51
|
Reichhardt M, Holmskov U, Meri S. SALSA—A dance on a slippery floor with changing partners. Mol Immunol 2017; 89:100-110. [DOI: 10.1016/j.molimm.2017.05.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 05/31/2017] [Accepted: 05/31/2017] [Indexed: 02/06/2023]
|
52
|
Garay J, Piazuelo MB, Lopez-Carrillo L, Leal YA, Majumdar S, Li L, Cruz-Rodriguez N, Serrano-Gomez SJ, Busso CS, Schneider BG, Delgado AG, Bravo LE, Crist AM, Meadows SM, Camargo MC, Wilson KT, Correa P, Zabaleta J. Increased expression of deleted in malignant brain tumors (DMBT1) gene in precancerous gastric lesions: Findings from human and animal studies. Oncotarget 2017; 8:47076-47089. [PMID: 28423364 PMCID: PMC5564545 DOI: 10.18632/oncotarget.16792] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/16/2017] [Indexed: 12/21/2022] Open
Abstract
Helicobacter pylori infection triggers a cascade of inflammatory stages that may lead to the appearance of non-atrophic gastritis, multifocal atrophic, intestinal metaplasia, dysplasia, and cancer. Deleted in malignant brain tumors 1 (DMBT1) belongs to the group of secreted scavenger receptor cysteine-rich proteins and is considered to be involved in host defense by binding to pathogens. Initial studies showed its deletion and loss of expression in a variety of tumors but the role of this gene in tumor development is not completely understood. Here, we examined the role of DMBT1 in gastric precancerous lesions in Caucasian, African American and Hispanic individuals as well as in the development of gastric pathology in a mouse model of H. pylori infection. We found that in 3 different populations, mucosal DMBT1 expression was significantly increased (2.5 fold) in individuals with dysplasia compared to multifocal atrophic gastritis without intestinal metaplasia; the increase was also observed in individuals with advanced gastritis and positive H. pylori infection. In our animal model, H. pylori infection of Dmbt1-/- mice resulted in significantly higher levels of gastritis, more extensive mucous metaplasia and reduced Il33 expression levels in the gastric mucosa compared to H. pylori-infected wild type mice. Our data in the animal model suggest that in response to H. pylori infection DMBT1 may mediate mucosal protection reducing the risk of developing gastric precancerous lesions. However, the increased expression in human gastric precancerous lesions points to a more complex role of DMBT1 in gastric carcinogenesis.
Collapse
Affiliation(s)
- Jone Garay
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, LA, USA
| | - M Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Yelda A Leal
- Unidad de Investigación Médica Yucatán de la Unidad Médica de Alta Especialidad (UMAE) del Instituto Mexicano del Seguro Social (IMSS), Yucatán, Mexico
| | - Sumana Majumdar
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, LA, USA
| | - Li Li
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, LA, USA
| | - Nataly Cruz-Rodriguez
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, LA, USA
- Pontificia Universidad Javeriana, Bogotá, Colombia
- Grupo de Investigacion en Biología del Cáncer, Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Silvia J Serrano-Gomez
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, LA, USA
- Pontificia Universidad Javeriana, Bogotá, Colombia
- Grupo de Investigacion en Biología del Cáncer, Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Carlos S Busso
- Department of Otorhinolaryngology, LSUHSC, New Orleans, LA, USA
| | - Barbara G Schneider
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alberto G Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Luis E Bravo
- Department of Pathology, Universidad del Valle, Cali, Colombia
| | - Angela M Crist
- Department of Cell and Molecular Biology Tulane University, New Orleans LA, USA
| | - Stryder M Meadows
- Department of Cell and Molecular Biology Tulane University, New Orleans LA, USA
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Keith T Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Pelayo Correa
