1155/2020/4607197] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 11/06/2020] [Accepted: 11/17/2020] [Indexed: 11/24/2022]
Abstract
Tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6), an E3 ubiquitin ligase, is a signal transduction molecule shared by the interleukin-1 receptor (IL-1R)/Toll-like receptor (TLR) family and the TNFR superfamily. TRAF6 has a unique TRAF domain and RING finger domain that mediate intracellular signaling events. In the immune system, TRAF6-mediated signaling has been shown to be critical for the development, homeostasis, and activation of a variety of immune cells, including B cells, T cells, dendritic cells, and macrophages. Although the pathogenesis and etiology of autoimmune diseases and cancer are not fully understood, it is worth noting that existing studies have shown that TRAF6 is involved in the pathogenesis and development of a variety of these diseases. Herein, we reviewed the role of TRAF6 in certain immune cells, as well as the function and potential effect of TRAF6 in autoimmune diseases and cancer. Our review indicates that TRAF6 may be a novel target for autoimmune diseases and cancer.
Collapse
|
39
|
Amina M, Al Musayeib NM, Alarfaj NA, El-Tohamy MF, Al-Hamoud GA. Antibacterial and Immunomodulatory Potentials of Biosynthesized Ag, Au, Ag-Au Bimetallic Alloy Nanoparticles Using the Asparagus racemosus Root Extract. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2453. [PMID: 33302432 PMCID: PMC7762544 DOI: 10.3390/nano10122453] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/04/2020] [Accepted: 12/06/2020] [Indexed: 12/15/2022]
Abstract
Two noble metals, such as silver and gold alloy nanoparticles, were successfully synthesized by the microwave assisted method in the presence of the Asparagus racemosus root extract and were used as an antibacterial and immunomodulatory agent. The nanostuctures of the synthesized nanoparticles were confirmed by various spectroscopic and microscopic techniques. The UV-vis spectrum exhibits a distinct absorption peak at 483 nm for the bimetallic alloy nanoparticles. The microscopic analysis revealed the spherical shaped morphology of the biosynthesized nanoparticles with a particle size of 10-50 nm. The antibacterial potential of the green synthesized single metal (AgNPs and AuNPs) and bimetallic alloy nanoparticles was tested against five bacterial strains. The bimetallic alloy nanoparticles displayed the highest zone of inhibition against P. aeurgnosia and S.aureus strains when compared to single metal nanoparticles and plant extract. In addition, the inmmunomodulatory potential of the root extract of A. racemosus, AgNPs, AuNPs, and Ag-Au alloy NPs is achieved by measuring the cytokine levels in macrophages (IL-1β, IL-6, and TNF-α) and NK cells (IFN-γ) of NK92 and THP1 cells using the solid phase sandwich ELISA technique. The results showed that the root extract of A. racemosus, AgNPs, and AuNPs can reduce the pro-inflammatory cytokine levels in the macrophages cells, while Ag-Au alloy NPs can reduce cytokine responses in NK92 cells. Overall, this study shows that the microwave assisted biogenic synthesized bimetallic nanoalloy nanoparticles could be further explored for the development of antibacterial and anti-inflammatory therapies.
Collapse
Affiliation(s)
- Musarat Amina
- Department of Pharmacognosy, Pharmacy College, King Saud University, Riyadh 11451, Saudi Arabia; (M.A.); (N.M.A.M.); (G.A.A.-H.)
| | - Nawal M. Al Musayeib
- Department of Pharmacognosy, Pharmacy College, King Saud University, Riyadh 11451, Saudi Arabia; (M.A.); (N.M.A.M.); (G.A.A.-H.)
| | - Nawal A. Alarfaj
- Department of Chemistry, College of Science, King Saud University, P.O. Box 22452, Riyadh 11451, Saudi Arabia;
| | - Maha F. El-Tohamy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 22452, Riyadh 11451, Saudi Arabia;
| | - Gadah A. Al-Hamoud
- Department of Pharmacognosy, Pharmacy College, King Saud University, Riyadh 11451, Saudi Arabia; (M.A.); (N.M.A.M.); (G.A.A.-H.)
