1
|
Ramirez GA, Cardamone C, Lettieri S, Fredi M, Mormile I. Clinical and Pathophysiological Tangles Between Allergy and Autoimmunity: Deconstructing an Old Dichotomic Paradigm. Clin Rev Allergy Immunol 2025; 68:13. [PMID: 39932658 PMCID: PMC11814061 DOI: 10.1007/s12016-024-09020-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2024] [Indexed: 02/14/2025]
Abstract
Allergic and autoimmune disorders are characterised by dysregulation of the immune responses to otherwise inert environmental substances and autoantigens, leading to inflammation and tissue damage. Their incidence has constantly increased in the last decades, and their co-occurrence defies current standards in patient care. For years, allergy and autoimmunity have been considered opposite conditions, with IgE and Th2 lymphocytes cascade driving canonical allergic manifestations and Th1/Th17-related pathways accounting for autoimmunity. Conversely, growing evidence suggests that these conditions not only share some common inciting triggers but also are subtended by overlapping pathogenic pathways. Permissive genetic backgrounds, along with epithelial barrier damage and changes in the microbiome, are now appreciated as common risk factors for both allergy and autoimmunity. Eosinophils and mast cells, along with autoreactive IgE, are emerging players in triggering and sustaining autoimmunity, while pharmacological modulation of B cells and Th17 responses has provided novel clues to the pathophysiology of allergy. By combining clinical and therapeutic evidence with data from mechanistic studies, this review provides a state-of-the-art update on the complex interplay between allergy and autoimmunity, deconstructing old dichotomic paradigms and offering potential clues for future research.
Collapse
Affiliation(s)
- Giuseppe A Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Chiara Cardamone
- Immunorheumatology Unit, University Hospital "San Giovanni Di Dio E Ruggi d'Aragona", Largo Città d'Ippocrate, Via San Leonardo 1, 84131, Salerno, Italy.
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy.
| | - Sara Lettieri
- Pulmonology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Micaela Fredi
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Ilaria Mormile
- Division of Internal Medicine and Clinical Immunology, Department of Internal Medicine and Clinical Complexity, AOU Federico II, Naples, Italy
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
2
|
Hamouda HA, Sayed RH, Eid NI, El-Sayeh BM. Azilsartan Attenuates 3-Nitropropinoic Acid-Induced Neurotoxicity in Rats: The Role of IĸB/NF-ĸB and KEAP1/Nrf2 Signaling Pathways. Neurochem Res 2024; 49:1017-1033. [PMID: 38184805 PMCID: PMC10901959 DOI: 10.1007/s11064-023-04083-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 01/08/2024]
Abstract
Huntington's disease (HD) is an autosomal-dominant neurodegenerative disorder characterized by motor, psychiatric and cognitive symptoms. Injection of 3-nitropropionic acid (3-NP) is a widely used experimental model for induction of HD. The current study aimed to inspect the potential neuroprotective properties of azilsartan (Azil), an angiotensin II type 1 receptor blocker (ATR1), in 3-NP-induced striatal neurotoxicity in rats. Rats were randomly allocated into five groups and treated for 14 days as follows: group I received normal saline; group II received Azil (10 mg/kg, p.o.); group III received 3-NP (10 mg/kg, i.p); group IV and V received Azil (5 or 10 mg/kg, p.o, respectively) 1 h prior to 3-NP injection. Both doses of Azil markedly attenuated motor and behavioural dysfunction as well as striatal histopathological alterations caused by 3-NP. In addition, Azil balanced striatal neurotransmitters levels as evidenced by the increase of striatal gamma-aminobutyric acid content and the decrease of glutamate content. Azil also amended neuroinflammation and oxidative stress via modulating IĸB/NF-ĸB and KEAP1/Nrf2 downstream signalling pathways, as well as reducing iNOS and COX2 levels. Moreover, Azil demonstrated an anti-apoptotic activity by reducing caspase-3 level and BAX/BCL2 ratio. In conclusion, the present study reveals the neuroprotective potential of Azil in 3-NP-induced behavioural, histopathological and biochemical changes in rats. These findings might be attributed to inhibition of ATR1/NF-κB signalling, modulation of Nrf2/KEAP1 signalling, anti-inflammatory, anti-oxidant and anti-apoptotic properties.
Collapse
Affiliation(s)
- Hend A Hamouda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt.
- School of Pharmacy, Newgiza University, Giza, Egypt.
| | - Nihad I Eid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt
| | - Bahia M El-Sayeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt
| |
Collapse
|
3
|
Franke K, Li Z, Bal G, Zuberbier T, Babina M. Synergism between IL-33 and MRGPRX2/FcεRI Is Primarily Due to the Complementation of Signaling Modules, and Only Modestly Supplemented by Prolonged Activation of Selected Kinases. Cells 2023; 12:2700. [PMID: 38067128 PMCID: PMC10705352 DOI: 10.3390/cells12232700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/16/2023] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
Skin mast cells (MCs) express high levels of MRGPRX2, FcεRI, and ST2, and vigorously respond to their ligands when triggered individually. IL-33/ST2 also potently synergizes with other receptors, but the molecular underpinnings are poorly understood. Human skin-derived MCs were stimulated via different receptors individually or jointly in the presence/absence of selective inhibitors. TNF was quantified by ELISA. Signaling cascades were studied by immunoblot. TNF was stimulated by FcεRI ≈ ST2 > MRGPRX2. Surprisingly, neither FcεRI nor MRGPRX2 stimulation elicited NF-κB activation (IκB degradation, p65 phosphorylation) in stark contrast to IL-33. Accordingly, TNF production did not depend on NF-κB in FcεRI- or MRGPRX2-stimulated MCs, but did well so downstream of ST2. Conversely, ERK1/2 and PI3K were the crucial modules upon FcεRI/MRGPRX2 stimulation, while p38 was key to the IL-33-elicited route. The different signaling prerequisites were mirrored by their activation patterns with potent pERK/pAKT after FcεRI/MRGPRX2, but preferential induction of pp38/NF-κB downstream of ST2. FcεRI/MRGPRX2 strongly synergized with IL-33, and some synergy was still observed upon inhibition of each module (ERK1/2, JNK, p38, PI3K, NF-κB). IL-33's contribution to synergism was owed to p38 > JNK > NF-κB, while the partner receptor contributed through ERK > PI3K ≈ JNK. Concurrent IL-33 led to slightly prolonged pERK (downstream of MRGPRX2) or pAKT (activated by FcεRI), while the IL-33-elicited modules (pp38/NF-κB) remained unaffected by co-stimulation of FcεRI/MRGPRX2. Collectively, the strong synergistic activity of IL-33 primarily results from the complementation of highly distinct modules following co-activation of the partner receptor rather than by altered signal strength of the same modules.
Collapse
Affiliation(s)
- Kristin Franke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Zhuoran Li
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Gürkan Bal
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Torsten Zuberbier
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Magda Babina
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| |
Collapse
|
4
|
Smith CF, Brandehoff NP, Pepin L, McCabe MC, Castoe TA, Mackessy SP, Nemkov T, Hansen KC, Saviola AJ. Feasibility of detecting snake envenomation biomarkers from dried blood spots. ANALYTICAL SCIENCE ADVANCES 2023; 4:26-36. [PMID: 38715579 PMCID: PMC10989584 DOI: 10.1002/ansa.202200050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/24/2023] [Accepted: 02/05/2023] [Indexed: 11/17/2024]
Abstract
Biofluid proteomics is a sensitive and high throughput technique that provides vast amounts of molecular data for biomarker discovery. More recently, dried blood spots (DBS) have gained traction as a stable, noninvasive, and relatively cheap source of proteomic data for biomarker identification in disease and injury. Snake envenomation is responsible for significant morbidity and mortality worldwide; however, much remains unknown about the systemic molecular response to envenomation and acquiring biological samples for analysis is a major hurdle. In this study, we utilized DBS acquired from a case of lethal rattlesnake envenomation to determine the feasibility of discovering biomarkers associated with human envenomation. We identified proteins that were either unique or upregulated in envenomated blood compared to non-envenomated blood and evaluated if physiological response pathways and protein markers that correspond to the observed syndromes triggered by envenomation could be detected. We demonstrate that DBS provide useful proteomic information on the systemic processes that resulted from envenomation in this case and find evidence for a massive and systemic inflammatory cascade, combined with coagulation dysregulation, complement system activation, hypoxia response activation, and apoptosis. We also detected potential markers indicative of lethal anaphylaxis, cardiac arrest, and brain death. Ultimately, DBS proteomics has the potential to provide stable and sensitive molecular data on envenomation syndromes and response pathways, which is particularly relevant in low-resource areas which may lack the materials for biofluid processing and storage.
Collapse
Affiliation(s)
- Cara F. Smith
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado DenverAuroraCOUSA
| | | | - Lesley Pepin
- Rocky Mountain Poison and Drug Safety, Denver Health and Hospital AuthorityDenverCOUSA
| | - Maxwell C. McCabe
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado DenverAuroraCOUSA
| | - Todd A. Castoe
- Department of BiologyUniversity of Texas at ArlingtonArlingtonTXUSA
| | - Stephen P. Mackessy
- Department of Biological SciencesUniversity of Northern ColoradoGreeleyCOUSA
| | - Travis Nemkov
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado DenverAuroraCOUSA
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado DenverAuroraCOUSA
| | - Anthony J. Saviola
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado DenverAuroraCOUSA
| |
Collapse
|
5
|
Qian G, Adeyanju O, Sunil C, Huang SK, Chen SY, Tucker TA, Idell S, Guo X. Dedicator of Cytokinesis 2 (DOCK2) Deficiency Attenuates Lung Injury Associated with Chronic High-Fat and High-Fructose Diet-Induced Obesity. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:226-238. [PMID: 34767813 PMCID: PMC8883439 DOI: 10.1016/j.ajpath.2021.10.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 09/21/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023]
Abstract
Obesity is a major risk factor for lung disease development. However, little is known about the impact of chronic high-fat and high-fructose (HFHF) diet-induced obesity on lung inflammation and subsequent pulmonary fibrosis. Herein we hypothesized that dedicator of cytokinesis 2 (DOCK2) promotes a proinflammatory phenotype of lung fibroblasts (LFs) to elicit lung injury and fibrosis in chronic HFHF diet-induced obesity. An HFHF diet for 20 weeks induced lung inflammation and profibrotic changes in wild-type C57BL/6 mice. CD68 and monocyte chemoattractant protein-1 (MCP-1) expression were notably increased in the lungs of wild-type mice fed an HFHF diet. An HFHF diet further increased lung DOCK2 expression that co-localized with fibroblast-specific protein 1, suggesting a role of DOCK2 in regulating proinflammatory phenotype of LFs. Importantly, DOCK2 knockout protected mice from lung inflammation and fibrosis induced by a HFHF diet. In primary human LFs, tumor necrosis factor-α (TNF-α) and IL-1β induced DOCK2 expression concurrent with MCP-1, IL-6, and matrix metallopeptidase 2. DOCK2 knockdown suppressed TNF-α-induced expression of these molecules and activation of phosphatidylinositol 3-kinase/AKT and NF-κB signaling pathways, suggesting a mechanism of DOCK2-mediated proinflammatory and profibrotic changes in human LFs. Taken together, these findings reveal a previously unrecognized role of DOCK2 in regulating proinflammatory phenotype of LFs, potentiation of lung inflammation, and pulmonary fibrosis in chronic HFHF diet-caused obesity.