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jovanny Zabaleta
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, LA, USA
- Department of Pediatrics, LSUHSC, New Orleans, LA, USA
| |
Collapse
|
53
|
Intraspecific differences in molecular stress responses and coral pathobiome contribute to mortality under bacterial challenge in Acropora millepora. Sci Rep 2017; 7:2609. [PMID: 28572677 PMCID: PMC5454005 DOI: 10.1038/s41598-017-02685-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/18/2017] [Indexed: 11/13/2022] Open
Abstract
Disease causes significant coral mortality worldwide; however, factors responsible for intraspecific variation in disease resistance remain unclear. We exposed fragments of eight Acropora millepora colonies (genotypes) to putatively pathogenic bacteria (Vibrio spp.). Genotypes varied from zero to >90% mortality, with bacterial challenge increasing average mortality rates 4–6 fold and shifting the microbiome in favor of stress-associated taxa. Constitutive immunity and subsequent immune and transcriptomic responses to the challenge were more prominent in high-mortality individuals, whereas low-mortality corals remained largely unaffected and maintained expression signatures of a healthier condition (i.e., did not launch a large stress response). Our results suggest that lesions appeared due to changes in the coral pathobiome (multiple bacterial species associated with disease) and general health deterioration after the biotic disturbance, rather than the direct activity of any specific pathogen. If diseases in nature arise because of weaknesses in holobiont physiology, instead of the virulence of any single etiological agent, environmental stressors compromising coral condition might play a larger role in disease outbreaks than is currently thought. To facilitate the diagnosis of compromised individuals, we developed and independently cross-validated a biomarker assay to predict mortality based on genes whose expression in asymptomatic individuals coincides with mortality rates.
Collapse
|
54
|
Goeppert B, Roessler S, Becker N, Zucknick M, Vogel MN, Warth A, Pathil-Warth A, Mehrabi A, Schirmacher P, Mollenhauer J, Renner M. DMBT1 expression in biliary carcinogenesis with correlation of clinicopathological data. Histopathology 2017; 70:1064-1071. [PMID: 28130841 DOI: 10.1111/his.13175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 01/18/2017] [Accepted: 01/24/2017] [Indexed: 12/31/2022]
Abstract
AIMS Deleted in malignant brain tumours 1 (DMBT1) exerts functions in the regulation of epithelial differentiation and inflammation and has been proposed as a tumour suppressor. Because chronic inflammation is a hallmark of cholangiocarcinogenesis, the aim of this study was to investigate the expression of DMBT1 in biliary tract cancer (BTC) and to correlate this expression with clinicopathological data. METHODS AND RESULTS The expression of DMBT1 protein was examined immunohistochemically in 157 BTC patients [41 intrahepatic (ICC), 60 extrahepatic cholangiocarcinomas (ECC) and 56 adenocarcinomas of the gallbladder (GBAC)]. Additionally, 56 samples of high-grade biliary intraepithelial neoplasia (BilIN 3) and 92 corresponding samples of histological non-neoplastic biliary tract tissues were included. DMBT1 expression was increased significantly in BilIN 3 compared to normal tissue (P < 0.0001) and BTC (P < 0.0001). BTC showed no significant difference in DMBT1 expression compared to non-neoplastic biliary tissue (P = 0.315). Absent DMBT1 expression in non-neoplastic biliary tissue of BTC patients was associated with poorer survival (P = 0.027). DMBT1 expression was correlated significantly with patients' age (P < 0.001). CONCLUSION DMBT1 is expressed differently in cholangiocarcinogenesis and poorer patients' survival rates are associated with absent DMBT1 expression in non-neoplastic biliary tissue, suggesting a tumour-suppressive role of DMBT1 in early cholangiocarcinogenesis.