| |
Collapse
|
40
|
Even-Or O, Avniel-Polak S, Barenholz Y, Nussbaum G. The cationic liposome CCS/C adjuvant induces immunity to influenza independently of the adaptor protein MyD88. Hum Vaccin Immunother 2020; 16:3146-3154. [PMID: 32401698 PMCID: PMC8641586 DOI: 10.1080/21645515.2020.1750247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/18/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023] Open
Abstract
Traditional non-living vaccines are often least effective in the populations that need them most, such as neonates and elderly adults. Vaccine adjuvants are one approach to boost the immunogenicity of antigens in populations with reduced immunity. Ideally, vaccine adjuvants will increase the seroconversion rates across the population, lead to stronger immune responses, and enable the administration of fewer vaccine doses. We previously demonstrated that a cationic liposomal formulation of the commercial influenza split virus vaccine (CCS/C-HA) enhanced cellular and humoral immunity to the virus, increased seroconversion rates, and improved survival after live virus challenge in a preclinical model, as compared to the commercial vaccine as is (F-HA). We now evaluated vaccine efficacy in different strains and sexes of mice and determined the role of innate immunity in the mechanism of action of the CCS/C adjuvant by testing the response of mice deficient in Toll-like receptors or the TLR/IL-1 adaptor protein MyD88 following immunization with CCS/C-HA vs. F-HA. Although TLR2- and TLR4-deficient mice responded to F-HA immunization, F-HA immunization failed to engender a significant immune response in the absence of MyD88. In contrast, immunization with the CCS/C-HA vaccine overcame the requirement for MyD88 in the response to the commercial vaccine and improved the immune responses and seroconversion rates in all strains of mice tested, including those deficient in TLR2 and TLR4.
Collapse
Affiliation(s)
- Orli Even-Or
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Shani Avniel-Polak
- Institute of Dental Sciences, The Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - Yechezkel Barenholz
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Gabriel Nussbaum
- Institute of Dental Sciences, The Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| |
Collapse
|
41
|
Muraleedharan A, Rotem-Dai N, Strominger I, Anto NP, Isakov N, Monsonego A, Livneh E. Protein kinase C eta is activated in reactive astrocytes of an Alzheimer's disease mouse model: Evidence for its immunoregulatory function in primary astrocytes. Glia 2020; 69:697-714. [PMID: 33068318 DOI: 10.1002/glia.23921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 09/24/2020] [Accepted: 10/01/2020] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is the primary cause of age-related dementia. Pathologically, AD is characterized by synaptic loss, the accumulation of β-amyloid peptides and neurofibrillary tangles, glial activation, and neuroinflammation. Whereas extensive studies focused on neurons and activation of microglia in AD, the role of astrocytes has not been well-characterized. Protein kinase C (PKC) was also implicated in AD; however, its role in astrocyte activation was not elucidated. Using the 5XFAD mouse model of AD, we show that PKC-eta (PKCη), an astrocyte-specific stress-activated and anti-apoptotic kinase, plays a role in reactive astrocytes. We demonstrate that PKCη staining is highly enriched in cortical astrocytes in a disease-dependent manner and in the vicinity of amyloid-β peptides plaques. Moreover, activation of PKCη, as indicated by its increased phosphorylation levels, is exhibited mainly in cortical astrocytes derived from adult 5XFAD mice. PKCη activation was associated with elevated levels of reactive astrocytic markers and upregulation of the pro-inflammatory cytokine interleukin 6 (IL-6) compared to littermate controls. Notably, inhibiting the kinase activity of PKCη in 5XFAD astrocyte cultures markedly increased the levels of secreted IL-6-a phenomenon that was also observed in wild-type astrocytes stimulated by inflammatory cytokines (e.g., TNFα, IL-1). Similar increase in the release of IL-6 was also observed upon inhibition of either the mammalian target of rapamycin (mTOR) or the protein phosphatase 2A (PP2A). Our findings suggest that the mTOR-PKCη-PP2A signaling cascade functions as a negative feedback loop of NF-κB-induced IL-6 release in astrocytes. Thus, we identify PKCη as a regulator of neuroinflammation in AD.
