51
|
Pollard HB, Shivakumar C, Starr J, Eidelman O, Jacobowitz DM, Dalgard CL, Srivastava M, Wilkerson MD, Stein MB, Ursano RJ. "Soldier's Heart": A Genetic Basis for Elevated Cardiovascular Disease Risk Associated with Post-traumatic Stress Disorder. Front Mol Neurosci 2016; 9:87. [PMID: 27721742 PMCID: PMC5033971 DOI: 10.3389/fnmol.2016.00087] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/05/2016] [Indexed: 11/13/2022] Open
Abstract
"Soldier's Heart," is an American Civil War term linking post-traumatic stress disorder (PTSD) with increased propensity for cardiovascular disease (CVD). We have hypothesized that there might be a quantifiable genetic basis for this linkage. To test this hypothesis we identified a comprehensive set of candidate risk genes for PTSD, and tested whether any were also independent risk genes for CVD. A functional analysis algorithm was used to identify associated signaling networks. We identified 106 PTSD studies that report one or more polymorphic variants in 87 candidate genes in 83,463 subjects and controls. The top upstream drivers for these PTSD risk genes are predicted to be the glucocorticoid receptor (NR3C1) and Tumor Necrosis Factor alpha (TNFA). We find that 37 of the PTSD candidate risk genes are also candidate independent risk genes for CVD. The association between PTSD and CVD is significant by Fisher's Exact Test (P = 3 × 10-54). We also find 15 PTSD risk genes that are independently associated with Type 2 Diabetes Mellitus (T2DM; also significant by Fisher's Exact Test (P = 1.8 × 10-16). Our findings offer quantitative evidence for a genetic link between post-traumatic stress and cardiovascular disease, Computationally, the common mechanism for this linkage between PTSD and CVD is innate immunity and NFκB-mediated inflammation.
Collapse
Affiliation(s)
- Harvey B. Pollard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health SciencesBethesda, MD, USA
- Collaborative Health Initiative Research Program, Uniformed Services University of the Health SciencesBethesda, MD, USA
| | - Chittari Shivakumar
- Collaborative Health Initiative Research Program, Uniformed Services University of the Health SciencesBethesda, MD, USA
| | - Joshua Starr
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health SciencesBethesda, MD, USA
| | - Ofer Eidelman
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health SciencesBethesda, MD, USA
| | - David M. Jacobowitz
- Collaborative Health Initiative Research Program, Uniformed Services University of the Health SciencesBethesda, MD, USA
| | - Clifton L. Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health SciencesBethesda, MD, USA
- Collaborative Health Initiative Research Program, Uniformed Services University of the Health SciencesBethesda, MD, USA
| | - Meera Srivastava
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health SciencesBethesda, MD, USA
| | - Matthew D. Wilkerson
- Collaborative Health Initiative Research Program, Uniformed Services University of the Health SciencesBethesda, MD, USA
| | - Murray B. Stein
- Department of Psychiatry, University of California, San DiegoSan Diego, CA, USA
| | - Robert J. Ursano
- Department of Psychiatry Uniformed Services University School of Medicine, Uniformed Services University of the Health SciencesBethesda, MD, USA
- Center for the Study of Traumatic stress, Uniformed Services University of the Health SciencesBethesda, MD, USA
| |
Collapse
|
52
|
Wei J, Hettinghouse A, Liu C. The role of progranulin in arthritis. Ann N Y Acad Sci 2016; 1383:5-20. [PMID: 27505256 DOI: 10.1111/nyas.13191] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/28/2016] [Accepted: 07/07/2016] [Indexed: 12/11/2022]
Abstract
Progranulin (PGRN) is a growth factor with a unique beads-on-a-string structure that is involved in multiple pathophysiological processes, including anti-inflammation, tissue repair, wound healing, neurodegenerative diseases, and tumorigenesis. This review presents up-to-date information concerning recent studies on the role of PGRN in inflammatory arthritis and osteoarthritis, with a special focus on the involvement of the interactions and interplay between PGRN and tumor necrosis factor receptor (TNFR) family members in regulating such musculoskeletal diseases. In addition, this paper highlights the applications of atsttrin, an engineered protein comprising three TNFR-binding fragments of PGRN, as a promising intervention in treating arthritis.
Collapse
Affiliation(s)
- Jianlu Wei
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York.,Department of Orthopaedic Surgery, Medical School of Shandong University, Jinan, Shandong, China
| | - Aubryanna Hettinghouse
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York
| | - Chuanju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York.,Department of Cell Biology, New York University School of Medicine, New York, New York
| |
Collapse
|
53
|
Tay C, Liu YH, Hosseini H, Kanellakis P, Cao A, Peter K, Tipping P, Bobik A, Toh BH, Kyaw T. B-cell-specific depletion of tumour necrosis factor alpha inhibits atherosclerosis development and plaque vulnerability to rupture by reducing cell death and inflammation. Cardiovasc Res 2016; 111:385-97. [PMID: 27492217 DOI: 10.1093/cvr/cvw186] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/08/2016] [Indexed: 12/31/2022] Open
Abstract
AIMS B2 lymphocytes promote atherosclerosis development but their mechanisms of action are unknown. Here, we investigated the role of tumour necrosis factor alpha (TNF-α) produced by B2 cells in atherogenesis. METHODS AND RESULTS We found that 50% of TNF-α-producing spleen lymphocytes were B2 cells and ∼20% of spleen and aortic B cells produced TNF-α in hyperlipidemic ApoE(-/-) mice. We generated mixed bone marrow (80% μMT/20% TNF-α(-/-)) chimeric LDLR(-/-) mice where only B cells did not express TNF-α. Atherosclerosis was reduced in chimeric LDLR(-/-) mice with TNF-α-deficient B cells. TNF-α expression in atherosclerotic lesions and in macrophages were also reduced accompanied by fewer apoptotic cells, reduced necrotic cores, and reduced lesion Fas, interleukin-1β and MCP-1 in mice with TNF-α-deficient B cells compared to mice with TNF-α-sufficient B cells. To confirm that the reduced atherosclerosis is attributable to B2 cells, we transferred wild-type and TNF-α-deficient B2 cells into ApoE(-/-) mice deficient in B cells or in lymphocytes. After 8 weeks of high fat diet, we found that atherosclerosis was increased by wild-type but not TNF-α-deficient B2 cells. Lesions of mice with wild-type B2 cells but not TNF-α-deficient B2 cells also had increased apoptotic cells and necrotic cores. Transferred B2 cells were found in lesions of recipient mice, suggesting that TNF-α-producing B2 cells promote atherosclerosis within lesions. CONCLUSION We conclude that TNF-α produced by B2 cells is a key mechanism by which B2 cells promote atherogenesis through augmenting macrophage TNF-α production to induce cell death and inflammation that promote plaque vulnerability.
