1
|
Mondell E, Nino G, Hong X, Wang X, Gutierrez MJ. Immune Biomarkers at Birth Predict Lower Respiratory Tract Infection Risk in a Large Birth Cohort. Pathogens 2024; 13:765. [PMID: 39338956 PMCID: PMC11435078 DOI: 10.3390/pathogens13090765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Lower respiratory tract infections (LRTIs) remain the leading cause of infant morbidity and mortality worldwide and affect long-term respiratory health. Identifying immunological determinants of LRTI susceptibility may help stratify disease risk and identify therapies. This study aimed to identify neonatal immunological factors predicting LRTI risk in infancy. Cord blood plasma from 191 neonates from the Boston Birth Cohort was analyzed for 28 soluble immune factors. LRTI was defined as bronchiolitis, bronchitis, or pneumonia during the first year of life. Welch's t-test demonstrated significantly higher log10 transformed concentrations of IL-17 and IFNγ in the LRTI group compared to neonates without LRTI in the first year of life (p < 0.05). Risk associations were determined using multivariate survival models. There were 29 infants with LRTIs. High cord blood levels of IFNγ (aHR = 2.35, 95% CI 1.07-5.17), TNF-β (aHR = 2.86, 95% CI 1.27-6.47), MIP-1α (aHR = 2.82, 95% CI 1.22-6.51), and MIP-1β (aHR = 2.34, 95% CI 1.05-5.20) were associated with a higher risk of LRTIs. RANTES was associated with a lower risk (aHR = 0.43, 95% CI 0.19-0.97). Soluble immune factors linked to antiviral immunity (IFNγ) and cytokines mediating inflammatory responses (TNF-β), and cell homing (MIP-1α/b), at birth were associated with an increased risk of LRTIs during infancy.
Collapse
Affiliation(s)
- Ethan Mondell
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Gustavo Nino
- Division of Pulmonary and Sleep Medicine, Children’s National Hospital, George Washington University, Washington, DC 20010, USA;
- Center for Genetic Medicine Research, Children’s Research Institute, Washington, DC 20010, USA
| | - Xiumei Hong
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (X.H.); (X.W.)
| | - Xiaobin Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (X.H.); (X.W.)
- Division of General Pediatrics and Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Maria J. Gutierrez
- Division of Pediatric Allergy, Immunology and Rheumatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
2
|
Grayck MR, McCarthy WC, Solar M, Balasubramaniyan N, Zheng L, Orlicky DJ, Wright CJ. Implications of neonatal absence of innate immune mediated NFκB/AP1 signaling in the murine liver. Pediatr Res 2024; 95:1791-1802. [PMID: 38396130 DOI: 10.1038/s41390-024-03071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/03/2024] [Accepted: 01/20/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND The developmental immaturity of the innate immune system helps explains the increased risk of infection in the neonatal period. Importantly, innate immune signaling pathways such as p65/NFκB and c-Jun/AP1 are responsible for the prevention of hepatocyte apoptosis in adult animals, yet whether developmental immaturity of these pathways increases the risk of hepatic injury in the neonatal period is unknown. METHODS Using a murine model of endotoxemia (LPS 5 mg/kg IP x 1) in neonatal (P3) and adult mice, we evaluated histologic evidence of hepatic injury and apoptosis, presence of p65/NFκB and c-Jun/AP1 activation and associated transcriptional regulation of apoptotic genes. RESULTS We demonstrate that in contrast to adults, endotoxemic neonatal (P3) mice exhibit a significant increase in hepatic apoptosis. This is associated with absent hepatic p65/NFκB signaling and impaired expression of anti-apoptotic target genes. Hepatic c-Jun/AP1 activity was attenuated in endotoxemic P3 mice, with resulting upregulation of pro-apoptotic factors. CONCLUSIONS These results demonstrate that developmental absence of innate immune p65/NFκB and c-Jun/AP1 signaling, and target gene expression is associated with apoptotic injury in neonatal mice. More work is needed to determine if this contributes to long-term hepatic dysfunction, and whether immunomodulatory approaches can prevent this injury. IMPACT Various aspects of developmental immaturity of the innate immune system may help explain the increased risk of infection in the neonatal period. In adult models of inflammation and infection, innate immune signaling pathways such as p65/NFκB and c-Jun/AP1 are responsible for a protective, pro-inflammatory transcriptome and regulation of apoptosis. We demonstrate that in contrast to adults, endotoxemic neonatal (P3) mice exhibit a significant increase in hepatic apoptosis associated with absent hepatic p65/NFκB signaling and c-Jun/AP1 activity. We believe that these results may explain in part hepatic dysfunction with neonatal sepsis, and that there may be unrecognized developmental and long-term hepatic implications of early life exposure to systemic inflammatory stress.
Collapse
Affiliation(s)
- Maya R Grayck
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - William C McCarthy
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Mack Solar
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Natarajan Balasubramaniyan
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lijun Zheng
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - David J Orlicky
- Dept of Pathology, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
3
|
Rodriguez BN, Huang H, Chia JJ, Hoffmann A. The noncanonical NFκB pathway: Regulatory mechanisms in health and disease. WIREs Mech Dis 2024:e1646. [PMID: 38634218 PMCID: PMC11486840 DOI: 10.1002/wsbm.1646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/19/2024]
Abstract
The noncanonical NFκB signaling pathway mediates the biological functions of diverse cell survival, growth, maturation, and differentiation factors that are important for the development and maintenance of hematopoietic cells and immune organs. Its dysregulation is associated with a number of immune pathologies and malignancies. Originally described as the signaling pathway that controls the NFκB family member RelB, we now know that noncanonical signaling also controls NFκB RelA and cRel. Here, we aim to clarify our understanding of the molecular network that mediates noncanonical NFκB signaling and review the human diseases that result from a deficient or hyper-active noncanonical NFκB pathway. It turns out that dysregulation of RelA and cRel, not RelB, is often implicated in mediating the resulting pathology. This article is categorized under: Immune System Diseases > Molecular and Cellular Physiology Cancer > Molecular and Cellular Physiology Immune System Diseases > Stem Cells and Development.
Collapse
Affiliation(s)
- Benancio N. Rodriguez
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, CA; Molecular Biology Institute, Los Angeles, CA
| | - Helen Huang
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, CA; Institute for Quantitative and Computational Biosciences, Los Angeles, CA
| | - Jennifer J. Chia
- Department of Microbiology, Immunology, and Molecular Genetics, Los Angeles, CA; Molecular Biology Institute, Los Angeles, Calif; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA
| | - Alexander Hoffmann
- Department of Microbiology, Immunology, and Molecular Genetics; Molecular Biology Institute; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA
| |
Collapse
|
4
|
Shynlova O, Nadeem L, Lye S. Progesterone control of myometrial contractility. J Steroid Biochem Mol Biol 2023; 234:106397. [PMID: 37683774 DOI: 10.1016/j.jsbmb.2023.106397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
During pregnancy, the primary function of the uterus is to be quiescent and not contract, which allows the growing fetus to develop and mature. A uterine muscle layer, myometrium, is composed of smooth muscle cells (SMCs). Before the onset of labor contractions, the uterine SMCs experience a complex biochemical and molecular transformation involving the expression of contraction-associated proteins. Labor is initiated when genes in SMCs are activated in response to a combination of hormonal, inflammatory and mechanical signals. In this review, we provide an overview of molecular mechanisms regulating the process of parturition in humans, focusing on the hormonal control of the myometrium, particularly the steroid hormone progesterone. The primary reason for discussing the regulation of myometrial contractility by progesterone is the importance of the clinical problem of preterm birth. It is thought that the hormonal mechanisms regulating premature uterine contractions represent an untimely triggering of the normal events occurring during term parturition. Yet, our knowledge of the complex and redundant hormonal pathways controlling uterine contractile activity leading to delivery of the neonate remains incomplete. Finally, we introduce recent animal studies using a novel class of drugs, Selective Progesterone Receptor Modulators, targeting progesterone signaling to prevent premature myometrial contractions.
Collapse
Affiliation(s)
- Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada; Department of Physiology, University of Toronto, M5S 1A1, Canada; Department of Obstetrics & Gynecology, University of Toronto, M5S 1A1, Canada.
| | - Lubna Nadeem
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada
| | - Stephen Lye
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada; Department of Physiology, University of Toronto, M5S 1A1, Canada; Department of Obstetrics & Gynecology, University of Toronto, M5S 1A1, Canada
| |
Collapse
|
5
|
Chatterjee T, Lewis TL, Arora I, Gryshyna AE, Underwood L, Masjoan Juncos JX, Aggarwal S. Sex-Based Disparities in Leukocyte Migration and Activation in Response to Inhalation Lung Injury: Role of SDF-1/CXCR4 Signaling. Cells 2023; 12:1719. [PMID: 37443753 PMCID: PMC10340292 DOI: 10.3390/cells12131719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of the study was to determine whether sex-related differences exist in immune response to inhalation lung injury. C57BL/6 mice were exposed to Cl2 gas (500 ppm for 15, 20, or 30 min). Results showed that male mice have higher rates of mortality and lung injury than females. The binding of the chemokine ligand C-X-C motif chemokine 12 (CXCL12), also called stromal-derived-factor-1 (SDF-1), to the C-X-C chemokine receptor type 4 (CXCR4) on lung cells promotes the migration of leukocytes from circulation to lungs. Therefore, the hypothesis was that elevated SDF-1/CXCR4 signaling mediates exaggerated immune response in males. Plasma, blood leukocytes, and lung cells were collected from mice post-Cl2 exposure. Plasma levels of SDF-1 and peripheral levels of CXCR4 in lung cells were higher in male vs. female mice post-Cl2 exposure. Myeloperoxidase (MPO) and elastase activity was significantly increased in leukocytes of male mice exposed to Cl2. Lung cells were then ex vivo treated with SDF-1 (100 ng/mL) in the presence or absence of the CXCR4 inhibitor, AMD3100 (100 nM). SDF-1 significantly increased migration, MPO, and elastase activity in cells obtained from male vs. female mice post-Cl2 exposure. AMD3100 attenuated these effects, suggesting that differential SDF-1/CXCR4 signaling may be responsible for sex-based disparities in the immune response to inhalation lung injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Saurabh Aggarwal
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35205-3703, USA; (T.C.); (T.L.L.); (I.A.); (A.E.G.); (L.U.); (J.X.M.J.)
| |
Collapse
|
6
|
Huyghe J, Priem D, Bertrand MJM. Cell death checkpoints in the TNF pathway. Trends Immunol 2023:S1471-4906(23)00105-9. [PMID: 37357102 DOI: 10.1016/j.it.2023.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 06/27/2023]
Abstract
Tumor necrosis factor (TNF) plays a central role in orchestrating mammalian inflammatory responses. It promotes inflammation either directly by inducing inflammatory gene expression or indirectly by triggering cell death. TNF-mediated cell death-driven inflammation can be beneficial during infection by providing cell-extrinsic signals that help to mount proper immune responses. Uncontrolled cell death caused by TNF is instead highly detrimental and is believed to cause several human autoimmune diseases. Death is not the default response to TNF sensing. Molecular brakes, or cell death checkpoints, actively repress TNF cytotoxicity to protect the organism from its detrimental consequences. These checkpoints therefore constitute essential safeguards against inflammatory diseases. Recent advances in the field have revealed the existence of several new and unexpected brakes against TNF cytotoxicity and pathogenicity.
Collapse
Affiliation(s)
- Jon Huyghe
- Cell Death and Inflammation Unit, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Dario Priem
- Cell Death and Inflammation Unit, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Mathieu J M Bertrand
- Cell Death and Inflammation Unit, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium.
| |
Collapse
|
7
|
van Muilekom DR, Collet B, Rebl H, Zlatina K, Sarais F, Goldammer T, Rebl A. Lost and Found: The Family of NF-κB Inhibitors Is Larger than Assumed in Salmonid Fish. Int J Mol Sci 2023; 24:10229. [PMID: 37373375 DOI: 10.3390/ijms241210229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/14/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
NF-κB signalling is largely controlled by the family of 'inhibitors of NF-κB' (IκB). The relevant databases indicate that the genome of rainbow trout contains multiple gene copies coding for iκbα (nfkbia), iκbε (nfkbie), iκbδ (nkfbid), iκbζ (nfkbiz), and bcl3, but it lacks iκbβ (nfkbib) and iκbη (ankrd42). Strikingly, three nfkbia paralogs are apparently present in salmonid fish, two of which share a high sequence identity, while the third putative nfkbia gene is significantly less like its two paralogs. This particular nfkbia gene product, iκbα, clusters with the human IκBβ in a phylogenetic analysis, while the other two iκbα proteins from trout associate with their human IκBα counterpart. The transcript concentrations were significantly higher for the structurally more closely related nfkbia paralogs than for the structurally less similar paralog, suggesting that iκbβ probably has not been lost from the salmonid genomes but has been incorrectly designated as iκbα. In the present study, two gene variants coding for iκbα (nfkbia) and iκbε (nfkbie) were prominently expressed in the immune tissues and, particularly, in a cell fraction enriched with granulocytes, monocytes/macrophages, and dendritic cells from the head kidney of rainbow trout. Stimulation of salmonid CHSE-214 cells with zymosan significantly upregulated the iκbα-encoding gene while elevating the copy numbers of the inflammatory markers interleukin-1-beta and interleukin-8. Overexpression of iκbα and iκbε in CHSE-214 cells dose-dependently quenched both the basal and stimulated activity of an NF-κB promoter suggesting their involvement in immune-regulatory processes. This study provides the first functional data on iκbε-versus the well-researched iκbα factor-in a non-mammalian model species.
