101
|
Xiao K, Zou WH, Yang Z, Rehman ZU, Ansari AR, Yuan HR, Zhou Y, Cui L, Peng KM, Song H. The role of visfatin on the regulation of inflammation and apoptosis in the spleen of LPS-treated rats. Cell Tissue Res 2014; 359:605-618. [PMID: 25358398 DOI: 10.1007/s00441-014-1997-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 08/21/2014] [Indexed: 01/01/2023]
Abstract
The purpose of the present study is to determine if visfatin is involved in inflammation or apoptosis induced by LPS in rat. Forty Wistar rats were divided into four groups: saline group, LPS group, visfatin group and Visfatin + LPS co-stimulated group. Spleen samples from each group of rats were collected for study. The spleen structure was examined by histological imaging. Apoptosis was evaluated with TUNEL reaction. Caspase-3 was detected with immunohistochemistry and western blot. The apoptosis-related genes were detected by qPCR and inflammatory cytokines were tested by ELISA. Our main findings were as follows. (1) Macrophages were markedly increased in the visfatin group compared with the saline group. This finding was confirmed when spleen samples were examined with western blot using CD68 antibody. (2) Visfatin promoted the expression of CD68 and caspase-3 in rat spleen, whereas visfatin could inhibit the expression of CD68 and activated caspase-3 in spleen of LPS-induced acute inflammation. (3) Visfatin had a pro-apoptotic effect on normal rat spleen, whereas it exerted an anti-apoptotic effect during LPS-induced lymphocytes apoptosis in rat spleen. Moreover, the effect of visfatin on cell apoptosis was mediated by the mitochondrial pathway. (4) Visfatin could modulate both the anti-inflammatory cytokines and pro-inflammatory cytokines in rat spleen, such as IL-10, IL-4, IL-6, TNF-α and IL-1β. Taken together, we demonstrate that visfatin could participate in the inflammatory process in rat spleen by modulating the macrophages and inflammatory cytokines. Also, visfatin plays a dual role in the apoptosis in rat spleen, which is mediated by the mitochondrial pathway.
Collapse
Affiliation(s)
- Ke Xiao
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wei-Hua Zou
- College of Plant Science & Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhi Yang
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zia Ur Rehman
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Abdur Rahman Ansari
- College of Veterinary and Animal Sciences, Jhang University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Huai-Rui Yuan
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ying Zhou
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lu Cui
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ke-Mei Peng
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hui Song
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
102
|
Paek J, Kang JH, Kim HS, Lee I, Seo KW, Yang MP. Serum adipokine concentrations in dogs with acute pancreatitis. J Vet Intern Med 2014; 28:1760-9. [PMID: 25312217 PMCID: PMC4895644 DOI: 10.1111/jvim.12437] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 06/11/2014] [Accepted: 07/23/2014] [Indexed: 01/15/2023] Open
Abstract
Background Limited information is available about the role of adipokines in the development and progression of acute pancreatitis (AP) in dogs. Objectives To determine whether the circulating concentrations of adipokines differed between healthy dogs and dogs with AP, and whether the circulating concentrations differed between AP survivors and AP nonsurvivors. Animals Twenty‐eight healthy dogs and 25 client‐owned dogs with AP. Methods Prospective observational cohort study of 25 client‐owned dogs with newly diagnosed AP and 28 otherwise healthy dogs with similar body condition scores. The serum concentrations of leptin, adiponectin, resistin, visfatin, interleukin (IL)‐1β, IL‐6, IL‐10, IL‐18, and tumor necrosis factor (TNF)‐α were measured. Results The serum concentrations of leptin (P = .0021), resistin (P = .0010), visfatin (P < .0001), IL‐1β (P < .0001), IL‐6 (P = .0002), IL‐10 (P < .0001), and IL‐18 (P < .0001) were significantly higher in dogs with AP than healthy dogs, whereas the adiponectin concentration (P = .0011) was significantly lower. There were significant differences in the serum concentrations of leptin (P = .028) and adiponectin (P = .046) in survivors and nonsurvivors. After the disappearance of clinical signs, the concentrations of resistin (P = .037) and IL‐1β (P = .027) decreased significantly, whereas the serum concentrations of leptin (P > .999), adiponectin (P = .11), visfatin (P = .83), IL‐6 (P = .82), IL‐10 (P = .82), IL‐18 (P = .56), and TNF‐α (P = .94) did not differ significantly. Conclusion and Clinical Importance This study showed that dysregulation of adipokines might be involved in the pathogenesis of AP. In addition, leptin and adiponectin are likely to be associated with mortality rate in AP.
Collapse
Affiliation(s)
- J Paek
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Korea
| | | | | | | | | | | |
Collapse
|
103
|
Moreno-Vinasco L, Quijada H, Sammani S, Siegler J, Letsiou E, Deaton R, Saadat L, Zaidi RS, Messana J, Gann PH, Machado RF, Ma W, Camp SM, Wang T, Garcia JGN. Nicotinamide phosphoribosyltransferase inhibitor is a novel therapeutic candidate in murine models of inflammatory lung injury. Am J Respir Cell Mol Biol 2014; 51:223-8. [PMID: 24588101 DOI: 10.1165/rcmb.2012-0519oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We previously identified the intracellular nicotinamide phosphoribosyltransferase (iNAMPT, aka pre-B-cell colony enhancing factor) as a candidate gene promoting acute respiratory distress syndrome (ARDS) and ventilator-induced lung injury (VILI) with circulating nicotinamide phosphoribosyltransferase potently inducing NF-κB signaling in lung endothelium. iNAMPT also synthesizes intracellular nicotinamide adenine dinucleotide (iNAD) in response to extracellular oxidative stress, contributing to the inhibition of apoptosis via ill-defined mechanisms. We now further define the role of iNAMPT activity in the pathogenesis of ARDS/VILI using the selective iNAMPT inhibitor FK-866. C57/B6 mice were exposed to VILI (40 ml/kg, 4 h) or LPS (1.5 mg/kg, 18 h) after osmotic pump delivery of FK-866 (100 mg/kg/d, intraperitoneally). Assessment of total bronchoalveolar lavage (BAL) protein, polymorphonuclear neutrophil (PMN) levels, cytokine levels (TNF-α, IL-6, IL-1α), lung iNAD levels, and injury scores revealed that FK-866-mediated iNAMPT inhibition successfully reduced lung tissue iNAD levels, BAL injury indices, inflammatory cell infiltration, and lung injury scores in LPS- and VILI-exposed mice. FK-866 further increased lung PMN apoptosis, as reflected by caspase-3 activation in BAL PMNs. These findings support iNAMPT inhibition via FK-866 as a novel therapeutic agent for ARDS via enhanced apoptosis in inflammatory PMNs.
Collapse
|
104
|
Meyer NJ. Beyond single-nucleotide polymorphisms: genetics, genomics, and other 'omic approaches to acute respiratory distress syndrome. Clin Chest Med 2014; 35:673-84. [PMID: 25453417 DOI: 10.1016/j.ccm.2014.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This article summarizes the contributions of high-throughput genomic, proteomic, metabolomic, and gene expression investigations to the understanding of inherited or acquired risk for acute respiratory distress syndrome (ARDS). Although not yet widely applied to a complex trait like ARDS, these techniques are now routinely used to study a variety of disease states. Omic applications hold great promise for identifying novel factors that may contribute to ARDS pathophysiology or may be appropriate for further development as biomarkers or surrogates in clinical studies. Opportunities and challenges of different techniques are discussed, and examples of successful applications in non-ARDS fields are used to illustrate the potential use of each technique.
Collapse
Affiliation(s)
- Nuala J Meyer
- Pulmonary, Allergy, and Critical Care Medicine, University of Pennsylvania, Perelman School of Medicine, 3600 Spruce Street, 5039 Maloney Building, Philadelphia, PA 19104, USA.
| |
Collapse
|
105
|
Lipocalin-2 test in distinguishing acute lung injury cases from septic mice without acute lung injury. ACTA ACUST UNITED AC 2014; 29:65-77. [PMID: 24998227 DOI: 10.1016/s1001-9294(14)60031-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To explore whether the amount of lipocalin-2 in the biofluid could reflect the onset of sepsis-induced acute lung injury (ALI) in mice. METHODS Lipopolysaccharide (LPS, 10 mg/kg) injection or cecal ligation and puncture (CLP) was performed to induce severe sepsis and ALI in C57 BL/6 male mice randomly divided into 5 groups (n=10 in each group): group A (intraperitoneal LPS injection), group B (intravenous LPS injection via tail vein), group C (CLP with 25% of the cecum ligated), group D (CLP with 75% of the cecum ligated), and the control group (6 sham-operation controls plus 4 saline controls). All the mice received volume resuscitation. Measurements of pulmonary morphological and functional alterations were used to identify the presence of experimental ALI. The expressions of lipocalin-2 and interleukin (IL)-6 in serum, bronchoalveolar lavage fluid (BALF), and lung tissue were quantified at both protein and mRNA levels. The overall abilities of lipocalin-2 and IL-6 tests to diagnose sepsis-induced ALI were evaluated by generating receiver operator characteristic curves (ROC) and computing area under curve (AUC). RESULTS In both group B and group D, most of the main features of experimental ALI were reproduced in mice, while group A and group C showed septic syndrome without definite evidence for the presence of ALI. Compared with septic mice without ALI (group A+group C), lipocalin-2 protein expression in septic mice with ALI (group B+group D) was significantly up-regulated in BALF (P<0.01) and in serum (P<0.01), and mRNA expression boosted in lung tissues (all P<0.05). Lipocalin-2 tests performed better than IL-6 tests in recognizing sepsis-induced ALI cases, evidenced by the larger AUC of the former (BALF tests, 0.8800 versus 0.6625; serum tests, 0.8500 versus 0.7000). Using a dual cutoff system to diagnose sepsis-induced ALI, BALF lipocalin-2 test exhibited the highest positive likelihood ratio (13.000) and the lowest negative likelihood ratio (0.077) among the tests of lipocalin-2 and IL-6 in blood and BALF. A statistically significant correlation was found between lipocalin-2 concentration in BALF and that in serum (Spearman r=0.8803, P<0.0001). CONCLUSIONS Lipocalin-2 expression is significantly up-regulated in septic ALI mice compared with those without ALI. Lipocalin-2 tests with a dual cutoff system could be an effective tool in distinguishing experimental ALI cases.
Collapse
|
106
|
Zhou T, Wang T, Garcia JGN. Expression of nicotinamide phosphoribosyltransferase-influenced genes predicts recurrence-free survival in lung and breast cancers. Sci Rep 2014; 4:6107. [PMID: 25146220 PMCID: PMC4141256 DOI: 10.1038/srep06107] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 07/30/2014] [Indexed: 01/16/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is a rate-limiting enzyme in the salvage pathway of nicotinamide adenine dinucleotide biosynthesis. NAMPT protein is a secreted plasma biomarker in inflammation and in cancer. The NAMPT enzymatic inhibitor, FK866, acts as an inducer of apoptosis and is a cancer therapeutic candidate, however, little is known regarding the influence of NAMPT on cancer biological mechanisms or on the prognosis of human cancers. We interrogated known microarray data sets to define NAMPT knockdown-influenced gene expression to demonstrate that reduced NAMPT expression strongly dysregulates cancer biology signaling pathways. Comparisons of gene expression datasets of four cancer types generated a N39 molecular signature exhibiting consistent dysregulated expression in multiple cancer tissues. The N39 signature provides a significant and independent prognostic tool of human recurrence-free survival in lung and breast cancers. Despite the absence of clear elucidation of molecular mechanisms, this study validates NAMPT as a novel “oncogene” with a central role in carcinogenesis. Furthermore, the N39 signature provides a potentially useful tool for prediction of recurrence-free survival in lung and breast cancer and validates NAMPT as a novel and effective therapeutic target in cancer.