Collapse
Affiliation(s)
- Christopher Tay
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Yu-Han Liu
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia
| | - Hamid Hosseini
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Peter Kanellakis
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia
| | - Anh Cao
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia
| | - Peter Tipping
- Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Alex Bobik
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Tin Kyaw
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| |
Collapse
|
54
|
Żera T, Nowiński A, Kwiatkowski P. Centrally administered TNF increases arterial blood pressure independently of nitric oxide synthase. Neuropeptides 2016; 58:67-72. [PMID: 27241175 DOI: 10.1016/j.npep.2016.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 04/15/2016] [Accepted: 05/23/2016] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Emerging evidence indicates that increased levels of TNF in the brain are associated with hypertension. Nitric oxide synthase (NOS) is involved in the central control of the cardiovascular system, exerting both pro- and antihypertensive effects. TNF induces hypothalamic synthesis of nitric oxide. AIM We checked if acutely administered TNF into the cerebral ventricles affects arterial blood pressure, heart rate and baroreflex sensitivity, and whether TNF actions are dependent on NOS in normotensive rats. METHODS We carried out hemodynamic measurements in 6 groups of freely moving, adult Sprague-Dawley male rats, intracerebroventricularly (ICV) infused with either: 1) saline (5μl/h); 2) TNF (200ng/5μl/h); 3) non-selective NO synthase inhibitor - l-NG-Nitroarginine Methyl Ester (l-NAME) (1mg/5μl/h); 4) TNF together with l-NAME (200ng and 1mg/5μl/h, respectively); 5) neuronal NO synthase inhibitor - 7-nitroindazole sodium salt (7-NI) (20μg/10μl/h); 6) or TNF together with 7-NI (200ng and 20μg/10μl/h, respectively). Mean arterial blood pressure (MABP), heart rate (HR) and spontaneous baroreflex sensitivity (sBRS) evaluated by the sequence method were analysed. RESULTS ICV infusion of TNF caused a significant increase in MABP accompanied by a transient increase in HR, and a decrease in sBRS. ICV infusion of l-NAME increased MABP, but it did not change HR, nor sBRS. ICV infusion of 7-NI did not affect MABP, nor HR, nor sBRS. TNF administered together with l-NAME increased MABP with a transient increase in HR without changes of sBRS. Similarly, ICV infusion of TNF with 7-NI increased MABP without changes in HR and sBRS. CONCLUSIONS Centrally administered TNF increases MABP and HR and blunts sBRS. The pressor effect of TNF appears to be independent of NOS activity in the brain. Inhibition of nNOS restores sBRS in TNF treated rats.
Collapse
Affiliation(s)
- Tymoteusz Żera
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, The Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland.
| | - Artur Nowiński
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, The Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Piotr Kwiatkowski
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, The Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| |
Collapse
|
55
|
The novel adipokine progranulin counteracts IL-1 and TLR4-driven inflammatory response in human and murine chondrocytes via TNFR1. Sci Rep 2016; 6:20356. [PMID: 26853108 PMCID: PMC4745010 DOI: 10.1038/srep20356] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/30/2015] [Indexed: 12/20/2022] Open
Abstract
Progranulin (PGRN) is a recently identified adipokine that is supposed to have anti-inflammatory actions. The proinflammatory cytokine interleukin-1β (IL1β) stimulates several mediators of cartilage degradation. Toll like receptor-4 (TLR4) can bind to various damage-associated molecular patterns, leading to inflammatory condition. So far, no data exist of PGRN effects in inflammatory conditions induced by IL1β or lipopolysaccharide (LPS). Here, we investigated the anti-inflammatory potential of PGRN in IL1β- or LPS-induced inflammatory responses of chondrocytes. Human osteoarthritic chondrocytes and ATDC-5 cells were treated with PGRN in presence or not of IL1β or LPS. First, we showed that recombinant PGRN had no effects on cell viability. We present evidence that PGRN expression was increased during the differentiation of ATDC-5 cell line. Moreover, PGRN mRNA and protein expression is increased in cartilage, synovial and infrapatellar fat pad tissue samples from OA patients. PGRN mRNA levels are upregulated under TNFα and IL1β stimulation. Our data showed that PGRN is able to significantly counteract the IL1β-induced expression of NOS2, COX2, MMP13 and VCAM-1. LPS-induced expression of NOS2 is also decreased by PGRN. These effects are mediated, at least in part, through TNFR1. Taken together, our results suggest that PGRN has a clear anti-inflammatory function.
Collapse
|
56
|
Zhang G, Sun Y, Wang Y, Liu R, Bao Y, Li Q. MiR-502-5p inhibits IL-1β-induced chondrocyte injury by targeting TRAF2. Cell Immunol 2016; 302:50-57. [PMID: 26861148 DOI: 10.1016/j.cellimm.2016.01.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/13/2016] [Accepted: 01/30/2016] [Indexed: 12/18/2022]
Abstract
Osteoarthritis (OA) is characterized by articular cartilage degradation and joint inflammation. MicroRNAs have been proven to play an important role in the regulation of chondrogenesis. The aim of the present study was to investigate the effect of miR-502-5p in OA. The results showed that miR-502-5p levels were significantly down-regulated in OA articular tissues and IL-1β-induced chondrocytes compared with control groups. MiR-502-5p overexpression inhibited IL-1β-induced reduction in cell viability and increase in cell apoptosis, and alleviated IL-1β-induced extracellular matrix (ECM) metabolic imbalance and pro-inflammatory cytokine production. MiR-502-5p targeted the 3'-untranslated region (UTR) of TRAF2 to inhibit its expression. The IL-1β-induced activation of NF-κB signaling pathway was inhibited by PDTC, an inhibitor of NF-κB, which was also suppressed by the miR-502-5p mimic and TRAF2 siRNA transfection. In conclusion, miR-502-5p may exhibit a protective effect on IL-1β-induced chondrocyte injury by targeting TRAF2 and inhibiting NF-κB signaling pathway.