Collapse
Affiliation(s)
- Doret R van Muilekom
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Bertrand Collet
- VIM, UVSQ, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Henrike Rebl
- Department of Cell Biology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Kristina Zlatina
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Fabio Sarais
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Tom Goldammer
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
- Faculty of Agriculture and Environmental Sciences, University of Rostock, 18059 Rostock, Germany
| | - Alexander Rebl
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| |
Collapse
|
8
|
Wang J, He L, Wang S, Zhao H, Chen J, Dong Y, Yasen S, Wang L, Zou H. Therapeutic effect of the total saponin from Panax Japonicus on experimental autoimmune encephalomyelitis by attenuating inflammation and regulating gut microbiota in mice. JOURNAL OF ETHNOPHARMACOLOGY 2023:116681. [PMID: 37230280 DOI: 10.1016/j.jep.2023.116681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rhizomes of Panax japonicus (RPJ), a traditional herbal medicine, was used for treating arthritis and physical weakness in China from the Ming dynasty. Triterpene saponins are the main bioactive components of RPJ. In this work, for the first time, we evaluate the therapeutic effect of the total saponin from RPJ (TSPJ) on experimental autoimmune encephalomyelitis (EAE) mice induced by myelin oligodendrocyte glycoprotein (MOG) 35-55, a commonly used animal model of Multiple sclerosis (MS). AIM OF THE STUDY To evaluate the therapeutic effect of TSPJ on EAE and explored its possible underlying mechanisms. MATERIALS AND METHODS EAE was induced by MOG 35-55. Mice were administrated with TSPJ (36.5 mg/kg, 73 mg/kg) and prednisone acetate (positive control) orally once daily up to 28 days postimmunization, and their neurological deficit was scored. Hematoxylin and Eosin (HE), Luxol Fast Blue (LFB), and transmission electron microscopy (TEM) were carried out to evaluate the EAE-induced pathological changes in the brain and spinal cord. IL-17a and Foxp3 levels in central nervous system(CNS)were evaluated by immunohistochemical staining. The changes in IL-1β, IL-6, and TNF-α levels in serum and CNS were measured with ELISA. Quantitative reverse transcription PCR (qRT-PCR) was used to access mRNA expression in CNS of the above indices. The percentages of Th1, Th2, Th17and Treg cells in spleen were determined by Flow Cytometry (FCM). Furthermore, 16S rDNA sequencing was used to detect the intestinal flora of mice in each group. In vitro studies, lipopolysaccharides (LPS)-induced BV2 microglia cells were used and the expression of TLR4, MyD88, p65, and p-p65 in cells was detected by Western blot. RESULTS TSPJ treatment significantly alleviated neurological impairment caused by EAE. Histological examination confirmed the protective effects of TSPJ on myelin sheath and the reduction of inflammatory cell infiltration in the brain and spinal cord of EAE mice. TSPJ notably downregulated the ratio of IL-17a/Foxp3 at protein and mRNA levels in CNS, as well as Th17/Treg and Th1/Th2 cell ratios in the spleen of EAE mice. The levels of TNF-α, IL-6, and IL-1β in CNS and peripheral serum also decreased post-TSPJ treatment. In vitro, TSPJ suppressed LPS-induced production of inflammatory factors in BV2 cells via TLR4-MyD88-NF-κB signaling pathway. More importantly, TSPJ interventions altered the composition of gut microbiota and restored the ratio of Firmicutes to Bacteroidetes in EAE mice. Furthermore, Spearman's correlation analysis revealed that a relationship existed between statistically significantly altered genera and CNS inflammatory indices. CONCLUSION Our results demonstrated TSPJ had therapeutic effects on EAE. Its anti-neuroinflammation property in EAE was related to modulating gut microbiota and inhibiting TLR4-MyD88-NF-κB signaling pathway. Our study indicated that TSPJ may be a potential candidate for the treatment of MS.
Collapse
Affiliation(s)
- Jing Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Liying He
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Siyuan Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Jie Chen
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Yixin Dong
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Subinuer Yasen
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Lei Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Haiyan Zou
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
9
|
Lv D, Zhao X. Evaluation of the anti-RANKL monoclonal antibody in rheumatoid arthritis rats. Arch Rheumatol 2023; 38:22-31. [DOI: 10.46497/archrheumatol.2023.9240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/30/2021] [Indexed: 03/18/2023] Open
Abstract
Objectives: In this study, we aimed to investigate the therapeutic effect of anti-receptor activator of nuclear factor kappa-κB ligand (RANKL) monoclonal antibodies R748-1-1-1, R748-1-1-2 and R748-1-1-3 on rheumatoid arthritis (RA) in a rat model.
Materials and methods: Gene cloning, hybridoma technology, affinity purification, enzyme-linked immunosorbent assay, general observation, hematoxylin-eosin staining, X-ray, and many other experimental techniques were used in this study.
Results: Improved collagen-induced arthritis (CIA) modeling was successfully constructed. The RANKL gene was cloned and the anti-RANKL monoclonal antibody was prepared. Following treatment with the anti-RANKL monoclonal antibody, the soft tissue swelling of the hind paws, the joint thickening, the narrowed joint gap, and the blurred edge of the bone joint were improved. The pathological changes such as synovial hyperplasia of fibrous tissue, cartilage and bone destruction were significantly decreased in the anti-RANKL monoclonal antibody-treated CIA group. Compared to the normal control group and phosphate buffer saline (PBS)-treated CIA group, the expression of tumor necrosis factor-alpha (TNF-α) and interleukin-1 (IL-1) in antibody-treated CIA group, positive drug-treated CIA group, and IgG-treated CIA group were decreased (p<0.05).
Conclusion: The anti-RANKL monoclonal antibody can promote the therapeutic effect of RA rats, indicating that the anti-RANKL monoclonal antibody has a certain potential value and may be beneficial to the further study of the mechanism of RA treatment.
Collapse
|
10
|
RIP1 post-translational modifications. Biochem J 2022; 479:929-951. [PMID: 35522161 DOI: 10.1042/bcj20210725] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 11/17/2022]
Abstract
Receptor interacting protein 1 (RIP1) kinase is a critical regulator of inflammation and cell death signaling, and plays a crucial role in maintaining immune responses and proper tissue homeostasis. Mounting evidence argues for the importance of RIP1 post-translational modifications in control of its function. Ubiquitination by E3 ligases, such as inhibitors of apoptosis (IAP) proteins and LUBAC, as well as the reversal of these modifications by deubiquitinating enzymes, such as A20 and CYLD, can greatly influence RIP1 mediated signaling. In addition, cleavage by caspase-8, RIP1 autophosphorylation, and phosphorylation by a number of signaling kinases can greatly impact cellular fate. Disruption of the tightly regulated RIP1 modifications can lead to signaling disbalance in TNF and/or TLR controlled and other inflammatory pathways, and result in severe human pathologies. This review will focus on RIP1 and its many modifications with an emphasis on ubiquitination, phosphorylation, and cleavage, and their functional impact on the RIP1's role in signaling pathways.
Collapse
|
11
|
Palmer CS, Kimmey JM. Neutrophil Recruitment in Pneumococcal Pneumonia. Front Cell Infect Microbiol 2022; 12:894644. [PMID: 35646729 PMCID: PMC9136017 DOI: 10.3389/fcimb.2022.894644] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/18/2022] [Indexed: 01/19/2023] Open
Abstract
Streptococcus pneumoniae (Spn) is the primary agent of community-acquired pneumonia. Neutrophils are innate immune cells that are essential for bacterial clearance during pneumococcal pneumonia but can also do harm to host tissue. Neutrophil migration in pneumococcal pneumonia is therefore a major determinant of host disease outcomes. During Spn infection, detection of the bacterium leads to an increase in proinflammatory signals and subsequent expression of integrins and ligands on both the neutrophil as well as endothelial and epithelial cells. These integrins and ligands mediate the tethering and migration of the neutrophil from the bloodstream to the site of infection. A gradient of host-derived and bacterial-derived chemoattractants contribute to targeted movement of neutrophils. During pneumococcal pneumonia, neutrophils are rapidly recruited to the pulmonary space, but studies show that some of the canonical neutrophil migratory machinery is dispensable. Investigation of neutrophil migration is necessary for us to understand the dynamics of pneumococcal infection. Here, we summarize what is known about the pathways that lead to migration of the neutrophil from the capillaries to the lung during pneumococcal infection.
Collapse
Affiliation(s)
| | - Jacqueline M. Kimmey
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
12
|
Hu F, Zhang Y, Liu Q, Wang Z. PurA facilitates Edwardsiella piscicida to escape NF-κB signaling activation. FISH & SHELLFISH IMMUNOLOGY 2022; 124:254-260. [PMID: 35395412 DOI: 10.1016/j.fsi.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/01/2022] [Accepted: 04/01/2022] [Indexed: 06/14/2023]
Abstract
The host NF-κB signaling pathway plays critical role in defensing against bacterial infection. However, bacteria also evolve strategies to escape from host clearance. Edwardsiella piscicida is a threatening pathogen in aquaculture, while the molecular mechanism of E. piscicida in inhibiting NF-κB signaling remains largely unknown. Herein, using E. piscicida transposon insertion mutant library combined with a NF-κB luciferase reporter system, we identified forty-six genes of E. piscicida, which were involved in inhibiting the NF-κB signaling activation in vitro. Moreover, we further explored the top 10 significantly changed mutants through zebrafish larvae infection model and validated that six genes were involved in inhibiting NF-κB activation in vivo. Specifically, we identified the adenylosuccinate synthase mutated strain (ΔpurA) infection exhibited a robust activation of NF-κB signaling, along with higher expression of cxcl8a and cxcl8b to mediate the recruitment of neutrophils in vivo. Taken together, these results identified the key factors of E. piscicida in inhibiting NF-κB activation, which will contribute to better understanding the pathogenesis of this important pathogen.
Collapse
Affiliation(s)
- Feizi Hu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yuanxing Zhang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, 200237, China
| | - Qin Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, 200237, China
| | - Zhuang Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
13
|
Li J, Zhang N, Li M, Hong T, Meng W, Ouyang T. The Emerging Role of OTUB2 in Diseases: From Cell Signaling Pathway to Physiological Function. Front Cell Dev Biol 2022; 10:820781. [PMID: 35309903 PMCID: PMC8926145 DOI: 10.3389/fcell.2022.820781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
Ovarian tumor (OTU) domain-containing ubiquitin aldehyde-binding protein Otubain2 (OTUB2) was a functional cysteine protease in the OTU family with deubiquitinase activity. In recent years, with the wide application of molecular biology techniques, molecular mechanism regulation at multiple levels of cell signaling pathways has been gradually known, such as ubiquitin-mediated protein degradation and phosphorylation-mediated protein activation. OTUB2 is involved in the deubiquitination of many key proteins in different cell signaling pathways, and the effect of OTUB2 on human health or disease is not clear. OTUB2 is likely to cause cancer and other malignant diseases while maintaining normal human development and physiological function. Therefore, it is of great value to comprehensively understand the regulatory mechanism of OTUB2 and regard it as a target for the treatment of diseases. This review makes a general description and appropriate analysis of OTUB2's regulation in different cell signaling pathways, and connects OTUB2 with cancer from the research hotspot perspective of DNA damage repair and immunity, laying the theoretical foundation for future research.