Collapse
Affiliation(s)
- Guoqing Qian
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Oluwaseun Adeyanju
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Christudas Sunil
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Steven K. Huang
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Shi-You Chen
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia,Department of Surgery, School of Medicine, The University of Missouri, Columbia, Missouri
| | - Torry A. Tucker
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Xia Guo
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas,Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia,Address correspondence to Xia Guo, Ph.D., Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, 11937 US Highway 271, Lab A-1, Tyler, TX 75708.
| |
Collapse
|
6
|
Mace EL, Zhao S, Lipscomb B, Wootten CT, Belcher RH. Clinical Significance of Mast Cells in the Supraglottic Larynx of Children With Aerodigestive Disease. Otolaryngol Head Neck Surg 2021; 167:375-381. [PMID: 34699295 DOI: 10.1177/01945998211055139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To analyze the association of mast cells found on supraglottic biopsy of pediatric patients with common aerodigestive diseases. STUDY DESIGN Cross-sectional study. SETTING Tertiary care children's hospital. METHODS A total of 461 pediatric patients undergoing otolaryngology aerodigestive procedures provided consent between 2014 and 2019, and biopsies of the supraglottic larynx were collected at the time of their surgery. Pathologists reviewed biopsies for the presence and number of mast cells per high-power field. The patients' electronic health records were reviewed for relevant demographic data and clinical diagnoses present at the time of biopsy. Multivariate logistic regression was used to assess the relationship of mast cells with odds of aerodigestive disease. RESULTS Patients with mast cells in their biopsy had significantly higher odds of asthma (odds ratio [OR], 2.02; 95% CI, 1.17-3.46), gastroesophageal reflux disease (OR, 2.36; 95% CI, 1.47-3.77), laryngomalacia (OR, 2.98; 95% CI, 1.80-4.94), laryngeal anomalies (OR, 2.32; 95% CI, 1.52-3.55), and obstructive sleep apnea (OR, 2.16; 95% CI, 1.35-3.45). When mast cells were evaluated as a continuous variable, there was a nonlinear relationship between increasing mast cell count and odds of disease. CONCLUSIONS Mast cells are known to be associated with inflammatory conditions, though little is known about their presence in laryngeal inflammation. Results from our study demonstrate an association between mast cells in the pediatric larynx and asthma, gastroesophageal reflux disease, laryngomalacia, laryngeal anomalies, and obstructive sleep apnea. Our study also showed a nonlinear relationship between number of mast cells and odds of disease diagnosis.
Collapse
Affiliation(s)
- Emily L Mace
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Brittany Lipscomb
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Christopher T Wootten
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ryan H Belcher
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
7
|
Nguyen SMT, Rupprecht CP, Haque A, Pattanaik D, Yusin J, Krishnaswamy G. Mechanisms Governing Anaphylaxis: Inflammatory Cells, Mediators, Endothelial Gap Junctions and Beyond. Int J Mol Sci 2021; 22:ijms22157785. [PMID: 34360549 PMCID: PMC8346007 DOI: 10.3390/ijms22157785] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
Anaphylaxis is a severe, acute, life-threatening multisystem allergic reaction resulting from the release of a plethora of mediators from mast cells culminating in serious respiratory, cardiovascular and mucocutaneous manifestations that can be fatal. Medications, foods, latex, exercise, hormones (progesterone), and clonal mast cell disorders may be responsible. More recently, novel syndromes such as delayed reactions to red meat and hereditary alpha tryptasemia have been described. Anaphylaxis manifests as sudden onset urticaria, pruritus, flushing, erythema, angioedema (lips, tongue, airways, periphery), myocardial dysfunction (hypovolemia, distributive or mixed shock and arrhythmias), rhinitis, wheezing and stridor. Vomiting, diarrhea, scrotal edema, uterine cramps, vaginal bleeding, urinary incontinence, dizziness, seizures, confusion, and syncope may occur. The traditional (or classical) pathway is mediated via T cells, Th2 cytokines (such as IL-4 and 5), B cell production of IgE and subsequent crosslinking of the high affinity IgE receptor (FcεRI) on mast cells and basophils by IgE-antigen complexes, culminating in mast cell and basophil degranulation. Degranulation results in the release of preformed mediators (histamine, heparin, tryptase, chymase, carboxypeptidase, cathepsin G and tumor necrosis factor alpha (TNF-α), and of de novo synthesized ones such as lipid mediators (cysteinyl leukotrienes), platelet activating factor (PAF), cytokines and growth factors such as vascular endothelial growth factor (VEGF). Of these, histamine, tryptase, cathepsin G, TNF-α, LTC4, PAF and VEGF can increase vascular permeability. Recent data suggest that mast cell-derived histamine and PAF can activate nitric oxide production from endothelium and set into motion a signaling cascade that leads to dilatation of blood vessels and dysfunction of the endothelial barrier. The latter, characterized by the opening of adherens junctions, leads to increased capillary permeability and fluid extravasation. These changes contribute to airway edema, hypovolemia, and distributive shock, with potentially fatal consequences. In this review, besides mechanisms (endotypes) underlying IgE-mediated anaphylaxis, we also provide a brief overview of IgG-, complement-, contact system-, cytokine- and mast cell-mediated reactions that can result in phenotypes resembling IgE-mediated anaphylaxis. Such classifications can lead the way to precision medicine approaches to the management of this complex disease.
Collapse
Affiliation(s)
| | | | - Aaisha Haque
- The Bill Hefner VA Medical Center, Salisbury, NC 27106, USA;
| | - Debendra Pattanaik
- Division of Allergy and Immunology, UT Memphis College of Medicine, Memphis, TN 38103, USA;
| | - Joseph Yusin
- The Division of Allergy and Immunology, Greater Los Angeles VA Medical Center, Los Angeles, CA 90011, USA;
| | - Guha Krishnaswamy
- Department of Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27106, USA;
- The Bill Hefner VA Medical Center, Salisbury, NC 27106, USA;
- Correspondence:
| |
Collapse
|
8
|
Anti-Inflammatory Effects of an Extract from Pseudomonas aeruginosa and Its Purified Product 1-Hydroxyphenazine on RAW264.7 Cells. Curr Microbiol 2021; 78:2762-2773. [PMID: 34043026 DOI: 10.1007/s00284-021-02544-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 05/17/2021] [Indexed: 12/28/2022]
Abstract
The purpose of this study was to discuss the effects of an extract from the culture medium of Pseudomonas aeruginosa (P. aeruginosa) 2016NX1 (chloroform extract of P. aeruginosa, CEPA) and its purified product 1-hydroxyphenazine on RAW264.7 cell inflammation. Cell viability was evaluated by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) method. TNF-α production was determined by an ELISA method. The effects of CEPA and its purified product 1-hydroxyphenazine on cell morphology were investigated using an inverted microscope. Quantitative real-time PCR was performed to determine mRNA expression levels. CEPA and 1-hydroxyphenazine had no obvious toxicity to cells when their concentrations were no more than 20 μg ml-1 and 5 μg ml-1, respectively. Both CEPA and 1-hydroxyphenazine suppressed the secretion of TNF-α and significantly reduced the mRNA expression levels of TNF-α, IL-1β, and IL-6. Both CEPA and 1-hydroxyphenazine inhibited M1 cell polarization after lipopolysaccharide (LPS) stimulation. The results in this article lay a good foundation for the biopharmaceutical applications of CEPA and 1-hydroxyphenazine in the future. CEPA and 1-hydroxyphenazine had certain anti-inflammatory activity, and inhibited LPS-induced RAW264.7 cell inflammation. Our findings suggest that CEPA and 1-hydroxyphenazine are potential chemicals with anti-inflammatory activity.
Collapse
|
9
|
Ge Q, Ying J, Shi Z, Mao Q, Jin H, Wang PE, Chen J, Yuan W, Tong P, Li J. Chlorogenic Acid retards cartilaginous endplate degeneration and ameliorates intervertebral disc degeneration via suppressing NF-κB signaling. Life Sci 2021; 274:119324. [PMID: 33711382 DOI: 10.1016/j.lfs.2021.119324] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/19/2021] [Accepted: 02/27/2021] [Indexed: 02/07/2023]
Abstract
AIMS Intervertebral Disc Degeneration (IDD) is a key factor involved in low back pain (LBP) which affects approximately 540 million individuals worldwide. Chlorogenic Acid (CGA), a natural compound, exerts anti-inflammatory property in several diseases. Here, we aim to investigate the biological effect of CGA on IDD and explore the underlying mechanism. MATERIALS AND METHODS Lumbar spine instability (LSI) model in mice was utilized to mimic process of IDD. The effects of CGA in response to LSI were evaluated by luminescent imaging, micro-CT, histomorphology, and immunohistochemistry in vivo. Besides, the cytotoxicity of CGA on chondrocytes was detected by cell counting kit-8 (CCK-8) and the biological effects were assessed by polymerase chain reaction (PCR) in vitro. KEY FINDINGS We found that CGA treatment dramatically suppressed the NF-κB activity in LSI mice. Moreover, administration of CGA mitigated cartilaginous endplate degeneration and postponed IDD development accompanying a decrease of inflammatory and catabolic mediators. Specifically, CGA ameliorated endplate degeneration might be related to its protective effects against endplate chondrocytes apoptosis and trans-differentiation. We further elucidated that CGA exerted these biological effects mainly by repressing NF-κB signaling in cartilage endplate. SIGNIFICANCE Our study has illustrated, for the first time, the curative effects as well as the latent mechanism of CGA in IDD and our results suggested that CGA administration might be used as an alternative therapy for IDD.
Collapse
Affiliation(s)
- Qinwen Ge
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jun Ying
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Zhenyu Shi
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiang Mao
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ping-Er Wang
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiali Chen
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wenhua Yuan
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Peijian Tong
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China.
| | - Ju Li
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China.
| |
Collapse
|
10
|
Ayo TE, Adhikari P, Sugita S, Xu H. TNF Production in Activated RBL-2H3 Cells Requires Munc13-4. Inflammation 2021; 43:744-751. [PMID: 31897916 DOI: 10.1007/s10753-019-01161-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mast cell activation triggers intricate signaling pathways that promote the expression and/or release of a wide range of mediators including tumor necrosis factor (TNF; also known as TNFα). In this study, we investigated the connection between TNF secretion and TNF production, exploiting RBL-2H3 cells (a tumor analog of mucosal mast cells) that are depleted of Munc13-4, a crucial component of the mast cell exocytic machinery. We showed that antigen/IgE elicited robust TNF production in RBL-2H3 cells, but not in Munc13-4 knockout cells. The production defect was corrected when Munc13-4 was reintroduced into the knockout cell line, suggesting that the phenotype was not caused by any secondary effect derived from the knockout approach. Furthermore, pre-incubation of RBL-2H3 cells with R-7050, an antagonist of TNF receptor-dependent signaling, was shown to block TNF production without inhibiting TNF release. These observations provide fresh evidence for a robust feed-back loop to boost TNF production in activated mast cells.
Collapse
Affiliation(s)
- Tolulope E Ayo
- Department of Cell and Molecular Biology, School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Pratikshya Adhikari
- Department of Cell and Molecular Biology, School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Shuzo Sugita
- Division of Fundamental Neurobiology, University Health Network, Toronto, ON, M5T 2S8, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Hao Xu
- Department of Cell and Molecular Biology, School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS, 39406, USA.
| |
Collapse
|
11
|
Choi YA, Lee S, Choi JK, Kang BC, Kim MJ, Dhakal H, Kwon TK, Khang D, Kim SH. The suppressive effect of dabrafenib, a therapeutic agent for metastatic melanoma, in IgE-mediated allergic inflammation. Int Immunopharmacol 2020; 83:106398. [PMID: 32197228 DOI: 10.1016/j.intimp.2020.106398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/21/2022]
Abstract
The functional inhibition of mast cells, which serve as a key effector cells in allergic reactions may be a specific target for treating immunoglobulin (Ig)E-mediated allergic reactions, which occur in various allergic diseases including anaphylaxis, asthma, and atopic dermatitis. In this study, we demonstrated the effects of dabrafenib, a therapeutic agent used to treat metastatic melanoma, with a focus on mast cell activation and local cutaneous anaphylaxis. In two types of mast cells (RBL-2H3 and mouse bone marrow-derived mast cells), dabrafenib (0.01, 0.1, 1 μM) pretreatment significantly decreased IgE-induced degranulation, intracellular calcium influx, and the activity of intracellular signaling molecules, such as Lyn, Syk, Akt, and PLCγ. Dabrafenib ameliorated mRNA and protein expression levels of interleukin-4 and tumor necrosis factor-α by the reduction of nuclear localization of nuclear factor-κB and nuclear factor of activated T-cells. In passive cutaneous anaphylaxis, oral administration of dabrafenib (0.1, 1, 10 mg/kg) reduced local pigmentation and ear thickness in a dose-dependent manner. Taken together, these results suggest that dabrafenib is a therapeutic drug candidate that controls IgE-mediated allergic inflammatory diseases through suppression of mast cell activity.