Collapse
Affiliation(s)
| | | | - Natalia Becker
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manuela Zucknick
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Oslo Center for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Monika N Vogel
- Diagnostic and Interventional Radiology, Thoraxklinik, University Hospital Heidelberg, Germany
| | - Arne Warth
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Anita Pathil-Warth
- Department of Internal Medicine IV, Gastroenterology and Hepatology, University Hospital Heidelberg, Germany
| | - Arianeb Mehrabi
- Department of General Visceral and Transplantation Surgery, University Hospital Heidelberg, Germany
| | | | - Jan Mollenhauer
- Molecular Oncology and Lundbeckfonden Center of Excellence NanoCAN, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Marcus Renner
- Institute of Pathology, University Hospital, Heidelberg, Germany
| |
Collapse
|
55
|
Loux SC, Scoggin KE, Troedsson MHT, Squires EL, Ball BA. Characterization of the cervical mucus plug in mares. Reproduction 2017; 153:197-210. [DOI: 10.1530/rep-16-0396] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/24/2016] [Accepted: 11/14/2016] [Indexed: 02/06/2023]
Abstract
The cervical mucus plug (CMP) is believed to play an integral role in the maintenance of pregnancy in the mare, primarily by inhibiting microbial entry. Unfortunately, very little is known about its composition or origin. To determine the proteomic composition of the CMP, we collected CMPs from mares (n = 4) at 9 months of gestation, and proteins were subsequently analyzed by nano-LC–MS/MS. Results were searched against EquCab2.0, and proteomic pathways were predicted by Ingenuity Pathway Analysis. Histologic sections of the CMP were stained with H&E and PAS. To identify the origin of highly abundant proteins in the CMP, we performed qPCR on endometrial and cervical mucosal mRNA from mares in estrus, diestrus as well as mares at 4 and 10 m gestation on transcripts for lactotransferrin, uterine serpin 14, uteroglobin, uteroferrin, deleted in malignant brain tumors 1 and mucins 4, 5b and 6. Overall, we demonstrated that the CMP is composed of a complex milieu of proteins during late gestation, many of which play an important role in immune function. Proteins traditionally considered to be endometrial proteins were found to be produced by the cervical mucosa suggesting that the primary source of the CMP is the cervical mucosa itself. In summary, composition of the equine CMP is specifically regulated not only during pregnancy but also throughout the estrous cycle. The structural and compositional changes serve to provide both a structural barrier as well as a physiological barrier during pregnancy to prevent infection of the fetus and fetal membranes.
Collapse
|
56
|
Martino JV, Van Limbergen J, Cahill LE. The Role of Carrageenan and Carboxymethylcellulose in the Development of Intestinal Inflammation. Front Pediatr 2017; 5:96. [PMID: 28507982 PMCID: PMC5410598 DOI: 10.3389/fped.2017.00096] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 04/13/2017] [Indexed: 12/19/2022] Open
Abstract
Although the exact pathophysiology remains unknown, the development of inflammatory bowel disease (IBD) is influenced by the interplay between genetics, the immune system, and environmental factors such as diet. The commonly used food additives, carrageenan and carboxymethylcellulose (CMC), are used to develop intestinal inflammation in animal models. These food additives are excluded from current dietary approaches to induce disease remission in Crohn's disease such as exclusive enteral nutrition (EEN) using a polymeric formula. By reviewing the existing scientific literature, this review aims to discuss the role that carrageenan and CMC may play in the development of IBD. Animal studies consistently report that carrageenan and CMC induce histopathological features that are typical of IBD while altering the microbiome, disrupting the intestinal epithelial barrier, inhibiting proteins that provide protection against microorganisms, and stimulating the elaboration of pro-inflammatory cytokines. Similar trials directly assessing the influence of carrageenan and CMC in humans are of course unethical to conduct, but recent studies of human epithelial cells and the human microbiome support the findings from animal studies. Carrageenan and CMC may trigger or magnify an inflammatory response in the human intestine but are unlikely to be identified as the sole environmental factor involved in the development of IBD or in disease recurrence after treatment. However, the widespread use of carrageenan and CMC in foods consumed by the pediatric population in a "Western" diet is on the rise alongside a corresponding increase in IBD incidence, and questions are being raised about the safety of frequent usage of these food additives. Therefore, further research is warranted to elucidate the role of carrageenan and CMC in intestinal inflammation, which may help identify novel nutritional strategies that hinder the development of the disease or prevent disease relapse post-EEN treatment.