Collapse
Affiliation(s)
- Jun Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Jiangxi, China.,Department of the Second Clinical Medical College of Nanchang University, Jiangxi, China
| | - Na Zhang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Wei Meng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Taohui Ouyang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| |
Collapse
|
14
|
Cerps S, Sverrild A, Ramu S, Nieto‐Fontarigo JJ, Akbarshahi H, Menzel M, Andersson C, Tillgren S, Hvidtfeldt M, Porsbjerg C, Uller L. House dust mite sensitization and exposure affects bronchial epithelial anti-microbial response to viral stimuli in patients with asthma. Allergy 2022; 77:2498-2508. [PMID: 35114024 PMCID: PMC9546181 DOI: 10.1111/all.15243] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/22/2021] [Accepted: 01/02/2022] [Indexed: 12/01/2022]
Abstract
Introduction Allergen exposure worsens viral‐triggered asthma exacerbations and could predispose the host to secondary bacterial infections. We have previously demonstrated that exposure to house dust mite (HDM) reduced TLR‐3‐induced IFN‐β in human bronchial epithelial cells (HBECs) from healthy donors. We hypothesize that HDM sensitization in different ways may be involved in both viral and bacterial resistance of HBECs in asthma. In this study, the role of HDM sensitization and effects of HDM exposure on viral stimulus‐challenged HBECs from asthmatic donors have been explored with regard to expression and release of molecules involved in anti‐viral and anti‐bacterial responses, respectively. Methods HBECs from HDM‐sensitized (HDM+) and unsensitized (HDM‐) patients with asthma were used. HBECs were exposed to HDM or heat inactivated (hi)‐HDM (20 μg/ml) for 24 h prior to stimulation with the viral infection mimic, Poly(I:C), for 3 or 24 h. Samples were analyzed with ELISA and RT‐qPCR for β‐defensin‐2, IFN‐β, TSLP, and neutrophil‐recruiting mediators: IL‐8 and TNF‐⍺. NFκB signaling proteins p105, p65, and IκB‐⍺ were analyzed by Western blot. Results Poly(I:C)‐induced IFN‐β expression was reduced in HBECs from HDM + compared to HDM‐ patients (p = 0.05). In vitro exposure of HBECs to HDM furthermore reduced anti‐microbial responses to Poly(I:C) including β‐defensin‐2, IL‐8, and TNF‐⍺, along with reduced NFκB activity. This was observed in HBECs from asthma patients sensitized to HDM, as well as in non‐sensitized patients. By contrast, Poly (I:C)‐induced release of TSLP, a driver of T2 inflammation, was not reduced with exposure to HDM. Conclusion Using HBECs challenged with viral infection mimic, Poly(I:C), we demonstrated that allergic sensitization to HDM was associated with impaired anti‐viral immunity and that HDM exposure reduced anti‐viral and anti‐bacterial defense molecules, but not TSLP, across non‐allergic as well as allergic asthma. These data suggest a role of HDM in the pathogenesis of asthma exacerbations evoked by viral infections including sequential viral‐bacterial and viral‐viral infections.
Collapse
Affiliation(s)
- Samuel Cerps
- Department of Experimental Medical Science BMC D12 Lund University Lund Sweden
| | - Asger Sverrild
- Department of Respiratory Medicine University Hospital Bispebjerg Copenhagen Denmark
| | - Sangeetha Ramu
- Department of Experimental Medical Science BMC D12 Lund University Lund Sweden
| | | | - Hamid Akbarshahi
- Department of Experimental Medical Science BMC D12 Lund University Lund Sweden
| | - Mandy Menzel
- Department of Experimental Medical Science BMC D12 Lund University Lund Sweden
| | - Cecilia Andersson
- Department of Experimental Medical Science BMC D12 Lund University Lund Sweden
| | - Sofia Tillgren
- Department of Experimental Medical Science BMC D12 Lund University Lund Sweden
| | - Morten Hvidtfeldt
- Department of Respiratory Medicine University Hospital Bispebjerg Copenhagen Denmark
| | - Celeste Porsbjerg
- Department of Respiratory Medicine University Hospital Bispebjerg Copenhagen Denmark
| | - Lena Uller
- Department of Experimental Medical Science BMC D12 Lund University Lund Sweden
| |
Collapse
|
15
|
Zhang Y, Huang K, Zhang Y, Han T, Li L, Ruan C, Sun YH, Shi W, Han W, Wu SQ, Song J, Liu J, Han J. A unique death pathway keeps RIPK1 D325A mutant mice in check at embryonic day 10.5. PLoS Biol 2021; 19:e3001304. [PMID: 34437534 PMCID: PMC8389420 DOI: 10.1371/journal.pbio.3001304] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/27/2021] [Indexed: 01/10/2023] Open
Abstract
Tumor necrosis factor receptor-1 (TNFR1) signaling, apart from its pleiotropic functions in inflammation, plays a role in embryogenesis as deficiency of varieties of its downstream molecules leads to embryonic lethality in mice. Caspase-8 noncleavable receptor interacting serine/threonine kinase 1 (RIPK1) mutations occur naturally in humans, and the corresponding D325A mutation in murine RIPK1 leads to death at early midgestation. It is known that both the demise of Ripk1D325A/D325A embryos and the death of Casp8-/- mice are initiated by TNFR1, but they are mediated by apoptosis and necroptosis, respectively. Here, we show that the defects in Ripk1D325A/D325A embryos occur at embryonic day 10.5 (E10.5), earlier than that caused by Casp8 knockout. By analyzing a series of genetically mutated mice, we elucidated a mechanism that leads to the lethality of Ripk1D325A/D325A embryos and compared it with that underlies Casp8 deletion-mediated lethality. We revealed that the apoptosis in Ripk1D325A/D325A embryos requires a scaffold function of RIPK3 and enzymatically active caspase-8. Unexpectedly, caspase-1 and caspase-11 are downstream of activated caspase-8, and concurrent depletion of Casp1 and Casp11 postpones the E10.5 lethality to embryonic day 13.5 (E13.5). Moreover, caspase-3 is an executioner of apoptosis at E10.5 in Ripk1D325A/D325A mice as its deletion extends life of Ripk1D325A/D325A mice to embryonic day 11.5 (E11.5). Hence, an unexpected death pathway of TNFR1 controls RIPK1 D325A mutation-induced lethality at E10.5.
Collapse
Affiliation(s)
- Yingying Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Kai Huang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yuxia Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Tao Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Lang Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chenchen Ruan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Ye-hsuan Sun
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Wenke Shi
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Wei Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Su-qin Wu
- Laboratory Animal Center, Xiamen University, Xiamen, Fujian, China
| | - Jing Song
- Laboratory Animal Center, Xiamen University, Xiamen, Fujian, China
| | - Jun Liu
- Laboratory Animal Center, Xiamen University, Xiamen, Fujian, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Laboratory Animal Center, Xiamen University, Xiamen, Fujian, China
- Research Unit of Cellular Stress of CAMS, Cancer Research Center of Xiamen University, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
- * E-mail:
| |
Collapse
|
16
|
Shen D, Chu F, Lang Y, Zheng C, Li C, Liu K, Zhu J. Nuclear factor kappa B inhibitor suppresses experimental autoimmune neuritis in mice via declining macrophages polarization to M1 type. Clin Exp Immunol 2021; 206:110-117. [PMID: 34118070 DOI: 10.1111/cei.13637] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/10/2021] [Accepted: 06/10/2021] [Indexed: 11/28/2022] Open
Abstract
Guillain-Barré syndrome (GBS) is an acute inflammatory and immune-mediated demyelinating disease of the peripheral nervous system (PNS). Macrophages play a central role in its animal model, experimental autoimmune neuritis (EAN), which has been well accepted. Additionally, nuclear factor (NF)-κB inhibitors have been used to treat cancers and have shown beneficial effects. Here, we investigated the therapeutic effect of M2 macrophage and the NF-κB pathway's correlation with macrophage activation in EAN in C57BL/6 mice. We demonstrate that M2 macrophage transfusion could alleviate the clinical symptoms of EAN by reducing the proportion of M1 macrophage in the peak period, inhibiting the phosphorylation of NF-κB p65. The NF-κB inhibitor (BAY-11-7082) could alleviate the clinical symptoms of EAN and shorten the duration of symptoms by reducing the proportion of M1 macrophages and the expression of proinflammatory cytokines. Consequently, BAY-11-7082 exhibits strong potential as a therapeutic strategy for ameliorating EAN by influencing the balance of M1/M2 macrophages and inflammatory cytokines.
Collapse
Affiliation(s)
- Donghui Shen
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Neuroscience Center, Department of Neurology, Qingdao Municipal Hospital, Qingdao, China
| | - Fengna Chu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Yue Lang
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Chao Zheng
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Chunrong Li
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Kangding Liu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
17
|
Williams TC, Jackson DJ, Maltby S, Walton RP, Ching YM, Glanville N, Singanayagam A, Brewins JJ, Clarke D, Hirsman AG, Loo SL, Wei L, Beale JE, Casolari P, Caramori G, Papi A, Belvisi M, Wark PAB, Johnston SL, Edwards MR, Bartlett NW. Rhinovirus-induced CCL17 and CCL22 in Asthma Exacerbations and Differential Regulation by STAT6. Am J Respir Cell Mol Biol 2021; 64:344-356. [PMID: 33264064 PMCID: PMC7909342 DOI: 10.1165/rcmb.2020-0011oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 10/19/2020] [Indexed: 12/17/2022] Open
Abstract
The interplay of type-2 inflammation and antiviral immunity underpins asthma exacerbation pathogenesis. Virus infection induces type-2 inflammation-promoting chemokines CCL17 and CCL22 in asthma; however, mechanisms regulating induction are poorly understood. By using a human rhinovirus (RV) challenge model in human airway epithelial cells in vitro and mice in vivo, we assessed mechanisms regulating CCL17 and CCL22 expression. Subjects with mild to moderate asthma and healthy volunteers were experimentally infected with RV and airway CCL17 and CCL22 protein quantified. In vitro airway epithelial cell- and mouse-RV infection models were then used to define STAT6- and NF-κB-mediated regulation of CCL17 and CCL22 expression. Following RV infection, CCL17 and CCL22 expression was higher in asthma, which differentially correlated with clinical and immunological parameters. Air-liquid interface-differentiated primary epithelial cells from donors with asthma also expressed higher levels of RV-induced CCL22. RV infection boosted type-2 cytokine-induced STAT6 activation. In epithelial cells, type-2 cytokines and STAT6 activation had differential effects on chemokine expression, increasing CCL17 and suppressing CCL22, whereas NF-κB promoted expression of both chemokines. In mice, RV infection activated pulmonary STAT6, which was required for CCL17 but not CCL22 expression. STAT6-knockout mice infected with RV expressed increased levels of NF-κB-regulated chemokines, which was associated with rapid viral clearance. Therefore, RV-induced upregulation of CCL17 and CCL22 was mediated by NF-κB activation, whereas expression was differentially regulated by STAT6. Together, these findings suggest that therapeutic targeting of type-2 STAT6 activation alone will not block all inflammatory pathways during RV infection in asthma.
Collapse
Affiliation(s)
- Teresa C. Williams
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - David J. Jackson
- Asthma UK Centre, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
- Guy’s Severe Asthma Centre, Guy’s & St. Thomas’ National Health Service Trust, London, United Kingdom
| | - Steven Maltby
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Ross P. Walton
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Yee-Mann Ching
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nicholas Glanville
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Aran Singanayagam
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jennifer J. Brewins
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Deborah Clarke
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Respiratory, Inflammation and Autoimmunity Department, MedImmune, Cambridge, United Kingdom
| | - Aurica G. Hirsman
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Su-Ling Loo
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Lan Wei
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Janine E. Beale
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Paolo Casolari
- Interdepartmental Study Center for Inflammatory and Smoke-Related Airway Diseases, Cardiorespiratory and Internal Medicine Section, University of Ferrara, Ferrara, Italy
| | - Gaetano Caramori
- Interdepartmental Study Center for Inflammatory and Smoke-Related Airway Diseases, Cardiorespiratory and Internal Medicine Section, University of Ferrara, Ferrara, Italy
- Dipartimento di Scienze Biomediche, Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali, Università degli Studi di Messina, Messina, Italy; and
| | - Alberto Papi
- Interdepartmental Study Center for Inflammatory and Smoke-Related Airway Diseases, Cardiorespiratory and Internal Medicine Section, University of Ferrara, Ferrara, Italy
| | - Maria Belvisi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Respiratory, Inflammation and Autoimmunity Department, MedImmune, Cambridge, United Kingdom
| | - Peter A. B. Wark
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | | | - Michael R. Edwards
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nathan W. Bartlett
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
18
|
Lin WC, Fessler MB. Regulatory mechanisms of neutrophil migration from the circulation to the airspace. Cell Mol Life Sci 2021; 78:4095-4124. [PMID: 33544156 PMCID: PMC7863617 DOI: 10.1007/s00018-021-03768-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/22/2020] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
The neutrophil, a short-lived effector leukocyte of the innate immune system best known for its proteases and other degradative cargo, has unique, reciprocal physiological interactions with the lung. During health, large numbers of ‘marginated’ neutrophils reside within the pulmonary vasculature, where they patrol the endothelial surface for pathogens and complete their life cycle. Upon respiratory infection, rapid and sustained recruitment of neutrophils through the endothelial barrier, across the extravascular pulmonary interstitium, and again through the respiratory epithelium into the airspace lumen, is required for pathogen killing. Overexuberant neutrophil trafficking to the lung, however, causes bystander tissue injury and underlies several acute and chronic lung diseases. Due in part to the unique architecture of the lung’s capillary network, the neutrophil follows a microanatomic passage into the distal airspace unlike that observed in other end-organs that it infiltrates. Several of the regulatory mechanisms underlying the stepwise recruitment of circulating neutrophils to the infected lung have been defined over the past few decades; however, fundamental questions remain. In this article, we provide an updated review and perspective on emerging roles for the neutrophil in lung biology, on the molecular mechanisms that control the trafficking of neutrophils to the lung, and on past and ongoing efforts to design therapeutics to intervene upon pulmonary neutrophilia in lung disease.