Collapse
Affiliation(s)
- Young-Ae Choi
- Cell & Matrix Research Institute, Department of Pharmacology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Soyoung Lee
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Jin Kyeong Choi
- Molecular Immunology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA; Department of Immunology, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Byeong-Cheol Kang
- Cell & Matrix Research Institute, Department of Pharmacology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Min-Jong Kim
- Cell & Matrix Research Institute, Department of Pharmacology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Hima Dhakal
- Cell & Matrix Research Institute, Department of Pharmacology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Dongwoo Khang
- Department of Physiology, School of Medicine, Gachon University, Incheon, Republic of Korea.
| | - Sang-Hyun Kim
- Cell & Matrix Research Institute, Department of Pharmacology, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
12
|
Li M, Mittal SK, Foulsham W, Amouzegar A, Sahu SK, Chauhan SK. Mast cells contribute to the induction of ocular mucosal alloimmunity. Am J Transplant 2019; 19:662-673. [PMID: 30129280 PMCID: PMC7941346 DOI: 10.1111/ajt.15084] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 08/05/2018] [Accepted: 08/12/2018] [Indexed: 01/25/2023]
Abstract
Beyond their historical role as the effector cells in allergic disorders, mast cells have been implicated in regulating both innate and adaptive immune responses. Possessing considerable functional plasticity, mast cells are abundant at mucosal surfaces, where the host and external environments interface. The purpose of this study was to evaluate the contribution of mast cells to allograft rejection at the ocular surface. Using a well-characterized murine model of corneal transplantation, we report that mast cells promote allosensitization. Our data show mast cell frequencies and activation are increased following transplantation. We demonstrate that mast cell inhibition (a) limits the infiltration of inflammatory cells and APC maturation at the graft site; (b) reduces allosensitization and the generation of Th1 cells in draining lymphoid tissues; (c) decreases graft infiltration of alloimmune-inflammatory cells; and (d) prolongs allograft survival. Our data demonstrate a novel function of mast cells in promoting allosensitization at the ocular surface.
Collapse
Affiliation(s)
- Mingshun Li
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA,Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Sharad K. Mittal
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - William Foulsham
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Afsaneh Amouzegar
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Srikant K. Sahu
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA,L.V. Prasad Eye Institute, Bhubaneswar, Odisha, India
| | - Sunil K. Chauhan
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Sahu SK, Mittal SK, Foulsham W, Li M, Sangwan VS, Chauhan SK. Mast Cells Initiate the Recruitment of Neutrophils Following Ocular Surface Injury. Invest Ophthalmol Vis Sci 2019; 59:1732-1740. [PMID: 29610857 PMCID: PMC5885762 DOI: 10.1167/iovs.17-23398] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Purpose The purpose of this study was to investigate the contribution of mast cells to early neutrophil recruitment during ocular inflammation. Methods In a murine model of corneal injury, the epithelium and anterior stroma were removed using a handheld motor brush. Cromolyn sodium (2% in PBS) eye drops were administered topically for mast cell inhibition. In vitro, bone marrow–derived mast cells were cultured alone or with corneal tissue. The frequencies of CD45+ inflammatory cells, CD11b+Ly6G+ neutrophils, and ckit+FcεR1+ mast cells in the cornea were assessed by flow cytometry. mRNA expression of CXCL2 was evaluated by real-time PCR and protein expression by ELISA. β-Hexosaminidase assays were performed to gauge mast cell activation. Results Neutrophil infiltration of the cornea was observed within 1 hour of injury, with neutrophil frequencies increasing over subsequent hours. Concurrent expansion of mast cell frequencies at the cornea were observed, with mast cell activation (assessed by β-hexosaminidase levels) peaking at 6 hours after injury. Evaluation of CXCL2 mRNA and protein expression levels demonstrated augmented expression by injured corneal tissue relative to naïve corneal tissue. Mast cells were observed to constitutively express CXCL2, with significantly higher expression of CXCL2 protein compared with naïve corneal tissue. Culture with harvested injured corneas further amplified CXCL2 expression by mast cells. In vivo, mast cell inhibition was observed to decrease CXCL2 expression, limit early neutrophil infiltration, and reduce inflammatory cytokine expression by the cornea. Conclusions Our data suggest that mast cell activation after corneal injury amplifies their secretion of CXCL2 and promotes the initiation of early neutrophil recruitment.
Collapse
Affiliation(s)
- Srikant K Sahu
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States.,L.V. Prasad Eye Institute, Bhubaneswar, Odisha, India
| | - Sharad K Mittal
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - William Foulsham
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Mingshun Li
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States.,Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Beijing, P.R. China
| | - Virender S Sangwan
- L.V. Prasad Eye Institute, Bhubaneswar, Odisha, India.,L.V. Prasad Eye Institute, Hyderabad, India
| | - Sunil K Chauhan
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
14
|
Green DP, Limjunyawong N, Gour N, Pundir P, Dong X. A Mast-Cell-Specific Receptor Mediates Neurogenic Inflammation and Pain. Neuron 2019; 101:412-420.e3. [PMID: 30686732 DOI: 10.1016/j.neuron.2019.01.012] [Citation(s) in RCA: 278] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/29/2018] [Accepted: 12/31/2018] [Indexed: 12/12/2022]
Abstract
Mast cells can be found in close proximity to peripheral nerve endings where, upon activation, they release a broad range of pro-inflammatory cytokines and chemokines. However, the precise mechanism underlying this so-called neurogenic inflammation and associated pain has remained elusive. Here we report that the mast-cell-specific receptor Mrgprb2 mediates inflammatory mechanical and thermal hyperalgesia and is required for recruitment of innate immune cells at the injury site. We also found that the neuropeptide substance P (SP), an endogenous agonist of Mrgprb2, facilitates immune cells' migration via Mrgprb2. Furthermore, SP activation of the human mast cell led to the release of multiple pro-inflammatory cytokines and chemokines via the human homolog MRGPRX2. Surprisingly, the SP-mediated inflammatory responses were independent of its canonical receptor, neurokinin-1 receptor (NK-1R). These results identify Mrgprb2/X2 as an important neuroimmune modulator and a potential target for treating inflammatory pain.
Collapse
Affiliation(s)
- Dustin P Green
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nathachit Limjunyawong
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Naina Gour
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Priyanka Pundir
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Howard Hughes Medical Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
15
|
Donaldson AR, Tanase CE, Awuah D, Vasanthi Bathrinarayanan P, Hall L, Nikkhah M, Khademhosseini A, Rose F, Alexander C, Ghaemmaghami AM. Photocrosslinkable Gelatin Hydrogels Modulate the Production of the Major Pro-inflammatory Cytokine, TNF-α, by Human Mononuclear Cells. Front Bioeng Biotechnol 2018; 6:116. [PMID: 30283776 PMCID: PMC6156527 DOI: 10.3389/fbioe.2018.00116] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022] Open
Abstract
Hydrogels are an attractive class of biomaterials in tissue engineering due to their inherently compatible properties for cell culture. Gelatin methacryloyl (GelMA) has shown significant promise in the fields of tissue engineering and drug delivery, as its physical properties can be precisely tuned depending on the specific application. There is a growing appreciation for the interaction between biomaterials and cells of the immune system with the increasing usage of biomaterials for in vivo applications. Here, we addressed the current lack of information regarding the immune-modulatory properties of photocrosslinked GelMA. We investigated the ability of human mononuclear cells to mount inflammatory responses in the context of a GelMA hydrogel platform. Using lipopolysaccharide to stimulate a pro-inflammatory immune response, we found tumor necrosis factor-α (TNF-α) expression was suppressed in GelMA culture conditions. Our findings have important implications on the future use of GelMA, and potentially similar hydrogels, and highlight the significance of investigating the potential immune-modulatory properties of biomaterials.
Collapse
Affiliation(s)
- Amy R Donaldson
- Immunology and Tissue Modelling Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Constantin Edi Tanase
- Immunology and Tissue Modelling Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Dennis Awuah
- Immunology and Tissue Modelling Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | - Laurence Hall
- Immunology and Tissue Modelling Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, CA, United States
| | - Felicity Rose
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Amir M Ghaemmaghami
- Immunology and Tissue Modelling Group, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
16
|
The Role of the Nuclear Factor κB Pathway in the Cellular Response to Low and High Linear Energy Transfer Radiation. Int J Mol Sci 2018; 19:ijms19082220. [PMID: 30061500 PMCID: PMC6121395 DOI: 10.3390/ijms19082220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Astronauts are exposed to considerable doses of space radiation during long-term space missions. As complete shielding of the highly energetic particles is impracticable, the cellular response to space-relevant radiation qualities has to be understood in order to develop countermeasures and to reduce radiation risk uncertainties. The transcription factor Nuclear Factor κB (NF-κB) plays a fundamental role in the immune response and in the pathogenesis of many diseases. We have previously shown that heavy ions with a linear energy transfer (LET) of 100–300 keV/µm have a nine times higher potential to activate NF-κB compared to low-LET X-rays. Here, chemical inhibitor studies using human embryonic kidney cells (HEK) showed that the DNA damage sensor Ataxia telangiectasia mutated (ATM) and the proteasome were essential for NF-κB activation in response to X-rays and heavy ions. NF-κB’s role in cellular radiation response was determined by stable knock-down of the NF-κB subunit RelA. Transfection of a RelA short-hairpin RNA plasmid resulted in higher sensitivity towards X-rays, but not towards heavy ions. Reverse Transcriptase real-time quantitative PCR (RT-qPCR) showed that after exposure to X-rays and heavy ions, NF-κB predominantly upregulates genes involved in intercellular communication processes. This process is strictly NF-κB dependent as the response is completely absent in RelA knock-down cells. NF-κB’s role in the cellular radiation response depends on the radiation quality.
Collapse
|
17
|
Ubale RV, Shastri PN, Oettinger C, D’Souza MJ. Pulmonary Administration of Microparticulate Antisense Oligonucleotide (ASO) for the Treatment of Lung Inflammation. AAPS PharmSciTech 2018; 19:1908-1919. [PMID: 29663290 DOI: 10.1208/s12249-018-1002-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 03/19/2018] [Indexed: 01/01/2023] Open
Abstract
Targeted delivery to the lung for controlling lung inflammation is an area that we have explored in this study. The purpose was to use microparticles containing an antisense oligonucleotide (ASO) to NF-κB to inhibit the production of proinflammatory cytokines. Microparticles were prepared using the B-290 Buchi Spray Dryer using albumin as the microparticle matrix. Physicochemical characterization of the microparticles showed the size ranged from 2 to 5 μm, the charge was - 38.4 mV, and they had a sustained release profile over 72 h. Uptake of FITC-labeled ASO-loaded microparticles versus FITC-labeled ASO solution by RAW264.7 murine macrophage cells was 5-10-fold higher. After pulmonary delivery of microparticles to Sprague-Dawley rats, the microparticles were uniformly distributed throughout the lung and were retained in the lungs until 48 h. Serum cytokine (TNF-α and IL-1β) levels of rats after induction of lung inflammation by lipopolysaccharide were measured until 72 h. Animals receiving ASO-loaded microparticles were successful in significantly controlling lung inflammation during this period as compared to animals receiving no treatment. This study was successful in proving that microparticulate ASO therapy was capable of controlling lung inflammation.