Collapse
Affiliation(s)
- Guoliang Zhang
- Orthopedical Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China; Department of Orthopedics, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Yanhong Sun
- Department of Physiology, Inner Mongolia Medical University, Hohhot 010110, China
| | - Yuewen Wang
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Rui Liu
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Yimin Bao
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Qi Li
- Orthopedical Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| |
Collapse
|
57
|
Khalfaoui S, Eichhorn V, Karagiannidis C, Bayh I, Brockmann M, Pieper M, Windisch W, Schildgen O, Schildgen V. Lung Infection by Human Bocavirus Induces the Release of Profibrotic Mediator Cytokines In Vivo and In Vitro. PLoS One 2016; 11:e0147010. [PMID: 26807786 PMCID: PMC4726461 DOI: 10.1371/journal.pone.0147010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/28/2015] [Indexed: 12/12/2022] Open
Abstract
Human Bocavirus subtype 1 (HBoV1) is associated with respiratory diseases and may contribute to chronic lung diseases by persisting in the infected host. Here the question was addressed if HBoV infections could contribute to fibrogenesis processes as suggested by previously published clinical observations. Cytokine profiles induced by HBoV infection in CuFi-8 air-liquid interphase cell cultures and in bronchoalveolar lavage fluid (BALF) of 20 HBoV-positive and 12 HBoV-negative patients were analysed by semi-quantitative Western spot blot analyses. Although lots of cytokines were regulated independently of HBoV status, several cytokines associated with lung fibrosis and tumour development, e.g., EGF, VEGF, TARC (CCL17), TNF-α, TNF-β, TIMP-1, were clearly upregulated in the HBoV-positive cohort. These findings suggest that the development of lung fibrosis might be triggered by HBoV induced cytokine expression.
Collapse
Affiliation(s)
- Soumaya Khalfaoui
- Kliniken der Stadt Köln gGmbH, University Hospital Witten/Herdecke, Cologne-Merheim, Ostmerheimer Strasse 200, Institute for Pathology, D-51109 Cologne, Germany
| | - Vivien Eichhorn
- Kliniken der Stadt Köln gGmbH, University Hospital Witten/Herdecke, Cologne-Merheim, Ostmerheimer Strasse 200, Institute for Pathology, D-51109 Cologne, Germany
| | - Christian Karagiannidis
- Kliniken der Stadt Köln gGmbH, University Hospital Witten/Herdecke, Cologne-Merheim, Ostmerheimer Strasse 200, Department of Pneumology and Critical Care Medicine, D-51109 Cologne, Germany
| | - Inga Bayh
- Institute for Medical Biometry and Epidemiology, Faculty of Health, Alfred-Herrhausen-Str. 50, Witten/Herdecke University, Witten, Germany
| | - Michael Brockmann
- Kliniken der Stadt Köln gGmbH, University Hospital Witten/Herdecke, Cologne-Merheim, Ostmerheimer Strasse 200, Institute for Pathology, D-51109 Cologne, Germany
| | - Monika Pieper
- Kliniken der Stadt Köln gGmbH, University Hospital Witten/Herdecke, Cologne-Merheim, Ostmerheimer Strasse 200, Institute for Pathology, D-51109 Cologne, Germany
| | - Wolfram Windisch
- Kliniken der Stadt Köln gGmbH, University Hospital Witten/Herdecke, Cologne-Merheim, Ostmerheimer Strasse 200, Department of Pneumology and Critical Care Medicine, D-51109 Cologne, Germany
| | - Oliver Schildgen
- Kliniken der Stadt Köln gGmbH, University Hospital Witten/Herdecke, Cologne-Merheim, Ostmerheimer Strasse 200, Institute for Pathology, D-51109 Cologne, Germany
| | - Verena Schildgen
- Kliniken der Stadt Köln gGmbH, University Hospital Witten/Herdecke, Cologne-Merheim, Ostmerheimer Strasse 200, Institute for Pathology, D-51109 Cologne, Germany
- * E-mail: ;
| |
Collapse
|
58
|
MAHMOOD A, TIWARI AK, ŞAHİN K, KÜÇÜK Ö, ALI S. Triterpenoid saponin-rich fraction of Centella asiatica decreases IL-1β andNF-κB, and augments tissue regeneration and excision wound repair. Turk J Biol 2016. [DOI: 10.3906/biy-1507-63] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
59
|
Protumoral TSP50 Regulates Macrophage Activities and Polarization via Production of TNF-α and IL-1β, and Activation of the NF-κB Signaling Pathway. PLoS One 2015; 10:e0145095. [PMID: 26684869 PMCID: PMC4684331 DOI: 10.1371/journal.pone.0145095] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 11/28/2015] [Indexed: 12/27/2022] Open
Abstract
Testes-specific protease 50 (TSP50) is abnormally overexpressed in many kinds of cancers and promotes cell proliferation and migration. However, whether TSP50 can influence the tumor microenvironment, especially the function of immune cells in the microenvironment, remains largely unknown. We demonstrated that exposure to the conditioned medium from TSP50-overexpressing cells, or co-culture with TSP50-overexpressing cells, enhanced the cytokine production and phagocytic activities of macrophages, and induced M2b polarization. Further investigation showed that production of TNF-α and IL-1β was strongly induced by TSP50 in TSP50-overexpressing cells. TSP50-induced TNF-α and IL-1β were main factors that mediated the effects of TSP50-overexpressing cells on macrophages. The NF-κB pathway could be activated in macrophages upon the treatment of conditioned medium of TSP50-overexpressing cells and its activation is necessary for the observed effects on macrophages. Taken together, our results suggested that oncogenic TSP50 expressed in cells could activate surrounding macrophages and induce M2b polarization, partly through inducing TNF-α/ IL-1β secretion and subsequent NF-κB pathway activation. This implies a potential mechanism by which oncogene TSP50 regulates tumor microenvironment to support tumor development.
Collapse
|
60
|
Uğuz AC, Öz A, Nazıroğlu M. Curcumin inhibits apoptosis by regulating intracellular calcium release, reactive oxygen species and mitochondrial depolarization levels in SH-SY5Y neuronal cells. J Recept Signal Transduct Res 2015; 36:395-401. [PMID: 26608462 DOI: 10.3109/10799893.2015.1108337] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Neurological diseases such as Alzheimer's and Parkinson's diseases are incurable progressive neurological disorders caused by the degeneration of neuronal cells and characterized by motor and non-motor symptoms. Curcumin, a turmeric product, is an anti-inflammatory agent and an effective reactive oxygen and nitrogen species scavenging molecule. Hydrogen peroxide (H2O2) is the main source of oxidative stress, which is claimed to be the major source of neurological disorders. Hence, in this study we aimed to investigate the effect of curcumin on Ca(2+) signaling, oxidative stress parameters, mitochondrial depolarization levels and caspase-3 and -9 activities that are induced by the H2O2 model of oxidative stress in SH-SY5Y neuronal cells. SH-SY5Y neuronal cells were divided into four groups namely, the control, curcumin, H2O2, and curcumin + H2O2 groups. The dose and duration of curcumin and H2O2 were determined from published data. The cells in the curcumin, H2O2, and curcumin + H2O2 groups were incubated for 24 h with 5 µM curcumin and 100 µM H2O2. Lipid peroxidation and cytosolic free Ca(2+) concentrations were higher in the H2O2 group than in the control group; however, their levels were lower in the curcumin and curcumin + H2O2 groups than in the H2O2 group alone. Reduced glutathione (GSH) and glutathione peroxidase (GSH-Px) values were lower in the H2O2 group although they were higher in the curcumin and curcumin + H2O2 groups than in the H2O2 group. Caspase-3 activity was lower in the curcumin group than in the H2O2 group. In conclusion, curcumin strongly induced modulator effects on oxidative stress, intracellular Ca(2+) levels, and the caspase-3 and -9 values in an experimental oxidative stress model in SH-SY5Y cells.