Collapse
Affiliation(s)
- John Vincent Martino
- Pediatric Gastroenterology, Hepatology and Nutrition, IWK Health Centre, Halifax, NS, Canada
| | - Johan Van Limbergen
- Pediatric Gastroenterology, Hepatology and Nutrition, IWK Health Centre, Halifax, NS, Canada.,Medicine, Dalhousie University, Halifax, NS, Canada
| | - Leah E Cahill
- Medicine, Dalhousie University, Halifax, NS, Canada.,Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
57
|
Abstract
Trefoil factor (TFF) peptides, with a 40-amino acid motif and including six conserved cysteine residues that form intramolecular disulfide bonds, are a family of mucin-associated secretory molecules mediating many physiological roles that maintain and restore gastrointestinal (GI) mucosal homeostasis. TFF peptides play important roles in response to GI mucosal injury and inflammation. In response to acute GI mucosal injury, TFF peptides accelerate cell migration to seal the damaged area from luminal contents, whereas chronic inflammation leads to increased TFF expression to prevent further progression of disease. Although much evidence supports the physiological significance of TFF peptides in mucosal defenses, the molecular and cellular mechanisms of TFF peptides in the GI epithelium remain largely unknown. In this review, we summarize the functional roles of TFF1, 2, and 3 and illustrate their action mechanisms, focusing on defense mechanisms in the GI tract.
Collapse
Affiliation(s)
- Eitaro Aihara
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| | - Kristen A Engevik
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| | - Marshall H Montrose
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| |
Collapse
|
58
|
Transient and Prolonged Response of Chicken Cecum Mucosa to Colonization with Different Gut Microbiota. PLoS One 2016; 11:e0163932. [PMID: 27685470 PMCID: PMC5042506 DOI: 10.1371/journal.pone.0163932] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/16/2016] [Indexed: 12/15/2022] Open
Abstract
In this study we determined protein and gene expression in the caeca of newly hatched chickens inoculated with cecal contents sourced from hens of different ages. Over 250 proteins exhibited modified expression levels in response to microbiota inoculation. The most significant inductions were observed for ISG12-2, OASL, ES1, LYG2, DMBT1-L, CDD, ANGPTL6, B2M, CUZD1, IgM and Ig lambda chain. Of these, ISG12-2, ES1 and both immunoglobulins were expressed at lower levels in germ-free chickens compared to conventional chickens. In contrast, CELA2A, BRT-2, ALDH1A1, ADH1C, AKR1B1L, HEXB, ALDH2, ALDOB, CALB1 and TTR were expressed at lower levels following inoculation of microbiota. When chicks were given microbiota preparations from different age donors, the recipients mounted differential responses to the inoculation which also differed from the response profile in naturally colonised birds. For example, B2M, CUZD1 and CELA2A responded differently to the inoculation with microbiota of 4- or 40-week-old hens. The increased or decreased gene expression could be recorded 6 weeks after the inoculation of newly hatched chickens. To characterise the proteins that may directly interact with the microbiota we characterised chicken proteins that co-purified with the microbiota and identified a range of host proteins including CDD, ANGPTL6, DMBT1-L, MEP1A and Ig lambda. We propose that induction of ISG12-2 results in reduced apoptosis of host cells exposed to the colonizing commensal microbiota and that CDD, ANGPTL6, DMBT1-L, MEP1A and Ig lambda reduce contact of luminal microbiota with the gut epithelium thereby reducing the inflammatory response.
Collapse
|
59
|
Müller H, Weiss C, Renner M, Felderhoff-Müser U, Mollenhauer J. DMBT1 promotes basal and meconium-induced nitric oxide production in human lung epithelial cells in vitro. Histochem Cell Biol 2016; 147:389-397. [DOI: 10.1007/s00418-016-1493-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2016] [Indexed: 10/21/2022]
|
60
|
Fuess LE, Pinzόn C JH, Weil E, Mydlarz LD. Associations between transcriptional changes and protein phenotypes provide insights into immune regulation in corals. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 62:17-28. [PMID: 27109903 DOI: 10.1016/j.dci.2016.04.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 06/05/2023]
Abstract
Disease outbreaks in marine ecosystems have driven worldwide declines of numerous taxa, including corals. Some corals, such as Orbicella faveolata, are particularly susceptible to disease. To explore the mechanisms contributing to susceptibility, colonies of O. faveolata were exposed to immune challenge with lipopolysaccharides. RNA sequencing and protein activity assays were used to characterize the response of corals to immune challenge. Differential expression analyses identified 17 immune-related transcripts that varied in expression post-immune challenge. Network analyses revealed several groups of transcripts correlated to immune protein activity. Several transcripts, which were annotated as positive regulators of immunity were included in these groups, and some were downregulated following immune challenge. Correlations between expression of these transcripts and protein activity results further supported the role of these transcripts in positive regulation of immunity. The observed pattern of gene expression and protein activity may elucidate the processes contributing to the disease susceptibility of species like O. faveolata.