Collapse
|
18
|
Parmar G, Pundarikakshudu K, Balaraman R. Anti-anaphylactic and antiasthmatic activity of Euphorbia thymifolia L. on experimental animals. J Tradit Complement Med 2018; 9:60-65. [PMID: 30671367 PMCID: PMC6335472 DOI: 10.1016/j.jtcme.2018.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 02/28/2018] [Accepted: 03/06/2018] [Indexed: 11/10/2022] Open
Abstract
In Ayurveda, Euphorbia thymifolia L. (Euphorbiaceae) prescribed in the treatment of various ailments like bronchial asthma, cough, diarrhea and bleeding piles. The present study was investigated to evaluate antianaphylactic, mast cell stabilizing and antiasthmatic activity of methanol and aqueous extract of E. thymifolia (ET) on experimental animals. Anaphylaxis was induced by administration of horse serum and triple antigen vaccine intraperitoneal (i.p.) in albino Wistar rats. Extracts of ET were administered to the rats in dose of 250 and 500 mg/kg orally for 14 days. At the end of treatment, asthma score was measured and various blood parameters like differential count (DC), total WBC count and IgE were estimated. Interleukin (IL)-4, IL-5 and TNF-α were measured by ELISA commercial kit from BALF. Histopathological changes of lungs were observed. Antiasthmatic activity of extracts of ET was also studied on histamine-induced bronchospasm in guinea pigs. In vitro mast cell stabilizing activity of extracts was evaluated on compound 48/80 challenged rat intestinal mesenteric mast cells. The treatment with extracts of ET produced significant decrease in asthma score and they also brought to normalcy the increased total WBC, DC counts, serum IgE, TNF-α, IL-4 and IL-5 in BALF. The histopathological study further supported the protective effect of ET extracts. The pretreatment with extracts of ET displayed significant reduction in degranulation of mesenteric mast cell numbers. The treatment with extracts of ET significantly increased in time of PCD. Thus, these findings concluded that E. thymifolia could be effectively used in the treatment of anaphylaxis and asthma.
Collapse
Affiliation(s)
- Ghanshyam Parmar
- Department of Pharmacy, Sumandeep Vidyapeeth, At post: Piparia, Taluka: Waghodia, Dist: Vadodara, 391760, Gujarat, India
| | - Kilambi Pundarikakshudu
- Department of Pharmacognosy, L. J. Institute of Pharmacy, Between Sarkhej Circle and Kataria Motors, S. G. Road, Ahmedabad, 382210, India
| | - R Balaraman
- Department of Pharmacy, Sumandeep Vidyapeeth, At post: Piparia, Taluka: Waghodia, Dist: Vadodara, 391760, Gujarat, India
| |
Collapse
|
19
|
Cantone M, Santos G, Wentker P, Lai X, Vera J. Multiplicity of Mathematical Modeling Strategies to Search for Molecular and Cellular Insights into Bacteria Lung Infection. Front Physiol 2017; 8:645. [PMID: 28912729 PMCID: PMC5582318 DOI: 10.3389/fphys.2017.00645] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/16/2017] [Indexed: 12/13/2022] Open
Abstract
Even today two bacterial lung infections, namely pneumonia and tuberculosis, are among the 10 most frequent causes of death worldwide. These infections still lack effective treatments in many developing countries and in immunocompromised populations like infants, elderly people and transplanted patients. The interaction between bacteria and the host is a complex system of interlinked intercellular and the intracellular processes, enriched in regulatory structures like positive and negative feedback loops. Severe pathological condition can emerge when the immune system of the host fails to neutralize the infection. This failure can result in systemic spreading of pathogens or overwhelming immune response followed by a systemic inflammatory response. Mathematical modeling is a promising tool to dissect the complexity underlying pathogenesis of bacterial lung infection at the molecular, cellular and tissue levels, and also at the interfaces among levels. In this article, we introduce mathematical and computational modeling frameworks that can be used for investigating molecular and cellular mechanisms underlying bacterial lung infection. Then, we compile and discuss published results on the modeling of regulatory pathways and cell populations relevant for lung infection and inflammation. Finally, we discuss how to make use of this multiplicity of modeling approaches to open new avenues in the search of the molecular and cellular mechanisms underlying bacterial infection in the lung.
Collapse
Affiliation(s)
| | | | | | | | - Julio Vera
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum ErlangenErlangen, Germany
| |
Collapse
|
20
|
Ono Y, Sakamoto K. Lipopolysaccharide inhibits myogenic differentiation of C2C12 myoblasts through the Toll-like receptor 4-nuclear factor-κB signaling pathway and myoblast-derived tumor necrosis factor-α. PLoS One 2017; 12:e0182040. [PMID: 28742154 PMCID: PMC5524356 DOI: 10.1371/journal.pone.0182040] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 07/11/2017] [Indexed: 12/21/2022] Open
Abstract
Background Circulating lipopolysaccharide (LPS) concentrations are often elevated in patients with sepsis or with various endogenous diseases that are associated with metabolic endotoxemia. Involuntary loss of skeletal muscle, termed muscle wasting, is commonly observed in these conditions, suggesting that circulating LPS might play an essential role in its development. Although impairment of muscle regeneration is an important determinant of skeletal muscle wasting, it is unclear whether LPS affects this process and, if so, by what mechanism. Here, we used the C2C12 myoblast cell line to investigate the effects of LPS on myogenesis. Methods C2C12 myoblasts were grown to 80% confluence and induced to differentiate in the absence or presence of LPS (0.1 or 1 μg/mL); TAK-242 (1 μM), a specific inhibitor of Toll-like receptor 4 (TLR4) signaling; and a tumor necrosis factor (TNF)-α neutralizing antibody (5 μg/mL). Expression of a skeletal muscle differentiation marker (myosin heavy chain II), two essential myogenic regulatory factors (myogenin and MyoD), and a muscle negative regulatory factor (myostatin) was analyzed by western blotting. Nuclear factor-κB (NF-κB) DNA-binding activity was measured using an enzyme-linked immunosorbent assay. Results LPS dose-dependently and significantly decreased the formation of multinucleated myotubes and the expression of myosin heavy chain II, myogenin, and MyoD, and increased NF-κB DNA-binding activity and myostatin expression. The inhibitory effect of LPS on myogenic differentiation was reversible, suggesting that it was not caused by nonspecific toxicity. Both TAK-242 and anti-TNF-α reduced the LPS-induced increase in NF-κB DNA-binding activity, downregulation of myogenic regulatory factors, and upregulation of myostatin, thereby partially rescuing the impairment of myogenesis. Conclusions Our data suggest that LPS inhibits myogenic differentiation via a TLR4–NF-κB-dependent pathway and an autocrine/paracrine TNF-α-induced pathway. These pathways may be involved in the development of muscle wasting caused by sepsis or metabolic endotoxemia.
Collapse
Affiliation(s)
- Yuko Ono
- Department of Pharmacology, School of Medicine, Fukushima Medical University, Fukushima, Japan
- Emergency and Critical Care Medical Center, Fukushima Medical University Hospital, Fukushima, Japan
| | - Kazuho Sakamoto
- Department of Pharmacology, School of Medicine, Fukushima Medical University, Fukushima, Japan
- * E-mail:
| |
Collapse
|
21
|
Müller-Calleja N, Manukyan D, Canisius A, Strand D, Lackner KJ. Hydroxychloroquine inhibits proinflammatory signalling pathways by targeting endosomal NADPH oxidase. Ann Rheum Dis 2017; 76:891-897. [PMID: 27903507 DOI: 10.1136/annrheumdis-2016-210012] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/28/2016] [Accepted: 11/05/2016] [Indexed: 01/18/2023]
Abstract
OBJECTIVES Hydroxychloroquine (HCQ) has been used for decades to treat patients with rheumatic diseases, for example, systemic lupus erythematosus (SLE), rheumatoid arthritis or the antiphospholipid syndrome (APS). We hypothesise that HCQ might target endosomal NADPH oxidase (NOX), which is involved in the signal transduction of cytokines as well as antiphospholipid antibodies (aPL). METHODS For in vitro experiments, monocytic cells were stimulated with tumour necrosis factor α (TNFα), interleukin-1β (IL-1β) or a human monoclonal aPL and the activity of NOX was determined by flow cytometry. The expression of genes known to be induced by these stimuli was quantified by quantitative reverse transcription PCR. Live cell imaging was performed by confocal laser scanning microscopy. Finally, the effects of HCQ on NOX-induced signal transduction were analysed in an in vivo model of venous thrombosis. RESULTS HCQ strongly reduces or completely prevents the induction of endosomal NOX by TNFα, IL-1β and aPL in human monocytes and MonoMac1 cells. As a consequence, induction of downstream genes by these stimuli is reduced or abrogated. This effect of HCQ is not mediated by direct interference with the agonists but by inhibiting the translocation of the catalytic subunit of NOX2 (gp91phox) into the endosome. In vivo, HCQ protects mice from aPL-induced and NOX2-mediated thrombus formation. CONCLUSIONS We describe here a novel mechanism of action of HCQ, that is, interference with the assembly of endosomal NOX2. Since endosomal NOX2 is involved in many inflammatory and prothrombotic signalling pathways, this activity of HCQ might explain many of its beneficial effects in rheumatic diseases including the APS.
Collapse
Affiliation(s)
- Nadine Müller-Calleja
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Davit Manukyan
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Antje Canisius
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Dennis Strand
- Department of Medicine 1, University Medical Center Mainz, Mainz, Germany
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
22
|
Kim A, Nam YJ, Shin YK, Lee MS, Sohn DS, Lee CS. Rotundarpene inhibits TNF-α-induced activation of the Akt, mTOR, and NF-κB pathways, and the JNK and p38 associated with production of reactive oxygen species. Mol Cell Biochem 2017; 434:113-125. [PMID: 28432555 DOI: 10.1007/s11010-017-3041-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/17/2017] [Indexed: 11/26/2022]
Abstract
Ilex Rotunda Thunb has been shown to have anti-inflammatory and antioxidant effects. In human keratinocytes, we investigated the effect of rotundarpene (4-caffeoyl-3-methyl-but-2-ene-1,4-diol) on the TNF-α-stimulated production of inflammatory mediators in relation to the Akt, mTOR, and NF-κB pathways, and the JNK and p38-MAPK. Rotundarpene, Akt inhibitor, Bay 11-7085, rapamycin, and N-acetylcysteine inhibited the TNF-α-stimulated production of cytokines and chemokines, increase in the levels of p-Akt and mTOR, activation of NF-κB, and production of reactive oxygen species in keratinocytes. TNF-α treatment induced phosphorylation of the JNK and p38-MAPK. Inhibitors of the c-JNK (SP600125) and p38-MAPK (SB203580) reduced the TNF-α-induced production of inflammatory mediators, binding of NF-κB to DNA, and activation of the JNK and p38-MAPK in keratinocytes. The results show that rotundarpene may reduce the TNF-α-stimulated inflammatory mediator production by suppressing the reactive oxygen species-dependent activation of the Akt, mTOR, and NF-κB pathways, and activation of the JNK and p38-MAPK in human keratinocytes. Additionally, rotundarpene appears to attenuate the Akt, mTOR, and NF-κB pathways and the JNK and p38-MAPK-mediated inflammatory skin diseases.
Collapse
Affiliation(s)
- Arum Kim
- Department of Pharmacology, College of Medicine, and The BK21plus Skin Barrier Network Human Resources Development Team, Chung-Ang University, Seoul, 156-756, South Korea
| | - Yoon Jeong Nam
- Department of Pharmacology, College of Medicine, and The BK21plus Skin Barrier Network Human Resources Development Team, Chung-Ang University, Seoul, 156-756, South Korea
| | - Yong Kyoo Shin
- Department of Pharmacology, College of Medicine, and The BK21plus Skin Barrier Network Human Resources Development Team, Chung-Ang University, Seoul, 156-756, South Korea
| | - Min Sung Lee
- Department of Internal Medicine, SoonChunHyang University Hospital, Bucheon, Kyung-Gi-Do, 420-767, South Korea
| | - Dong Suep Sohn
- Department of Thoracic and Cardiovascular Surgery, Chung-Ang University Hospital, Seoul, 156-755, South Korea
| | - Chung Soo Lee
- Department of Pharmacology, College of Medicine, and The BK21plus Skin Barrier Network Human Resources Development Team, Chung-Ang University, Seoul, 156-756, South Korea.
| |
Collapse
|
23
|
Yu H, Aravindan N, Xu J, Natarajan M. Inter- and intra-cellular mechanism of NF-kB-dependent survival advantage and clonal expansion of radio-resistant cancer cells. Cell Signal 2017; 31:105-111. [DOI: 10.1016/j.cellsig.2017.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/21/2016] [Accepted: 01/04/2017] [Indexed: 11/29/2022]
|
24
|
Abstract
Mast cells (MCs) play a central role in tissue homoeostasis, sensing the local environment through numerous innate cell surface receptors. This enables them to respond rapidly to perceived tissue insults with a view to initiating a co-ordinated programme of inflammation and repair. However, when the tissue insult is chronic, the ongoing release of multiple pro-inflammatory mediators, proteases, cytokines and chemokines leads to tissue damage and remodelling. In asthma, there is strong evidence of ongoing MC activation, and their mediators and cell-cell signals are capable of regulating many facets of asthma pathophysiology. This article reviews the evidence behind this.