Collapse
Affiliation(s)
- Abdülhadi Cihangir Uğuz
- a Department of Biophysics , School of Medicine, Süleyman Demirel University , Isparta , Turkey and.,b Neuroscience Research Center, Süleyman Demirel University , Isparta , Turkey
| | - Ahmi Öz
- a Department of Biophysics , School of Medicine, Süleyman Demirel University , Isparta , Turkey and
| | - Mustafa Nazıroğlu
- a Department of Biophysics , School of Medicine, Süleyman Demirel University , Isparta , Turkey and.,b Neuroscience Research Center, Süleyman Demirel University , Isparta , Turkey
| |
Collapse
|
61
|
Gong Z, Zhou J, Li H, Gao Y, Xu C, Zhao S, Chen Y, Cai W, Wu J. Curcumin suppresses NLRP3 inflammasome activation and protects against LPS-induced septic shock. Mol Nutr Food Res 2015; 59:2132-42. [PMID: 26250869 DOI: 10.1002/mnfr.201500316] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/09/2015] [Accepted: 07/24/2015] [Indexed: 01/13/2023]
Abstract
SCOPE The NLRP3 inflammasome responds to various pathogen-derived factors and danger-associated molecules, mediating IL-1β maturation, therefore is involved in multiple inflammatory diseases. Curcumin has been shown to possess strong anti-inflammatory activity, but the underlying mechanism is not fully understood. Here, we sought to investigate the role and mechanism of curcumin on the inhibition of mature IL-1β production via the regulation of NLRP3 inflammasome. METHODS AND RESULTS Curcumin dramatically inhibited the production of mature IL-1β in LPS-primed macrophages triggered by multiple NLRP3 inflammasome activators, and also reduced the level of cleaved caspase-1 as measured by western blot and ELISA. Curcumin prevented K(+) efflux, the common trigger for NLRP3 inflammasome activation, and attenuated lysosomes disruption and intracellular ROS formation as well. The inhibition of NLRP3 inflammasome by curcumin was in part mediated via the suppression of extracellular regulated protein kinases phosphorylation. Furthermore, administration of curcumin significantly reduced peritoneal IL-1β and HMGB-1 concentration induced by LPS and improved the survival of mice suffering from lethal endotoxic shock. CONCLUSION Curcumin potently inhibits the activation of NLRP3 inflammasome which may contribute to its anti-inflammatory activity. Our finding offers a mechanistic basis for the therapeutic potential of curcumin in septic shock and other NLRP3 inflammasome-driven diseases.
Collapse
Affiliation(s)
- Zizhen Gong
- Department of pediatric Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China.,Shanghai Institute for Pediatric Research, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, P. R. China
| | - Jiefei Zhou
- Department of pediatric Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China.,Shanghai Institute for Pediatric Research, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, P. R. China
| | - Hui Li
- Department of Pathology, Shanghai institute of Health Science, Shanghai, P. R. China
| | - Yanhong Gao
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China
| | - Congfeng Xu
- Shanghai Institute of Immunology, Institutes of Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China
| | - Shengnan Zhao
- Department of pediatric Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China.,Shanghai Institute for Pediatric Research, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, P. R. China
| | - Yingwei Chen
- Shanghai Institute for Pediatric Research, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, P. R. China
| | - Wei Cai
- Department of pediatric Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China.,Shanghai Institute for Pediatric Research, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, P. R. China
| | - Jin Wu
- Department of pediatric Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China.,Shanghai Institute for Pediatric Research, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, P. R. China
| |
Collapse
|
62
|
Tumor necrosis factor alpha and its receptors in behaviour and neurobiology of adult mice, in the absence of an immune challenge. Behav Brain Res 2015; 290:51-60. [PMID: 25934492 DOI: 10.1016/j.bbr.2015.04.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/21/2015] [Accepted: 04/23/2015] [Indexed: 11/21/2022]
Abstract
Tumor necrosis factor alpha (TNF-α) is a vital component of the immune system and CNS. We previously showed that 3-month-old TNF-α and TNF-α receptor knockout mice had impaired cognition, whilst at 12-months-old mice had better cognition. To extend these findings on possible age-dependent TNF-α effects in the brain, we investigated the behaviour of 6-month-old TNF-α knockout mice and their neurobiological correlates. 6-month-old TNF(-/-), TNF-R1(-/-) and TNF-R2(-/-) mice were compared to age-matched WT mice and tested for various behaviours. ELISA hippocampal levels of nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF) and qPCR mRNA levels of Tnfa, Tnfr1, Tnfr2, Il10 and Il1β were measured. TNF-R1(-/-) and TNF(-/-) mice were found to have lesser exploratory behaviour than WT mice, while TNF-R1(-/-) mice displayed better memory than WT and TNF-R2(-/-) mice. Both TNF(-/-) and TNF-R2(-/-) mice exhibited significantly lower immobility on the depression test than WT mice. Additionally, TNF(-/-) mice expressed significantly lower levels of BDNF than WT mice in the hippocampus while TNF-R1(-/-) mice displayed significantly lower BDNF levels compared to both WT and TNF-R2(-/-) mice. TNF-R2(-/-) mice also displayed significantly higher levels of NGF compared to TNF-R1(-/-) mice. These results illustrate that TNF-α and its receptors mediate several behavioural phenotypes. Finally, BDNF and NGF levels appear to be regulated by TNF-α and its receptors even under immunologically unchallenged conditions.