Collapse
Affiliation(s)
- Lauren E Fuess
- Department of Biology, University of Texas Arlington, Arlington, TX, USA
| | - Jorge H Pinzόn C
- Department of Biology, University of Texas Arlington, Arlington, TX, USA
| | - Ernesto Weil
- Department of Marine Sciences, University of Puerto Rico, Mayagüez, PR, USA
| | - Laura D Mydlarz
- Department of Biology, University of Texas Arlington, Arlington, TX, USA.
| |
Collapse
|
61
|
Abstract
Survival after lung transplantation is limited in large part due to the high incidence of chronic rejection, known as chronic lung allograft dysfunction (CLAD). Pulmonary infections are a frequent complication in lung transplant recipients, due both to immunosuppressive medications and constant exposure of the lung allograft to the external environment via the airways. Infection is a recognized risk factor for the development of CLAD, and both acute infection and chronic lung allograft colonization with microorganisms increase the risk for CLAD. Acute infection by community acquired respiratory viruses, and the bacteria Pseudomonas aeruginosa and Staphylococcus aureus are increasingly recognized as important risk factors for CLAD. Colonization by the fungus Aspergillus may also augment the risk of CLAD. Fostering this transition from healthy lung to CLAD in each of these infectious episodes is the persistence of an inflammatory lung allograft environment.
Collapse
Affiliation(s)
- Aric L Gregson
- Division of Infectious Diseases, Department of Medicine, University of California, Box 957119, Warren Hall 14-154, Los Angeles, CA, 90995-7119, USA.
| |
Collapse
|
62
|
Gregson AL, Hoji A, Injean P, Poynter ST, Briones C, Palchevskiy V, Weigt SS, Shino MY, Derhovanessian A, Sayah D, Saggar R, Ross D, Ardehali A, Lynch JP, Belperio JA. Altered Exosomal RNA Profiles in Bronchoalveolar Lavage from Lung Transplants with Acute Rejection. Am J Respir Crit Care Med 2016. [PMID: 26308930 DOI: 10.1164/rccm.201503-0558oc].] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The mechanism by which acute allograft rejection leads to chronic rejection remains poorly understood despite its common occurrence. Exosomes, membrane vesicles released from cells within the lung allograft, contain a diverse array of biomolecules that closely reflect the biologic state of the cell and tissue from which they are released. Exosome transcriptomes may provide a better understanding of the rejection process. Furthermore, biomarkers originating from this transcriptome could provide timely and sensitive detection of acute cellular rejection (AR), reducing the incidence of severe AR and chronic lung allograft dysfunction and improving outcomes. OBJECTIVES To provide an in-depth analysis of the bronchoalveolar lavage fluid exosomal shuttle RNA population after lung transplantation and evaluate for differential expression between acute AR and quiescence. METHODS Serial bronchoalveolar lavage specimens were ultracentrifuged to obtain the exosomal pellet for RNA extraction, on which RNA-Seq was performed. MEASUREMENTS AND MAIN RESULTS AR demonstrates an intense inflammatory environment, skewed toward both innate and adaptive immune responses. Novel, potential upstream regulators identified offer potential therapeutic targets. CONCLUSIONS Our findings validate bronchoalveolar lavage fluid exosomal shuttle RNA as a source for understanding the pathophysiology of AR and for biomarker discovery in lung transplantation.