Collapse
Affiliation(s)
- Wan-Chi Lin
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC, 27709, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
19
|
Rombauts A, Abelenda-Alonso G, Cuervo G, Gudiol C, Carratalà J. Role of the inflammatory response in community-acquired pneumonia: clinical implications. Expert Rev Anti Infect Ther 2021; 20:1261-1274. [PMID: 33034228 DOI: 10.1080/14787210.2021.1834848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Despite adequate antibiotic coverage, community-acquired pneumonia (CAP) remains a leading cause of hospitalization and mortality worldwide. It induces both a local pulmonary and a systemic inflammatory response, particularly significant in severe cases. The intensity of the dysregulated host response varies from patient to patient and has a negative impact on survival and other outcomes. AREAS COVERED This comprehensive review summarizes the pathophysiological aspects of the inflammatory response in CAP, briefly discusses the usefulness of biomarkers, and assesses the clinical evidence for modulating the inflammatory pathways. We searched PubMed for the most relevant studies, reviews, and meta-analysis until August 2020. EXPERT OPINION Notable efforts have been made to identify biomarkers that can accurately differentiate between viral and bacterial etiology, and indeed, to enhance risk stratification in CAP. However, none has proven ideal and no recommended biomarker-guided algorithms exist. Biomarker signatures from proteomic and metabolomic studies could be more useful for such assessments. To date, most studies have produced contradictory results concerning the role of immunomodulatory agents (e.g. corticosteroids, macrolides, and statins) in CAP. Adequately identifying the population who may benefit most from effective modulation of the inflammatory response remains a challenge.
Collapse
Affiliation(s)
- Alexander Rombauts
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Gabriela Abelenda-Alonso
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Guillermo Cuervo
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain
| | - Carlota Gudiol
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain.,Spanish Network for Research in Infectious Disease (REIPI), Instituto de Salud Carlos III, Madrid, Spain.,University of Barcelona, Barcelona, Spain.,Institut Català d'Oncologia (ICO), Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Carratalà
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain.,Spanish Network for Research in Infectious Disease (REIPI), Instituto de Salud Carlos III, Madrid, Spain.,University of Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Wang LY, Liu ZX, Zhao LM, Huang LX, Qin YX, Su YQ, Zheng WQ, Wang F, Yan QP. Dual RNA-seq provides novel insight into the roles of dksA from Pseudomonas plecoglossicida in pathogen-host interactions with large yellow croakers ( Larimichthys crocea). Zool Res 2020; 41:410-422. [PMID: 32521576 PMCID: PMC7340521 DOI: 10.24272/j.issn.2095-8137.2020.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
Pseudomonas plecoglossicida is a rod-shaped, gram-negative bacterium with flagella. It causes visceral white spot disease and high mortality in Larimichthys crocea during culture, resulting in serious economic loss. Analysis of transcriptome and quantitative real-time polymerase chain reaction (PCR) data showed that dksA gene expression was significantly up-regulated after 48 h of infection with Epinephelus coioides (log 2FC=3.12, P<0.001). RNAi of five shRNAs significantly reduced the expression of dksA in P. plecoglossicida, and the optimal silencing efficiency was 96.23%. Compared with wild-type strains, the symptoms of visceral white spot disease in L. crocea infected with RNAi strains were reduced, with time of death delayed by 48 h and mortality reduced by 25%. The dksA silencing led to a substantial down-regulation in cellular component-, flagellum-, and ribosome assembly-related genes in P. plecoglossicida, and the significant up-regulation of fliC may be a way in which virulence is maintained in P. plecoglossicida. The GO and KEGG results showed that RNAi strain infection in L. crocea led to the down-regulation of inflammatory factor genes in immune-related pathways, which were associated with multiple immune response processes. Results also showed that dksA was a virulence gene in P. plecoglossicida. Compared with the wild-type strains, RNAi strain infection induced a weaker immune response in L. crocea.
Collapse
Affiliation(s)
- Lu-Ying Wang
- Fisheries College, Jimei University, Xiamen, Fujian 361021, China
| | - Zi-Xu Liu
- Fisheries College, Jimei University, Xiamen, Fujian 361021, China
| | - Ling-Min Zhao
- Fisheries College, Jimei University, Xiamen, Fujian 361021, China
| | - Li-Xing Huang
- Fisheries College, Jimei University, Xiamen, Fujian 361021, China
| | - Ying-Xue Qin
- Fisheries College, Jimei University, Xiamen, Fujian 361021, China
| | - Yong-Quan Su
- State Key Laboratory of Large Yellow Croaker Breeding, Ningde Fufa Aquatic Products Co., Ltd., Ningde, Fujian 352000, China
| | - Wei-Qiang Zheng
- State Key Laboratory of Large Yellow Croaker Breeding, Ningde Fufa Aquatic Products Co., Ltd., Ningde, Fujian 352000, China
| | - Fan Wang
- Fujian Provincial Fishery Technical Extension Center, Fuzhou, Fujian 350003, China
| | - Qing-Pi Yan
- Fisheries College, Jimei University, Xiamen, Fujian 361021, China
- State Key Laboratory of Large Yellow Croaker Breeding, Ningde Fufa Aquatic Products Co., Ltd., Ningde, Fujian 352000, China. E-mail:
| |
Collapse
|
21
|
Ouyang G, Liao Q, Zhang D, Rong F, Cai X, Fan S, Zhu J, Wang J, Liu X, Liu X, Xiao W. Zebrafish NF-κB/p65 Is Required for Antiviral Responses. THE JOURNAL OF IMMUNOLOGY 2020; 204:3019-3029. [PMID: 32321758 DOI: 10.4049/jimmunol.1900309] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 03/30/2020] [Indexed: 01/08/2023]
Abstract
Transcriptional programs regulated by the NF-κB family are essential for the inflammatory response as well as for innate and adaptive immunity. NF-κB activation occurs via two major signaling pathways: the canonical and the noncanonical. The canonical NF-κB pathway responds to diverse immune stimulations and leads to rapid but transient activation. As a member of the canonical NF-κB family, p65 is thought to be a key regulator of viral infection. Because of the embryonic lethality of p65-null mice, the physiological role of p65 in the antiviral immune response is still unclear. In this study, we generated p65-null zebrafish, which were viable and indistinguishable from their wildtype (WT) siblings under normal conditions. However, p65-null zebrafish were more sensitive to spring viremia of carp virus infection than their WT siblings. Further assays indicated that proinflammatory and antiviral genes, including IFN, were downregulated in p65-null zebrafish after spring viremia of carp virus infection compared with their WT siblings. Our results thus suggested that p65 is required for the antiviral response, activating not only proinflammatory genes but also antiviral genes (including IFN).
Collapse
Affiliation(s)
- Gang Ouyang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Qian Liao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Dawei Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Fangjing Rong
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Xiaolian Cai
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Sijia Fan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Junji Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Jing Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Xing Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Xueqin Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Wuhan Xiao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; .,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| |
Collapse
|
22
|
Aboonabi A, Aboonabi A. Anthocyanins reduce inflammation and improve glucose and lipid metabolism associated with inhibiting nuclear factor-kappaB activation and increasing PPAR-γ gene expression in metabolic syndrome subjects. Free Radic Biol Med 2020; 150:30-39. [PMID: 32061902 DOI: 10.1016/j.freeradbiomed.2020.02.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/31/2020] [Accepted: 02/09/2020] [Indexed: 02/06/2023]
Abstract
Anthocyanins exhibit antioxidant and anti-inflammatory activities via a multitude of biochemical mechanisms. However, the signaling pathways involved in the actions of anthocyanins against chronic inflammation are not fully understood. The effects of berry-rich anthocyanin supplements (320 mg/day) for four weeks were examined on features of metabolic syndrome components and the expression of PPAR-γ, Nrf2, and NF-κB dependent genes in MetS and healthy subjects. Total RNA was isolated from whole blood with the PAXgene proprietary blood collection system. Four weeks anthocyanin consumption significantly decreased fasting blood glucose (15.7% vs 3.2%), TG (18.2% vs -1.39%), cholesterol (33.5% vs 1.56%) and LDL (28.4% vs -15.6%) in the MetS compared to Control group (P-value < 0.05, 95% CI). There was a significant up regulation in the expression PPAR-γ gene associated with the lipid and glucose metabolism in MetS subjects which negatively correlated (P-value < 0.01) with the change in the FBG (r = -0.488), Cholesterol (r = -0.496), TG (r = -0.513) and LDL (r = -0.519). Moreover, anthocyanin supplementation decreases serum hs-CRP (-36.3% vs 6.25%) in MetS in compared to Control group (P-value < 0.05). Anthocyanin supplementation also down-regulated the expression of NF-κB dependent genes including TNF-α (-28% and -15%), IL-6 (-16.1% and -13.6%), IL-1A (-21.5% and -12.9%), PCAM-1 (-15% and -17.5%), and COX-2(-26% and -27%) in both MetS and Control group respectively (P-value < 0.05). The study results suggested that berry supplements improved selected features of metabolic syndrome and related cardiovascular risk factors. These benefits may be due to the inhibition of NF-κB dependent gene expression and enhancement of PPAR-γ.
Collapse
Affiliation(s)
- Anahita Aboonabi
- School of Medical Science, Gold Coast Campus, Griffith University, Parklands Drive, Southport, Queensland, 4222, Australia.
| | - Arta Aboonabi
- West Center of Tehran, Payam Noor University, Shahid Bagheri Town, Tehran, Iran.
| |
Collapse
|
23
|
T. cruzi infection among aged rats: Melatonin as a promising therapeutic molecule. Exp Gerontol 2020; 135:110922. [PMID: 32151734 DOI: 10.1016/j.exger.2020.110922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/12/2020] [Accepted: 03/05/2020] [Indexed: 12/12/2022]
Abstract
Although T. cruzi was identified as the cause of Chagas disease more than 100 years ago, satisfactory treatments still do not exist, especially for chronic disease. Here we review work suggesting that melatonin could have promise as a Chagas therapeutic. Melatonin has remarkably diverse actions. It is an immunomodulator, an anti-inflammatory, an antioxidant, a free radical scavenger, and has antiapoptotic and anti-aging effects. The elderly (aged 60 years or more) as a group are growing faster than any other age group. Here we discuss the major effects and the mechanisms of action of melatonin on aged T. cruzi-infected rats. Melatonin's protective effects may be consequences of its cooperative antioxidant and immunomodulatory actions. Melatonin modulates oxidative damage, inducing an antioxidant response and reversing age-related thymus regression. Its protective actions could be the result of its anti-apoptotic activity, and by its counteracting the excessive production of corticosterone. This review describes our work showing that host age plays an important and variable influence on the progression of systemic T. cruzi infection and supporting the hypothesis that melatonin should be considered as a powerful therapeutic compound with multiple activities that can improve host homeostasis during experimental T. cruzi infection.