Collapse
|
63
|
Potuck AN, Weed BL, Leifer CA, Chu CC. Electrostatically Self-Assembled Biodegradable Microparticles from Pseudoproteins and Polysaccharide: Fabrication, Characterization, and Biological Properties. Biomacromolecules 2015; 16:564-77. [DOI: 10.1021/bm5016255] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
| | | | - Cynthia A. Leifer
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, New York 14853-4401, United States
| | | |
Collapse
|
64
|
Zhou T, Cui X, Zhou Y, Guo J, Rong Y, Chen W. Switch regulation of interleukin-1 beta in downstream of inflammatory cytokines induced by two micro-sized silica particles on differentiated THP-1 macrophages. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:457-466. [PMID: 25596480 DOI: 10.1016/j.etap.2014.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 12/20/2014] [Accepted: 12/24/2014] [Indexed: 06/04/2023]
Abstract
To investigate the regulated role of IL-1β in initiating and maintaining inflammation, PMA-differentiated THP-1 macrophages were exposed to two micro-sized crystalline silica particles (Si3-5μm and Si1μm) from 3h to 24h, respectively. Cytotoxicity and inflammatory cytokines (IL-1β, TNF-α and IL-6) expressions measured showed that they were induced by both silica particles in positive dose-dependent manners. The levels of inflammatory cytokines induced by Si1μm were higher than those induced by Si3-5μm at low concentration. When pretreated with anti-human IL-1β, not only the high levels of IL-1β but also elevated TNF-α and IL-6 induced by both silica particles were remarkably blocked, especially Si1μm particle. In addition, recombinant human IL-1β protein could induce macrophages to strikingly augment TNF-α and IL-6 expressions. Our data suggest that IL-1β could play a critical role of switching regulation in the downstream inflammation induced by micro-sized silica particles.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China
| | - Xiuqing Cui
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yun Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jiali Guo
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yi Rong
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Weihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
65
|
Yang X, Zhao C, Gao Z, Su X. A novel regulator of lung inflammation and immunity: pulmonary parasympathetic inflammatory reflex. QJM 2014; 107:789-92. [PMID: 24440925 DOI: 10.1093/qjmed/hcu005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In this review, we first analyzed the current status of cholinergic anti-inflammatory pathway and then put forward a novel regulatory machinery-pulmonary parasympathetic inflammatory reflex, which is composed by lung vagal sensors at afferent arm, α7 nAChR (α7 nicotinic acetylcholine receptors)-expressing cells at efferent arm and the brain information integrating center. This modulatory circuit might loop the lungs, immune and nervous systems and play a very important role in regulating lung infection, inflammation and immunity through the neural innervations and signals when the lungs encounter pathogenic challenges.
Collapse
Affiliation(s)
- X Yang
- From the Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - C Zhao
- From the Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Z Gao
- From the Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - X Su
- From the Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
66
|
Phospholipase cε, an effector of ras and rap small GTPases, is required for airway inflammatory response in a mouse model of bronchial asthma. PLoS One 2014; 9:e108373. [PMID: 25269075 PMCID: PMC4182471 DOI: 10.1371/journal.pone.0108373] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 08/20/2014] [Indexed: 11/29/2022] Open
Abstract
Background Phospholipase Cε (PLCε) is an effector of Ras and Rap small GTPases and expressed in non-immune cells. It is well established that PLCε plays an important role in skin inflammation, such as that elicited by phorbol ester painting or ultraviolet irradiation and contact dermatitis that is mediated by T helper (Th) 1 cells, through upregulating inflammatory cytokine production by keratinocytes and dermal fibroblasts. However, little is known about whether PLCε is involved in regulation of inflammation in the respiratory system, such as Th2-cells-mediated allergic asthma. Methods We prepared a mouse model of allergic asthma using PLCε+/+ mice and PLCεΔX/ΔX mutant mice in which PLCε was catalytically-inactive. Mice with different PLCε genotypes were immunized with ovalbumin (OVA) followed by the challenge with an OVA-containing aerosol to induce asthmatic response, which was assessed by analyzing airway hyper-responsiveness, bronchoalveolar lavage fluids, inflammatory cytokine levels, and OVA-specific immunoglobulin (Ig) levels. Effects of PLCε genotype on cytokine production were also examined with primary-cultured bronchial epithelial cells. Results After OVA challenge, the OVA-immunized PLCεΔX/ΔX mice exhibited substantially attenuated airway hyper-responsiveness and broncial inflammation, which were accompanied by reduced Th2 cytokine content in the bronchoalveolar lavage fluids. In contrast, the serum levels of OVA-specific IgGs and IgE were not affected by the PLCε genotype, suggesting that sensitization was PLCε-independent. In the challenged mice, PLCε deficiency reduced proinflammatory cytokine production in the bronchial epithelial cells. Primary-cultured bronchial epithelial cells prepared from PLCεΔX/ΔX mice showed attenuated pro-inflammatory cytokine production when stimulated with tumor necrosis factor-α, suggesting that reduced cytokine production in PLCεΔX/ΔX mice was due to cell-autonomous effect of PLCε deficiency. Conclusions PLCε plays an important role in the pathogenesis of bronchial asthma through upregulating inflammatory cytokine production by the bronchial epithelial cells.
Collapse
|
67
|
Kim HK. Role of ERK/MAPK signalling pathway in anti-inflammatory effects of Ecklonia cava in activated human mast cell line-1 cells. ASIAN PAC J TROP MED 2014. [DOI: 10.1016/s1995-7645(14)60120-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
68
|
Extrusion process enhances the anti-inflammatory effect of Acanthopanax senticosus leaves. Food Sci Biotechnol 2014. [DOI: 10.1007/s10068-014-0122-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
69
|
Complex immune cell interplay in the gamma interferon response during Toxoplasma gondii infection. Infect Immun 2014; 82:3090-7. [PMID: 24866795 DOI: 10.1128/iai.01722-14] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Toxoplasma gondii is an obligate intracellular parasite of clinical importance, especially in immunocompromised patients. Investigations into the immune response to the parasite found that T cells are the primary effector cells regulating gamma interferon (IFN-γ)-mediated host resistance. However, recent studies have revealed a critical role for the innate immune system in mediating host defense independently of the T cell responses to the parasite. This body of knowledge is put into perspective by the unifying theme that immunity to the protozoan parasite requires a strong IFN-γ host response. In the following review, we discuss the role of IFN-γ-producing cells and the signals that regulate IFN-γ production during T. gondii infection.