Collapse
Affiliation(s)
- P Bradding
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK
| | - G Arthur
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK
| |
Collapse
|
25
|
Sato K, Mihara Y, Kanai K, Yamashita Y, Kimura Y, Itoh N. Tyrosol ameliorates lipopolysaccharide-induced ocular inflammation in rats via inhibition of nuclear factor (NF)-κB activation. J Vet Med Sci 2016; 78:1429-1438. [PMID: 27238160 PMCID: PMC5059370 DOI: 10.1292/jvms.16-0166] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
We evaluated the anti-inflammatory effect of tyrosol (Tyr) on endotoxin-induced uveitis (EIU) in rats. EIU was induced in male Lewis rats by subcutaneous injection of lipopolysaccharide (LPS). Tyr (10, 50 or 100 mg/kg) was intravenously injected 2 hr before, simultaneously and 2 hr after LPS injection. The aqueous humor (AqH) was collected 24 hr after LPS injection; the infiltrating cell number, protein concentration, and tumor necrosis factor (TNF)-α, prostaglandin (PG)-E2 and nitric oxide (NO) levels were determined. Histopathologic examination and immunohistochemical studies for nuclear factor (NF)-κB, inhibitor of κB (IκB)-α, cyclooxygenase (COX)-2 and inducible NO synthase (iNOS) in the iris-ciliary body (ICB) were performed at 3 or 24 hr after LPS injection. To further clarify the anti-inflammatory effects, RAW264.7 macrophages were stimulated with LPS in the presence or absence of Tyr. Tyr reduced, in a dose-dependent manner, the infiltrating cell number, protein concentration, and TNF-α, PGE2 and NO levels in AqH and improved histopathologic scores of EIU. Tyr also inhibited LPS-induced COX-2 and iNOS expression, IκB-α degradation and nuclear translocation of activated NF-κB in ICB. Tyr significantly suppressed inflammatory mediator production in the culture medium and COX-2 and iNOS expression and activated NF-κB translocation in LPS-stimulated RAW264.7 cells. These results suggest that Tyr suppresses ocular inflammation of EIU by inhibiting NF-κB activation and subsequent proinflammatory mediator production.
Collapse
Affiliation(s)
- Kazuaki Sato
- Department of Small Animal Internal Medicine I, School of Veterinary Medicine, University of Kitasato, 35-1, Towada, Aomori 034-8628, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Kim YH, Choi YR, Kim JY, Kwak SH. Anti-Allergic Effect of 1,2,3,4,6-Penta-O-Galloyl-β-D-Glucose on RBL-2H3 Cells. ACTA ACUST UNITED AC 2016. [DOI: 10.3746/jkfn.2016.45.4.613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
27
|
The anti-inflammatory and pro-resolution effects of aspirin-triggered RvD1 (AT-RvD1) on peripheral blood mononuclear cells from patients with severe asthma. Int Immunopharmacol 2016; 35:142-148. [PMID: 27044027 DOI: 10.1016/j.intimp.2016.03.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 03/03/2016] [Accepted: 03/11/2016] [Indexed: 12/19/2022]
Abstract
Asthma is an inflammatory disease that is characterized by a predominance of eosinophils and/or neutrophils in the airways. In the resolution of inflammation, lipid mediators such as resolvin D1 (RvD1) and its epimer aspirin-triggered RvD1 (AT-RvD1) are produced and demonstrate anti-inflammatory and pro-resolution effects. In experimental models such as airway allergic inflammation induced by ovalbumin in mice, RvD1 and AT-RvD1 alleviate some of the most important phenotypes of asthma. Here, we demonstrated the effects of AT-RvD1 on peripheral blood mononuclear cells (PBMCs) from healthy individuals and patients with severe asthma stimulated with lipopolysaccharide (LPS) or Dermatophagoides pteronyssinus (DM). AT-RvD1 (100nM) reduced the concentration of TNF-α in PBMCs from healthy individuals and patients with severe asthma stimulated with LPS or DM. In addition, AT-RvD1 lowered the production of IL-10 only in PBMCs from patients with severe asthma stimulated with LPS. These effects were associated in part with decreasing NF-κB activation. Moreover, AT-RvD1 significantly increased phagocytosis of apoptotic neutrophils by monocytes from patients with severe asthma. In conclusion, AT-RvD1 demonstrated both anti-inflammatory and pro-resolution effects in PBMCs from patients with severe asthma and could become in the future an alternative treatment for asthma.
Collapse
|
28
|
Bugajev V, Halova I, Draberova L, Bambouskova M, Potuckova L, Draberova H, Paulenda T, Junyent S, Draber P. Negative regulatory roles of ORMDL3 in the FcεRI-triggered expression of proinflammatory mediators and chemotactic response in murine mast cells. Cell Mol Life Sci 2016; 73:1265-85. [PMID: 26407610 PMCID: PMC11108389 DOI: 10.1007/s00018-015-2047-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 08/19/2015] [Accepted: 09/17/2015] [Indexed: 10/23/2022]
Abstract
Single-nucleotide polymorphism studies have linked the chromosome 17q12-q21 region, where the human orosomucoid-like (ORMDL)3 gene is localized, to the risk of asthma and several other inflammatory diseases. Although mast cells are involved in the development of these diseases, the contribution of ORMDL3 to the mast cell physiology is unknown. In this study, we examined the role of ORMDL3 in antigen-induced activation of murine mast cells with reduced or enhanced ORMDL3 expression. Our data show that in antigen-activated mast cells, reduced expression of the ORMDL3 protein had no effect on degranulation and calcium response, but significantly enhanced phosphorylation of AKT kinase at Ser 473 followed by enhanced phosphorylation and degradation of IκBα and translocation of the NF-κB p65 subunit into the nucleus. These events were associated with an increased expression of proinflammatory cytokines (TNF-α, IL-6, and IL-13), chemokines (CCL3 and CCL4), and cyclooxygenase-2 dependent synthesis of prostaglandin D2. Antigen-mediated chemotaxis was also enhanced in ORMDL3-deficient cells, whereas spreading on fibronectin was decreased. On the other hand, increased expression of ORMDL3 had no significant effect on the studied signaling events, except for reduced antigen-mediated chemotaxis. These data were corroborated by increased IgE-antigen-dependent passive cutaneous anaphylaxis in mice with locally silenced ORMDL3 using short interfering RNAs. Our data also show that antigen triggers suppression of ORMDL3 expression in the mast cells. In summary, we provide evidence that downregulation of ORMDL3 expression in mast cells enhances AKT and NF-κB-directed signaling pathways and chemotaxis and contributes to the development of mast cell-mediated local inflammation in vivo.
Collapse
Affiliation(s)
- Viktor Bugajev
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Ivana Halova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Lubica Draberova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Monika Bambouskova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Lucie Potuckova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Helena Draberova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Tomas Paulenda
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Sergi Junyent
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Petr Draber
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic.
| |
Collapse
|
29
|
Solano-Aguilar G, Molokin A, Botelho C, Fiorino AM, Vinyard B, Li R, Chen C, Urban J, Dawson H, Andreyeva I, Haverkamp M, Hibberd PL. Transcriptomic Profile of Whole Blood Cells from Elderly Subjects Fed Probiotic Bacteria Lactobacillus rhamnosus GG ATCC 53103 (LGG) in a Phase I Open Label Study. PLoS One 2016; 11:e0147426. [PMID: 26859761 PMCID: PMC4747532 DOI: 10.1371/journal.pone.0147426] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 12/31/2015] [Indexed: 02/07/2023] Open
Abstract
We examined gene expression of whole blood cells (WBC) from 11 healthy elderly volunteers participating on a Phase I open label study before and after oral treatment with Lactobacillus rhamnosus GG-ATCC 53103 (LGG)) using RNA-sequencing (RNA-Seq). Elderly patients (65–80 yrs) completed a clinical assessment for health status and had blood drawn for cellular RNA extraction at study admission (Baseline), after 28 days of daily LGG treatment (Day 28) and at the end of the study (Day 56) after LGG treatment had been suspended for 28 days. Treatment compliance was verified by measuring LGG-DNA copy levels detected in host fecal samples. Normalized gene expression levels in WBC RNA were analyzed using a paired design built within three analysis platforms (edgeR, DESeq2 and TSPM) commonly used for gene count data analysis. From the 25,990 transcripts detected, 95 differentially expressed genes (DEGs) were detected in common by all analysis platforms with a nominal significant difference in gene expression at Day 28 following LGG treatment (FDR<0.1; 77 decreased and 18 increased). With a more stringent significance threshold (FDR<0.05), only two genes (FCER2 and LY86), were down-regulated more than 1.5 fold and met the criteria for differential expression across two analysis platforms. The remaining 93 genes were only detected at this threshold level with DESeq2 platform. Data analysis for biological interpretation of DEGs with an absolute fold change of 1.5 revealed down-regulation of overlapping genes involved with Cellular movement, Cell to cell signaling interactions, Immune cell trafficking and Inflammatory response. These data provide evidence for LGG-induced transcriptional modulation in healthy elderly volunteers because pre-treatment transcription levels were restored at 28 days after LGG treatment was stopped. To gain insight into the signaling pathways affected in response to LGG treatment, DEG were mapped using biological pathways and genomic data mining packages to indicate significant biological relevance. Trial Registration: ClinicalTrials.gov NCT01274598
Collapse
Affiliation(s)
- Gloria Solano-Aguilar
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
- * E-mail:
| | - Aleksey Molokin
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Christine Botelho
- Division of Global Health, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Anne-Maria Fiorino
- Division of Global Health, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Bryan Vinyard
- Statistics Group, Northeast Area, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Robert Li
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Celine Chen
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Joseph Urban
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Harry Dawson
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Irina Andreyeva
- Division of Global Health, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Miriam Haverkamp
- Division of Global Health, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Patricia L. Hibberd
- Division of Global Health, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| |
Collapse
|
30
|
Kumase F, Takeuchi K, Morizane Y, Suzuki J, Matsumoto H, Kataoka K, Al-Moujahed A, Maidana DE, Miller JW, Vavvas DG. AMPK-Activated Protein Kinase Suppresses Ccr2 Expression by Inhibiting the NF-κB Pathway in RAW264.7 Macrophages. PLoS One 2016; 11:e0147279. [PMID: 26799633 PMCID: PMC4723067 DOI: 10.1371/journal.pone.0147279] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 01/02/2016] [Indexed: 01/08/2023] Open
Abstract
C-C chemokine receptor 2 (Ccr2) is a key pro-inflammatory marker of classic (M1) macrophage activation. Although Ccr2 is known to be expressed both constitutively and inductively, the full regulatory mechanism of its expression remains unclear. AMP-activated protein kinase (AMPK) is not only a master regulator of energy homeostasis but also a central regulator of inflammation. In this study, we sought to assess AMPK's role in regulating RAW264.7 macrophage Ccr2 protein levels in resting (M0) or LPS-induced M1 states. In both M0 and M1 RAW264.7 macrophages, knockdown of the AMPKα1 subunit by siRNA led to increased Ccr2 levels whereas pharmacologic (A769662) activation of AMPK, attenuated LPS-induced increases in Ccr2 expression in an AMPK dependent fashion. The increases in Ccr2 levels by AMPK downregulation were partially reversed by NF-κB inhibition whereas TNF-a inhibition had minimal effects. Our results indicate that AMPK is a negative regulator of Ccr2 expression in RAW264.7 macrophages, and that the mechanism of action of AMPK inhibition of Ccr2 is mediated, in part, through the NF-κB pathway.