Collapse
Affiliation(s)
- Tong Zhou
- 1] Arizona Respiratory Center and Department of Medicine, The University of Arizona, Tucson, Arizona, USA [2]
| | - Ting Wang
- 1] Arizona Respiratory Center and Department of Medicine, The University of Arizona, Tucson, Arizona, USA [2]
| | - Joe G N Garcia
- Arizona Respiratory Center and Department of Medicine, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
107
|
Mitra S, Wade MS, Sun X, Moldobaeva N, Flores C, Ma SF, Zhang W, Garcia JGN, Jacobson JR. GADD45a promoter regulation by a functional genetic variant associated with acute lung injury. PLoS One 2014; 9:e100169. [PMID: 24940746 PMCID: PMC4062486 DOI: 10.1371/journal.pone.0100169] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 05/22/2014] [Indexed: 01/28/2023] Open
Abstract
Rationale Growth arrest DNA damage inducible alpha (GADD45a) is a stress-induced gene we have shown to participate in the pathophysiology of ventilator-induced lung injury (VILI) via regulation of mechanical stress-induced Akt ubiquitination and phosphorylation. The regulation of GADD45a expression by mechanical stress and its relationship with acute lung injury (ALI) susceptibility and severity, however, remains unknown. Objectives We examined mechanical stress-dependent regulatory elements (MSRE) in the GADD45a promoter and the contribution of promoter polymorphisms in GADD45a expression and ALI susceptibility. Methods and Results Initial studies in GADD45a knockout and heterozygous mice confirmed the relationship of GADD45a gene dose to VILI severity. Human lung endothelial cells (EC) transfected with a luciferase vector containing the full length GADD45a promoter sequence (−771 to +223) demonstrated a >4 fold increase in GADD45a expression in response to 18% cyclic stretch (CS, 4 h) compared to static controls while specific promoter regions harboring CS-dependent MSRE were identified using vectors containing serial deletion constructs of the GADD45a promoter. In silico analyses of GADD45a promoter region (−371 to −133) revealed a potential binding site for specificity protein 1 (SP1), a finding supported by confirmed SP1 binding with the GADD45a promoter and by the significant attenuation of CS-dependent GADD45a promoter activity in response to SP1 silencing. Separately, case-control association studies revealed a significant association of a GADD45a promoter SNP at −589 (rs581000, G>C) with reduced ALI susceptibility. Subsequently, we found allelic variation of this SNP is associated with both differential GADD45a expression in mechanically stressed EC (18% CS, 4 h) and differential binding site of interferon regulatory factor 7 (IRF7) at this site. Conclusion These results strongly support a functional role for GADD45a in ALI/VILI and identify a specific gene variant that confers risk for ALI.
Collapse
Affiliation(s)
- Sumegha Mitra
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Michael S. Wade
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Xiaoguang Sun
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Nurgul Moldobaeva
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Carlos Flores
- Centro de Investigacion Biomedica en red Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Nuestra Senora de Candelaria, Tenerife, Spain
| | - Shwu-Fan Ma
- Division of Pulmonary, Critical Care and Sleep, University of Chicago, Chicago, Illinois, United States of America
| | - Wei Zhang
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Joe G. N. Garcia
- Arizona Health Sciences Center, University of Arizona, Tucson, Arizona, United States of America
| | - Jeffrey R. Jacobson
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
108
|
Leivo-Korpela S, Lehtimäki L, Hämälainen M, Vuolteenaho K, Kööbi L, Järvenpää R, Kankaanranta H, Saarelainen S, Moilanen E. Adipokines NUCB2/nesfatin-1 and visfatin as novel inflammatory factors in chronic obstructive pulmonary disease. Mediators Inflamm 2014; 2014:232167. [PMID: 24891763 PMCID: PMC4033393 DOI: 10.1155/2014/232167] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/22/2014] [Accepted: 04/22/2014] [Indexed: 12/23/2022] Open
Abstract
COPD (chronic obstructive pulmonary disease) is a common lung disease characterized by airflow limitation and systemic inflammation. Recently, adipose tissue mediated inflammation has gathered increasing interest in the pathogenesis of the disease. In this study, we investigated the role of novel adipocytokines nesfatin-1 and visfatin in COPD by measuring if they are associated with the inflammatory activity, lung function, or symptoms. Plasma levels of NUCB2/nesfatin-1 and visfatin were measured together with IL-6, IL-8, TNF- α , and MMP-9, lung function, exhaled nitric oxide, and symptoms in 43 male patients with emphysematous COPD. The measurements were repeated in a subgroup of the patients after four weeks' treatment with inhaled fluticasone. Both visfatin and NUCB2/nesfatin-1 correlated positively with plasma levels of IL-6 (r = 0.341, P = 0.027 and rho = 0.401, P = 0.008, resp.) and TNF- α (r = 0.305, P = 0.052 and rho = 0.329, P = 0.033, resp.) and NUCB2/nesfatin-1 also with IL-8 (rho = 0.321, P = 0.036) in patients with COPD. Further, the plasma levels of visfatin correlated negatively with pulmonary diffusing capacity (r = -0.369, P = 0.016). Neither of the adipokines was affected by fluticasone treatment and they were not related to steroid-responsiveness. The present results introduce adipocytokines NUCB2/nesfatin-1 and visfatin as novel factors associated with systemic inflammation in COPD and suggest that visfatin may mediate impaired pulmonary diffusing capacity.
Collapse
Affiliation(s)
- Sirpa Leivo-Korpela
- Department of Respiratory Medicine, Tampere University Hospital, PL 2000, 33521 Tampere, Finland
- The Immunopharmacology Research Group, School of Medicine, University of Tampere and Tampere University Hospital, 33014 Tampere, Finland
| | - Lauri Lehtimäki
- The Immunopharmacology Research Group, School of Medicine, University of Tampere and Tampere University Hospital, 33014 Tampere, Finland
- Allergy Centre, Tampere University Hospital, PL 2000, 33521 Tampere, Finland
| | - Mari Hämälainen
- The Immunopharmacology Research Group, School of Medicine, University of Tampere and Tampere University Hospital, 33014 Tampere, Finland
| | - Katriina Vuolteenaho
- The Immunopharmacology Research Group, School of Medicine, University of Tampere and Tampere University Hospital, 33014 Tampere, Finland
| | - Lea Kööbi
- Medical Imaging Centre, Tampere University Hospital, PL 2000, 33521 Tampere, Finland
| | - Ritva Järvenpää
- Medical Imaging Centre, Tampere University Hospital, PL 2000, 33521 Tampere, Finland
| | - Hannu Kankaanranta
- Department of Respiratory Medicine, Seinäjoki Central Hospital, 60220 Seinäjoki, Finland
| | - Seppo Saarelainen
- Department of Respiratory Medicine, Tampere University Hospital, PL 2000, 33521 Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, School of Medicine, University of Tampere and Tampere University Hospital, 33014 Tampere, Finland
| |
Collapse
|
109
|
Matsuda A, Yang WL, Jacob A, Aziz M, Matsuo S, Matsutani T, Uchida E, Wang P. FK866, a visfatin inhibitor, protects against acute lung injury after intestinal ischemia-reperfusion in mice via NF-κB pathway. Ann Surg 2014; 259:1007-17. [PMID: 24169192 DOI: 10.1097/sla.0000000000000329] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To determine whether administration of FK866, a competitive inhibitor of visfatin, attenuates acute lung injury induced by intestinal ischemia-reperfusion (I/R). BACKGROUND Acute lung injury, a frequent complication of intestinal I/R, is an inflammatory disorder of the lung, which is characterized by an overproduction of proinflammatory cytokines and increased permeability of the alveolar-capillary barrier, resulting in multiple organ dysfunction. Therefore, the development of novel and effective therapies for intestinal I/R is critical for the improvement of patient outcome. Visfatin, a 54-kDa secretory protein, is known as a proinflammatory cytokine and plays a deleterious role in inflammatory diseases. METHODS Male C57BL/6J mice were subjected to intestinal I/R induced by occlusion of the superior mesenteric artery for 90 minutes, followed by reperfusion. During reperfusion period, mice were treated with vehicle or FK866 (10 mg/kg of body weight) by an intraperitoneal injection. The levels of visfatin, proinflammatory mediators, and other markers were assessed 4 hours after reperfusion. In addition, survival study was conducted in intestinal I/R mice with or without FK866 treatment. RESULTS Plasma and lung visfatin protein levels were significantly increased after intestinal I/R. FK866 treatment significantly attenuated intestinal and lung injury by inhibiting proinflammatory cytokine production, cellular apoptosis, and NF-κB activation, hence improving survival rate. In vitro studies showed that macrophages treated with lipopolysaccharides upregulated visfatin expression, whereas FK866 inhibited proinflammatory cytokine production via modulation of the NF-κB pathway. CONCLUSIONS Collectively, these findings implicate FK866 as a novel therapeutic compound for intestinal I/R-induced attenuates acute lung injury via modulation of innate immune functions.
Collapse
Affiliation(s)
- Akihisa Matsuda
- *Department of Surgery, Hofstra North Shore-LIJ School of Medicine and Center for Translational Research, The Feinstein Institute for Medical Research, Manhasset, NY †Department of Surgery, Nippon Medical School, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
110
|
Adyshev DM, Elangovan VR, Moldobaeva N, Mapes B, Sun X, Garcia JGN. Mechanical stress induces pre-B-cell colony-enhancing factor/NAMPT expression via epigenetic regulation by miR-374a and miR-568 in human lung endothelium. Am J Respir Cell Mol Biol 2014; 50:409-18. [PMID: 24053186 DOI: 10.1165/rcmb.2013-0292oc] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Increased lung vascular permeability and alveolar edema are cardinal features of inflammatory conditions such as acute respiratory distress syndrome (ARDS) and ventilator-induced lung injury (VILI). We previously demonstrated that pre-B-cell colony-enhancing factor (PBEF)/NAMPT, the proinflammatory cytokine encoded by NAMPT, participates in ARDS and VILI inflammatory syndromes. The present study evaluated posttranscriptional regulation of PBEF/NAMPT gene expression in human lung endothelium via 3'-untranslated region (UTR) microRNA (miRNA) binding. In silico analysis identified hsa-miR-374a and hsa-miR-568 as potential miRNA candidates. Increased PBEF/NAMPT transcription (by RT-PCR) and expression (by Western blotting) induced by 18% cyclic stretch (CS) (2 h: 3.4 ± 0.06 mRNA fold increase (FI); 10 h: 1.5 ± 0.06 protein FI) and by LPS (4 h: 3.8 ± 0.2 mRNA FI; 48 h: 2.6 ± 0.2 protein FI) were significantly attenuated by transfection with mimics of hsa-miR-374a or hsa-miR-568 (40-60% reductions each). LPS and 18% CS increased the activity of a PBEF/NAMPT 3'-UTR luciferase reporter (2.4-3.25 FI) with induction reduced by mimics of each miRNA (44-60% reduction). Specific miRNA inhibitors (antagomirs) for each PBEF/NAMPT miRNA significantly increased the endogenous PBEF/NAMPT mRNA (1.4-3.4 ± 0.1 FI) and protein levels (1.2-1.4 ± 0.1 FI) and 3'-UTR luciferase activity (1.4-1.7 ± 0.1 FI) compared with negative antagomir controls. Collectively, these data demonstrate that increased PBEF/NAMPT expression induced by bioactive agonists (i.e., excessive mechanical stress, LPS) involves epigenetic regulation with hsa-miR-374a and hsa-miR-568, representing novel therapeutic strategies to reduce inflammatory lung injury.