Collapse
|
70
|
Ogunbileje JO, Nawgiri RS, Anetor JI, Akinosun OM, Farombi EO, Okorodudu AO. Particles internalization, oxidative stress, apoptosis and pro-inflammatory cytokines in alveolar macrophages exposed to cement dust. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 37:1060-1070. [PMID: 24769344 DOI: 10.1016/j.etap.2014.03.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/24/2014] [Accepted: 03/30/2014] [Indexed: 06/03/2023]
Abstract
Exposure to cement dust is one of the most common occupational dust exposures worldwide, but the mechanism of toxicity has not been fully elucidated. Cement dust (N) and clinker (C) samples collected from Nigeria and another sample of cement dust (U) collected from USA were evaluated using alveolar macrophage (NR8383) cell culture to determine the contribution of different sources of cement dust in the severity of cement dust toxicity. Cement dust particles internalization and morphologic alterations using transmission electron microscopy (TEM), cytotoxicity, apoptotic cells induction, intracellular reactive oxygen species generation, glutathione reduction, TNF-α, IL-1β, and CINC-3 secretion in alveolar macrophages (NR8383) exposed to cement dust and clinker samples were determined. Particles were internalized into the cytoplasmic vacuoles, with cells exposed to U showing increased cell membrane blebbing. Also, NR8383 exposed to U show more significant ROS generation, apoptotic cells induction and decreased glutathione. Interleukin-1β and TNF-α secretion were significantly more in cells exposed to both cement dust samples compared with clinker, while CINC-3 secretion was significantly more in cells exposed to clinker (p < 0.05). Endocytosis, oxidative stress induced-apoptosis and induction of pro-inflammatory cytokines may be key mechanisms of cement dust immunotoxicity in the lung and toxicity may be factory dependent.
Collapse
Affiliation(s)
- J O Ogunbileje
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Department of Chemical Pathology and Immunology, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - R S Nawgiri
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - J I Anetor
- Department of Chemical Pathology and Immunology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - O M Akinosun
- Department of Chemical Pathology and Immunology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - E O Farombi
- Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - A O Okorodudu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
71
|
Immunophenotype expressions and cytokine profiles of influenza A H1N1 virus infection in pediatric patients in 2009. DISEASE MARKERS 2014; 2014:195453. [PMID: 24696530 PMCID: PMC3948652 DOI: 10.1155/2014/195453] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 12/26/2013] [Accepted: 01/13/2014] [Indexed: 02/07/2023]
Abstract
Background. A novel swine-origin influenza A H1N1 virus (S-OIV) caused human infection and acute respiratory illness in 2009, resulting in an influenza pandemic. Objectives. This study characterized the immune responses of S-OIV infection in pediatric patients at risk of pulmonary complications. Methods. All enrolled pediatric patients were confirmed virologically for S-OIV infection in 2009-2010, prospectively. Changes in cellular immunophenotypes were analyzed using flow cytometry. Plasma cytokine levels associated with S-OIV infection by pulmonary and without pulmonary complications were measured using cytokine cytometric bead assay kits. Results. A total of 85 patients with a mean age of 10.3 years were recruited. The level of C-reactive protein (CRP) was high in patients exhibiting pulmonary complications. The percentage of cellular immunophenotypes did not change between patients with and without pulmonary complications. The absolute numbers of peripheral blood mononuclear cells (PBMC), CD3, CD8, and CD16CD56 decreased with acute S-OIV pulmonary complications. Acute influenza infection with pulmonary complications was associated with high plasma concentrations of IL-1β, IL-6, IL-12, and IFN-γ. Conclusion. Immunophenotype studies have reported variability in immune response to the severity of S-OIV infections. Acute phase cytokine profiles of the 2009 S-OIV infection might have contributed to the pathogenesis of the pulmonary complications.
Collapse
|
72
|
Liang Y, Ma S, Zhang Y, Wang Y, Cheng Q, Wu Y, Jin Y, Zheng D, Wu D, Liu H. IL-1β and TLR4 signaling are involved in the aggravated murine acute graft-versus-host disease caused by delayed bortezomib administration. THE JOURNAL OF IMMUNOLOGY 2013; 192:1277-85. [PMID: 24363427 DOI: 10.4049/jimmunol.1203428] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
It was shown that the proteasome inhibitor, bortezomib, administered immediately following allogeneic bone marrow transplantation resulted in marked inhibition of acute graft-versus-host disease (aGVHD), with retention of graft-versus-tumor effects. However, continuous bortezomib administration resulted in significant acceleration of graft-versus-host disease-dependent morbidity. We carried out studies to dissect the mechanisms of aggravated aGVHD caused by delayed bortezomib administration. First, we demonstrated that IL-1β was critically involved, and the subsequent aGVHD could be alleviated by IL-1β blockade. Bortezomib treatment after dendritic cell (DC) activation resulted in drastically elevated IL-1β production, whereas bortezomib treatment before DC activation inhibited IL-1β production, suggesting that the timing of bortezomib administration significantly affected IL-1β production by DCs. We further demonstrated that delayed administration of bortezomib accelerated aGVHD through TLR4 signaling. Because the LPS levels were much lower with reduced-intensity conditioning compared with high-dose irradiation, the accelerated graft-versus-host disease-dependent morbidity with delayed bortezomib administration could be rescued by reduced-intensity conditioning. Our studies suggested that TLR4 pathway activation and delayed bortezomib administration amplified the production of IL-1β and other inflammatory cytokines, which resulted in accelerated aGVHD-dependent morbidity. These results indicated that decreased toxicity of continuous bortezomib administration could be achieved by reduced-intensity conditioning or by inhibiting IL-1β.
Collapse
Affiliation(s)
- Yong Liang
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Karki R, Igwe OJ. Toll-like receptor 4-mediated nuclear factor kappa B activation is essential for sensing exogenous oxidants to propagate and maintain oxidative/nitrosative cellular stress. PLoS One 2013; 8:e73840. [PMID: 24058497 PMCID: PMC3776800 DOI: 10.1371/journal.pone.0073840] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 07/31/2013] [Indexed: 11/19/2022] Open
Abstract
The mechanism(s) by which cells can sense exogenous oxidants that may contribute to intracellular oxidative/nitrosative stress is not clear. The objective of this study was to determine how cells might respond to exogenous oxidants to potentially initiate, propagate and/or maintain inflammation associated with many human diseases through NF-κB activation. First, we used HEK-Blue cells that are stably transfected with mouse toll-like receptor 4 (mTLR4) or mouse TLR2. These cells also express optimized secreted embryonic alkaline phosphatase (SEAP) reporter gene under the control of a promoter inducible by NF-κB transcription factor. These cells were challenged with their respective receptor-specific ligands, different pro-oxidants and/or inhibitors that act at different levels of the receptor signaling pathways. A neutralizing antibody directed against TLR4 inhibited responses to both TLR4-specific agonist and a prooxidant, which confirmed that both agents act through TLR4. We used the level of SEAP released into the culture media due to NF-κB activation as a measure of TLR4 or TLR2 stimulation. Pro-oxidants evoked increased release of SEAP from HEK-Blue mTLR4 cells at a much lower concentration compared with release from the HEK-Blue mTLR2 cells. Specific TLR4 signaling pathway inhibitors and oxidant scavengers (anti-oxidants) significantly attenuated oxidant-induced SEAP release by TLR4 stimulation. Furthermore, a novel pro-oxidant that decays to produce the same reactants as activated phagocytes induced inflammatory pain responses in the mouse orofacial region with increased TLR4 expression, and IL-1β and TNFα tissue levels. EUK-134, a synthetic serum-stable scavenger of oxidative species decreased these effects. Our data provide in vitro and related in vivo evidence that exogenous oxidants can induce and maintain inflammation by acting mainly through a TLR4-dependent pathway, with implications in many chronic human ailments.