Collapse
Affiliation(s)
- Fumiaki Kumase
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kimio Takeuchi
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yuki Morizane
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Ophthalmology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jun Suzuki
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Ophthalmology, Tokyo Medical University, Tokyo, Japan
| | - Hidetaka Matsumoto
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Keiko Kataoka
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ahmad Al-Moujahed
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel E. Maidana
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joan W. Miller
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Demetrios G. Vavvas
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
31
|
Srivastava AK, Nagar H, Srivastava R, Ahirwar V, Chandel HS. Evaluation of antitussive and anti-asthmatic activity of Tabernaemontana divaricata(L.) R. Br. Ex Roem. and Schult. Ayu 2016; 37:256-263. [PMID: 29491680 PMCID: PMC5822985 DOI: 10.4103/ayu.ayu_35_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background: The study was aimed to investigate the antitussive and anti-asthmatic activities of ethanolic extract of Tabernaemontana divaricata (TDEE) leaves by in vivo and in vitro models. Recently, indole alkaloids (monoterpenoid indole alkaloids) have been approved as investigational new drug for clinical trial in respiratory diseases, and T. divaricata has already proven its potential for the presence of indole alkaloids. Materials and Methods: Acute toxicity studies of TDEE were performed in accordance with the Organization for Economic Cooperation and Development guidelines no. 425. The sensitized guinea pigs were screened out and divided into control, standard, and TDEE-treated groups. Anti-asthmatic activity of TDEE was assessed by in vitro guinea pig tracheal chain method and in vivo bronchoprotective test method using aminophylline as a standard drug. Taken codeine as standard, antitussive activity was evaluated by in vivo citric acid-induced tussive response. Results: TDEE was found to be safe up to 2000 mg/kg, body weight. TDEE exhibits maximum bronchi relaxation of 91.66% and 92.83% against acetylcholine and histamine-induced contraction, respectively. TDEE exhibited maximum and significant (P < 0.001) bronchoprotection of 42.28% at the dose level of 200 mg/kg, body weight. TDEE at aerosolic dose of 6% (w/v) exhibited decreased average cough frequency (4.83 ± 0.30) which is quite significant (P < 0.001) and effective as compared to standard drug codeine. Based on the histopathological evidences, TDEE-treated groups showed reduced inflammatory cell infiltration and had restored epithelial damage. Conclusion: The results of the study revealed the potent antitussive and anti-asthmatic activities of T. divaricata, which support its further implication for the treatment of cough-associated complications such as cough variant asthma.
Collapse
Affiliation(s)
- Amit Kumar Srivastava
- Department of Pharmacology, Truba Institute of Pharmacy, Bhopal, Madhya Pradesh, India
| | - Hemant Nagar
- Department of Pharmacology, Truba Institute of Pharmacy, Bhopal, Madhya Pradesh, India
| | - Rajnish Srivastava
- Department of Pharmacology, Moradabad Educational Trust Group of Institutions, Faculty of Pharmacy, Moradabad, Uttar Pradesh, India
| | - Varsha Ahirwar
- Department of Pharmacology, Truba Institute of Pharmacy, Bhopal, Madhya Pradesh, India
| | | |
Collapse
|
32
|
Kim OK, Nam DE, Jun W, Lee J. Anti-Inflammatory and Gastroprotective Activities of C
udrania Tricuspidata
Leaf Extract Against Acute HCl/Ethanol-Induced Gastric Mucosal Injury in Sprague-Dawley Rats. J Food Biochem 2015. [DOI: 10.1111/jfbc.12149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ok-Kyung Kim
- Department of Medical Nutrition; Kyung Hee University; Yongin 446-701 Korea
| | - Da-Eun Nam
- Department of Medical Nutrition; Kyung Hee University; Yongin 446-701 Korea
| | - Woojin Jun
- Department of Food and Nutrition; Chonnam National University; Gwangju Korea
| | - Jeongmin Lee
- Department of Medical Nutrition; Kyung Hee University; Yongin 446-701 Korea
- Research Institute of Clinical Nutrition; Kyung Hee University; Seoul 130-701 Korea
| |
Collapse
|
33
|
He YQ, Zhang WT, Shi CH, Wang FM, Tian XJ, Ma LL. Phloroglucinol protects the urinary bladder via inhibition of oxidative stress and inflammation in a rat model of cyclophosphamide-induced interstitial cystitis. Chin Med J (Engl) 2015; 128:956-62. [PMID: 25836618 PMCID: PMC4834014 DOI: 10.4103/0366-6999.154316] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background: Phloroglucinol plays an important role in oxidative stress and inflammatory responses. The effects of phloroglucinol have been proven in various disease models. The aim of the present study was to investigate the efficacy and possible mechanisms of phloroglucinol in the treatment of interstitial cystitis (IC). Methods: Thirty-two female Sprague-Dawley (SD) rats were used in this study. IC was induced by intraperitoneal injection of cyclophosphamide (CYP). Rats were randomly allocated to one of four groups (n = 8 per group): A control group, which was injected with saline (75 mg/kg; i.p.) instead of CYP on days 1, 4, and 7; a chronic IC group, which was injected with CYP (75 mg/kg; i.p.) on days 1, 4, and 7; a high-dose (30 mg/kg) phloroglucinol-treated group; and a low-dose (15 mg/kg) phloroglucinol-treated group. On day 8, the rats in each group underwent cystometrography (CMG), and the bladders were examined for evidence of oxidative stress and inflammation. Statistical analysis was performed by analysis of variance (ANOVA) followed by least square difference multiple comparison post-hoc test. Results: Histological evaluation showed that bladder inflammation in CYP-treated rats was suppressed by phloroglucinol. CMG revealed that the CYP treatment induced overactive bladder in rats that was reversed by phloroglucinol. Up-regulated tumor necrosis factor-α and interleukin-6 expression in the CYP-treated rats were also suppressed in the phloroglucinol treated rats. CYP treatment significantly increased myeloperoxidase activity as well as the decreased activities of catalase of the bladder, which was reversed by treatment with phloroglucinol. Conclusions: The application of phloroglucinol suppressed oxidative stress, inflammation, and overactivity in the bladder. This may provide a new treatment strategy for IC.
Collapse
Affiliation(s)
| | | | | | | | | | - Lu-Lin Ma
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
34
|
Yang Y, Gorzelanny C, Bauer AT, Halter N, Komljenovic D, Bäuerle T, Borsig L, Roblek M, Schneider SW. Nuclear heparanase-1 activity suppresses melanoma progression via its DNA-binding affinity. Oncogene 2015; 34:5832-42. [PMID: 25745999 DOI: 10.1038/onc.2015.40] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 01/09/2015] [Accepted: 01/14/2015] [Indexed: 12/13/2022]
Abstract
Heparanase-1 (HPSE) plays a pivotal role in structural remodeling of the ECM and the glycocalyx, thus conferring protumorigenic, proangiogenic and prometastatic properties to many cancer entities. In addition to its extracellular function, recent studies suggest an intracellular activity of HPSE with a largely unknown significance during tumor progression. Therefore, we investigated the relevance of the dual functions of HPSE to malignant melanoma in vitro, as well as in different mouse melanoma models based on the intradermal or intravenous injection of melanoma cells. Consistent with its extracellular action, an HPSE deficiency led to a reduced shedding of the glycocalyx accompanied by a reduced availability of vascular endothelial growth factor, affecting tumor growth and vascularization. In contrast, we measured an elevated expression of the protumorigenic factors pentraxin-3, tissue factor, TNF-α and most prominently, MMP-9, upon HPSE knockdown. In vivo, an HPSE deficiency was related to increased lymph node metastasis. Since the inhibition of its extracellular function with heparin was unable to block the gene regulatory impact of HPSE, we proposed an intracellular mechanism. Immunostaining revealed a counter-staining of HPSE and NF-κB in the nucleus, suggesting a close relationship between both proteins. This finding was further supported by the discovery of a direct charge-driven molecular interaction between HPSE and DNA by using atomic force microscopy and a co-precipitation approach. Our findings are novel and point towards a dual function for HPSE in malignant melanoma with a protumorigenic extracellular activity and a tumor-suppressive nuclear action. The identification of molecular strategies to shuttle extracellular HPSE into the nuclei of cancer cells could provide new therapeutic options.
Collapse
Affiliation(s)
- Y Yang
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - C Gorzelanny
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - A T Bauer
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - N Halter
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - D Komljenovic
- Division of Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - T Bäuerle
- Institute of Radiology, University Hospital Erlangen, Erlangen, Germany
| | - L Borsig
- Institute of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, Zürich, Switzerland
| | - M Roblek
- Institute of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, Zürich, Switzerland
| | - S W Schneider
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
35
|
Caldwell AB, Cheng Z, Vargas JD, Birnbaum HA, Hoffmann A. Network dynamics determine the autocrine and paracrine signaling functions of TNF. Genes Dev 2014; 28:2120-33. [PMID: 25274725 PMCID: PMC4180974 DOI: 10.1101/gad.244749.114] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A hallmark of the inflammatory response to pathogen exposure is the production of tumor necrosis factor (TNF) that coordinates innate and adaptive immune responses by functioning in an autocrine or paracrine manner. Numerous molecular mechanisms contributing to TNF production have been identified, but how they function together in macrophages remains unclear. Here, we pursued an iterative systems biology approach to develop a quantitative understanding of the regulatory modules that control TNF mRNA synthesis and processing, mRNA half-life and translation, and protein processing and secretion. By linking the resulting model of TNF production to models of the TLR-, the TNFR-, and the NFκB signaling modules, we were able to study TNF's functions during the inflammatory response to diverse TLR agonists. Contrary to expectation, we predicted and then experimentally confirmed that in response to lipopolysaccaride, TNF does not have an autocrine function in amplifying the NFκB response, although it plays a potent paracrine role in neighboring cells. However, in response to CpG DNA, autocrine TNF extends the duration of NFκB activity and shapes CpG-induced gene expression programs. Our systems biology approach revealed that network dynamics of MyD88 and TRIF signaling and of cytokine production and response govern the stimulus-specific autocrine and paracrine functions of TNF.
Collapse
Affiliation(s)
- Andrew B Caldwell
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, and San Diego Center for Systems Biology, University of California at San Diego, La Jolla, California 92093, USA
| | - Zhang Cheng
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, and San Diego Center for Systems Biology, University of California at San Diego, La Jolla, California 92093, USA; Institute for Quantitative and Computational Biosciences, Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California 90025, USA
| | - Jesse D Vargas
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, and San Diego Center for Systems Biology, University of California at San Diego, La Jolla, California 92093, USA; Institute for Quantitative and Computational Biosciences, Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California 90025, USA
| | - Harry A Birnbaum
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, and San Diego Center for Systems Biology, University of California at San Diego, La Jolla, California 92093, USA; Institute for Quantitative and Computational Biosciences, Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California 90025, USA
| | - Alexander Hoffmann
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, and San Diego Center for Systems Biology, University of California at San Diego, La Jolla, California 92093, USA; Institute for Quantitative and Computational Biosciences, Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California 90025, USA
| |
Collapse
|
36
|
Sasi SP, Song J, Park D, Enderling H, McDonald JT, Gee H, Garrity B, Shtifman A, Yan X, Walsh K, Natarajan M, Kishore R, Goukassian DA. TNF-TNFR2/p75 signaling inhibits early and increases delayed nontargeted effects in bone marrow-derived endothelial progenitor cells. J Biol Chem 2014; 289:14178-93. [PMID: 24711449 DOI: 10.1074/jbc.m114.567743] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
TNF-α, a pro-inflammatory cytokine, is highly expressed after being irradiated (IR) and is implicated in mediating radiobiological bystander responses (RBRs). Little is known about specific TNF receptors in regulating TNF-induced RBR in bone marrow-derived endothelial progenitor cells (BM-EPCs). Full body γ-IR WT BM-EPCs showed a biphasic response: slow decay of p-H2AX foci during the initial 24 h and increase between 24 h and 7 days post-IR, indicating a significant RBR in BM-EPCs in vivo. Individual TNF receptor (TNFR) signaling in RBR was evaluated in BM-EPCs from WT, TNFR1/p55KO, and TNFR2/p75KO mice, in vitro. Compared with WT, early RBR (1-5 h) were inhibited in p55KO and p75KO EPCs, whereas delayed RBR (3-5 days) were amplified in p55KO EPCs, suggesting a possible role for TNFR2/p75 signaling in delayed RBR. Neutralizing TNF in γ-IR conditioned media (CM) of WT and p55KO BM-EPCs largely abolished RBR in both cell types. ELISA protein profiling of WT and p55KO EPC γ-IR-CM over 5 days showed significant increases in several pro-inflammatory cytokines, including TNF-α, IL-1α (Interleukin-1 alpha), RANTES (regulated on activation, normal T cell expressed and secreted), and MCP-1. In vitro treatments with murine recombinant (rm) TNF-α and rmIL-1α, but not rmMCP-1 or rmRANTES, increased the formation of p-H2AX foci in nonirradiated p55KO EPCs. We conclude that TNF-TNFR2 signaling may induce RBR in naïve BM-EPCs and that blocking TNF-TNFR2 signaling may prevent delayed RBR in BM-EPCs, conceivably, in bone marrow milieu in general.