Collapse
Affiliation(s)
- Djanybek M Adyshev
- Institute for Personalized Respiratory Medicine, Department of Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | | | | | | | | | | |
Collapse
|
111
|
Visfatin concentrations in children with leukemia before and after stem cell transplantation. Exp Hematol 2014; 42:252-60. [DOI: 10.1016/j.exphem.2013.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/16/2013] [Accepted: 12/16/2013] [Indexed: 11/23/2022]
|
112
|
Ooi SQ, Chan RME, Poh LKS, Loke KY, Heng CK, Chan YH, Gan SU, Lee KO, Lee YS. Visfatin and its genetic variants are associated with obesity-related morbidities and cardiometabolic risk in severely obese children. Pediatr Obes 2014; 9:81-91. [PMID: 23447513 DOI: 10.1111/j.2047-6310.2013.00149.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Revised: 12/28/2012] [Accepted: 01/10/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND Visfatin is an adipokine, associated with obesity and possibly glucose regulation. OBJECTIVE The aim of this study was to examine the association of visfatin and its genetic variants with adiposity, cardiometabolic risk factors and obesity-related morbidities in obese children. METHODS Anthropometric measurements, dual energy X-ray absorptiometry scan, fasting blood samples and oral glucose tolerance tests were performed for 243 obese children. We screened the visfatin gene of 24 obese subjects and then performed genotyping of identified genetic variants in other 219 obese children through direct DNA sequencing. RESULTS Fasting serum visfatin correlated with measures of obesity and liver enzymes and was elevated in obese children with abnormal glucose tolerance and non-alcoholic fatty liver disease. The two upstream single nucleotide polymorphisms, -3187G>A (rs11977021) and -1537C>T (rs61330082), were at complete linkage disequilibrium. The AA genotype of -3187G>A was associated with higher serum visfatin (6.17 ± 0.76 ng mL(-1) vs. 3.92 ± 0.44 ng mL(-1)) and higher triglyceride (1.39 ± 0.08 mmol L(-1) vs. 1.19 ± 0.07 mmol L(-1)) as compared with the GG genotype. There was also a significant linear increase in serum visfatin across GG to GA to AA genotype of -3187G>A, indicating possible additive effect of A allele. The dominant GA + AA genotype model of +21426G>A (rs2302559) was associated with lower serum visfatin (3.83 ± 0.56 ng mL(-1) vs. 5.13 ± 0.34 ng mL(-1)) and lower plasma glucose (4.37 ± 0.08 mmol L(-1) vs. 4.77 ± 0.12 mmol L(-1)) as compared with the GG genotype. CONCLUSION Visfatin and its genetic variants were associated with adiposity, obesity-related morbidities and adverse cardiometabolic parameters. This supported our hypothesis that visfatin plays a significant role in the development of obesity-related morbidities and cardiometabolic risk.
Collapse
Affiliation(s)
- S Q Ooi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Enli Y, Balci YI, Gönen C, Uzun E, Polat A. Adipocytokine concentrations in children with different types of beta-thalassemia. Scandinavian Journal of Clinical and Laboratory Investigation 2014; 74:306-11. [DOI: 10.3109/00365513.2014.883639] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
114
|
Kruis T, Batra A, Siegmund B. Bacterial translocation - impact on the adipocyte compartment. Front Immunol 2014; 4:510. [PMID: 24432024 PMCID: PMC3881001 DOI: 10.3389/fimmu.2013.00510] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/24/2013] [Indexed: 12/29/2022] Open
Abstract
Over the last decade it became broadly recognized that adipokines and thus the fat tissue compartment exert a regulatory function on the immune system. Our own group described the pro-inflammatory function of the adipokine leptin within intestinal inflammation in a variety of animal models. Following-up on this initial work, the aim was to reveal stimuli and mechanisms involved in the activation of the fat tissue compartment and the subsequent release of adipokines and other mediators paralleled by the infiltration of immune cells. This review will summarize the current literature on the possible role of the mesenteric fat tissue in intestinal inflammation with a focus on Crohn’s disease (CD). CD is of particular interest in this context since the transmural intestinal inflammation has been associated with a characteristic hypertrophy of the mesenteric fat, a phenomenon called “creeping fat.” The review will address three consecutive questions: (i) What is inducing adipocyte activation, (ii) which factors are released after activation and what are the consequences for the local fat tissue compartment and infiltrating cells; (iii) do the answers generated before allow for an explanation of the role of the mesenteric fat tissue within intestinal inflammation? With this review we will provide a working model indicating a close interaction in between bacterial translocation, activation of the adipocytes, and subsequent direction of the infiltrating immune cells. In summary, the models system mesenteric fat indicates a unique way how adipocytes can directly interact with the immune system.
Collapse
Affiliation(s)
- Tassilo Kruis
- Department of Medicine I (Gastroenterology, Rheumatology, Infectious Diseases), Charité - Universitätsmedizin Berlin , Berlin , Germany
| | - Arvind Batra
- Department of Medicine I (Gastroenterology, Rheumatology, Infectious Diseases), Charité - Universitätsmedizin Berlin , Berlin , Germany
| | - Britta Siegmund
- Department of Medicine I (Gastroenterology, Rheumatology, Infectious Diseases), Charité - Universitätsmedizin Berlin , Berlin , Germany
| |
Collapse
|
115
|
Yun MR, Seo JM, Park HY. Visfatin contributes to the differentiation of monocytes into macrophages through the differential regulation of inflammatory cytokines in THP-1 cells. Cell Signal 2013; 26:705-15. [PMID: 24378536 DOI: 10.1016/j.cellsig.2013.12.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 12/11/2013] [Accepted: 12/22/2013] [Indexed: 12/31/2022]
Abstract
Visfatin is a novel multifunctional adipocytokine with inflammatory properties. Although a link between visfatin and atherosclerosis has recently been suggested, its actions in the development of atherosclerosis remain unknown. Therefore, we investigated a potential role and underlying mechanism(s) of visfatin in monocytes/macrophages differentiation, a critical early step in atherogenesis, using phorbol-12-myristate-13-acetate (PMA)-stimulated THP-1 cell models. The co-incubation of PMA with visfatin-induced CD36 expression with a concomitant increase in the phagocytosis of latex beads compared with PMA alone treatment. Moreover, visfatin markedly increased interleukin (IL)-1β secretion by enhancing IL-1β mRNA stability in a short-term incubation. Visfatin also significantly elevated the secretion of IL-6 as well as IL-1β in a longer incubation period, which was partially suppressed by nuclear factor-κB (NF-κB) inhibitor, BAY11-7082, and c-Jun-N-terminal kinase (JNK) inhibitor, SP600125. Furthermore, silencing IL-1β successfully blocked IL-6 secretion, CD36 expression, and NF-κB activation in response to visfatin. Collectively, these results suggest that visfatin enhances the IL-1β-dependent induction of IL-6 and CD36 via distinct signaling pathways mediated by JNK and NF-κB, respectively, and consequently, leading to the acceleration of monocytes/macrophages differentiation.
Collapse
Affiliation(s)
- Mi Ran Yun
- Division of Cardiovascular and Rare Diseases, Center for Biomedical Sciences, Korea National Institute of Health, Republic of Korea; JE-UK Laboratory of Molecular Cancer Therapeutics, Yonsei Cancer Research Institute, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Jeong Mi Seo
- Division of Cardiovascular and Rare Diseases, Center for Biomedical Sciences, Korea National Institute of Health, Republic of Korea
| | - Hyun Young Park
- Division of Cardiovascular and Rare Diseases, Center for Biomedical Sciences, Korea National Institute of Health, Republic of Korea.
| |
Collapse
|
116
|
Fox EJ, Leeder JS, Ye SQ, Becker ML. Decreased expression of nicotinamide phosphoribosyltransferase in patients with juvenile idiopathic arthritis receiving methotrexate. J Rheumatol 2013; 40:741-2. [PMID: 23637381 DOI: 10.3899/jrheum.120639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
117
|
Abstract
Mechanical ventilation (MV) is, by definition, the application of external forces to the lungs. Depending on their magnitude, these forces can cause a continuum of pathophysiological alterations ranging from the stimulation of inflammation to the disruption of cell-cell contacts and cell membranes. These side effects of MV are particularly relevant for patients with inhomogeneously injured lungs such as in acute lung injury (ALI). These patients require supraphysiological ventilation pressures to guarantee even the most modest gas exchange. In this situation, ventilation causes additional strain by overdistension of the yet non-injured region, and additional stress that forms because of the interdependence between intact and atelectatic areas. Cells are equipped with elaborate mechanotransduction machineries that respond to strain and stress by the activation of inflammation and repair mechanisms. Inflammation is the fundamental response of the host to external assaults, be they of mechanical or of microbial origin and can, if excessive, injure the parenchymal tissue leading to ALI. Here, we will discuss the forces generated by MV and how they may injure the lungs mechanically and through inflammation. We will give an overview of the mechanotransduction and how it leads to inflammation and review studies demonstrating that ventilator-induced lung injury can be prevented by blocking pathways of mechanotransduction or inflammation.
Collapse
Affiliation(s)
- Ulrike Uhlig
- Department of Pharmacology & Toxicology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | | |
Collapse
|
118
|
Procaccini C, De Rosa V, Galgani M, Carbone F, La Rocca C, Formisano L, Matarese G. Role of adipokines signaling in the modulation of T cells function. Front Immunol 2013; 4:332. [PMID: 24151494 PMCID: PMC3799205 DOI: 10.3389/fimmu.2013.00332] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/01/2013] [Indexed: 12/15/2022] Open
Abstract
The field that links immunity and metabolism is rapidly expanding. Apparently non-immunological disorders such as obesity and type 2 diabetes have been linked to immune dysregulation, suggesting that metabolic alterations can be induced by or be consequence of an altered self-immune tolerance. In this context, adipose tissue produces and releases a variety of pro-inflammatory and anti-inflammatory factors, termed "adipokines," which can be considered as the bridge between obesity-related exogenous factors, such as nutrition and lifestyle, and the molecular events leading to metabolic syndrome, inflammatory, and/or autoimmune conditions. In obesity, increased production of most adipokines impacts on multiple functions such as appetite and energy balance, modulation of immune responses, insulin sensitivity, angiogenesis, blood pressure, lipid metabolism, and so on. This report aims to discuss some of the recent topics of adipocytokine research and their related signaling pathways, that may be of particular importance as could lead to effective therapeutic strategies for obesity-associated diseases.
Collapse
Affiliation(s)
- Claudio Procaccini
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Salerno, Italy
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
- Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia, Roma, Italy
| | - Mario Galgani
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Fortunata Carbone
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Salerno, Italy
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Claudia La Rocca
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Salerno, Italy
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Luigi Formisano
- Department of Biological, Geological and Environmental Sciences, Division of Pharmacology, University of Sannio, Benevento, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Salerno, Italy
- IRCCS MultiMedica, Milano, Italy
| |
Collapse
|
119
|
Romacho T, Villalobos LA, Cercas E, Carraro R, Sánchez-Ferrer CF, Peiró C. Visfatin as a novel mediator released by inflamed human endothelial cells. PLoS One 2013; 8:e78283. [PMID: 24130902 PMCID: PMC3795064 DOI: 10.1371/journal.pone.0078283] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 09/11/2013] [Indexed: 11/25/2022] Open
Abstract
Background Visfatin is a multifaceted adipokine whose circulating levels are enhanced in different metabolic diseases. Extracellular visfatin can exert various deleterious effects on vascular cells, including inflammation and proliferation. Limited evidence exists, however, on the capacity of human vascular cells to synthesize and release visfatin by themselves, under basal or pro-inflammatory conditions. Methods and Results Intracellular visfatin was detected by Western blot in non-stimulated human umbilical vein endothelial cells (HUVEC). However, exposing HUVEC for 18 h to a series of pro-inflammatory stimulus, such as interleukin (IL)-1β (1 to 10 ng/mL), tumor necrosis factor-α (1 to 10 ng/mL) or angiotensin II (10 pmol/L to 1 μmol/L) markedly enhanced intracellular visfatin content. Using IL-1β (10 ng/mL; 18 h), it was determined that the increase in intracellular visfatin, which was paralleled by enhanced visfatin mRNA levels, relied on a signalling mechanism involving both nuclear factor-κB and poly (ADP ribose) polymerase-1 activation. Moreover, IL-1β modified the sub-cellular localization of visfatin; while in non-stimulated HUVEC immunoreactive visfatin predominantly showed an intra-nuclear granular pattern, in IL-1β-inflamed cells an extra-nuclear filamentous staining, co-localising with F-actin fibers and suggesting a secretory pattern, was mainly found. Indeed, IL-1β promoted visfatin secretion, as determined by both ELISA and immunocytochemistry. Conclusions Human endothelial cells synthesize and release visfatin, particularly in response to inflammation. We suggest that the inflamed endothelium can be a source of visfatin, which arises as a local inflammatory mediator and a potential therapeutic target to interfere with vascular inflammation.