Collapse
Affiliation(s)
- Amitha Muraleedharan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The National Institute of Biotechnology in the Negev, Zlotowski Neuroscience Center, and Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Noa Rotem-Dai
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Itai Strominger
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The National Institute of Biotechnology in the Negev, Zlotowski Neuroscience Center, and Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nikhil Ponnoor Anto
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Noah Isakov
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Monsonego
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The National Institute of Biotechnology in the Negev, Zlotowski Neuroscience Center, and Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Etta Livneh
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
42
|
Pastor-Fernández G, Mariblanca IR, Navarro MN. Decoding IL-23 Signaling Cascade for New Therapeutic Opportunities. Cells 2020; 9:cells9092044. [PMID: 32906785 PMCID: PMC7563346 DOI: 10.3390/cells9092044] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/24/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
The interleukin 23 (IL-23) is a key pro-inflammatory cytokine in the development of chronic inflammatory diseases, such as psoriasis, inflammatory bowel diseases, multiple sclerosis, or rheumatoid arthritis. The pathological consequences of excessive IL-23 signaling have been linked to its ability to promote the production of inflammatory mediators, such as IL-17, IL-22, granulocyte-macrophage colony-stimulating (GM-CSF), or the tumor necrosis factor (TNFα) by target populations, mainly Th17 and IL-17-secreting TCRγδ cells (Tγδ17). Due to their pivotal role in inflammatory diseases, IL-23 and its downstream effector molecules have emerged as attractive therapeutic targets, leading to the development of neutralizing antibodies against IL-23 and IL-17 that have shown efficacy in different inflammatory diseases. Despite the success of monoclonal antibodies, there are patients that show no response or partial response to these treatments. Thus, effective therapies for inflammatory diseases may require the combination of multiple immune-modulatory drugs to prevent disease progression and to improve quality of life. Alternative strategies aimed at inhibiting intracellular signaling cascades using small molecule inhibitors or interfering peptides have not been fully exploited in the context of IL-23-mediated diseases. In this review, we discuss the current knowledge about proximal signaling events triggered by IL-23 upon binding to its membrane receptor to bring to the spotlight new opportunities for therapeutic intervention in IL-23-mediated pathologies.
Collapse
|
43
|
Ivanovska J, Kang NYC, Ivanovski N, Nagy A, Belik J, Gauda EB. Recombinant adiponectin protects the newborn rat lung from lipopolysaccharide-induced inflammatory injury. Physiol Rep 2020; 8:e14553. [PMID: 32889775 PMCID: PMC7507528 DOI: 10.14814/phy2.14553] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 02/06/2023] Open
Abstract
Preterm infants are at high risk for developing bronchopulmonary dysplasia and pulmonary hypertension from inflammatory lung injury. In adult models, adiponectin (APN)—an adipocyte‐derived hormone—protects the lung from inflammatory injury and pulmonary vascular remodeling. Cord blood APN levels in premature infants born < 26 weeks gestation are 5% of the level in infants born at term. We previously reported the expression profile of APN and its receptors in neonatal rat lung homogenates during the first 3 weeks of postnatal development. Here, we characterize the expression profile of APN and its receptors in specific lung cells and the effects of exogenous recombinant APN (rAPN) on lipopolysaccharide‐(LPS)‐induced cytokine and chemokine production in total lung homogenates and specific lung cells. In vitro, rAPN added to primary cultures of pulmonary artery smooth muscle cells attenuated the expression of LPS‐induced pro‐inflammatory cytokines while increasing the expression of anti‐inflammatory cytokines. In vivo, intraperitoneal rAPN (2 mg/kg), given 4 hr prior to intrapharyngeal administration of LPS (5 mg/kg) to newborn rats at postnatal day 4, significantly reduced gene and protein expression of the pro‐inflammatory cytokine IL‐1ß and reduced protein expression of the chemokines monocyte chemoattractant protein (MCP‐1) and macrophage inflammatory protein‐1 alpha (MIP‐1α) in the lung. LPS‐induced histopathological changes in the lung were also decreased. Moreover, rAPN given 20 hr after intrapharyngeal LPS had a similar effect on lung inflammation. These findings suggest a role for APN in protecting the lung from inflammation during early stages of lung development.