Collapse
Affiliation(s)
- Sharath P Sasi
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135
| | - Jin Song
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135
| | - Daniel Park
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135
| | - Heiko Enderling
- the Center of Cancer Systems Biology, GeneSys Research Institute, Boston, Massachusetts 02135, Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - J Tyson McDonald
- the Center of Cancer Systems Biology, GeneSys Research Institute, Boston, Massachusetts 02135, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Hannah Gee
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135
| | - Brittany Garrity
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135
| | - Alexander Shtifman
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Xinhua Yan
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135, the Center of Cancer Systems Biology, GeneSys Research Institute, Boston, Massachusetts 02135, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Kenneth Walsh
- the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Mohan Natarajan
- the University of Texas Health Science Center, San Antonio, Texas 78229, and
| | - Raj Kishore
- the Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois 60611
| | - David A Goukassian
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135, Tufts University School of Medicine, Boston, Massachusetts 02111, the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts 02118,
| |
Collapse
|
37
|
Xiao S, Jiang J, Shim DW, Kim TK, Kang TB, Lee KH. Anti-allergic Effect of Ethanolic Extract of Flos Sophora japonica L. on Ca++Ionophore Stimulated Murine RBL-2H3 Cells. ACTA ACUST UNITED AC 2014. [DOI: 10.3746/jkfn.2014.43.3.349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
38
|
Pękalski J, Zuk PJ, Kochańczyk M, Junkin M, Kellogg R, Tay S, Lipniacki T. Spontaneous NF-κB activation by autocrine TNFα signaling: a computational analysis. PLoS One 2013; 8:e78887. [PMID: 24324544 PMCID: PMC3855823 DOI: 10.1371/journal.pone.0078887] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 09/16/2013] [Indexed: 11/18/2022] Open
Abstract
NF-κB is a key transcription factor that regulates innate immune response. Its activity is tightly controlled by numerous feedback loops, including two negative loops mediated by NF-κB inducible inhibitors, IκBα and A20, which assure oscillatory responses, and by positive feedback loops arising due to the paracrine and autocrine regulation via TNFα, IL-1 and other cytokines. We study the NF-κB system of interlinked negative and positive feedback loops, combining bifurcation analysis of the deterministic approximation with stochastic numerical modeling. Positive feedback assures the existence of limit cycle oscillations in unstimulated wild-type cells and introduces bistability in A20-deficient cells. We demonstrated that cells of significant autocrine potential, i.e., cells characterized by high secretion of TNFα and its receptor TNFR1, may exhibit sustained cytoplasmic-nuclear NF-κB oscillations which start spontaneously due to stochastic fluctuations. In A20-deficient cells even a small TNFα expression rate qualitatively influences system kinetics, leading to long-lasting NF-κB activation in response to a short-pulsed TNFα stimulation. As a consequence, cells with impaired A20 expression or increased TNFα secretion rate are expected to have elevated NF-κB activity even in the absence of stimulation. This may lead to chronic inflammation and promote cancer due to the persistent activation of antiapoptotic genes induced by NF-κB. There is growing evidence that A20 mutations correlate with several types of lymphomas and elevated TNFα secretion is characteristic of many cancers. Interestingly, A20 loss or dysfunction also leaves the organism vulnerable to septic shock and massive apoptosis triggered by the uncontrolled TNFα secretion, which at high levels overcomes the antiapoptotic action of NF-κB. It is thus tempting to speculate that some cancers of deregulated NF-κB signaling may be prone to the pathogen-induced apoptosis.
Collapse
Affiliation(s)
- Jakub Pękalski
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel J. Zuk
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
- Institute of Theoretical Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Marek Kochańczyk
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Michael Junkin
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Ryan Kellogg
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Savaş Tay
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Tomasz Lipniacki
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
- Department of Statistics, Rice University, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
39
|
Kim D, Kim JY. Anti-CD14 antibody reduces LPS responsiveness via TLR4 internalization in human monocytes. Mol Immunol 2013; 57:210-5. [PMID: 24172224 DOI: 10.1016/j.molimm.2013.09.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 09/07/2013] [Accepted: 09/28/2013] [Indexed: 10/26/2022]
Abstract
CD14 is an LPS sensing receptor that is primarily expressed in monocytes. CD14 binds and transfers bacterial LPS to the surface TLR4:MD-2 complex to enable its recognition. After recognizing LPS, this complex produces the first intracellular signals via TIRAP and MyD88, after which surface TLR4/LPS complex is rapidly internalized and produces additional signals via TRAM and TRIF. It was recently suggested that CD14 is a key regulator of LPS-induced TLR4 endocytosis and second signaling. In the present study, we showed that surface TLR4 expressions of human primary monocytes and cell line THP-1 were significantly reduced after treatment with anti-CD14 Ab. Among three anti-CD14 Abs with different epitope specificities used in this study, My4, which has an epitope specificity for LPS binding domain of the CD14 molecule, was found to be the most potent at reduction of surface expression of TLR4 as well as CD14. To test the reason for this reduction, we performed an in vitro internalization assay using anti-TLR4 Ab conjugated with toxin. The results of this analysis indicated surface CD14 ligation-mediated TLR4 internalization, and the mechanism of the internalization was found to be partially clathrin-dependent. We next examined NF-κB/AP-1 activation and TNF-α production of THP-1XBlue-CD14 cells in response to LPS challenge with or without My4 pre-treatment. The results revealed that NF-κB/AP-1 activation and TNF-α production of cells treated with My4 were significantly impaired when compared to the control. Our results suggest that membrane CD14 ligation-mediated TLR4 internalization is a novel mechanism for effective down-regulation of surface expression of TLR4 and subsequent reduction of LPS response of human monocytes.
Collapse
Affiliation(s)
- Donghee Kim
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | | |
Collapse
|
40
|
Golikova EA, Lopatnikova JA, Kovalevskaya-Kucheryavenko TV, Nepomnyashih VM, Sennikov SV. Levels of TNF, TNF autoantibodies and soluble TNF receptors in patients with bronchial asthma. J Asthma 2013; 50:705-11. [PMID: 23638975 DOI: 10.3109/02770903.2013.796972] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES The aim of the study was to evaluate the potential contribution made by tumor necrosis factor (TNF) autoantibodies to the pathogenesis of bronchial asthma (BA). METHODS We used affinity chromatography methods and a magnetic separation procedure to purify human autoantibodies specific to TNF. The autoantibodies were used as a calibration material to determine the absolute content of autoantibodies to TNF using enzyme-linked immunosorbent assay (ELISA). TNF content and levels of soluble receptors to TNF were determined using the ELISA commercial test kits. RESULTS We demonstrated significant increases in the levels of TNF and soluble TNF receptors in the sera of patients with uncontrolled and controlled BA, as compared with healthy donors. Levels of autoantibodies of the IgG2 and IgG4 subclasses were significantly higher in sera from patients with uncontrolled BA than in healthy donors. Levels of IgG2 autoantibodies were significantly higher in sera from patients with uncontrolled BA than in patients with controlled BA. CONCLUSIONS BA is associated with changes in the levels of not only TNF and soluble receptors for TNF, but also autoantibodies to TNF. Given the magnitude of the changes in the levels of different subclasses of autoantibodies to TNF, we propose that these autoantibodies might contribute to the pathogenesis of BA.
Collapse
Affiliation(s)
- Elena Alexeevna Golikova
- Laboratory of Molecular Immunology, Federal State Budgetary Institution Research Institute of Clinical Immunology, Russian Academy of Medical Sciences Siberian Branch, Novosibirsk, Russia
| | | | | | | | | |
Collapse
|
41
|
Lee KE, Kim EY, Kim CS, Choi JS, Bae EH, Ma SK, Park JS, Jung YD, Kim SH, Lee JU, Kim SW. Macrophage-stimulating protein attenuates hydrogen peroxide-induced apoptosis in human renal HK-2 cells. Eur J Pharmacol 2013; 715:304-11. [PMID: 23726950 DOI: 10.1016/j.ejphar.2013.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 04/30/2013] [Accepted: 05/11/2013] [Indexed: 01/12/2023]
Abstract
Macrophage-stimulating protein (MSP) and its receptor, recepteur d'origine nantais (RON), play an important role in cell proliferation and migration. We have investigated the role of MSP in hydrogen peroxide (H2O2)-induced renal tubular apoptosis. Human renal proximal tubular (HK-2) cells were incubated with H2O2 for 24h in the presence of different concentrations of MSP, and cell viability was measured by MTT assay. The protein expression of Bax, Bcl-2, caspase-3, mitogen-activated protein kinases (MAPKs), phosphatidylinositol-3-kinase (PI3K)/Akt, and nuclear factor-kappa B (NF-κB) was determined by semiquantitative immunoblotting. Apoptosis was assessed by flow cytometry analysis after HK-2 cells were stained with fluorescein isothiocyanate-conjugated annexin V protein and propidium iodide. H2O2 treatment decreased cell viability in HK-2 cells; this was counteracted by MSP pretreatment. H2O2 treatment induced an increased ratio of Bax/Bcl-2, cleaved caspase-3, and the number of condensed nuclei, which was also counteracted by MSP. Flow cytometry analysis showed H2O2-induced apoptosis, and its prevention by MSP treatment. Increased protein expression of phospho-p38 MAPK was attenuated by MSP, while phospho-extracellular signal-regulated kinase and c-Jun-N-terminal kinase were not affected. H2O2 induced NF-κB activation and IκB-α degradation, but the increased nuclear NF-κB activation was counteracted by MSP or by a p38 MAPK inhibitor. H2O2 treatment decreased expression of phospho-PI3K and phospho-Akt, which was reversed by MSP pretreatment. These findings suggest that MSP attenuates H2O2-induced apoptosis in HK-2 cells by modulating the p38 and NF-κB, as well as PI3K/Akt, signaling pathways.
Collapse
Affiliation(s)
- Ko Eun Lee
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Typical cell signaling response to ionizing radiation: DNA damage and extranuclear damage. Chin J Cancer Res 2013; 24:83-9. [PMID: 23357898 DOI: 10.1007/s11670-012-0083-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Accepted: 01/15/2012] [Indexed: 12/15/2022] Open
Abstract
To treat many types of cancer, ionizing radiation (IR) is primarily used as external-beam radiotherapy, brachytherapy, and targeted radionuclide therapy. Exposure of tumor cells to IR can induce DNA damage as well as generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) which can cause non-DNA lesions or extracellular damage like lipid perioxidation. The initial radiation-induced cell responses to DNA damage and ROS like the proteolytic processing, as well as synthesis and releasing ligands (such as growth factors, cytokines, and hormone) can cause the delayed secondary responses in irradiated and unirradiated bystander cells through paracrine and autocrine pathways.