Collapse
Affiliation(s)
- Tania Romacho
- Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital de La Princesa, Madrid, Spain
| | - Laura A. Villalobos
- Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital de La Princesa, Madrid, Spain
| | - Elena Cercas
- Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital de La Princesa, Madrid, Spain
| | - Raffaele Carraro
- Servicio de Endocrinología, Hospital Universitario de La Princesa and Departamento de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital de La Princesa, Madrid, Spain
| | - Carlos F. Sánchez-Ferrer
- Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital de La Princesa, Madrid, Spain
| | - Concepción Peiró
- Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital de La Princesa, Madrid, Spain
- * E-mail:
| |
Collapse
|
120
|
Garcia JGN, Sznajder JI. Healthcare Disparities in Patients with Acute Respiratory Distress Syndrome. Toward Equity. Am J Respir Crit Care Med 2013; 188:631-2. [DOI: 10.1164/rccm.201307-1394ed] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
121
|
Meyer NJ. Future clinical applications of genomics for acute respiratory distress syndrome. THE LANCET RESPIRATORY MEDICINE 2013; 1:793-803. [PMID: 24461759 DOI: 10.1016/s2213-2600(13)70134-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute respiratory distress syndrome remains a substantial cause of morbidity and mortality in intensive care units, yet no specific pharmacotherapy has proven useful in reducing the duration of mechanical ventilation or improving survival. One factor that might hamper the development of treatment for acute respiratory distress syndrome is the heterogeneous nature of the population who present with the syndrome. In this Review, the potential of genomic approaches-genetic association, gene expression, metabolomic, proteomic, and systems biology applications-for the identification of molecular endotypes within acute respiratory distress syndrome and potentially for the prediction, diagnosis, prognosis, and treatment of this difficult disorder are discussed.
Collapse
Affiliation(s)
- Nuala J Meyer
- Department of Medicine Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
122
|
Sun X, Ma SF, Wade MS, Acosta-Herrera M, Villar J, Pino-Yanes M, Zhou T, Liu B, Belvitch P, Moitra J, Han YJ, Machado R, Noth I, Natarajan V, Dudek SM, Jacobson JR, Flores C, Garcia JGN. Functional promoter variants in sphingosine 1-phosphate receptor 3 associate with susceptibility to sepsis-associated acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol 2013; 305:L467-77. [PMID: 23911438 DOI: 10.1152/ajplung.00010.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The genetic mechanisms underlying the susceptibility to acute respiratory distress syndrome (ARDS) are poorly understood. We previously demonstrated that sphingosine 1-phosphate (S1P) and the S1P receptor S1PR3 are intimately involved in lung inflammatory responses and vascular barrier regulation. Furthermore, plasma S1PR3 protein levels were shown to serve as a biomarker of severity in critically ill ARDS patients. This study explores the contribution of single nucleotide polymorphisms (SNPs) of the S1PR3 gene to sepsis-associated ARDS. S1PR3 SNPs were identified by sequencing the entire gene and tagging SNPs selected for case-control association analysis in African- and ED samples from Chicago, with independent replication in a European case-control study of Spanish individuals. Electrophoretic mobility shift assays, luciferase activity assays, and protein immunoassays were utilized to assess the functionality of associated SNPs. A total of 80 variants, including 29 novel SNPs, were identified. Because of limited sample size, conclusive findings could not be drawn in African-descent ARDS subjects; however, significant associations were found for two promoter SNPs (rs7022797 -1899T/G; rs11137480 -1785G/C), across two ED samples supporting the association of alleles -1899G and -1785C with decreased risk for sepsis-associated ARDS. In addition, these alleles significantly reduced transcription factor binding to the S1PR3 promoter; reduced S1PR3 promoter activity, a response particularly striking after TNF-α challenge; and were associated with lower plasma S1PR3 protein levels in ARDS patients. These highly functional studies support S1PR3 as a novel ARDS candidate gene and a potential target for individualized therapy.
Collapse
Affiliation(s)
- Xiaoguang Sun
- Institute for Personalize Respiratory Medicine, Univ. of Illinois at Chicago, 3099 COMRB (MC719 909 S. Wolcott Ave., Chicago, IL 60612.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Kolgazi M, Uslu U, Yuksel M, Velioglu-Ogunc A, Ercan F, Alican I. The role of cholinergic anti-inflammatory pathway in acetic acid-induced colonic inflammation in the rat. Chem Biol Interact 2013; 205:72-80. [PMID: 23810507 DOI: 10.1016/j.cbi.2013.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 05/23/2013] [Accepted: 06/11/2013] [Indexed: 12/23/2022]
Abstract
The "cholinergic anti-inflammatory pathway" provides neurological modulation of cytokine synthesis to limit the magnitude of the immune response. This study aimed to evaluate the impact of the cholinergic anti-inflammatory pathway on the extent of tissue integrity, oxidant-antioxidant status and neutrophil infiltration to the inflamed organ in a rat model of acetic acid-induced colitis. Colitis was induced by intrarectal administration of 5% acetic acid (1ml) to Sprague-Dawley rats (200-250g; n=7-8 per group). Control group received an equal volume of saline intrarectally. The rats were treated with either nicotine (1mg/kg/day) or huperzine A (0.1mg/kg/day) intraperitoneally for 3 days. After decapitation, the distal colon was scored macroscopically and microscopically. Tissue samples were used for the measurement of malondialdehyde (MDA) and glutathione (GSH) levels, and myeloperoxidase (MPO) activity. Formation of reactive oxygen species was monitored by using chemiluminescence (CL). Nuclear factor (NF)-κB expression was evaluated in colonic samples via immunohistochemical analysis. Trunk blood was collected for the assessment of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-10, resistin and visfatin levels. Both nicotine and huperzine A reduced the extent of colonic lesions, increased colonic MDA level, high MPO activity and NF-κB expression in the colitis group. Elevation of serum IL-1β level due to colitis was also attenuated by both treatments. Additionally, huperzine A was effective to reverse colitis-induced high lucigenin-enhanced CL values and serum TNF-α levels. Colitis group revealed decreased serum visfatin levels compared to control group which was completely reversed by nicotine. In conclusion, modulation of the cholinergic system either by nicotine or ACh esterase inhibition improved acetic acid-induced colonic inflammation as confirmed by macroscopic and microscopic examination and biochemical assays.
Collapse
Affiliation(s)
- Meltem Kolgazi
- Marmara University School of Medicine, Department of Physiology, Istanbul, Turkey
| | | | | | | | | | | |
Collapse
|
124
|
Sun Z, Lei H, Zhang Z. Pre-B cell colony enhancing factor (PBEF), a cytokine with multiple physiological functions. Cytokine Growth Factor Rev 2013; 24:433-42. [PMID: 23787158 DOI: 10.1016/j.cytogfr.2013.05.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 05/12/2013] [Accepted: 05/21/2013] [Indexed: 02/07/2023]
Abstract
Pre-B cell colony enhancing factor (PBEF) is regarded as a proinflammatory cytokine. Named for its first discovered function as a pre-B cell colony enhancing factor, it has since been found to have many other functions relating to cell metabolism, inflammation, and immune modulation. It has also been found to have intracellular and extracellular forms, with the two overlapping in function. Most of the intracellular functions of PBEF are due to its role as a nicotinamide phosphoribosyltransferase (Nampt). It has been found in human endothelial cells, where it is able to induce angiogenesis through upregulation of VEGF and VEGFR and secretion of MCP-1. In human umbilical endothelial cells, PBEF increases levels of the protease MMP 2/9. PBEF has also been found in a variety of immune cells other than B cells and has been shown to inhibit apoptosis of macrophages. Extracellular PBEF has been shown to increase inflammatory cytokines, such as TNF-α, IL-1β, IL-16, and TGF-β1, and the chemokine receptor CCR3. PBEF also increases the production of IL-6, TNF-α, and IL-1β in CD14(+) monocyctes, macrophages, and dendritic cells, enhances the effectiveness of T cells, and is vital to the development of both B and T lymphocytes. The purpose of this review is to summarize the recent advances in PBEF research.
Collapse
Affiliation(s)
- Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Cardiology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China.
| | | | | |
Collapse
|
125
|
Visfatin/Nampt: an adipokine with cardiovascular impact. Mediators Inflamm 2013; 2013:946427. [PMID: 23843684 PMCID: PMC3697395 DOI: 10.1155/2013/946427] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 05/20/2013] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue is acknowledged as an endocrine organ that releases bioactive factors termed adipokines. Visfatin was initially identified as a novel adipokine with insulin-mimetic properties in mice. This adipokine was identical to two previously described molecules, namely, pre-B cell colony-enhancing factor (PBEF) and the enzyme nicotinamide phosphoribosyltransferase (Nampt). Enhanced circulating visfatin/Nampt levels have been reported in metabolic diseases, such as obesity and type 2 diabetes. Moreover, visfatin/Nampt circulating levels correlate with markers of systemic inflammation. In cardiovascular diseases, visfatin/Nampt was initially proposed as a clinical marker of atherosclerosis, endothelial dysfunction, and vascular damage, with a potential prognostic value. Nevertheless, beyond being a surrogate clinical marker, visfatin/Nampt is an active player promoting vascular inflammation, and atherosclerosis. Visfatin/Nampt effects on cytokine and chemokine secretion, macrophage survival, leukocyte recruitment by endothelial cells, vascular smooth muscle inflammation and plaque destabilization make of this adipokine an active factor in the development and progression of atherosclerosis. Further research is required to fully understand the mechanisms mediating the cellular actions of this adipokine and to better characterize the factors regulating visfatin/Nampt expression and release in all these pathologic scenarios. Only then, we will be able to conclude whether visfatin/Nampt is a therapeutical target in cardiometabolic diseases.