Collapse
|
24
|
Chen T, Zhu J, Hang CH, Wang YH. The Potassium SK Channel Activator NS309 Protects Against Experimental Traumatic Brain Injury Through Anti-Inflammatory and Immunomodulatory Mechanisms. Front Pharmacol 2019; 10:1432. [PMID: 31849677 PMCID: PMC6895208 DOI: 10.3389/fphar.2019.01432] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 11/08/2019] [Indexed: 12/22/2022] Open
Abstract
Neuroinflammation plays important roles in neuronal cell death and functional deficits after TBI. Small conductance Ca2+-activated K+ channels (SK) have been shown to be potential therapeutic targets for treatment of neurological disorders, such as stroke and Parkinson’s disease (PD). The aim of the present study was to investigate the role of SK channels in an animal model of TBI induced by controlled cortical impact (CCI). The SK channels activator NS309 at a concentration of 2 mg/kg was administered by intraperitoneal injection, and no obviously organ-related toxicity of NS309 was found in Sprague-Dawley (SD) rats. Treatment with NS309 significantly reduced brain edema after TBI, but had no effect on contusion volume. This protection can be observed even when the administration was delayed by 4 h after injury. NS309 attenuated the TBI-induced deficits in neurological function, which was accompanied by the reduced neuronal apoptosis. The results of immunohistochemistry showed that NS309 decreased the number of neutrophils, lymphocytes, and microglia cells, with no effect on astrocytes. In addition, NS309 markedly decreased the levels of pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α) and chemokines (MCP-1, MIP-2, and RANTES), but increased the levels of anti-inflammatory cytokines (IL-4, IL-10, and TGF-β1) after TBI. The results of RT-PCR and western blot showed that NS309 increased TSG-6 expression and inhibited NF-κB activation. Furthermore, knockdown of TSG-6 using in vivo transfection with TSG-6 specific shRNA partially reversed the protective and anti-inflammatory effects of NS309 against TBI. In summary, our results indicate that the SK channel activator NS309 could modulate inflammation-associated immune cells and cytokines via regulating the TSG-6/NF-κB pathway after TBI. The present study offers a new sight into the mechanisms responsible for SK channels activation with implications for the treatment of TBI.
Collapse
Affiliation(s)
- Tao Chen
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, China.,Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jie Zhu
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, China
| | - Chun-Hua Hang
- Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yu-Hai Wang
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, China
| |
Collapse
|
25
|
Abstract
Pneumonia is an important cause of morbidity and mortality. However, pneumonia is an unusual outcome of respiratory infection. Most of the time, microbes in the lung can be controlled by a combination of constitutive and recruited defense mechanisms. Inflammation is a key component of recruited defenses. Variations in inflammation that influence pneumonia susceptibility and severity are considered here.
Collapse
Affiliation(s)
- Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Abstract
This review by Shan et al. discusses necroptosis, a form of regulated necrotic cell death mediated by RIPK1 kinase activity, RIPK3, and MLKL, which can be activated under apoptosis-deficient conditions. Both necroptosis and apoptosis can be activated in response to various mutations that result in the abortion of defective embryos and during human inflammatory and neurodegenerative pathologies. Necroptosis, a form of regulated necrotic cell death mediated by RIPK1 (receptor-interacting protein kinase 1) kinase activity, RIPK3, and MLKL (mixed-lineage kinase domain-like pseudokinase), can be activated under apoptosis-deficient conditions. Modulating the activation of RIPK1 by ubiquitination and phosphorylation is critical to control both necroptosis and apoptosis. Mutant mice with kinase-dead RIPK1 or RIPK3 and MLKL deficiency show no detrimental phenotype in regard to development and adult homeostasis. However, necroptosis and apoptosis can be activated in response to various mutations that result in the abortion of the defective embryos and human inflammatory and neurodegenerative pathologies. RIPK1 inhibition represents a key therapeutic strategy for treatment of diseases where blocking both necroptosis and apoptosis can be beneficial.
Collapse
Affiliation(s)
- Bing Shan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PuDong District, Shanghai 201203, China
| | - Heling Pan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PuDong District, Shanghai 201203, China
| | - Ayaz Najafov
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PuDong District, Shanghai 201203, China.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
27
|
Hattori Y, Hattori K, Suzuki T, Palikhe S, Matsuda N. Nucleic-acid based gene therapy approaches for sepsis. Eur J Pharmacol 2018; 833:403-410. [PMID: 29935173 DOI: 10.1016/j.ejphar.2018.06.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/06/2018] [Accepted: 06/19/2018] [Indexed: 12/15/2022]
Abstract
Despite advances in overall medical care, sepsis and its sequelae continue to be an embarrassing clinical entity with an unacceptably high mortality rate. The central reason for high morbidity and high mortality of sepsis and its sequelae is the lack of an effective treatment. Previous clinical trials have largely failed to identify an effective therapeutic target to improve clinical outcomes in sepsis. Thus, the key goal favoring the outcome of septic patients is to devise innovative and evolutionary therapeutic strategies. Gene therapy can be considered as one of the most promising novel therapeutic approaches for nasty disorders. Since a number of transcription factors, such as nuclear factor-κB (NF-κB) and activator protein-1 (AP-1), play a pivotal role in the pathophysiology of sepsis that can be characterized by the induction of multiple genes and their products, sepsis may be regarded as a gene-related disorder and gene therapy may be considered a promising novel therapeutic approach for treatment of sepsis. In this review article, we provide an up-to-date summary of the gene-targeting approaches, which have been developed in animal models of sepsis. Our review sheds light on the molecular basis of sepsis pathology for the development of novel gene therapy approaches and leads to the conclusion that future research efforts may fully take into account gene therapy for the treatment of sepsis.
Collapse
Affiliation(s)
- Yuichi Hattori
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Kohshi Hattori
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Tokiko Suzuki
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Sailesh Palikhe
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Naoyuki Matsuda
- Department of Emergency and Critical Care Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
28
|
Song JH, Kang HB, Park SH, Jeong JH, Park J, You Y, Lee YH, Lee J, Kim E, Choi KC, Jun W. Extracts of Porphyra tenera (Nori Seaweed) Activate the Immune Response in Mouse RAW264.7 Macrophages via NF-κB Signaling. J Med Food 2018; 20:1152-1159. [PMID: 29243967 DOI: 10.1089/jmf.2017.4014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Porphyra tenera, also known as nori, is a red algal species of seaweed. It is cultivated in Asia for culinary purposes. We report that P. tenera extract (PTE) enhances the immune response in mouse macrophages. We found that P. tenera extract regulates the NF-κB IκB kinase (IKK) signaling pathway, and we assessed the expression and translocation of p65, a subunit of NF-κB, in RAW264.7 mouse macrophage cells after treatment with PTE. We also investigated the effects of 10% ethanol PTE (PTE10) in RAW264.7 cells. The production of IL-10, IL-6, TNF-α, and IFN-γ was induced by PTE treatment of the macrophages, and PTE also enhanced p-IκB and p-AKT. PTE10 showed no cytotoxicity at 10-20 μg/mL in RAW264.7 cells. PTE10, in fact, increased cell viability at 24 h, stimulated macrophage cells, and induced the phosphorylation of Akt. Akt stimulates IKK activity through the phosphorylation of IKKα and enhances immune activity through the activation of NF-κB. In this study, NF-κB activation was induced by increasing p-NF-κB and p-IKK. A subunit of NF-κB, p65, was located in the nucleus and increased the expression of various cytokines. PTE thus enhanced the immune response through IκB-α immunostimulation signaling in RAW264.7 cells. PTE10 has potential therefore for development of future treatments requiring immune system stimulation.
Collapse
Affiliation(s)
- Ji-Hye Song
- 1 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, South Korea .,2 Department of Pharmacology, University of Ulsan College of Medicine , Seoul, Korea
| | - Hee-Bum Kang
- 1 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, South Korea .,2 Department of Pharmacology, University of Ulsan College of Medicine , Seoul, Korea
| | - Seung-Ho Park
- 1 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, South Korea .,2 Department of Pharmacology, University of Ulsan College of Medicine , Seoul, Korea
| | - Ji-Hoon Jeong
- 1 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, South Korea .,2 Department of Pharmacology, University of Ulsan College of Medicine , Seoul, Korea
| | - Jeongjin Park
- 3 Division of Food and Nutrition, Chonnam National University , Gwangju, Korea.,4 Research Institute for Human Ecology, Chonnam National University , Gwangju, Korea
| | - Yanghee You
- 3 Division of Food and Nutrition, Chonnam National University , Gwangju, Korea.,4 Research Institute for Human Ecology, Chonnam National University , Gwangju, Korea
| | - Yoo-Hyun Lee
- 5 Department of Food Science and Nutrition, The University of Suwon , Suwon, Korea
| | - Jeongmin Lee
- 6 Department of Medical Nutrition, Kyung Hee University , Yongin, Korea
| | - Eungpil Kim
- 7 Marine Biotechnology Research Center , Jeonnam Bioindustry Foundation, Wando, Korea
| | - Kyung-Chul Choi
- 1 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, South Korea .,2 Department of Pharmacology, University of Ulsan College of Medicine , Seoul, Korea
| | - Woojin Jun
- 3 Division of Food and Nutrition, Chonnam National University , Gwangju, Korea.,4 Research Institute for Human Ecology, Chonnam National University , Gwangju, Korea
| |
Collapse
|
29
|
Slotta C, Storm J, Pfisterer N, Henkel E, Kleinwächter S, Pieper M, Ruiz-Perera LM, Greiner JFW, Kaltschmidt B, Kaltschmidt C. IKK1/2 protect human cells from TNF-mediated RIPK1-dependent apoptosis in an NF-κB-independent manner. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1025-1033. [PMID: 29630899 DOI: 10.1016/j.bbamcr.2018.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 03/30/2018] [Accepted: 04/05/2018] [Indexed: 01/19/2023]
Abstract
TNF signaling is directly linked to cancer development and progression. A broad range of tumor cells is able to evade cell death induced by TNF impairing the potential anti-cancer value of TNF in therapy. Although sensitizing cells to TNF-induced death therefore has great clinical implications, detailed mechanistic insights into TNF-mediated human cell death still remain unknown. Here, we analyzed human cells by applying CRISPR/Cas9n to generate cells deficient of IKK1, IKK2, IKK1/2 and RELA. Despite stimulation with TNF resulted in impaired NF-κB activation in all genotypes compared to wildtype cells, increased cell death was observable only in IKK1/2-double-deficient cells. Cell death could be detected by Caspase-3 activation and binding of Annexin V. TNF-induced programmed cell death in IKK1/2-/- cells was further shown to be mediated via RIPK1 in a predominantly apoptotic manner. Our findings demonstrate the IKK complex to protect from TNF-induced cell death in human cells independently to NF-κB RelA suggesting IKK1/2 to be highly promising targets for cancer therapy.
Collapse
Affiliation(s)
- Carsten Slotta
- Department of Cell Biology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany; Molecular Neurobiology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany
| | - Jonathan Storm
- Department of Cell Biology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany
| | - Nina Pfisterer
- Department of Cell Biology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany
| | - Elena Henkel
- Department of Cell Biology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany
| | - Svenja Kleinwächter
- Department of Cell Biology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany
| | - Maren Pieper
- Department of Cell Biology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany
| | - Lucia M Ruiz-Perera
- Molecular Neurobiology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany
| | - Johannes F W Greiner
- Department of Cell Biology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany
| | - Barbara Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany; Molecular Neurobiology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Universitaetsstr. 25, 33501 Bielefeld, Germany.
| |
Collapse
|
30
|
Zhu F, Hu Y. Integrity of IKK/NF-κB Shields Thymic Stroma That Suppresses Susceptibility to Autoimmunity, Fungal Infection, and Carcinogenesis. Bioessays 2018. [PMID: 29522649 DOI: 10.1002/bies.201700131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A pathogenic connection between autoreactive T cells, fungal infection, and carcinogenesis has been demonstrated in studies of human autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) as well as in a mouse model in which kinase-dead Ikkα knock-in mice develop impaired central tolerance, autoreactive T cell-mediated autoimmunity, chronic fungal infection, and esophageal squamous cell carcinoma, which recapitulates APECED. IκB kinase α (IKKα) is one subunit of the IKK complex required for NF-κB activation. IKK/NF-κB is essential for central tolerance establishment by regulating the development of medullary thymic epithelial cells (mTECs) that facilitate the deletion of autoreactive T cells in the thymus. In this review, we extensively discuss the pathogenic roles of inborn errors in the IKK/NF-κB loci in the phenotypically related diseases APECED, immune deficiency syndrome, and severe combined immunodeficiency; differentiate how IKK/NF-κB components, through mTEC (stroma), T cells/leukocytes, or epithelial cells, contribute to the pathogenesis of infectious diseases, autoimmunity, and cancer; and highlight the medical significance of IKK/NF-κB in these diseases.