Collapse
Affiliation(s)
- Rajendra Karki
- Division of Pharmacology and Toxicology, University of Missouri-Kansas City, Missouri, United States of America
| | - Orisa J. Igwe
- Division of Pharmacology and Toxicology, University of Missouri-Kansas City, Missouri, United States of America
| |
Collapse
|
74
|
Jayaraman P, Sada-Ovalle I, Nishimura T, Anderson AC, Kuchroo VK, Remold HG, Behar SM. IL-1β promotes antimicrobial immunity in macrophages by regulating TNFR signaling and caspase-3 activation. THE JOURNAL OF IMMUNOLOGY 2013; 190:4196-204. [PMID: 23487424 DOI: 10.4049/jimmunol.1202688] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In vivo control of Mycobacterium tuberculosis reflects the balance between host immunity and bacterial evasion strategies. Effector Th1 cells that mediate protective immunity by depriving the bacterium of its intracellular niche are regulated to prevent overexuberant inflammation. One key immunoregulatory molecule is Tim3. Although Tim3 is generally recognized to downregulate Th1 responses, we recently described that its interaction with Galectin-9 expressed by M. tuberculosis-infected macrophages stimulates IL-1β secretion, which is essential for survival in the mouse model. Why IL-1β is required for host resistance to M. tuberculosis infection is unknown. In this article, we show that IL-1β directly kills M. tuberculosis in murine and human macrophages and does so through the recruitment of other antimicrobial effector molecules. IL-1β directly augments TNF signaling in macrophages through the upregulation of TNF secretion and TNFR1 cell surface expression, and results in activation of caspase-3. Thus, IL-1β and downstream TNF production lead to caspase-dependent restriction of intracellular M. tuberculosis growth.
Collapse
Affiliation(s)
- Pushpa Jayaraman
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
75
|
Interleukin-19 mediates tissue damage in murine ischemic acute kidney injury. PLoS One 2013; 8:e56028. [PMID: 23468852 PMCID: PMC3582636 DOI: 10.1371/journal.pone.0056028] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 01/04/2013] [Indexed: 02/07/2023] Open
Abstract
Inflammation and renal tubular injury are major features of acute kidney injury (AKI). Many cytokines and chemokines are released from injured tubular cells and acts as proinflammatory mediators. However, the role of IL-19 in the pathogenesis of AKI is not defined yet. In bilateral renal ischemia/reperfusion injury (IRI)-induced and HgCl2-induced AKI animal models, real-time quantitative (RTQ)-PCR showed that the kidneys, livers, and lungs of AKI mice expressed significantly higher IL-19 and its receptors than did sham control mice. Immunohistochemical staining showed that IL-19 and its receptors were strongly stained in the kidney, liver, and lung tissue of AKI mice. In vitro, IL-19 upregulated MCP-1, TGF-β1, and IL-19, and induced mitochondria-dependent apoptosis in murine renal tubular epithelial M-1 cells. IL-19 upregulated TNF-α and IL-10 in cultured HepG2 cells, and it increased IL-1β and TNF-α expression in cultured A549 cells. In vivo, after renal IRI or a nephrotoxic dose of HgCl2 treatment, IL-20R1-deficient mice (the deficiency blocks IL-19 signaling) showed lower levels of blood urea nitrogen (BUN) in serum and less tubular damage than did wild-type mice. Therefore, we conclude that IL-19 mediates kidney, liver, and lung tissue damage in murine AKI and that blocking IL-19 signaling may provide a potent therapeutic strategy for treating AKI.
Collapse
|
76
|
Qin J, Shang L, Ping AS, Li J, Li XJ, Yu H, Magdalou J, Chen LB, Wang H. TNF/TNFR signal transduction pathway-mediated anti-apoptosis and anti-inflammatory effects of sodium ferulate on IL-1β-induced rat osteoarthritis chondrocytes in vitro. Arthritis Res Ther 2012; 14:R242. [PMID: 23134577 PMCID: PMC3674623 DOI: 10.1186/ar4085] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Accepted: 11/06/2012] [Indexed: 01/25/2023] Open
Abstract
Introduction Sodium ferulate (SF) is a natural component of traditional Chinese herbs. Our previous study shows that SF has a protective effect on osteoarthritis (OA). The objective of this study was to investigate the effect of SF on the TNF/TNF receptor (TNFR) signal transduction pathway of rat OA chondrocytes. Methods Primary rat articular chondrocytes were co-treated with IL-1β and SF. Chondrocyte apoptosis was assessed by fluorescein isothiocyanate-annexin V/propidium iodide assay. The PCR array was used to screen the expression of 84 key genes involved in apoptosis. The release of TNFα and prostaglandin E2 were analyzed by ELISA. Expressions of proteins were assessed by western blotting. The activity of NF-κB was determined by electrophoretic mobility shift assay (EMSA). Gene expression of inducible nitric oxide synthase (iNOS) was evaluated by real-time quantitative PCR. The nitric oxide content was measured with the Griess method. Results After treatment with SF, the apoptosis rate of chondrocytes significantly attenuated (P < 0.01). Results of the apoptosis PCR array suggested that mRNA expression of some core proteins in the TNF/TNFR pathway showed valuable regulation. The protein expressions of TNFα, TNFR-1, TNF receptor-associated death domain, caspase-8 and caspase-3 were prevented by SF in a concentration-dependent manner. SF also inhibited activities of caspase-8 and caspase-3 compared with the OA model control (P < 0.01). TNF receptor-associated factor-2 expression, phosphorylations of inhibitor of NF-κB kinase (IKK) subunits alpha and beta, and NF-κB inhibitor, alpha (IκBα) were all concentration-dependently suppressed by SF treatment. The results of EMSA showed that SF inhibited the activity of NF-κB. In addition, the expressions of cycloxygenase-2 and iNOS and the contents of prostaglandin E2 and NO were attenuated with the treatment of SF (P < 0.01). Conclusion SF has anti-apoptosis and anti-inflammatory effects on an OA model induced by IL-1β in vitro, which were due to inhibitory actions on the caspase-dependent apoptosis pathway and the IKK/NF-κB signal transduction pathway of the TNF/TNFR pathway.