Collapse
|
126
|
Lee K, Huh JW, Lim CM, Koh Y, Hong SB. Clinical role of serum pre-B cell colony-enhancing factor in ventilated patients with sepsis and acute respiratory distress syndrome. ACTA ACUST UNITED AC 2013; 45:760-5. [DOI: 10.3109/00365548.2013.797600] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
127
|
Hu W, Liu CW, Su J, Lu J, Zhu Y, Liu BW. Elevated plasma visfatin concentrations in patients with community-acquired pneumonia. Peptides 2013; 43:8-12. [PMID: 23466352 DOI: 10.1016/j.peptides.2013.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 02/21/2013] [Accepted: 02/21/2013] [Indexed: 01/18/2023]
Abstract
Visfatin has been associated with some inflammatory disease. This study aimed to compare plasma visfatin levels in patients with community-acquired pneumonia and healthy controls and to furthermore investigate the relationship between their concentrations and 30-day mortality in patients. Plasma visfatin concentrations were measured in 176 patients and 95 healthy controls. The admission visfatin levels were significantly increased in all patients, survivals and non-survivals with community-acquired pneumonia compared with healthy control individuals, associated with pneumonia severity index score, Acute Physiology and Chronic Health Evaluation II score, white blood cell count, and plasma C-reactive protein level, and identified as an independent predictor for 30-day mortality. Its predictive value was similar to those of pneumonia severity index score and Acute Physiology and Chronic Health Evaluation II score. However, visfatin did not statistically significantly improve the predictive values of pneumonia severity index score and Acute Physiology and Chronic Health Evaluation II score. Thus, higher plasma visfatin level correlates with disease severity and markers of system inflammation and represent a novel biomarker for predicting 30-day mortality in patients with community-acquired pneumonia.
Collapse
Affiliation(s)
- Wei Hu
- Department of Intensive Care Unit, The First People's Hospital of Hangzhou, Nanjing Medical University, 261 Huansha Road, Hangzhou 310006, China.
| | | | | | | | | | | |
Collapse
|
128
|
Meyer NJ, Feng R, Li M, Zhao Y, Sheu CC, Tejera P, Gallop R, Bellamy S, Rushefski M, Lanken PN, Aplenc R, O'Keefe GE, Wurfel MM, Christiani DC, Christie JD. IL1RN coding variant is associated with lower risk of acute respiratory distress syndrome and increased plasma IL-1 receptor antagonist. Am J Respir Crit Care Med 2013; 187:950-9. [PMID: 23449693 PMCID: PMC3707367 DOI: 10.1164/rccm.201208-1501oc] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 03/01/2013] [Indexed: 12/21/2022] Open
Abstract
RATIONALE Acute respiratory distress syndrome (ARDS) behaves as a complex genetic trait, yet knowledge of genetic susceptibility factors remains incomplete. OBJECTIVES To identify genetic risk variants for ARDS using large scale genotyping. METHODS A multistage genetic association study was conducted of three critically ill populations phenotyped for ARDS. Stage I, a trauma cohort study (n = 224), was genotyped with a 50K gene-centric single-nucleotide polymorphism (SNP) array. We tested SNPs associated with ARDS at P < 5 × 10(-4) for replication in stage II, a trauma case-control population (n = 778). SNPs replicating their association in stage II (P < 0.005) were tested in a stage III nested case-control population of mixed subjects in the intensive care unit (n = 2,063). Logistic regression was used to adjust for potential clinical confounders. We performed ELISA to test for an association between ARDS-associated genotype and plasma protein levels. MEASUREMENTS AND MAIN RESULTS A total of 12 SNPs met the stage I threshold for an association with ARDS. rs315952 in the IL1RN gene encoding IL-1 receptor antagonist (IL1RA) replicated its association with reduced ARDS risk in stages II (P < 0.004) and III (P < 0.02), and was robust to clinical adjustment (combined odds ratio = 0.81; P = 4.2 × 10(-5)). Plasma IL1RA level was associated with rs315952C in a subset of critically ill subjects. The effect of rs315952 was independent from the tandem repeat variant in IL1RN. CONCLUSIONS The IL1RN SNP rs315952C is associated with decreased risk of ARDS in three populations with heterogeneous ARDS risk factors, and with increased plasma IL1RA response. IL1RA may attenuate ARDS risk.
Collapse
Affiliation(s)
- Nuala J Meyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Weng JF, Chen J, Hong WC, Luo LF, Yu W, Luo SD. Plasma visfatin, associated with a genetic polymorphism -1535C>T, is correlated with C-reactive protein in Chinese Han patients with traumatic brain injury. Peptides 2013; 40:8-12. [PMID: 23270673 DOI: 10.1016/j.peptides.2012.12.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 12/14/2012] [Accepted: 12/14/2012] [Indexed: 01/08/2023]
Abstract
Visfatin is a newly identified pro-inflammatory adipokine and a genetic polymorphism -1535 C>T located in the visfatin gene promoter has been suggested to be associated with the regulation of visfatin expression in some inflammatory illness. However, there were some conflicting results regarding whether this variant is functional or not. This study aimed to examine the relations of the -1535 C>T single nucleotide polymorphism (SNP) of visfatin gene to the plasma visfatin and C-reactive protein concentrations in traumatic brain injury (TBI). 318 Chinese Han patients with TBI were recruited in this study. Plasma visfatin and C-reactive protein levels were significantly different between the genotypes in the SNP-1535 C>T even after adjustment for age, sex and body mass index. The genotype C-C had the highest plasma visfatin and C-reactive protein concentrations. The plasma visfatin and C-reactive protein concentrations between the variant genotypes C-T and T-T did not differ significantly. Plasma visfatin level was significantly associated with plasma C-reactive protein level using multivariate linear regression. Thus, the SNP-1535 C>T of visfatin gene seemed to be potentially involved in the inflammatory component of TBI through a decreased production of visfatin.
Collapse
Affiliation(s)
- Jian-Feng Weng
- Department of Neurosurgery, the Second People's Hospital of Cixi City, Cixi 315315, China
| | | | | | | | | | | |
Collapse
|
130
|
Preyat N, Leo O. Sirtuin deacylases: a molecular link between metabolism and immunity. J Leukoc Biol 2013; 93:669-80. [PMID: 23325925 DOI: 10.1189/jlb.1112557] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Lysine deacetylation by the NAD(+)-dependent family of sirtuins has been recognized as an important post-translational modification regulating a wide range of cellular processes. These lysine deacetylases have attracted much interest based on their ability to promote survival in response to stress. Sirtuins require NAD(+) for their enzymatic activity, suggesting that these enzymes may represent molecular links between cell metabolism and several human disorders, including diabetes and cancer. Inflammation represents a pathological situation with clear connections to metabolism and aging in humans, raising the possibility that sirtuins may also play an important role during a normal and/or a pathological immune response. A growing body of data has confirmed the immunomodulatory properties of sirtuins, although often with contrasting and opposing conclusions. These observations will be summarized herein and the possible strategies that may lead to the development of novel therapeutic approaches to treat inflammation briefly discussed.
Collapse
Affiliation(s)
- Nicolas Preyat
- Laboratory of Immunobiology, Institute for Molecular Biology and Medicine, Université Libre de Bruxelles, Gosselies, Belgium
| | | |
Collapse
|
131
|
O'Mahony DS, Glavan BJ, Holden TD, Fong C, Black RA, Rona G, Tejera P, Christiani DC, Wurfel MM. Inflammation and immune-related candidate gene associations with acute lung injury susceptibility and severity: a validation study. PLoS One 2012; 7:e51104. [PMID: 23251429 PMCID: PMC3522667 DOI: 10.1371/journal.pone.0051104] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Accepted: 10/31/2012] [Indexed: 02/06/2023] Open
Abstract
Introduction Common variants in genes related to inflammation, innate immunity, epithelial cell function, and angiogenesis have been reported to be associated with risks for Acute Lung Injury (ALI) and related outcomes. We tested whether previously-reported associations can be validated in an independent cohort at risk for ALI. Methods We identified 37 genetic variants in 27 genes previously associated with ALI and related outcomes. We prepared allelic discrimination assays for 12 SNPs from 11 genes with MAF>0.05 and genotyped these SNPs in Caucasian subjects from a cohort of critically ill patients meeting criteria for the systemic inflammatory response syndrome (SIRS) followed for development of ALI, duration of mechanical ventilation, and in-hospital death. We tested for associations using additive and recessive genetic models. Results Among Caucasian subjects with SIRS (n = 750), we identified a nominal association between rs2069832 in IL6 and ALI susceptibility (ORadj 1.61; 95% confidence interval [CI], 1.04–2.48, P = 0.03). In a sensitivity analysis limiting ALI cases to those who qualified for the Acute Respiratory Distress Syndrome (ARDS), rs61330082 in NAMPT was nominally associated with risk for ARDS. In terms of ALI outcomes, SNPs in MBL2 (rs1800450) and IL8 (rs4073) were nominally associated with fewer ventilator-free days (VFDs), and SNPs in NFE2L2 (rs6721961) and NAMPT (rs61330082) were nominally associated with 28-day mortality. The directions of effect for these nominal associations were in the same direction as previously reported but none of the associations survived correction for multiple hypothesis testing. Conclusion Although our primary analyses failed to statistically validate prior associations, our results provide some support for associations between SNPs in IL6 and NAMPT and risk for development of lung injury and for SNPs in IL8, MBL2, NFE2L2 and NAMPT with severity in ALI outcomes. These associations provide further evidence that genetic factors in genes related to immunity and inflammation contribute to ALI pathogenesis.
Collapse
Affiliation(s)
- D. Shane O'Mahony
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Bradford J. Glavan
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Tarah D. Holden
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Christie Fong
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - R. Anthony Black
- Institute of Translational Health Sciences (ITHS), University of Washington, Seattle, Washington, United States of America
| | - Gail Rona
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Paula Tejera
- Harvard School of Public Health, Harvard University, Boston, Massachusetts, United States of America
| | - David C. Christiani
- Harvard School of Public Health, Harvard University, Boston, Massachusetts, United States of America
| | - Mark M. Wurfel
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
132
|
Terra X, Auguet T, Quesada I, Aguilar C, Luna AM, Hernández M, Sabench F, Porras JA, Martínez S, Lucas A, Pellitero S, Llutart J, del Castillo D, Richart C. Increased levels and adipose tissue expression of visfatin in morbidly obese women: the relationship with pro-inflammatory cytokines. Clin Endocrinol (Oxf) 2012; 77:691-8. [PMID: 22182404 DOI: 10.1111/j.1365-2265.2011.04327.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The controversial results on the physiopathological role of visfatin led us to examine both circulating visfatin levels and gene expression in visceral (VAT) and subcutaneous fat (SAT) in a homogeneous group of morbidly obese women. DESIGN, PATIENTS AND MEASUREMENTS We analysed circulating levels of several adipo/cytokines in 133 Spanish women: 40 lean (C) [body mass index (BMI) < 25 kg/m(2) ] and 93 morbidly obese (MO) (BMI > 40 kg/m(2) ). In the MO group, we found 31 diabetic and 62 nondiabetic subjects. We obtained follow-up blood samples at 6 and 12 months after bariatric surgery from 30 MO patients. We determined the circulating levels of visfatin, adiponectin, interleukin-6 (IL6), C-reactive protein (CRP), resistin and tumour necrosis factor-α (TNFα) by ELISA, and visfatin, adiponectin, IL6, resistin and TNFα gene expression in SAT and VAT by real-time RT-PCR. RESULTS Circulating visfatin levels were higher in MO women compared with lean controls (C = 1·43 ± 0·14 μg/l, MO = 3·60 ± 0·29 μg/l, P < 0·001). After bariatric surgery-induced weight loss, visfatin levels were reduced significantly over 12 months. Visfatin expression in SAT and VAT was similar, but significantly higher in MO compared to C and independent of the presence of diabetes mellitus. Circulating visfatin levels were positively related to IL6 and CRP levels. Visfatin gene expression in VAT and SAT was strongly related to IL6 and TNFα expression. CONCLUSION In a homogeneous cohort of morbidly obese women, our findings show that visfatin has a strong relationship with pro-inflammatory factors in severe obesity.