Collapse
|
43
|
Suppression of NF-κB and NF-κB regulated oxidative stress and neuroinflammation by BAY 11-7082 (IκB phosphorylation inhibitor) in experimental diabetic neuropathy. Biochimie 2012; 94:1158-65. [DOI: 10.1016/j.biochi.2012.01.023] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 01/31/2012] [Indexed: 12/22/2022]
|
44
|
Effect of paeonol on antioxidant and immune regulatory activity in hepatocellular carcinoma rats. Molecules 2012; 17:4672-83. [PMID: 22522397 PMCID: PMC6268820 DOI: 10.3390/molecules17044672] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 03/27/2012] [Accepted: 04/05/2012] [Indexed: 12/26/2022] Open
Abstract
The study investigated the immunity and antioxidant potential of paeonol by employing a hepatocellular carcinoma (HCC) rat model. Three doses of paeonol (20, 40, 60 mg/kg b.w. orally) were administrated to diethylnitrosamine (DEN)-induced HCC rats. Results showed that paeonol significantly reduced the serum AST, ALT, ALP, GGT, AFU and liver MDA levels, increased serum WBC, TP, ALB, A/G, TNF-α and IFN-γ and liver antioxidant enzymes activities (SOD, CAT, GSH-Px, GR) in HCC rats. Altogether, these results suggest that the paeonol could effectively decrease oxidative injury and improve immunity function in HCC rats.
Collapse
|
45
|
Sjögren's syndrome pathological neovascularization is regulated by VEGF-A-stimulated TACE-dependent crosstalk between VEGFR2 and NF-κB. Genes Immun 2012; 13:411-20. [PMID: 22513453 DOI: 10.1038/gene.2012.9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We explore the involvement of tumor necrosis factor α (TNF-α)-converting enzyme (TACE) in vascular endothelial growth factor (VEGF) and its receptor 2 (VEGFR2) (VEGF-A/VEGFR2)-mediated angiogenesis in Sjögren's syndrome (SS), one of the most common autoimmune rheumatic diseases. To test the hypothesis that SS autoantibodies (Abs) regulate VEGF-A/VEGFR2 expression by a TACE-dependent nuclear factor-κB (NF-κB) activation pathway, their effects on the expression and activation of the VEGF-A/TACE/VEGFR2/NF-κB pathway were determined in human salivary gland epithelial cells (SGEC). An enhanced angiogenesis in SS salivary gland biopsies was observed, associated with an increased VEGF-A expression and activation of VEGF-A/VEGFR2 signaling. Human cytokine array analysis of the pro-inflammatory and pro-angiogenic protein response in SGEC treated with SS Abs revealed an overexpression of multiple pro-angiogenic factors. TACE RNA knockdown, the use of anti-VEGF-A monoclonal antibody and the inhibition of NF-κB activity significantly abrogated the release of pro-angiogenic factors, demonstrating that VEGF-A/TACE/VEGFR2/NF-κB axis dysfunction may be contributory to pathogenesis and exacerbation of this autoimmune condition.
Collapse
|
46
|
Lisi S, Sisto M, Lofrumento DD, D'Amore M. Sjögren's syndrome autoantibodies provoke changes in gene expression profiles of inflammatory cytokines triggering a pathway involving TACE/NF-κB. J Transl Med 2012; 92:615-24. [PMID: 22157716 DOI: 10.1038/labinvest.2011.190] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
We explore the association of the inflammatory gene expression profile observed in the chronic inflammatory autoimmune disorder Sjögren's syndrome (SS) with changes in TNF-α converting enzyme (TACE), tumor necrosis factor (TNF)-α and nuclear factor (NF)-κB levels showing that pathways that include TNF-α signaling converge on NF-κB contributing to exacerbate the diseases. The treatment of human salivary gland epithelial cells (SGECs) with SS anti-Ro/SSA autoantibodies (Abs) result in a progressive increase in NF-κB-DNA binding, that includes a marked enhancement in NF-κB subunit p65 protein-DNA binding. A human cytokine multi-analyte array demonstrated that the NF-κB proinflammatory target genes, increased by anti-Ro/SSA Abs treatment, includes CXC chemokines (CXCL1, CXCL6 and CXCL9), CC chemokines (CCL2, CCL13 and CCL20), interleukins (IL-1α, IL-1β, IL-1F8, IL-6, IL-8, IL-9, IL-13, IL-17 and IL-22) and their receptors (IL-1RN, IL-10Rα, IL-13Rα, CCR1, CCR2, CCR3, CCR4 and CXCR1). Blockade of TACE through the use of the specific inhibitor TAPI-1 regulates proinflammatory cytokines production in SGEC treated with anti-Ro/SSA Abs inhibiting NF-κB nuclear translocation and activation. To further investigate the role of NF-κB on anti-Ro/SSA Abs-determined proinflammatory gene expression, we used the inhibitory protein IκB-α dominant negative super-repressor as inhibitor of NF-κB-DNA binding, demonstrating that transfection with dominant-negative IκB-α in anti-Ro/SSA-treated SGEC determined a marked reduction of proinflammatory cytokines gene expression. Although further studies are needed to clarify the mechanisms underlying SS, our results demonstrate that SS Abs exert their pathogenic effects via triggering the TACE/TNF-α/NF-κB axis.
Collapse
Affiliation(s)
- Sabrina Lisi
- Department of Human Anatomy and Histology, Laboratory of Cell Biology, University of Bari Medical School, Bari, Italy.
| | | | | | | |
Collapse
|
47
|
Agarwal D, Elks CM, Reed SD, Mariappan N, Majid DS, Francis J. Chronic exercise preserves renal structure and hemodynamics in spontaneously hypertensive rats. Antioxid Redox Signal 2012; 16:139-52. [PMID: 21895524 PMCID: PMC3222098 DOI: 10.1089/ars.2011.3967] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 08/09/2011] [Accepted: 08/09/2011] [Indexed: 12/24/2022]
Abstract
AIMS Exercise training (ExT) is a recommended adjunct to many pharmaceutical antihypertensive therapies. The effects of chronic ExT on the development of hypertension-induced renal injury remain unknown. We examined whether ExT would preserve renal hemodynamics and structure in the spontaneously hypertensive rat (SHR), and whether these effects were mediated by improved redox status and decreased inflammation. Normotensive WKY rats and SHR underwent moderate-intensity ExT for 16 weeks. One group of SHR animals was treated with hydralazine to investigate the pressure-dependent/independent effects of ExT. Acute renal clearance experiments were performed prior to sacrifice. Tissue free radical production rates were measured by electron paramagnetic resonance; gene and protein expression were measured by real time RT-PCR and Western blot or immunofluorescence, respectively. Plasma angiotensin II levels and kidney antioxidants were assessed. Training efficacy was assessed by citrate synthase activity assay in hind-limb muscle. RESULTS ExT delayed hypertension, prevented oxidative stress and inflammation, preserved antioxidant status, prevented an increase in circulating AngII levels, and preserved renal hemodynamics and structure in SHR. In addition, exercise-induced effects, at least, in part, were found to be pressure-independent. INNOVATION This study is the first to provide mechanistic evidence for the renoprotective benefits of ExT in a model of hypertension. Our results demonstrate that initiation of ExT in susceptible patients can delay the development of hypertension and provide renoprotection at the functional and ultrastructural level. CONCLUSION Chronic ExT preserves renal hemodynamics and structure in SHR; these effects are partially mediated by improved redox status and decreased inflammation.
Collapse
Affiliation(s)
- Deepmala Agarwal
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Carrie M. Elks
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Scott D. Reed
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Nithya Mariappan
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Dewan S.A. Majid
- Department of Physiology, Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - Joseph Francis
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| |
Collapse
|
48
|
Babu SK, Puddicombe SM, Arshad HH, Wilson SJ, Ward J, Gozzard N, Higgs G, Holgate ST, Davies DE. Tumor necrosis factor alpha (TNF-α) autoregulates its expression and induces adhesion molecule expression in asthma. Clin Immunol 2011; 140:18-25. [PMID: 21459047 DOI: 10.1016/j.clim.2011.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 02/27/2011] [Accepted: 03/08/2011] [Indexed: 01/28/2023]
Abstract
Subjects with mild asthma underwent repeated low-dose allergen exposure and bronchial biopsies were examined for the expression of TNF-α and adhesion molecules. Bronchial biopsies from moderately severe asthmatics were then tested in an explant culture system to assess the effect of Der p and CDP-870, a TNF-α blocking pegylated-antibody Fab, on expression of TNF-α and adhesion molecules. Low-dose allergen challenge significantly upregulated sub-mucosal mast cells, TNF-α(+) cells, and VCAM. When bronchial explants were exposed to Der p and CDP 870 for 24h, CDP 870 caused a significant reduction in TNF-α release both at baseline and following stimulation with Der p allergen. The bronchial biopsies showed significant upregulation of TNF-α positive cells and ICAM-1 following exposure to Der p (p=0.03) and this was reduced in the presence of CDP-870. So, allergen exposure up-regulates TNF-α expression in asthma and down-stream targets, including adhesion molecules that contribute to airway inflammation.
Collapse
Affiliation(s)
- Suresh K Babu
- The Brooke Laboratories, Division of Infection, Inflammation, and Repair, University of Southampton School of Medicine, Southampton General Hospital, Southampton, SO16 6YD, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Quantification of mast cells and blood vessels in the skin of patients with cutaneous mucinosis. Am J Dermatopathol 2010; 32:453-8. [PMID: 20442641 DOI: 10.1097/dad.0b013e3181b1c593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent studies have suggested that mast cell numbers are increased in the skin of patients with cutaneous mucinosis and that these cells may have an important role in angiogenesis and production of mucin. Then, skin biopsies from 30 patients with cutaneous mucinosis (papular mucinosis, focal mucinosis, and mucinosis associated with lupus erythematosus) and from 10 healthy subjects were analyzed. Mast cells and blood vessels were immunolabeled with anti-tryptase and anti-CD34 antibodies, respectively, and then quantified stereologically. Counting was performed in papillary and reticular dermis. An increase in the number of mast cells was observed in the skin of patients with cutaneous mucinosis compared with the control group. Only minimal differences were observed in vessel stereology. There was no correlation between the increase in the number of mast cells and the number of blood vessels in the patients studied. There was no significant difference in the numbers of mast cells or blood vessels between the 3 subgroups of cutaneous mucinosis. Although many clinical forms of mucinosis have been described, neither mast cell number nor vessel distribution seems to distinguish the 3 different forms studied here.
Collapse
|
50
|
Turner DA, Paszek P, Woodcock DJ, Nelson DE, Horton CA, Wang Y, Spiller DG, Rand DA, White MRH, Harper CV. Physiological levels of TNFalpha stimulation induce stochastic dynamics of NF-kappaB responses in single living cells. J Cell Sci 2010; 123:2834-43. [PMID: 20663918 DOI: 10.1242/jcs.069641] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Nuclear factor kappa B (NF-kappaB) signalling is activated by cellular stress and inflammation and regulates cytokine expression. We applied single-cell imaging to investigate dynamic responses to different doses of tumour necrosis factor alpha (TNFalpha). Lower doses activated fewer cells and those responding showed an increasingly variable delay in the initial NF-kappaB nuclear translocation and associated IkappaBalpha degradation. Robust 100 minute nuclear:cytoplasmic NF-kappaB oscillations were observed over a wide range of TNFalpha concentrations. The result is supported by computational analyses, which identified a limit cycle in the system with a stable 100 minute period over a range of stimuli, and indicated no co-operativity in the pathway activation. These results suggest that a stochastic threshold controls functional all-or-nothing responses in individual cells. Deterministic and stochastic models simulated the experimentally observed activation threshold and gave rise to new predictions about the structure of the system and open the way for better mechanistic understanding of physiological TNFalpha activation of inflammatory responses in cells and tissues.
Collapse
Affiliation(s)
- David A Turner
- Centre for Cell Imaging, School of Biological Sciences, Bioscience Research Building, Liverpool, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|