Collapse
Affiliation(s)
- Julijana Ivanovska
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, University of Toronto, Toronto, ON, Canada
| | - Na-Young Cindy Kang
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, University of Toronto, Toronto, ON, Canada
| | - Nikola Ivanovski
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, University of Toronto, Toronto, ON, Canada
| | - Avita Nagy
- Department of Pediatric Laboratory Medicine, University of Toronto, Toronto, ON, Canada
| | - Jaques Belik
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, University of Toronto, Toronto, ON, Canada
| | - Estelle B Gauda
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
44
|
Zhang J, Fu L, Shen B, Liu Y, Wang W, Cai X, Kong L, Yan Y, Meng R, Zhang Z, Chen YNP, Liu Q, Wan ZK, Zhou T, Wang X, Gavine P, Del Rosario A, Ahn K, Philippar U, Attar R, Yang J, Xu Y, Edwards JP, Dai X. Assessing IRAK4 Functions in ABC DLBCL by IRAK4 Kinase Inhibition and Protein Degradation. Cell Chem Biol 2020; 27:1500-1509.e13. [PMID: 32888499 DOI: 10.1016/j.chembiol.2020.08.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/29/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
The interleukin-1 receptor-activated kinase 4 (IRAK4) belongs to the IRAK family of serine/threonine kinases and plays a central role in the innate immune response. However, the function of IRAK4 in tumor growth and progression remains elusive. Here we sought to determine the enzymatic and scaffolding functions of IRAK4 in activated B-cell-like diffuse large B cell lymphoma (ABC DLBCL). We chose a highly selective IRAK4 kinase inhibitor to probe the biological effects of kinase inhibition and developed a series of IRAK4 degraders to evaluate the effects of protein degradation in ABC DLBCL cells. Interestingly, the results demonstrated that neither IRAK4 kinase inhibition nor protein degradation led to cell death or growth inhibition, suggesting a redundant role for IRAK4 in ABC DLBCL cell survival. IRAK4 degraders characterized in this study provide useful tools for understanding IRAK4 protein scaffolding function, which was previously unachievable using pharmacological perturbation.
Collapse
Affiliation(s)
- Jing Zhang
- Oncology Biology, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Liqiang Fu
- Medicinal Chemistry, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Bin Shen
- Oncology Biology, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Yingtao Liu
- Medicinal Chemistry, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Wenqian Wang
- Oncology Biology, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Xin Cai
- Biomarker, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Linglong Kong
- Medicinal Chemistry, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Yilin Yan
- Biomarker, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Ryan Meng
- Nonclinical Safety, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Zhuming Zhang
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Ying-Nan P Chen
- Oncology Biology, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Qian Liu
- Medicinal Chemistry, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Zhao-Kui Wan
- Medicinal Chemistry, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Tianyuan Zhou
- Oncology Biology, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Xiaotao Wang
- Biomarker, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Paul Gavine
- Oncology Biology, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Amanda Del Rosario
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Kay Ahn
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Ulrike Philippar
- Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Ricardo Attar
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Jennifer Yang
- Oncology Biology, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - Yanping Xu
- Medicinal Chemistry, Janssen (China) Research & Development Center, Shanghai 201210, China
| | - James P Edwards
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Xuedong Dai
- Medicinal Chemistry, Janssen (China) Research & Development Center, Shanghai 201210, China.