Collapse
Affiliation(s)
- Ximena Terra
- Grup de Recerca en Medicina Aplicada Hospital Joan XXIII, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, IISPV, Tarragona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Adya R, Tan BK, Chen J, Randeva HS. Visfatin and endothelial angiogenesis: AUTHORS' RETROSPECTIVE. Cardiovasc Res 2012. [DOI: 10.1093/cvr/cvs207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
134
|
Hu P, Wang X, Haitsma JJ, Furmli S, Masoom H, Liu M, Imai Y, Slutsky AS, Beyene J, Greenwood CMT, dos Santos C. Microarray meta-analysis identifies acute lung injury biomarkers in donor lungs that predict development of primary graft failure in recipients. PLoS One 2012; 7:e45506. [PMID: 23071521 PMCID: PMC3470558 DOI: 10.1371/journal.pone.0045506] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2011] [Accepted: 08/23/2012] [Indexed: 11/19/2022] Open
Abstract
Objectives To perform a meta-analysis of gene expression microarray data from animal studies of lung injury, and to identify an injury-specific gene expression signature capable of predicting the development of lung injury in humans. Methods We performed a microarray meta-analysis using 77 microarray chips across six platforms, two species and different animal lung injury models exposed to lung injury with or/and without mechanical ventilation. Individual gene chips were classified and grouped based on the strategy used to induce lung injury. Effect size (change in gene expression) was calculated between non-injurious and injurious conditions comparing two main strategies to pool chips: (1) one-hit and (2) two-hit lung injury models. A random effects model was used to integrate individual effect sizes calculated from each experiment. Classification models were built using the gene expression signatures generated by the meta-analysis to predict the development of lung injury in human lung transplant recipients. Results Two injury-specific lists of differentially expressed genes generated from our meta-analysis of lung injury models were validated using external data sets and prospective data from animal models of ventilator-induced lung injury (VILI). Pathway analysis of gene sets revealed that both new and previously implicated VILI-related pathways are enriched with differentially regulated genes. Classification model based on gene expression signatures identified in animal models of lung injury predicted development of primary graft failure (PGF) in lung transplant recipients with larger than 80% accuracy based upon injury profiles from transplant donors. We also found that better classifier performance can be achieved by using meta-analysis to identify differentially-expressed genes than using single study-based differential analysis. Conclusion Taken together, our data suggests that microarray analysis of gene expression data allows for the detection of “injury" gene predictors that can classify lung injury samples and identify patients at risk for clinically relevant lung injury complications.
Collapse
Affiliation(s)
- Pingzhao Hu
- The Centre for Applied Genomics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xinchen Wang
- Keenan Research Center at the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada
| | - Jack J. Haitsma
- Keenan Research Center at the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada
| | - Suleiman Furmli
- Keenan Research Center at the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada
| | - Hussain Masoom
- Keenan Research Center at the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Thoracic Surgery Research Laboratory, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Yumiko Imai
- Biological Informatics and Experimental Therapeutics Akita University Graduate School of Medicine, Akita City, Akita, Japan
| | - Arthur S. Slutsky
- Keenan Research Center at the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada
| | - Joseph Beyene
- Program in Population Genomics, Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - Celia M. T. Greenwood
- Centre for Clinical Epidemiology, Lady Davis Institute and Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Claudia dos Santos
- Keenan Research Center at the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
135
|
PBEF/NAMPT/visfatin: a promising drug target for treating rheumatoid arthritis? Future Med Chem 2012; 4:751-69. [PMID: 22530639 DOI: 10.4155/fmc.12.34] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
NAMPT, also known as pre-B-cell colony-enhancing factor and visfatin, has been proposed to be involved in preventing apoptosis in cancer cells and, as such, has received a great deal of attention in recent years and stimulated the development to specific inhibitors for treating cancer. The role of NAMPT inhibitors as potential therapeutic agents for other diseases has not been studied extensively. Here, we describe their applicability for treating rheumatoid arthritis. We summarize current knowledge of NAMPT expression in healthy and diseased tissues, thereafter, we focus on pathological mechanisms relevant to rheumatoid arthritis that involve the NAMPT pathway and review the current status of NAMPT inhibitors being evaluated in clinical trials.
Collapse
|
136
|
Rivara MB, Bajwa EK, Januzzi JL, Gong MN, Thompson BT, Christiani DC. Prognostic significance of elevated cardiac troponin-T levels in acute respiratory distress syndrome patients. PLoS One 2012; 7:e40515. [PMID: 22808179 PMCID: PMC3395687 DOI: 10.1371/journal.pone.0040515] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 06/11/2012] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Elevated levels of biochemical markers of myocardial necrosis have been associated with worsened outcomes in Acute Respiratory Distress Syndrome (ARDS), but there are few prospective data on this relationship. We investigated elevated cardiac troponin T (cTnT) levels and their relationship with outcome in patients with ARDS. METHODS A prospective cohort study of patients with ARDS was conducted at a tertiary-care academic medical center. Patients had blood taken within 48 hours of ARDS onset and assayed for cTnT. Patients were followed for the outcomes of 60-day mortality, number of organ failures, and days free of mechanical ventilation. Echocardiographic and electrocardiographic (ECG) data were analyzed for signs of myocardial ischemia, infarction, or other myocardial dysfunction. RESULTS 177 patients were enrolled, 70 of whom died (40%). 119 patients had detectable cTnT levels (67%). Median cTnT level was 0.03 ng/mL, IQR 0-0.10 ng/mL, and levels were higher among non-survivors (P = .008). Increasing cTnT level was significantly associated with increasing mortality (P = .008). The association between increasing cTnT level and mortality remained significant after adjustment in a multivariate model (HR(adj) = 1.45, 95% CI 1.17-1.81, P = .001). Elevated cTnT level was also associated with increased number of organ failures (P = .002), decreased number of days free of mechanical ventilation (P = .03), echocardiographic wall motion abnormalities (P = 0.001), and severity of tricuspid regurgitation (P = .04). There was no association between ECG findings of myocardial ischemia or infarction and elevated cTnT. CONCLUSIONS Elevated cTnT levels are common in patients with ARDS, and are associated with worsened clinical outcomes and certain echocardiographic abnormalities. No association was seen between cTnT levels and ECG evidence of coronary ischemia.
Collapse
Affiliation(s)
- Matthew B. Rivara
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Ednan K. Bajwa
- Pulmonary and Critical Care Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| | - James L. Januzzi
- Cardiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michelle N. Gong
- Critical Care Division, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - B. Taylor Thompson
- Pulmonary and Critical Care Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David C. Christiani
- Pulmonary and Critical Care Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| |
Collapse
|
137
|
Abstract
Nicotinamide phosphoribosyltransferase is the rate-limiting enzyme that catalyzes the first step in the biosynthesis of nicotinamide adenine dinucleotide from nicotinamide. This protein was originally cloned as a putative pre-B cell colony-enhancing factor and also found to be a visceral fat-derived adipokine (visfatin). As a multifunctional protein, visfatin plays an important role in immunity, metabolism, aging, inflammation, and responses to stress. Visfatin also participates in several pathophysiological processes contributing to cardio-cerebro-vascular diseases, including hypertension, atherosclerosis, ischemic heart disease, and ischemic stroke. However, whether visfatin is a friend or a foe in these diseases remains uncertain. This brief review focuses on the current understanding of the complex role of visfatin in the cardio-cerebro-vascular system under normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Pei Wang
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | | | | |
Collapse
|
138
|
Fang X, Bai C, Wang X. Bioinformatics insights into acute lung injury/acute respiratory distress syndrome. Clin Transl Med 2012; 1:9. [PMID: 23369517 PMCID: PMC3560991 DOI: 10.1186/2001-1326-1-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 05/23/2012] [Indexed: 02/08/2023] Open
Abstract
Bioinformatics is the application of omics science, information technology, mathematics and statistics in the field of biomarker detection. Clinical bioinformatics can be applied for identification and validation of new biomarkers to improve current methods of monitoring disease activity and identify new therapeutic targets. Acute lung injurt (ALI)/Acute respiratory distress syndrome (ARDS) affects a large number of patients with a poor prognosis. The present review mainly focused on the progress in understanding disease heterogeneity through the use of evolving biological, genomic, and genetic approaches and the role of clinical bioinformatics in the pathogenesis and treatment of ALI/ARDS. The remarkable advances in clinical bioinformatics can be a new way for understanding disease pathogenesis, diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaocong Fang
- Department of Pulmonary MedicineZhongshan Hospital, Fudan University, Shanghai, China.
| | | | | |
Collapse
|
139
|
Dahl TB, Holm S, Aukrust P, Halvorsen B. Visfatin/NAMPT: a multifaceted molecule with diverse roles in physiology and pathophysiology. Annu Rev Nutr 2012; 32:229-43. [PMID: 22462624 DOI: 10.1146/annurev-nutr-071811-150746] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Visfatin/NAMPT (nicotinamide phosphoribosyltransferase) is a protein with several suggested functions. Although the first discovery of this molecule as a pre-B-cell colony-enhancing factor suggested primarily a cytokine function, its rediscovery as the key enzyme in nicotinamide adenine dinucleotide generation has considerably widened its potential biological activities. Although originally thought to be produced in adipose tissue (i.e., adipocytes and infiltrating macrophages), its production seems to involve other cells and tissues such as skeletal muscle, liver, immune cells, cardiomyocytes, and the brain. Visfatin/NAMPT has both intracellular and extracellular effects influencing several signaling pathways. Its broad spectrum of effects is mirrored by its potential involvement in a wide range of disorders including human immunodeficiency virus infection, septicemia, myocardial failure, atherosclerosis, metabolic disorders, inflammatory diseases, malignancies, and neurodegenerative disorders and aging. Moreover, studies on visfatin/NAMPT in atherosclerotic disorders suggest a rather complex role of this molecule in pathophysiology, potentially mediating both adaptive and maladaptive responses.
Collapse
Affiliation(s)
- Tuva B Dahl
- Research Institute for Internal Medicine, Faculty of Medicine, University of Oslo, Oslo University Hospital Rikshospitalet, Oslo, Norway.
| | | | | | | |
Collapse
|
140
|
Leikauf GD, Pope-Varsalona H, Concel VJ, Liu P, Bein K, Berndt A, Martin TM, Ganguly K, Jang AS, Brant KA, Dopico RA, Upadhyay S, Di YPP, Li Q, Hu Z, Vuga LJ, Medvedovic M, Kaminski N, You M, Alexander DC, McDunn JE, Prows DR, Knoell DL, Fabisiak JP. Integrative assessment of chlorine-induced acute lung injury in mice. Am J Respir Cell Mol Biol 2012; 47:234-44. [PMID: 22447970 DOI: 10.1165/rcmb.2012-0026oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The genetic basis for the underlying individual susceptibility to chlorine-induced acute lung injury is unknown. To uncover the genetic basis and pathophysiological processes that could provide additional homeostatic capacities during lung injury, 40 inbred murine strains were exposed to chlorine, and haplotype association mapping was performed. The identified single-nucleotide polymorphism (SNP) associations were evaluated through transcriptomic and metabolomic profiling. Using ≥ 10% allelic frequency and ≥ 10% phenotype explained as threshold criteria, promoter SNPs that could eliminate putative transcriptional factor recognition sites in candidate genes were assessed by determining transcript levels through microarray and reverse real-time PCR during chlorine exposure. The mean survival time varied by approximately 5-fold among strains, and SNP associations were identified for 13 candidate genes on chromosomes 1, 4, 5, 9, and 15. Microarrays revealed several differentially enriched pathways, including protein transport (decreased more in the sensitive C57BLKS/J lung) and protein catabolic process (increased more in the resistant C57BL/10J lung). Lung metabolomic profiling revealed 95 of the 280 metabolites measured were altered by chlorine exposure, and included alanine, which decreased more in the C57BLKS/J than in the C57BL/10J strain, and glutamine, which increased more in the C57BL/10J than in the C57BLKS/J strain. Genetic associations from haplotype mapping were strengthened by an integrated assessment using transcriptomic and metabolomic profiling. The leading candidate genes associated with increased susceptibility to acute lung injury in mice included Klf4, Sema7a, Tns1, Aacs, and a gene that encodes an amino acid carrier, Slc38a4.