Collapse
Affiliation(s)
- Feng Zhu
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, 21701, Maryland, USA
| | - Yinling Hu
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, 21701, Maryland, USA
| |
Collapse
|
31
|
Involvement of Alveolar Macrophages and Neutrophils in Acute Lung Injury After Scorpion Envenomation: New Pharmacological Targets. Inflammation 2018; 41:773-783. [DOI: 10.1007/s10753-018-0731-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
32
|
Varfolomeev E, Vucic D. Intracellular regulation of TNF activity in health and disease. Cytokine 2018; 101:26-32. [DOI: 10.1016/j.cyto.2016.08.035] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 01/27/2023]
|
33
|
Simulation of Cellular Energy Restriction in Quiescence (ERiQ)-A Theoretical Model for Aging. BIOLOGY 2017; 6:biology6040044. [PMID: 29231906 PMCID: PMC5745449 DOI: 10.3390/biology6040044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/08/2017] [Accepted: 12/09/2017] [Indexed: 02/07/2023]
Abstract
Cellular responses to energy stress involve activation of pro-survival signaling nodes, compensation in regulatory pathways and adaptations in organelle function. Specifically, energy restriction in quiescent cells (ERiQ) through energetic perturbations causes adaptive changes in response to reduced ATP, NAD+ and NADP levels in a regulatory network spanned by AKT, NF-κB, p53 and mTOR. Based on the experimental ERiQ platform, we have constructed a minimalistic theoretical model consisting of feedback motifs that enable investigation of stress-signaling pathways. The computer simulations reveal responses to acute energetic perturbations, promoting cellular survival and recovery to homeostasis. We speculated that the very same stress mechanisms are activated during aging in post-mitotic cells. To test this hypothesis, we modified the model to be deficient in protein damage clearance and demonstrate the formation of energy stress. Contrasting the network’s pro-survival role in acute energetic challenges, conflicting responses in aging disrupt mitochondrial maintenance and contribute to a lockstep progression of decline when chronically activated. The model was analyzed by a local sensitivity analysis with respect to lifespan and makes predictions consistent with inhibitory and gain-of-function experiments in aging.
Collapse
|
34
|
Xu C, Wu X, Zhang X, Xie Q, Fan C, Zhang H. Embryonic Lethality and Host Immunity of RelA-Deficient Mice Are Mediated by Both Apoptosis and Necroptosis. THE JOURNAL OF IMMUNOLOGY 2017; 200:271-285. [PMID: 29167229 DOI: 10.4049/jimmunol.1700859] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/26/2017] [Indexed: 01/10/2023]
Abstract
In mammalian cells, signaling pathways triggered by TNF can be switched from NF-κB activation to apoptosis and/or necroptosis. The in vivo mechanisms underlying the mutual regulation of these three signaling pathways are poorly understood. In this article, we report that the embryonic lethality of RelA-deficient mice is partially prevented by the deletion of Rip3 or Mlkl, but it is fully rescued by the combined ablation of Fadd and Rip3 or Mlkl or by blocking RIP1 kinase activity (RIP1K45A). RelA-/-Fadd-/-Rip3-/- triple-knockout (TKO) and RelA-/-Rip1K45A/K45A mice displayed bacterial pneumonia leading to death ∼2 wk after birth. Moreover, RelA-/-Rip1K45A/K45A mice, but not TKO mice, developed severe inflammation associated with inflammatory skin lesion. Antibiotic treatment improved bacterial pneumonia, extended the lifespan of TKO and RelA-/-Rip1K45A/K45A mice, and alleviated skin inflammation in RelA-/-Rip1K45A/K45A mice. These results show the mechanisms underlying the in vivo mutual regulation between NF-κB activation and the cell death pathway and provide new insights into this interplay in embryonic development and host immune homeostasis.
Collapse
Affiliation(s)
- Chengxian Xu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Xiaoxia Wu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Xixi Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Qun Xie
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and.,Department of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Cunxian Fan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Haibing Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| |
Collapse
|
35
|
Mitchell JP, Carmody RJ. NF-κB and the Transcriptional Control of Inflammation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 335:41-84. [PMID: 29305014 DOI: 10.1016/bs.ircmb.2017.07.007] [Citation(s) in RCA: 312] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The NF-κB transcription factor was discovered 30 years ago and has since emerged as the master regulator of inflammation and immune homeostasis. It achieves this status by means of the large number of important pro- and antiinflammatory factors under its transcriptional control. NF-κB has a central role in inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease, and autoimmunity, as well as diseases comprising a significant inflammatory component such as cancer and atherosclerosis. Here, we provide an overview of the studies that form the basis of our understanding of the role of NF-κB subunits and their regulators in controlling inflammation. We also describe the emerging importance of posttranslational modifications of NF-κB in the regulation of inflammation, and highlight the future challenges faced by researchers who aim to target NF-κB transcriptional activity for therapeutic benefit in treating chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jennifer P Mitchell
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Ruaidhrí J Carmody
- Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|
36
|
Coleman FT, Blahna MT, Kamata H, Yamamoto K, Zabinski MC, Kramnik I, Wilson AA, Kotton DN, Quinton LJ, Jones MR, Pelton SI, Mizgerd JP. Capacity of Pneumococci to Activate Macrophage Nuclear Factor κB: Influence on Necroptosis and Pneumonia Severity. J Infect Dis 2017; 216:425-435. [PMID: 28368460 DOI: 10.1093/infdis/jix159] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 03/23/2017] [Indexed: 12/13/2022] Open
Abstract
During pneumococcal pneumonia, antibacterial defense requires the orchestrated expression of innate immunity mediators, initiated by alveolar macrophages and dependent on transcription driven by nuclear factor κB (NF-κB). Such immune pressure may select for pneumococci, which avoid or subvert macrophage NF-κB activation. Analyzing pneumococci collected from children in Massachusetts, we found that the activation of macrophage NF-κB by Streptococcus pneumoniae is highly diverse, with a preponderance of low NF-κB activators that associate particularly with complicated pneumonia. Low NF-κB activators cause more severe lung infections in mice, and they drive macrophages toward an alternate and detrimental cell fate of necroptosis. Both outcomes can be reversed by activation of macrophages with pneumococci that are high NF-κB activators. These results suggest that low NF-κB activation is a virulence property of pneumococci and that the appropriate activation of macrophages, including NF-κB, may hold promise as an adjunct therapeutic avenue for pneumococcal pneumonia.
Collapse
Affiliation(s)
| | | | | | | | | | - Igor Kramnik
- Pulmonary Center.,Department of Microbiology.,Deparment of Medicine
| | | | | | - Lee J Quinton
- Pulmonary Center.,Deparment of Medicine.,Department of Pathology and Laboratory Medicine
| | | | | | - Joseph P Mizgerd
- Pulmonary Center.,Department of Microbiology.,Deparment of Medicine.,Department of Biochemistry, Boston University School of Medicine, Massachusetts
| |
Collapse
|
37
|
Traber KE, Symer EM, Allen E, Kim Y, Hilliard KL, Wasserman GA, Stewart CL, Jones MR, Mizgerd JP, Quinton LJ. Myeloid-epithelial cross talk coordinates synthesis of the tissue-protective cytokine leukemia inhibitory factor during pneumonia. Am J Physiol Lung Cell Mol Physiol 2017; 313:L548-L558. [PMID: 28522567 PMCID: PMC5625259 DOI: 10.1152/ajplung.00482.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 11/22/2022] Open
Abstract
In bacterial pneumonia, lung damage resulting from epithelial cell injury is a major contributor to the severity of disease and, in some cases, can lead to long-term sequelae, especially in the setting of severe lung injury or acute respiratory distress syndrome. Leukemia inhibitory factor (LIF), a member of the IL-6 cytokine family, is a critical determinant of lung tissue protection during pneumonia, but the cellular sources of LIF and the signaling pathways leading to its production in the infected lung are not known. Here, we demonstrate that lung epithelium, specifically alveolar type II cells, is the predominant site of LIF transcript induction in pneumonic mouse lungs. Epithelial cell cultures were induced to express LIF by bacteria and by sterile bronchoalveolar lavage fluid from pneumonic mice. Reciprocal bone marrow chimera studies demonstrated that LIF deficiency in the nonhematopoietic compartment, but not LIF deficiency in hematopoietic cells, eliminated LIF induction during pneumonia. Although NF-κB RelA (p65) is essential for the expression of many cytokines during pneumonia, its targeted mutation in the lung epithelium was inconsequential for pneumonia-driven LIF induction. However, maximal expression of this epithelial-derived cytokine was dependent on NF-κB RelA in myeloid cells. Overall, our data suggest a signaling axis whereby activation of NF-κB RelA in myeloid cells promotes epithelial LIF induction during lung infections, representing a means through which these two cell types collaborate to improve tissue resilience during pneumonia.
Collapse
Affiliation(s)
- Katrina E Traber
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Elise M Symer
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Eri Allen
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Yuri Kim
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Kristie L Hilliard
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts
| | - Gregory A Wasserman
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts
| | | | - Matthew R Jones
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts; and
| | - Lee J Quinton
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts;
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
38
|
Badran YR, Dedeoglu F, Leyva Castillo JM, Bainter W, Ohsumi TK, Bousvaros A, Goldsmith JD, Geha RS, Chou J. Human RELA haploinsufficiency results in autosomal-dominant chronic mucocutaneous ulceration. J Exp Med 2017; 214:1937-1947. [PMID: 28600438 PMCID: PMC5502421 DOI: 10.1084/jem.20160724] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 02/25/2017] [Accepted: 05/09/2017] [Indexed: 11/09/2022] Open
Abstract
Badran et al. demonstrate an essential contribution of biallelic RELA expression in protecting stromal and epithelial cells from TNF-mediated cell death in patients with chronic mucocutaneous ulceration. The treatment of chronic mucocutaneous ulceration is challenging, and only some patients respond selectively to inhibitors of tumor necrosis factor-α (TNF). TNF activates opposing pathways leading to caspase-8–mediated apoptosis as well as nuclear factor κB (NF-κB)–dependent cell survival. We investigated the etiology of autosomal-dominant, mucocutaneous ulceration in a family whose proband was dependent on anti-TNF therapy for sustained remission. A heterozygous mutation in RELA, encoding the NF-κB subunit RelA, segregated with the disease phenotype and resulted in RelA haploinsufficiency. The patients’ fibroblasts exhibited increased apoptosis in response to TNF, impaired NF-κB activation, and defective expression of NF-κB–dependent antiapoptotic genes. Rela+/− mice have similarly impaired NF-κB activation, develop cutaneous ulceration from TNF exposure, and exhibit severe dextran sodium sulfate–induced colitis, ameliorated by TNF inhibition. These findings demonstrate an essential contribution of biallelic RELA expression in protecting stromal cells from TNF-mediated cell death, thus delineating the mechanisms driving the effectiveness of TNF inhibition in this disease.
Collapse
Affiliation(s)
- Yousef R Badran
- Division of Immunology, Boston Children's Hospital, Boston, MA
| | - Fatma Dedeoglu
- Division of Immunology, Boston Children's Hospital, Boston, MA
| | | | - Wayne Bainter
- Division of Immunology, Boston Children's Hospital, Boston, MA
| | - Toshiro K Ohsumi
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA
| | - Athos Bousvaros
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA
| | - Jeffrey D Goldsmith
- Department of Pathology, Boston Children's Hospital, Boston, MA.,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Boston, MA
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Boston, MA
| |
Collapse
|
39
|
Nandhu MS, Kwiatkowska A, Bhaskaran V, Hayes J, Hu B, Viapiano MS. Tumor-derived fibulin-3 activates pro-invasive NF-κB signaling in glioblastoma cells and their microenvironment. Oncogene 2017; 36:4875-4886. [PMID: 28414309 PMCID: PMC5570669 DOI: 10.1038/onc.2017.109] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 02/19/2017] [Accepted: 03/04/2017] [Indexed: 12/19/2022]
Abstract
Molecular profiling of glioblastomas has revealed the presence of key signaling hubs that contribute to tumor progression and acquisition of resistance. One of these main signaling mechanisms is the NF-κB pathway, which integrates multiple extracellular signals into transcriptional programs for tumor growth, invasion, and maintenance of the tumor-initiating population. We show here that an extracellular protein released by glioblastoma cells, fibulin-3, drives oncogenic NF-κB in the tumor and increases NF-κB activation in peritumoral astrocytes. Fibulin-3 expression correlates with a NF-κB-regulated “invasive signature” linked to poorer survival, being a possible tissue marker for regions of active tumor progression. Accordingly, fibulin-3 promotes glioblastoma invasion in a manner that requires NF-κB activation both in the tumor cells and their microenvironment. Mechanistically, we found that fibulin-3 activates the metalloprotease ADAM17 by competing with its endogenous inhibitor, TIMP3. This results in sustained release of soluble TNFα by ADAM17, which in turn activates TNF receptors and canonical NF-κB signaling. Taken together, our results underscore fibulin-3 as a novel extracellular signal with strong activating effect on NF-κB in malignant gliomas. Because fibulin-3 is produced de novo in these tumors and is absent from normal brain we propose that targeting the fibulin-3/NF-κB axis may provide a novel avenue to disrupt oncogenic NF-κB signaling in combination therapies for malignant brain tumors.