Collapse
|
77
|
Horiba M, Qutna N, Gendapodi P, Agrawal S, Sapkota K, Abel P, Townley RG. Effect of IL-1β and TNF-α vs IL-13 on bronchial hyperresponsiveness, β2-adrenergic responses and cellularity of bronchial alveolar lavage fluid. ACTA ACUST UNITED AC 2012; 31:37-49. [PMID: 21951583 DOI: 10.1111/j.1474-8673.2011.00465.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
1 Levels of IL-13, IL-1β and TNF-α are increased in bronchial lavage fluid of asthmatics and induce certain significant features of bronchial asthma including airway hyper-responsiveness (AHR). In this study, we have investigated the effect of these cytokines in naïve mice and those sensitized to ovalbumin (OVA) on bronchoconstrictions to methacholine (MCh) and the functional antagonism induced by β2 -adrenoceptor agonism. 2 Naïve or OVA-sensitized mice were treated for 3 days with IL-1β (250 U), TNF-α (150 ng), IL-13 (5 μg) or combinations of IL-1β with TNF-α or IL-1β with IL-13. MCh-induced bronchoconstriction and its sensitivity to albuterol, a β2-adrenoceptor agonist, was assessed 24 h after the last cytokine administration. 3 In naïve mice, responsiveness to MCh was significantly increased by the combination of IL-1β and TNF-α, IL-13 alone or in combination with IL-1β, but not by treatment with IL-1β or TNF-α alone. Similar results were obtained in OVA-sensitized mice except that treatment with IL-13 alone did not increase sensitivity to MCh. 4 In naïve mice, albuterol sensitivity was only significantly attenuated by treatment with IL-1β and TNF-α in combination. In mice sensitized to OVA, albuterol sensitivity was significantly attenuated by treatment with TNF-α, IL-13 or IL-13 in combination with IL-1β. 5 Inflammatory cell influx was increased by all cytokines and combinations except IL-13 in OVA-sensitized mice. 6 Our data do not support a link between inflammatory cell influx and AHR. In addition, the mechanism of IL-13-induced AHR might involve decreased β2-adrenoceptor responsiveness.
Collapse
Affiliation(s)
- M Horiba
- Division of Asthma, Allergy and Immunology, Creighton University School of Medicine, Omaha, NE, USA
| | | | | | | | | | | | | |
Collapse
|
78
|
Bastarache JA, Sebag SC, Grove BS, Ware LB. Interferon-γ and tumor necrosis factor-α act synergistically to up-regulate tissue factor in alveolar epithelial cells. Exp Lung Res 2012; 37:509-17. [PMID: 21913843 DOI: 10.3109/01902148.2011.605512] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Fibrin deposition mediated through activation of tissue factor (TF) in the airspace is central to the pathogenesis of acute lung injury. Defining the mechanisms of TF regulation in the lung is critical to understanding pulmonary fibrin formation. Tumor necrosis factor-α (TNF-α) up-regulates TF in the injured lung, and there is emerging evidence that another cytokine, interferon-γ (IFN-γ), also modulates expression. The effects of TNF-α and IFN-γ on regulation of TF were studied in alveolar epithelial A549 cells. In addition, potential mechanisms of modulation of TF expression by the 2 cytokines were analyzed with the hypothesis that IFN-γ acts synergistically with TNF-α to up-regulate alveolar epithelial TF through modulation of TNF receptor (TNFR) expression. TNF-α but not IFN-γ treatment increased TF mRNA, protein, and cell surface TF activity. The combination of IFN-γ and TNF-α treatment augmented the effects of TNF-α on TF up-regulation and also increased release of procoagulant microparticles (MPs) from A549 cells. IFN-γ modulated expression of both TNF-α receptors. Studies utilizing neutralizing antibodies against the two TNF receptors showed that the TF effects were mediated primarily through augmentation of TNFR1-dependent cellular responses. These findings have important implications for regulation of fibrin formation in the lung in the setting of acute inflammation.
Collapse
Affiliation(s)
- Julie A Bastarache
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| | | | | | | |
Collapse
|
79
|
Blinded, controlled field trial of two commercially available Mycoplasma bovis bacterin vaccines in veal calves. Vaccine 2011; 29:5347-54. [DOI: 10.1016/j.vaccine.2011.05.092] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 05/12/2011] [Accepted: 05/25/2011] [Indexed: 11/19/2022]
|
80
|
The effects of interleukin-1beta in tumor necrosis factor-alpha-induced acute pulmonary inflammation in mice. Mediators Inflamm 2009; 2009:958658. [PMID: 19901996 PMCID: PMC2773377 DOI: 10.1155/2009/958658] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 07/08/2009] [Accepted: 08/12/2009] [Indexed: 12/21/2022] Open
Abstract
We determined the role of interleukin-1β (IL-1β) signaling on tumor necrosis factor alpha-induced (TNF-α) lung neutrophil influx as well as neutrophil chemoattractant macrophage inflammatory protein (MIP-2) and KC and soluble TNF-α receptor (TNFR) levels utilizing wildtype (WT), TNF receptor double knockout (TNFR1/TNFR2 KO), and IL-1β KO mice after oropharyngeal instillation with TNF-α. A significant increase in neutrophil accumulation in bronchoalveolar lavage fluid (BALF) and lung interstitium was detected in the WT mice six hours after TNF-α exposure. This correlated with an increase in BALF MIP-2. In contrast, BALF neutrophil numbers were not increased by TNF-α treatment of IL-1β KOs, correlating with a failure to induce BALF MIP-2 and a trend toward increased BALF soluble TNFR1. TNF-α-instillation increased lavage and serum KC and soluble TNFR2 irrespective of IL-1β expression. These results suggest IL-1β contributes, in part, to TNF-α-mediated, chemokine release, and neutrophil recruitment to the lung, potentially associated with altered soluble TNFR1 release into the BALF.
Collapse
|