Collapse
Affiliation(s)
- George D Leikauf
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, PA 15219-3130, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Maloney JP, Stearman RS, Bull TM, Calabrese DW, Tripp-Addison ML, Wick MJ, Broeckel U, Robbins IM, Wheeler LA, Cogan JD, Loyd JE. Loss-of-function thrombospondin-1 mutations in familial pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2012; 302:L541-54. [PMID: 22198906 PMCID: PMC3311532 DOI: 10.1152/ajplung.00282.2011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 12/15/2011] [Indexed: 12/31/2022] Open
Abstract
Most patients with familial pulmonary arterial hypertension (FPAH) carry mutations in the bone morphogenic protein receptor 2 gene (BMPR2). Yet carriers have only a 20% risk of disease, suggesting that other factors influence penetrance. Thrombospondin-1 (TSP1) regulates activation of TGF-β and inhibits endothelial and smooth muscle cell proliferation, pathways coincidentally altered in pulmonary arterial hypertension (PAH). To determine whether a subset of FPAH patients also have mutations in the TSP1 gene (THBS1) we resequenced the type I repeats of THBS1 encoding the TGF-β regulation and cell growth inhibition domains in 60 FPAH probands, 70 nonfamilial PAH subjects, and in large control groups. We identified THBS1 mutations in three families: a novel missense mutation in two (Asp362Asn), and an intronic mutation in a third (IVS8+255 G/A). Neither mutation was detected in population controls. Mutant 362Asn TSP1 had less than half of the ability of wild-type TSP1 to activate TGF-β. Mutant 362Asn TSP1 also lost the ability to inhibit growth of pulmonary arterial smooth muscle cells and was over threefold less effective at inhibiting endothelial cell growth. The IVS8+255 G/A mutation decreased and/or eliminated local binding of the transcription factors SP1 and MAZ but did not affect RNA splicing. These novel mutations implicate THBS1 as a modifier gene in FPAH. These THBS1 mutations have implications in the genetic evaluation of FPAH patients. However, since FPAH is rare, these data are most relevant as evidence for the importance of TSP1 in pulmonary vascular homeostasis. Further examination of THBS1 in the pathogenesis of PAH is warranted.
Collapse
Affiliation(s)
- James P Maloney
- Division of Pulmonary and Critical Care Medicine, University of Colorado, Aurora, 80045, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Jacques C, Holzenberger M, Mladenovic Z, Salvat C, Pecchi E, Berenbaum F, Gosset M. Proinflammatory actions of visfatin/nicotinamide phosphoribosyltransferase (Nampt) involve regulation of insulin signaling pathway and Nampt enzymatic activity. J Biol Chem 2012; 287:15100-8. [PMID: 22399297 DOI: 10.1074/jbc.m112.350215] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Visfatin (also termed pre-B-cell colony-enhancing factor (PBEF) or nicotinamide phosphoribosyltransferase (Nampt)) is a pleiotropic mediator acting on many inflammatory processes including osteoarthritis. Visfatin exhibits both an intracellular enzymatic activity (nicotinamide phosphoribosyltransferase, Nampt) leading to NAD synthesis and a cytokine function via the binding to its hypothetical receptor. We recently reported the role of visfatin in prostaglandin E(2) (PGE(2)) synthesis in chondrocytes. Here, our aim was to characterize the signaling pathways involved in this response in exploring both the insulin receptor (IR) signaling pathway and Nampt activity. IR was expressed in human and murine chondrocytes, and visfatin triggered Akt phosphorylation in murine chondrocytes. Blocking IR expression with siRNA or activity using the hydroxy-2-naphthalenyl methyl phosphonic acid tris acetoxymethyl ester (HNMPA-(AM)(3)) inhibitor diminished visfatin-induced PGE(2) release in chondrocytes. Moreover, visfatin-induced IGF-1R(-/-) chondrocytes released higher concentration of PGE(2) than IGF-1R(+/+) cells, a finding confirmed with an antibody that blocked IGF-1R. Using RT-PCR, we found that visfatin did not regulate IR expression and that an increased insulin release was also unlikely to be involved because insulin was unable to increase PGE(2) release. Inhibition of Nampt activity using the APO866 inhibitor gradually decreased PGE(2) release, whereas the addition of exogenous nicotinamide increased it. We conclude that the proinflammatory actions of visfatin in chondrocytes involve regulation of IR signaling pathways, possibly through the control of Nampt enzymatic activity.
Collapse
Affiliation(s)
- Claire Jacques
- UR4, Pierre and Marie Curie University, 75252 Paris, France
| | | | | | | | | | | | | |
Collapse
|
143
|
Schilling E, Hauschildt S. Extracellular ATP induces P2X7-dependent nicotinamide phosphoribosyltransferase release in LPS-activated human monocytes. Innate Immun 2012; 18:738-44. [PMID: 22377803 DOI: 10.1177/1753425912439614] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT), an enzyme involved in NAD biosynthesis, has recently been identified as a novel mediator of innate immunity. In the present study, we report that treatment of LPS-primed monocytes with ATP greatly enhanced the secretion of NAMPT in a time- and concentration-dependent manner without displaying any cytotoxic effect. NAMPT release was suppressed by pretreatment with the P2X(7) receptor (P2X(7)R) inhibitors oxidized ATP (oxATP) and KN-62, indicating the engagement of P2X(7)Rs. Furthermore, P2X(7)R was found to be involved in mediating cell permeability caused by the addition of ATP. To define a role of endogenous ATP in NAMPT secretion, LPS-primed monocytes were incubated in the presence of oxATP and KN-62, as well as the ATP-hydrolyzing enzymes apyrase and hexokinase. With the exception of oxATP, neither substance led to a decrease in NAMPT release, suggesting that autocrine/paracrine ATP is unlikely to be responsible for the LPS-induced release of NAMPT. In conclusion, the enhanced release of NAMPT by extracellular ATP described here indicates the requirement of a second stimulus for the efficient secretion of NAMPT. This mode of secretion, which also applies to IL-1β, might represent a general mechanism for the release of leaderless secretory proteins at locally restricted sites.
Collapse
Affiliation(s)
- Erik Schilling
- Institute of Biology, University of Leipzig, Leipzig, Germany
| | | |
Collapse
|
144
|
Lederer DJ, Kawut SM, Wickersham N, Winterbottom C, Bhorade S, Palmer SM, Lee J, Diamond JM, Wille KM, Weinacker A, Lama VN, Crespo M, Orens JB, Sonett JR, Arcasoy SM, Ware LB, Christie JD. Obesity and primary graft dysfunction after lung transplantation: the Lung Transplant Outcomes Group Obesity Study. Am J Respir Crit Care Med 2012; 184:1055-61. [PMID: 21799077 DOI: 10.1164/rccm.201104-0728oc] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
RATIONALE Obesity has been linked to acute lung injury and is a risk factor for early mortality after lung transplantation. OBJECTIVES To examine the associations of obesity and plasma adipokines with the risk of primary graft dysfunction after lung transplantation. METHODS We performed a prospective cohort study of 512 adult lung transplant recipients with chronic obstructive pulmonary disease or interstitial lung disease enrolled in the Lung Transplant Outcomes Group Study. In a nested case-control study, we measured plasma leptin, adiponectin, and resistin before lung transplantation and 6 and 24 hours after lung transplantation in 40 cases of primary graft dysfunction and 80 control subjects. Generalized linear mixed models and logistic regression were used to estimate risk ratios and odds ratios. MEASUREMENTS AND MAIN RESULTS Grade 3 primary graft dysfunction developed within 72 hours of transplantation in 29% participants. Obesity was associated with a twofold increased risk of primary graft dysfunction (adjusted risk ratio 2.1; 95% confidence interval, 1.7-2.6). The risk of primary graft dysfunction increased by 40% (confidence interval, 30–50%) for each 5 kg/m(2) increase in body mass index after accounting for center, diagnosis, cardiopulmonary bypass, and transplant procedure. Higher plasma leptin levels were associated with a greater risk of primary graft dysfunction (sex-adjusted P = 0.02). The associations of both obesity and leptin with primary graft dysfunction tended to be stronger among those who did not undergo cardiopulmonary bypass. CONCLUSIONS Obesity is an independent risk factor for primary graft dysfunction after lung transplantation.
Collapse
Affiliation(s)
- David J Lederer
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Prognostic utility of changes in N-terminal pro-brain natriuretic Peptide combined with sequential organ failure assessment scores in patients with acute lung injury/acute respiratory distress syndrome concomitant with septic shock. Shock 2012; 36:109-14. [PMID: 21478812 DOI: 10.1097/shk.0b013e31821d8f2d] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We investigated the prognostic utility of changes in N-terminal pro-brain natriuretic peptide (NT-proBNP) in combination with Sequential Organ Failure Assessment (SOFA) score in patients with acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) concomitant with septic shock. Forty-nine mechanically ventilated patients with ALI/ARDS concomitant with septic shock were studied. N-terminal pro-brain natriuretic peptide levels were measured on the first 3 days (days 0, 1, and 2) in the intensive care unit. The median NT-proBNP levels in survivors and nonsurvivors were 3,999 vs. 2,819 pg/mL on day 0 (P = 0.719); 4,495 vs. 5,397 pg/mL on day 1 (P = 0.543); and 2,325 vs. 14,173 pg/mL on day 2 (P = 0.028). N-terminal pro-brain natriuretic peptide levels increased significantly from baseline values in nonsurvivors only. We observed a monotonic increase in 28-day mortality associated with increasing quartiles of percent change in NT-proBNP on day 2 (P < 0.0001). Kaplan-Meier survival analysis revealed that mortality was significantly higher in patients with a change in NT-proBNP of 30% or more (log-rank P < 0.0001). On day 2, areas under the receiver operating characteristic curves for predicting 28-day mortality were 0.74 for SOFA alone and 0.85 (P = 0.028) for SOFA combined with percent change in NT-proBNP. In conclusion, in patients with ALI/ARDS concomitant with septic shock, a rising trend (high percent change) in NT-proBNP levels had better prognostic utility than absolute levels. The combination of percent change in NT-proBNP with SOFA may provide superior prognostic accuracy to SOFA alone.
Collapse
|
146
|
Christie JD, Wurfel MM, Feng R, O'Keefe GE, Bradfield J, Ware LB, Christiani DC, Calfee CS, Cohen MJ, Matthay M, Meyer NJ, Kim C, Li M, Akey J, Barnes KC, Sevransky J, Lanken PN, May AK, Aplenc R, Maloney JP, Hakonarson H, for the Trauma ALI SNP Consortium (TASC) investigators. Genome wide association identifies PPFIA1 as a candidate gene for acute lung injury risk following major trauma. PLoS One 2012; 7:e28268. [PMID: 22295056 PMCID: PMC3266233 DOI: 10.1371/journal.pone.0028268] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 11/04/2011] [Indexed: 12/29/2022] Open
Abstract
Acute Lung Injury (ALI) is a syndrome with high associated mortality characterized by severe hypoxemia and pulmonary infiltrates in patients with critical illness. We conducted the first investigation to use the genome wide association (GWA) approach to identify putative risk variants for ALI. Genome wide genotyping was performed using the Illumina Human Quad 610 BeadChip. We performed a two-stage GWA study followed by a third stage of functional characterization. In the discovery phase (Phase 1), we compared 600 European American trauma-associated ALI cases with 2266 European American population-based controls. We carried forward the top 1% of single nucleotide polymorphisms (SNPs) at p<0.01 to a replication phase (Phase 2) comprised of a nested case-control design sample of 212 trauma-associated ALI cases and 283 at-risk trauma non-ALI controls from ongoing cohort studies. SNPs that replicated at the 0.05 level in Phase 2 were subject to functional validation (Phase 3) using expression quantitative trait loci (eQTL) analyses in stimulated B-lymphoblastoid cell lines (B-LCL) in family trios. 159 SNPs from the discovery phase replicated in Phase 2, including loci with prior evidence for a role in ALI pathogenesis. Functional evaluation of these replicated SNPs revealed rs471931 on 11q13.3 to exert a cis-regulatory effect on mRNA expression in the PPFIA1 gene (p = 0.0021). PPFIA1 encodes liprin alpha, a protein involved in cell adhesion, integrin expression, and cell-matrix interactions. This study supports the feasibility of future multi-center GWA investigations of ALI risk, and identifies PPFIA1 as a potential functional candidate ALI risk gene for future research.