| |
Collapse
|
45
|
Barter MJ, Cheung K, Falk J, Panagiotopoulos AC, Cosimini C, O'Brien S, Teja-Putri K, Neill G, Deehan DJ, Young DA. Dynamic chromatin accessibility landscape changes following interleukin-1 stimulation. Epigenetics 2020; 16:106-119. [PMID: 32741307 PMCID: PMC7889151 DOI: 10.1080/15592294.2020.1789266] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Dynamic modifications of chromatin allow rapid access of the gene regulatory machinery to condensed genomic regions facilitating subsequent gene expression. Inflammatory cytokine stimulation of cells can cause rapid gene expression changes through direct signalling pathway-mediated transcription factor activation and regulatory element binding. Here we used the Assay for Transposase Accessible Chromatin with high-throughput sequencing (ATAC-seq) to assess regions of the genome that are differentially accessible following treatment of cells with interleukin-1 (IL-1). We identified 126,483 open chromatin regions, with 241 regions significantly differentially accessible following stimulation, with 64 and 177 more or less accessible, respectively. These differentially accessible regions predominantly correspond to regions of the genome marked as enhancers. Motif searching identified an overrepresentation of a number of transcription factors, most notably RelA, in the regions becoming more accessible, with analysis of ChIP-seq data confirmed RelA binding to these regions. A significant correlation in differential chromatin accessibility and gene expression was also observed. Functionality in regulating gene expression was confirmed using CRISPR/Cas9 genome-editing to delete regions that became more accessible following stimulation in the genes MMP13, IKBKE and C1QTNF1. These same regions were also accessible for activation using a dCas9-transcriptional activator and showed enhancer activity in a cellular model. Together, these data describe and functionally validate a number of dynamically accessible chromatin regions involved in inflammatory signalling.
Collapse
Affiliation(s)
- Matt J Barter
- Faculty of Medical Sciences, Skeletal Research Group, Biosciences Institute, Newcastle University , Newcastle upon Tyne, UK
| | - Kathleen Cheung
- Faculty of Medical Sciences, Skeletal Research Group, Biosciences Institute, Newcastle University , Newcastle upon Tyne, UK.,Faculty of Medical Sciences, Bioinformatics Support Unit, Newcastle University , Newcastle upon Tyne, UK
| | - Julia Falk
- Faculty of Medical Sciences, Skeletal Research Group, Biosciences Institute, Newcastle University , Newcastle upon Tyne, UK
| | - Andreas C Panagiotopoulos
- Faculty of Medical Sciences, Skeletal Research Group, Biosciences Institute, Newcastle University , Newcastle upon Tyne, UK
| | - Caitlin Cosimini
- Faculty of Medical Sciences, Skeletal Research Group, Biosciences Institute, Newcastle University , Newcastle upon Tyne, UK
| | - Siobhan O'Brien
- Faculty of Medical Sciences, Skeletal Research Group, Biosciences Institute, Newcastle University , Newcastle upon Tyne, UK
| | - Karina Teja-Putri
- Faculty of Medical Sciences, Skeletal Research Group, Biosciences Institute, Newcastle University , Newcastle upon Tyne, UK
| | - Graham Neill
- Faculty of Medical Sciences, Skeletal Research Group, Biosciences Institute, Newcastle University , Newcastle upon Tyne, UK
| | - David J Deehan
- Department of Orthopaedics, Freeman Hospital, Orthopaedics , UK
| | - David A Young
- Faculty of Medical Sciences, Skeletal Research Group, Biosciences Institute, Newcastle University , Newcastle upon Tyne, UK
| |
Collapse
|
46
|
Molecular characterization of TLR3 and TRIL in silvery pomfret (Pampus argenteus) and their expression profiles in response to bacterial components. Int J Biol Macromol 2020; 155:805-813. [DOI: 10.1016/j.ijbiomac.2020.03.246] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/15/2020] [Accepted: 03/29/2020] [Indexed: 01/14/2023]
|
47
|