Collapse
Affiliation(s)
- M S Nandhu
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY, USA
| | - A Kwiatkowska
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - V Bhaskaran
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - J Hayes
- Department of Neurological Surgery, Helen Diller Family Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - B Hu
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - M S Viapiano
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
40
|
Inflammatory Activation of Microglia and Astrocytes in Manganese Neurotoxicity. ADVANCES IN NEUROBIOLOGY 2017; 18:159-181. [PMID: 28889267 DOI: 10.1007/978-3-319-60189-2_8] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurotoxicity due to excessive exposure to manganese (Mn) has been described as early as 1837 (Couper, Br Ann Med Pharm Vital Stat Gen Sci 1:41-42, 1837). Extensive research over the past two decades has revealed that Mn-induced neurological injury involves complex pathophysiological signaling mechanisms between neurons and glial cells. Glial cells are an important target of Mn in the brain, both for sequestration of the metal, as well as for activating inflammatory signaling pathways that damage neurons through overproduction of numerous reactive oxygen and nitrogen species and inflammatory cytokines. Understanding how these pathways are regulated in glial cells during Mn exposure is critical to determining the mechanisms underlying permanent neurological dysfunction stemming from excess exposure. The subject of this review will be to delineate mechanisms by which Mn interacts with glial cells to perturb neuronal function, with a particular emphasis on neuroinflammation and neuroinflammatory signaling between distinct populations of glial cells.
Collapse
|
41
|
Rubin N, Harrison MR, Krainock M, Kim R, Lien CL. Recent advancements in understanding endogenous heart regeneration-insights from adult zebrafish and neonatal mice. Semin Cell Dev Biol 2016; 58:34-40. [PMID: 27132022 PMCID: PMC5028242 DOI: 10.1016/j.semcdb.2016.04.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/13/2016] [Accepted: 04/17/2016] [Indexed: 02/06/2023]
Abstract
Enhancing the endogenous regenerative capacity of the mammalian heart is a promising strategy that can lead to potential treatment of injured cardiac tissues. Studies on heart regeneration in zebrafish and neonatal mice have shown that cardiomyocyte proliferation is essential for replenishing myocardium. We will review recent advancements that have demonstrated the importance of Neuregulin 1/ErbB2 and innervation in regulating cardiomyocyte proliferation using both adult zebrafish and neonatal mouse heart regeneration models. Emerging findings suggest that different populations of macrophages and inflammation might contribute to regenerative versus fibrotic responses. Finally, we will discuss variation in the severity of the cardiac injury and size of the wound, which may explain the range of outcomes observed in different injury models.
Collapse
Affiliation(s)
- Nicole Rubin
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Michael R Harrison
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Michael Krainock
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Richard Kim
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Ching-Ling Lien
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States; Department of Biochemistry & Molecular Biology, Keck School of Medicine, University of Southern California, United States.
| |
Collapse
|
42
|
Vlantis K, Wullaert A, Polykratis A, Kondylis V, Dannappel M, Schwarzer R, Welz P, Corona T, Walczak H, Weih F, Klein U, Kelliher M, Pasparakis M. NEMO Prevents RIP Kinase 1-Mediated Epithelial Cell Death and Chronic Intestinal Inflammation by NF-κB-Dependent and -Independent Functions. Immunity 2016; 44:553-567. [PMID: 26982364 PMCID: PMC4803910 DOI: 10.1016/j.immuni.2016.02.020] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 10/25/2015] [Accepted: 12/14/2015] [Indexed: 12/25/2022]
Abstract
Intestinal epithelial cells (IECs) regulate gut immune homeostasis, and impaired epithelial responses are implicated in the pathogenesis of inflammatory bowel diseases (IBD). IEC-specific ablation of nuclear factor κB (NF-κB) essential modulator (NEMO) caused Paneth cell apoptosis and impaired antimicrobial factor expression in the ileum, as well as colonocyte apoptosis and microbiota-driven chronic inflammation in the colon. Combined RelA, c-Rel, and RelB deficiency in IECs caused Paneth cell apoptosis but not colitis, suggesting that NEMO prevents colon inflammation by NF-κB-independent functions. Inhibition of receptor-interacting protein kinase 1 (RIPK1) kinase activity or combined deficiency of Fas-associated via death domain protein (FADD) and RIPK3 prevented epithelial cell death, Paneth cell loss, and colitis development in mice with epithelial NEMO deficiency. Therefore, NEMO prevents intestinal inflammation by inhibiting RIPK1 kinase activity-mediated IEC death, suggesting that RIPK1 inhibitors could be effective in the treatment of colitis in patients with NEMO mutations and possibly in IBD.
Collapse
Affiliation(s)
- Katerina Vlantis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Andy Wullaert
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Apostolos Polykratis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Vangelis Kondylis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Marius Dannappel
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Robin Schwarzer
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Patrick Welz
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Teresa Corona
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6D, UK
| | - Falk Weih
- Leibniz-Institute for Age Research, Fritz-Lipmann-Institute, 07745 Jena, Germany
| | - Ulf Klein
- Herbert Irving Comprehensive Cancer Center, Departments of Pathology & Cell Biology and Microbiology & Immunology, Columbia University, New York, NY 10032, USA
| | - Michelle Kelliher
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
43
|
Peltzer N, Darding M, Walczak H. Holding RIPK1 on the Ubiquitin Leash in TNFR1 Signaling. Trends Cell Biol 2016; 26:445-461. [DOI: 10.1016/j.tcb.2016.01.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/13/2016] [Accepted: 01/19/2016] [Indexed: 12/22/2022]
|
44
|
B-cell survival and development controlled by the coordination of NF-κB family members RelB and cRel. Blood 2016; 127:1276-86. [PMID: 26773039 PMCID: PMC4786837 DOI: 10.1182/blood-2014-10-606988] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 12/29/2015] [Indexed: 11/20/2022] Open
Abstract
Targeted deletion of BAFF causes severe deficiency of splenic B cells. BAFF-R is commonly thought to signal to nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB)-inducing kinase dependent noncanonical NF-κB RelB. However, RelB-deficient mice have normal B-cell numbers. Recent studies showed that BAFF also signals to the canonical NF-κB pathway, and we found that both RelB and cRel are persistently activated, suggesting BAFF signaling coordinates both pathways to ensure robust B-cell development. Indeed, we report now that combined loss of these 2 NF-κB family members leads to impaired BAFF-mediated survival and development in vitro. Although single deletion of RelB and cRel was dispensable for normal B-cell development, double knockout mice displayed an early B-cell developmental blockade and decreased mature B cells. Despite disorganized splenic architecture in Relb(-/-)cRel(-/-) mice, generation of mixed-mouse chimeras established the developmental phenotype to be B-cell intrinsic. Together, our results indicate that BAFF signals coordinate both RelB and cRel activities to ensure survival during peripheral B-cell maturation.
Collapse
|
45
|
Hilliard KL, Allen E, Traber KE, Yamamoto K, Stauffer NM, Wasserman GA, Jones MR, Mizgerd JP, Quinton LJ. The Lung-Liver Axis: A Requirement for Maximal Innate Immunity and Hepatoprotection during Pneumonia. Am J Respir Cell Mol Biol 2015; 53:378-90. [PMID: 25607543 DOI: 10.1165/rcmb.2014-0195oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The hepatic acute-phase response (APR), stimulated by injury or inflammation, is characterized by significant changes in circulating acute-phase protein (APP) concentrations. Although individual functions of liver-derived APPs are known, the net consequence of APP changes is unclear. Pneumonia, which induces the APR, causes an inflammatory response within the airspaces that is coordinated largely by alveolar macrophages and is typified by cytokine production, leukocyte recruitment, and plasma extravasation, the latter of which may enable delivery of hepatocyte-derived APPs to the infection site. To determine the functional significance of the hepatic APR during pneumonia, we challenged APR-null mice lacking hepatocyte signal transducer and activator of transcription 3 (STAT3) and v-rel avian reticuloendotheliosis viral oncogene homolog A (RelA) with Escherichia coli in the airspaces. APR-null mice displayed ablated APP induction, significantly increased mortality, liver injury and apoptosis, and a trend toward increased bacterial burdens. TNF-α neutralization reversed hepatotoxicity, but not mortality, suggesting that APR-dependent survival is not solely due to hepatoprotection. After a milder (nonlethal) E. coli infection, hepatocyte-specific mutations decreased APP concentrations and pulmonary inflammation in bronchoalveolar lavage fluid. Cytokine expression in airspace macrophages, but not other airspace or circulating cells, was significantly dependent on APP extravasation into the alveoli. These data identify a novel signaling axis whereby the liver response enhances macrophage activation and pulmonary inflammation during pneumonia. Although hepatic acute-phase changes directly curb injury induced by TNF-α in the liver itself, APPs downstream of these same signals promote survival in association with innate immunity in the lungs, thus demonstrating a critical role for the lung-liver axis during pneumonia.
Collapse
Affiliation(s)
- Kristie L Hilliard
- Departments of 1 Microbiology.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Eri Allen
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Katrina E Traber
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Kazuko Yamamoto
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Nicole M Stauffer
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Gregory A Wasserman
- Departments of 1 Microbiology.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Matthew R Jones
- 3 Medicine.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Joseph P Mizgerd
- Departments of 1 Microbiology.,3 Medicine.,4 Biochemistry, and.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Lee J Quinton
- 3 Medicine.,5 Pathology and Laboratory Medicine, and.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
46
|
Kilzheimer M, Quandt J, Langhans J, Weihrich P, Wirth T, Brunner C. NF-κB-dependent signals control BOB.1/OBF.1 and Oct2 transcriptional activity in B cells. Eur J Immunol 2015; 45:3441-53. [DOI: 10.1002/eji.201545475] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 07/30/2015] [Accepted: 09/12/2015] [Indexed: 12/18/2022]
Affiliation(s)
| | - Jasmin Quandt
- Institute of Physiological Chemistry; Ulm University; Ulm Germany
| | - Julia Langhans
- Department of Otorhinolaryngology; Ulm University; Ulm Germany
| | - Petra Weihrich
- Institute of Physiological Chemistry; Ulm University; Ulm Germany
- Department of Otorhinolaryngology; Ulm University; Ulm Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry; Ulm University; Ulm Germany
| | - Cornelia Brunner
- Institute of Physiological Chemistry; Ulm University; Ulm Germany
- Department of Otorhinolaryngology; Ulm University; Ulm Germany
| |
Collapse
|
47
|
de la Fuente V, Federman N, Zalcman G, Salles A, Freudenthal R, Romano A. NF-κB transcription factor role in consolidation and reconsolidation of persistent memories. Front Mol Neurosci 2015; 8:50. [PMID: 26441513 PMCID: PMC4563083 DOI: 10.3389/fnmol.2015.00050] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/24/2015] [Indexed: 12/16/2022] Open
Abstract
Transcriptional regulation is an important molecular process required for long-term neural plasticity and long-term memory (LTM) formation. Thus, one main interest in molecular neuroscience in the last decades has been the identification of transcription factors that are involved in memory processes. Among them, the nuclear factor κB (NF-κB) family of transcription factors has gained interest due to a significant body of evidence that supports a key role of these proteins in synaptic plasticity and memory. In recent years, the interest was particularly reinforced because NF-κB was characterized as an important regulator of synaptogenesis. This function may be explained by its participation in synapse to nucleus communication, as well as a possible local role at the synapse. This review provides an overview of experimental work obtained in the last years, showing the essential role of this transcription factor in memory processes in different learning tasks in mammals. We focus the review on the consolidation and reconsolidation memory phases as well as on the regulation of immediate-early and late genes by epigenetic mechanisms that determine enduring forms of memories.