Collapse
Affiliation(s)
- Jason D. Christie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Biostatistics and Epidemiology, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - Mark M. Wurfel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Rui Feng
- Department of Biostatistics and Epidemiology, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Grant E. O'Keefe
- Department of Surgery, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Jonathan Bradfield
- Division of Human Genetics, Center for Applied Genomics, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - David C. Christiani
- Department of Environmental Health, Harvard School of Public Health and Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Carolyn S. Calfee
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California San Francisco, San Francisco, California, United States of America
| | - Mitchell J. Cohen
- Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
| | - Michael Matthay
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California San Francisco, San Francisco, California, United States of America
| | - Nuala J. Meyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Cecilia Kim
- Division of Human Genetics, Center for Applied Genomics, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Mingyao Li
- Department of Biostatistics and Epidemiology, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Joshua Akey
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Kathleen C. Barnes
- Division of Pulmonary, Allergy, and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jonathan Sevransky
- Division of Pulmonary, Allergy, and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Paul N. Lanken
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Addison K. May
- Department of Surgical Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Richard Aplenc
- Division of Oncology, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - James P. Maloney
- Division of Pulmonary and Critical Care Medicine, University of Colorado Health Sciences Center, Denver, Colorado, United States of America
| | - Hakon Hakonarson
- Division of Human Genetics, Center for Applied Genomics, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | | |
Collapse
|
147
|
Lucking SE, Maffei FA, Tamburro RF, Thomas NJ. Genetic Predisposition to Critical Illness in the Pediatric Intensive Care Unit. PEDIATRIC CRITICAL CARE STUDY GUIDE 2012. [PMCID: PMC7178837 DOI: 10.1007/978-0-85729-923-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Much progress has been made in the past decade in the understanding of the genetic contribution to the development of human disease in general, and critical care illness specifically. With the mapping of the human genome and on-going mapping of genetic polymorphisms and haplotypes in humans, the field of critical care is now in prime position to study the impact of genetics on common illnesses that affect children who require critical care, to examine how differences of the host defense response lead to variable outcomes in outwardly appearing similar disease states, and to study how genetic differences in response to therapy will help practitioners tailor therapeutic interventions to an individual child’s genetic composition. While we are still years away from true individualized medicine, we are now closer than ever to understanding why two might children respond to the same environmental insult in vastly different ways.
Collapse
Affiliation(s)
- Steven E. Lucking
- Children's Heart Group, Div. Pediatric Critical Care, Penn State Children's Hospital, University Drive 500, Hershey, 17078 Pennsylvania USA
| | - Frank A. Maffei
- Janet Weis Children's Hospital @ Geising, Pediatric Critical Care Medicine, Temple University School of Medicine, N. Academy Ave 100, Danville, 17822 Pennsylvania USA
| | - Robert F. Tamburro
- Milton S. Hershey Medical Center, Penn State College of Medicine, University Drive 500, Hershey, 17033-2390 Pennsylvania USA
| | - Neal J. Thomas
- College of Medicine, Penn State Children's Hospital, Pennsylvania State University, University Drive 500, Hershey, 17078 Pennsylvania USA
| |
Collapse
|
148
|
Mechanistic insights into the link between visfatin gene C-1535T polymorphism and coronary artery disease: an in vitro study. Mol Cell Biochem 2011; 363:315-22. [PMID: 22147201 DOI: 10.1007/s11010-011-1184-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Accepted: 11/24/2011] [Indexed: 12/17/2022]
Abstract
Visfatin, a pro-inflammatory cytokine predominantly released from leucocytes, is correlated with coronary artery disease (CAD). We have previously reported that the -1535C>T polymorphism (rs1330082), which located on the promoter region of visfatin, was associated with decreased risk of CAD. Here, we investigated the underlying mechanism by which this polymorphism affects the genetic susceptibility to CAD. The difference of the promoter activities between -1535T variant and -1535C allele was tested by luciferase reporter gene assay. The difference of transcription factor binding activities between T and C allele was evaluated by electrophoretic mobility shift assay. In reporter gene assay, we showed that the T variant had a significantly reduced transcriptional activity compared with the C allele. The T-variant significantly attenuated the promoter binding affinity to nuclear transcription factors and this effect became much obvious after treatment with TNF-α. Moreover, competition experiment revealed that the retarded complex formed by T-1535- or C-1535-probe binding to nuclear extracts was nearly completely inhibited by unlabeled activator protein-1 (AP-1) specific probe, indicating that AP-1 might be the target nuclear effector. Taken together, our data provided potential mechanistic link between the visfatin -1535C>T polymorphism and reduced CAD risk.
Collapse
|
149
|
Ma SF, Xie L, Pino-Yanes M, Sammani S, Wade MS, Letsiou E, Siegler J, Wang T, Infusino G, Kittles RA, Flores C, Zhou T, Prabhakar BS, Moreno-Vinasco L, Villar J, Jacobson JR, Dudek SM, Garcia JGN. Type 2 deiodinase and host responses of sepsis and acute lung injury. Am J Respir Cell Mol Biol 2011; 45:1203-11. [PMID: 21685153 PMCID: PMC3262665 DOI: 10.1165/rcmb.2011-0179oc] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 06/10/2011] [Indexed: 12/21/2022] Open
Abstract
The role of thyroid hormone metabolism in clinical outcomes of the critically ill remains unclear. Using preclinical models of acute lung injury (ALI), we assessed the gene and protein expression of type 2 deiodinase (DIO2), a key driver for synthesis of biologically active triiodothyronine, and addressed potential association of DIO2 genetic variants with ALI in a multiethnic cohort. DIO2 gene and protein expression levels in murine lung were validated by microarrays and immunoblotting. Lung injury was assessed by levels of bronchoalveolar lavage protein and leukocytes. Single-nucleotide polymorphisms were genotyped and ALI susceptibility association assessed. Significant increases in both DIO2 gene and D2 protein expression were observed in lung tissues from murine ALI models (LPS- and ventilator-induced lung injury), with expression directly increasing with the extent of lung injury. Mice with reduced levels of DIO2 expression (by silencing RNA) demonstrated reduced thyroxine levels in plasma and increased lung injury (increased bronchoalveolar lavage protein and leukocytes), suggesting a protective role for DIO2 in ALI. The G (Ala) allele of the Thr92Ala coding single-nucleotide polymorphism (rs225014) was protective in severe sepsis and severe sepsis-associated ALI after adjustments for age, sex, and genetic ancestry in a logistic regression model in European Americans. Our studies indicate that DIO2 is a novel ALI candidate gene, the nonsynonymous Thr92Ala coding variant of which confers ALI protection. Increased DIO2 expression may dampen the ALI inflammatory response, thereby strengthening the premise that thyroid hormone metabolism is intimately linked to the integrated response to inflammatory injury in critically ill patients.
Collapse
Affiliation(s)
- Shwu-Fan Ma
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Lishi Xie
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Maria Pino-Yanes
- Centro de Investigacion Biomedica en Red Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Tenerife, Spain
| | - Saad Sammani
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Michael S. Wade
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Eleftheria Letsiou
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Jessica Siegler
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Ting Wang
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Giovanni Infusino
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Rick A. Kittles
- Section of Hematology and Oncology, Department of Medicine, and
| | - Carlos Flores
- Centro de Investigacion Biomedica en Red Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Tenerife, Spain
| | - Tong Zhou
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Bellur S. Prabhakar
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois
| | - Liliana Moreno-Vinasco
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Jesus Villar
- Centro de Investigacion Biomedica en Red Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain; and
- Keenan Research Center at the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Canada
| | - Jeffrey R. Jacobson
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Steven M. Dudek
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Joe G. N. Garcia
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
150
|
Mitra S, Sammani S, Wang T, Boone DL, Meyer NJ, Dudek SM, Moreno-Vinasco L, Garcia JGN, Jacobson JR. Role of growth arrest and DNA damage-inducible α in Akt phosphorylation and ubiquitination after mechanical stress-induced vascular injury. Am J Respir Crit Care Med 2011; 184:1030-40. [PMID: 21816939 PMCID: PMC3763933 DOI: 10.1164/rccm.201103-0447oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 07/29/2011] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The stress-induced growth arrest and DNA damage-inducible a (GADD45a) gene is up-regulated by mechanical stress with GADD45a knockout (GADD45a(-/-)) mice demonstrating both increased susceptibility to ventilator-induced lung injury (VILI) and reduced levels of the cell survival and vascular permeability signaling effector (Akt). However, the functional role of GADD45a in the pathogenesis of VILI is unknown. OBJECTIVES We sought to define the role of GADD45a in the regulation of Akt activation induced by mechanical stress. METHODS VILI-challenged GADD45a(-/-) mice were administered a constitutively active Akt1 vector and injury was assessed by bronchoalveolar lavage cell counts and protein levels. Human pulmonary artery endothelial cells (EC) were exposed to 18% cyclic stretch (CS) under conditions of GADD45a silencing and used for immunoprecipitation, Western blotting or immunofluoresence. EC were also transfected with mutant ubiquitin vectors to characterize site-specific Akt ubiquitination. DNA methylation was measured using methylspecific polymerase chain reaction assay. MEASUREMENTS AND MAIN RESULTS Studies exploring the linkage of GADD45a with mechanical stress and Akt regulation revealed VILI challenged GADD45a(-/-) mice to have significantly reduced lung injury on overexpression of Akt1 transgene. Increased mechanical stress with 18% CS in EC induced Akt phosphorylation via E3 ligase tumor necrosis factor receptor–associated factor 6 (TRAF6)–mediated Akt K63 ubiquitination resulting in Akt trafficking and activation at the membrane. GADD45a is essential to this process because GADD45a silenced endothelial cells and GADD45a(-/-) mice exhibited increased Akt K48 ubiquitination leading to proteasomal degradation. These events involve loss of ubiquitin carboxyl terminal hydrolase 1(UCHL1), a deubiquitinating enzyme that normally removes K48 polyubiquitin chains bound to Akt thus promoting Akt K63 ubiquitination. Loss of GADD45a significantly reduces UCHL1 expression via UCHL1 promoter methylation resulting in increased Akt K48 ubiquitination and reduced Akt levels. CONCLUSIONS These studies highlight a novel role for GADD45a in the regulation of site-specific Akt ubiquitination and activation and implicate a significant functional role for GADD45a in the clinical predisposition to VILI.
Collapse
Affiliation(s)
- Sumegha Mitra
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Saad Sammani
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Ting Wang
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - David L. Boone
- Section of Gastroenterology, University of Chicago, Chicago, Illinois; and
| | - Nuala J. Meyer
- Division of Pulmonary, Allergy, and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven M. Dudek
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Liliana Moreno-Vinasco
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Joe G. N. Garcia
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Jeffrey R. Jacobson
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|