Hong OK, Lee SS, Yoo SJ, Lee MK, Kim MK, Baek KH, Song KH, Kwon HS. Gemigliptin Inhibits Interleukin-1β-Induced Endothelial-Mesenchymal Transition via Canonical-Bone Morphogenetic Protein Pathway. Endocrinol Metab (Seoul) 2020; 35:384-395. [PMID: 32615723 PMCID: PMC7386109 DOI: 10.3803/enm.2020.35.2.384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/04/2020] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Endothelial-to-mesenchymal transition (EndMT) contributes to inflammatory conditions inducing conversion of endothelial cells (ECs) into activated fibroblasts, promoting fibrotic diseases. Pro-inflammatory cytokine is the most potent inducer of EndMT. We investigated inhibition of interleukin-1β (IL-1β)-induced EndMT by gemigliptin, a dipeptidyl peptidase-IV inhibitor. METHODS We exposed human umbilical vein endothelial cells (HUVECs) to 10 ng/mL IL-1β/20 μM gemigliptin and analyzed the expression of endothelial, smooth muscle, mesenchymal, and osteoblastic markers, bone morphogenetic protein (BMP), Smad, and non-Smad signaling pathway proteins. RESULTS Morphological changes showed gemigliptin blocked IL-1β-induced EndMT, upregulated EC markers, and downregulated smooth muscle and mesenchymal markers. IL-1β activation of HUVECs is initiated by the BMP/Smad and non-smad BMP signaling pathways. Gemigliptin inhibited IL-1β induction of BMP2 and 7, activin receptor type IA, BMP receptor type IA, and BMP receptor type II. Reversal of IL-1β-mediated inhibition of BMP-induced Smad1/5/8, Smad2, and Smad3 phosphorylation by gemigliptin suggests involvement of the Smad pathway in gemigliptin action. In the non-Smad BMP pathway, gemigliptin treatment significantly increased the deactivation of extracellular regulated protein kinase (ERK), p38, and JNK by IL-1β. Gemigliptin treatment suppressed BMP-2-induced expression of key osteoblastic markers including osterix, runt-related transcription factor 2, and hepcidin during IL-1β-induced EndMT. CONCLUSION We demonstrated a novel protective mechanism of gemigliptin against fibrosis by suppressing IL-1β-induced EndMT.
Collapse
Affiliation(s)
- Oak-Kee Hong
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Seong-Su Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Bucheon,
Korea
| | - Soon Jib Yoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Bucheon,
Korea
| | - Min-Kyung Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Myongji Hospital, Hanyang University Medical Center, Goyang,
Korea
| | - Mee-Kyoung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Ki-Hyun Baek
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Ki-Ho Song
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Hyuk-Sang Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| |
Collapse
|
48
|
Li Q, Wang F, Wang Q, Zhang N, Zheng J, Zheng M, Liu R, Cui H, Wen J, Zhao G. SPOP promotes ubiquitination and degradation of MyD88 to suppress the innate immune response. PLoS Pathog 2020; 16:e1008188. [PMID: 32365080 PMCID: PMC7224567 DOI: 10.1371/journal.ppat.1008188] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/14/2020] [Accepted: 04/15/2020] [Indexed: 02/03/2023] Open
Abstract
As a canonical adaptor for the Toll-like receptor (TLR) family, myeloid differentiation primary response protein 88 (MyD88) has crucial roles in host defense against infection by microbial pathogens, and its dysregulation might induce autoimmune diseases. Here, we demonstrate that the chicken Cullin 3-based ubiquitin ligase adaptor Speckle-type BTB-POZ protein (chSPOP) recognizes the intermediate domain of chicken MyD88 (chMyD88) and degrades it through the proteasome pathway. Knockdown or genetic ablation of chSPOP leads to aberrant elevation of chMyD88 protein. Through this interaction, chSPOP negatively regulates NF-κB pathway activity and thus the production of IL-1β upon LPS challenge in chicken macrophages. Furthermore, Spop-deficient mice are more susceptible to infection with Salmonella typhimurium. Collectively, these findings demonstrate MyD88 as a bona fide substrate of SPOP and uncover a mechanism by which SPOP regulates MyD88 abundance and disease susceptibility.