Collapse
Affiliation(s)
- Verónica de la Fuente
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| | - Noel Federman
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| | - Gisela Zalcman
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| | - Angeles Salles
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| | - Ramiro Freudenthal
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| | - Arturo Romano
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| |
Collapse
|
48
|
Baig MS, Zaichick SV, Mao M, de Abreu AL, Bakhshi FR, Hart PC, Saqib U, Deng J, Chatterjee S, Block ML, Vogel SM, Malik AB, Consolaro MEL, Christman JW, Minshall RD, Gantner BN, Bonini MG. NOS1-derived nitric oxide promotes NF-κB transcriptional activity through inhibition of suppressor of cytokine signaling-1. ACTA ACUST UNITED AC 2015; 212:1725-38. [PMID: 26324446 PMCID: PMC4577833 DOI: 10.1084/jem.20140654] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 08/06/2015] [Indexed: 11/04/2022]
Abstract
The NF-κB pathway is central to the regulation of inflammation. Here, we demonstrate that the low-output nitric oxide (NO) synthase 1 (NOS1 or nNOS) plays a critical role in the inflammatory response by promoting the activity of NF-κB. Specifically, NOS1-derived NO production in macrophages leads to proteolysis of suppressor of cytokine signaling 1 (SOCS1), alleviating its repression of NF-κB transcriptional activity. As a result, NOS1(-/-) mice demonstrate reduced cytokine production, lung injury, and mortality when subjected to two different models of sepsis. Isolated NOS1(-/-) macrophages demonstrate similar defects in proinflammatory transcription on challenge with Gram-negative bacterial LPS. Consistently, we found that activated NOS1(-/-) macrophages contain increased SOCS1 protein and decreased levels of p65 protein compared with wild-type cells. NOS1-dependent S-nitrosation of SOCS1 impairs its binding to p65 and targets SOCS1 for proteolysis. Treatment of NOS1(-/-) cells with exogenous NO rescues both SOCS1 degradation and stabilization of p65 protein. Point mutation analysis demonstrated that both Cys147 and Cys179 on SOCS1 are required for its NO-dependent degradation. These findings demonstrate a fundamental role for NOS1-derived NO in regulating TLR4-mediated inflammatory gene transcription, as well as the intensity and duration of the resulting host immune response.
Collapse
Affiliation(s)
- Mirza Saqib Baig
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Sofia V Zaichick
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Mao Mao
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Andre L de Abreu
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Programa de Biociencias Aplicadas a Farmacia (PBF), Universidade Estadual de Maringa, Maringa 87020-900, Brazil
| | - Farnaz R Bakhshi
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Peter C Hart
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Anatomy and Cell Biology, Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN 46202
| | - Uzma Saqib
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Jing Deng
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Saurabh Chatterjee
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Michelle L Block
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Stephen M Vogel
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Asrar B Malik
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Marcia E L Consolaro
- Programa de Biociencias Aplicadas a Farmacia (PBF), Universidade Estadual de Maringa, Maringa 87020-900, Brazil
| | - John W Christman
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Richard D Minshall
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Environmental Health Sciences, University of South Carolina, Columbia, SC 29208
| | - Benjamin N Gantner
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Marcelo G Bonini
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Anatomy and Cell Biology, Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN 46202
| |
Collapse
|
49
|
Banoth B, Chatterjee B, Vijayaragavan B, Prasad MVR, Roy P, Basak S. Stimulus-selective crosstalk via the NF-κB signaling system reinforces innate immune response to alleviate gut infection. eLife 2015; 4. [PMID: 25905673 PMCID: PMC4432492 DOI: 10.7554/elife.05648] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 04/22/2015] [Indexed: 01/16/2023] Open
Abstract
Tissue microenvironment functions as an important determinant of the inflammatory response elicited by the resident cells. Yet, the underlying molecular mechanisms remain obscure. Our systems-level analyses identified a duration code that instructs stimulus specific crosstalk between TLR4-activated canonical NF-κB pathway and lymphotoxin-β receptor (LTβR) induced non-canonical NF-κB signaling. Indeed, LTβR costimulation synergistically enhanced the late RelA/NF-κB response to TLR4 prolonging NF-κB target gene-expressions. Concomitant LTβR signal targeted TLR4-induced newly synthesized p100, encoded by Nfkb2, for processing into p52 that not only neutralized p100 mediated inhibitions, but potently generated RelA:p52/NF-κB activity in a positive feedback loop. Finally, Nfkb2 connected lymphotoxin signal within the intestinal niche in reinforcing epithelial innate inflammatory RelA/NF-κB response to Citrobacter rodentium infection, while Nfkb2−/− mice succumbed to gut infections owing to stromal defects. In sum, our results suggest that signal integration via the pleiotropic NF-κB system enables tissue microenvironment derived cues in calibrating physiological responses. DOI:http://dx.doi.org/10.7554/eLife.05648.001 The innate immune system is the body's first line of defense against infection and disease. Innate immune cells are found in every tissue type, poised to respond immediately to damaged, stressed, or infected host cells. When innate immune cells recognize any injury or infection, one of the first things they do is trigger the inflammatory response. Fluid and other immune cells then move from the blood into the injured tissues. This movement can cause redness and swelling. But the response helps to establish a physical barrier against the spread of infection, promotes the elimination of both invading microbes and damaged host cells, and encourages the repair of the tissue. Inflammation is tightly controlled. If the response is too weak, it could leave an individual prone to serious infection. On the other hand, excessive inflammation can severely damage healthy cells and tissues. Inflammation is regulated differently in different tissue types, and the environment within the tissue itself influences the activity of local innate immune cells and the inflammatory response. However, the molecular mechanisms responsible for receiving and interpreting the signals derived from the host tissue remain unknown. Now, Banoth et al., have revealed that the integration of inflammation-provoking signals, such as injury or infection and cues from the tissue environment occurs via the so-called ‘NF-κB signaling system’. NF-κB is a protein found in almost all cell types, and when activated it is able to switch on the expression of many different genes. Banoth et al. explain that signal integration via the NF-κB system enables cues from the tissue environment to tune a cell's responses. Further experiments confirmed the importance of this signal integration by showing how a signal coming from intestinal tissue can influence the activity of innate immune cells and inflammation in the gut. These findings suggest that a breakdown in the NF-κB signaling system's ability to integrate multiple signals, including those derived from the tissue environment, may be responsible for many inflammatory disorders, and in particular those that involve the gut. Future work is now needed to explore this possibility. DOI:http://dx.doi.org/10.7554/eLife.05648.002
Collapse
Affiliation(s)
- Balaji Banoth
- Systems Immunology Laboratory, National Institute of Immunology, New Delhi, India
| | | | | | - M V R Prasad
- Systems Immunology Laboratory, National Institute of Immunology, New Delhi, India
| | - Payel Roy
- Systems Immunology Laboratory, National Institute of Immunology, New Delhi, India
| | - Soumen Basak
- Systems Immunology Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
50
|
Greco E, Aita A, Galozzi P, Gava A, Sfriso P, Negm OH, Tighe P, Caso F, Navaglia F, Dazzo E, De Bortoli M, Rampazzo A, Obici L, Donadei S, Merlini G, Plebani M, Todd I, Basso D, Punzi L. The novel S59P mutation in the TNFRSF1A gene identified in an adult onset TNF receptor associated periodic syndrome (TRAPS) constitutively activates NF-κB pathway. Arthritis Res Ther 2015; 17:93. [PMID: 25888769 PMCID: PMC4416318 DOI: 10.1186/s13075-015-0604-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 03/20/2015] [Indexed: 11/11/2022] Open
Abstract
Introduction Mutations in the TNFRSF1A gene, encoding tumor necrosis factor receptor 1 (TNF-R1), are associated with the autosomal dominant autoinflammatory disorder, called TNF receptor associated periodic syndrome (TRAPS). TRAPS is clinically characterized by recurrent episodes of long-lasting fever and systemic inflammation. A novel mutation (c.262 T > C; S59P) in the TNFRSF1A gene at residue 88 of the mature protein was recently identified in our laboratory in an adult TRAPS patient. The aim of this study was to functionally characterize this novel TNFRSF1A mutation evaluating its effects on the TNF-R1-associated signaling pathways, firstly NF-κB, under particular conditions and comparing the results with suitable control mutations. Methods HEK-293 cell line was transfected with pCMV6-AC construct expressing wild-type (WT) or c.262 T > C (S59P), c.362G > A (R92Q), c.236C > T (T50M) TNFRSF1A mutants. Peripheral blood mononuclear cells (PBMCs) were instead isolated from two TRAPS patients carrying S59P and R92Q mutations and from five healthy subjects. Both transfected HEK-293 and PBMCs were stimulated with tumor necrosis factor (TNF) or interleukin 1β (IL-1β) to evaluate the expression of TNF-R1, the activation of TNF-R1-associated downstream pathways and the pro-inflammatory cytokines by means of immunofluorescent assay, array-based technique, immunoblotting and immunometric assay, respectively. Results TNF induced cytoplasmic accumulation of TNF-R1 in all mutant cells. Furthermore, all mutants presented a particular set of active TNF-R1 downstream pathways. S59P constitutively activated IL-1β, MAPK and SRC/JAK/STAT3 pathways and inhibited apoptosis. Also, NF-κB pathway involvement was demonstrated in vitro by the enhancement of p-IκB-α and p65 nuclear subunit of NF-κB expression in all mutants in the presence of TNF or IL-1β stimulation. These in vitro results correlated with patients’ data from PBMCs. Concerning the pro-inflammatory cytokines secretion, mainly IL-1β induced a significant and persistent enhancement of IL-6 and IL-8 in PBMCs carrying the S59P mutation. Conclusions The novel S59P mutation leads to defective cellular trafficking and to constitutive activation of TNF-R1. This mutation also determines constitutive activation of the IL-1R pathway, inhibition of apoptosis and enhanced and persistent NF-κB activation and cytokine secretion in response to IL-1β stimulation. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0604-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eliana Greco
- University of Padova, Rheumatology Unit, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy. .,University of Padova, Laboratory Medicine, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy.
| | - Ada Aita
- University of Padova, Rheumatology Unit, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy. .,University of Padova, Laboratory Medicine, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy.
| | - Paola Galozzi
- University of Padova, Rheumatology Unit, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy.
| | - Alessandra Gava
- University of Padova, Rheumatology Unit, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy.
| | - Paolo Sfriso
- University of Padova, Rheumatology Unit, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy.
| | - Ola H Negm
- School of Life Sciences, The University of Nottingham, Queen's Medical Centre, Derby road, NG7 2UH, Nottingham, UK. .,Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Elgomhouria Street, 35516, Mansoura City, Egypt.
| | - Patrick Tighe
- School of Life Sciences, The University of Nottingham, Queen's Medical Centre, Derby road, NG7 2UH, Nottingham, UK.
| | - Francesco Caso
- University of Padova, Rheumatology Unit, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy.
| | - Filippo Navaglia
- University of Padova, Laboratory Medicine, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy.
| | - Emanuela Dazzo
- Institute of Neuroscience of the National Research Council, Section of Padova, Corso Stati Uniti, 4, 3512, Padova, Italy.
| | - Marzia De Bortoli
- Department of Biology, University of Padova, Via U. Bassi, 58/B, 35121, Padova, Italy.
| | - Alessandra Rampazzo
- Department of Biology, University of Padova, Via U. Bassi, 58/B, 35121, Padova, Italy.
| | - Laura Obici
- Amyloidosis Research and Treatment Center, Biotechnology Research laboratories, Fondazione IRCSS Policlinico San Matteo and University of Pavia, Viale Camillo Golgi 19, 27100, Pavia, Italy.
| | - Simona Donadei
- Amyloidosis Research and Treatment Center, Biotechnology Research laboratories, Fondazione IRCSS Policlinico San Matteo and University of Pavia, Viale Camillo Golgi 19, 27100, Pavia, Italy.
| | - Giampaolo Merlini
- Amyloidosis Research and Treatment Center, Biotechnology Research laboratories, Fondazione IRCSS Policlinico San Matteo and University of Pavia, Viale Camillo Golgi 19, 27100, Pavia, Italy.
| | - Mario Plebani
- University of Padova, Laboratory Medicine, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy.
| | - Ian Todd
- School of Life Sciences, The University of Nottingham, Queen's Medical Centre, Derby road, NG7 2UH, Nottingham, UK.
| | - Daniela Basso
- University of Padova, Laboratory Medicine, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy.
| | - Leonardo Punzi
- University of Padova, Rheumatology Unit, Department of Medicine - DIMED, Via Giustiniani 2, 35128, Padova, Italy.
| |
Collapse
|