Collapse
Affiliation(s)
- Qinghe Li
- Institute of Animal Sciences; State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Fei Wang
- Institute of Animal Sciences; State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qiao Wang
- Institute of Animal Sciences; State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Na Zhang
- Institute of Animal Sciences; State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jumei Zheng
- Institute of Animal Sciences; State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Maiqing Zheng
- Institute of Animal Sciences; State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ranran Liu
- Institute of Animal Sciences; State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Huanxian Cui
- Institute of Animal Sciences; State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jie Wen
- Institute of Animal Sciences; State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, China
- * E-mail: (JW); (GZ)
| | - Guiping Zhao
- Institute of Animal Sciences; State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, China
- * E-mail: (JW); (GZ)
| |
Collapse
|
49
|
Jin R, Xu J, Gao Q, Mao X, Yin J, Lu K, Guo Y, Zhang M, Cheng R. IL-33-induced neutrophil extracellular traps degrade fibronectin in a murine model of bronchopulmonary dysplasia. Cell Death Discov 2020; 6:33. [PMID: 32377396 PMCID: PMC7198621 DOI: 10.1038/s41420-020-0267-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/06/2020] [Accepted: 04/16/2020] [Indexed: 12/30/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the leading cause of chronic lung disease in preterm neonates. Extracellular matrix (ECM) abnormalities reshape lung development, contributing to BPD progression. In the present study, we first discovered that the ECM component fibronectin was reduced in the pulmonary tissues of model mice with BPD induced by lipopolysaccharide (LPS) and hyper-oxygen. Meanwhile, interleukin-33 (IL-33) and other inflammatory cytokines were elevated in BPD lung tissues. LPS stimulated the production of IL-33 in alveolar epithelial cells via myeloid differentiation factor 88 (MyD88), protein 38 (p38), and nuclear factor-kappa B (NF-κB) protein 65 (p65). Following the knockout of either IL-33 or its receptor suppression of tumorigenicity 2 (ST2) in mice, BPD disease severity was improved, accompanied by elevated fibronectin. ST2 neutralization antibody also relieved BPD progression and restored the expression of fibronectin. IL-33 induced the formation of neutrophil extracellular traps (NETs), which degraded fibronectin in alveolar epithelial cells. Moreover, DNase-mediated degradation of NETs was protective against BPD. Finally, a fibronectin inhibitor directly decreased fibronectin and caused BPD-like disease in the mouse model. Our findings may shed light on the roles of IL-33-induced NETs and reduced fibronectin in the pathogenesis of BPD.
Collapse
Affiliation(s)
- Rui Jin
- Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Junjie Xu
- Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Qianqian Gao
- Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Xiaonan Mao
- Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Jiao Yin
- Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Keyu Lu
- Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Yan Guo
- Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Mingshun Zhang
- NHC Key Laboratory of Antibody Technique, Department of Immunology, Nanjing Medical University, 211166 Nanjing, China
| | - Rui Cheng
- Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, 210008 Nanjing, China
| |
Collapse
|
50
|
p38 MAPK signalling regulates cytokine production in IL-33 stimulated Type 2 Innate Lymphoid cells. Sci Rep 2020; 10:3479. [PMID: 32103032 PMCID: PMC7044202 DOI: 10.1038/s41598-020-60089-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 01/27/2020] [Indexed: 12/20/2022] Open
Abstract
Type 2 Innate lymphoid cells (ILC2s) are implicated in helminth infections and asthma where they play a role in the production of Th2-type cytokines. ILC2s express the IL-33 receptor and are a major cell type thought to mediate the effects of this cytokine in vivo. To study the signalling pathways that mediate IL-33 induced cytokine production, a culture system was set up to obtain pure populations of ILC2s from mice. Inhibitors of the p38α/β and ERK1/2 MAPK pathways reduced the production of IL-5, IL-6, IL-9, IL-13 and GM-CSF by ILC2 in response to IL-33, with inhibition of p38 having the greatest effect. MK2 and 3 are kinases activated by p38α; MK2/3 inhibitors or knockout of MK2/3 in mice reduced the production of IL-6 and IL-13 (two cytokines implicated in asthma) but not IL-5, IL-9 or GM-CSF in response to IL-33. MK2/3 inhibition also suppressed IL-6 and IL-13 production by human ILC2s. MK2/3 were required for maximal S6 phosphorylation, suggesting an input from the p38α-MK2/3 pathway to mTOR1 activation in ILC2s. The mTORC1 inhibitor rapamycin also reduced IL-6 and IL-13 production, which would be consistent with a model in which MK2/3 regulate IL-6 and IL-13 via mTORC1 activation in ILC2s.
Collapse
|