Collapse
Affiliation(s)
- Aric L Gregson
- 1 Division of Infectious Diseases, Department of Medicine
| | - Aki Hoji
- 2 Department of Transplantation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Patil Injean
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Steven T Poynter
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Claudia Briones
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Vyacheslav Palchevskiy
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - S Sam Weigt
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Michael Y Shino
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Ariss Derhovanessian
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - David Sayah
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Rajan Saggar
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - David Ross
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Abbas Ardehali
- 4 Division of Cardiothoracic Surgery, Department of Surgery, University of California, Los Angeles, California; and
| | - Joseph P Lynch
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - John A Belperio
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| |
Collapse
|
63
|
Gregson AL, Hoji A, Injean P, Poynter ST, Briones C, Palchevskiy V, Weigt SS, Shino MY, Derhovanessian A, Sayah D, Saggar R, Ross D, Ardehali A, Lynch JP, Belperio JA. Altered Exosomal RNA Profiles in Bronchoalveolar Lavage from Lung Transplants with Acute Rejection. Am J Respir Crit Care Med 2016; 192:1490-503. [PMID: 26308930 DOI: 10.1164/rccm.201503-0558oc] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
RATIONALE The mechanism by which acute allograft rejection leads to chronic rejection remains poorly understood despite its common occurrence. Exosomes, membrane vesicles released from cells within the lung allograft, contain a diverse array of biomolecules that closely reflect the biologic state of the cell and tissue from which they are released. Exosome transcriptomes may provide a better understanding of the rejection process. Furthermore, biomarkers originating from this transcriptome could provide timely and sensitive detection of acute cellular rejection (AR), reducing the incidence of severe AR and chronic lung allograft dysfunction and improving outcomes. OBJECTIVES To provide an in-depth analysis of the bronchoalveolar lavage fluid exosomal shuttle RNA population after lung transplantation and evaluate for differential expression between acute AR and quiescence. METHODS Serial bronchoalveolar lavage specimens were ultracentrifuged to obtain the exosomal pellet for RNA extraction, on which RNA-Seq was performed. MEASUREMENTS AND MAIN RESULTS AR demonstrates an intense inflammatory environment, skewed toward both innate and adaptive immune responses. Novel, potential upstream regulators identified offer potential therapeutic targets. CONCLUSIONS Our findings validate bronchoalveolar lavage fluid exosomal shuttle RNA as a source for understanding the pathophysiology of AR and for biomarker discovery in lung transplantation.
Collapse
Affiliation(s)
- Aric L Gregson
- 1 Division of Infectious Diseases, Department of Medicine
| | - Aki Hoji
- 2 Department of Transplantation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Patil Injean
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Steven T Poynter
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Claudia Briones
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Vyacheslav Palchevskiy
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - S Sam Weigt
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Michael Y Shino
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Ariss Derhovanessian
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - David Sayah
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Rajan Saggar
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - David Ross
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Abbas Ardehali
- 4 Division of Cardiothoracic Surgery, Department of Surgery, University of California, Los Angeles, California; and
| | - Joseph P Lynch
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - John A Belperio
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| |
Collapse
|
64
|
Reichhardt MP, Meri S. SALSA: A Regulator of the Early Steps of Complement Activation on Mucosal Surfaces. Front Immunol 2016; 7:85. [PMID: 27014265 PMCID: PMC4781872 DOI: 10.3389/fimmu.2016.00085] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/22/2016] [Indexed: 12/11/2022] Open
Abstract
Complement is present mainly in blood. However, following mechanical damage or inflammation, serous exudates enter the mucosal surfaces. Here, the complement proteins interact with other endogenous molecules to keep microbes from entering the parenteral tissues. One of the mucosal proteins known to interact with the early complement components of both the classical and the lectin pathway is the salivary scavenger and agglutinin (SALSA). SALSA is also known as deleted in malignant brain tumors 1 and gp340. It is found both attached to the epithelium and secreted into the surrounding fluids of most mucosal surfaces. SALSA has been shown to bind directly to C1q, mannose-binding lectin, and the ficolins. Through these interactions SALSA regulates activation of the complement system. In addition, SALSA interacts with surfactant proteins A and D, secretory IgA, and lactoferrin. Ulcerative colitis and Crohn's disease are examples of diseases, where complement activation in mucosal tissues may occur. This review describes the latest advances in our understanding of how the early complement components interact with the SALSA molecule. Furthermore, we discuss how these interactions may affect disease propagation on mucosal surfaces in immunological and inflammatory diseases.
Collapse
Affiliation(s)
- Martin Parnov Reichhardt
- Immunobiology Research Program, Research Programs Unit, Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki , Helsinki , Finland
| | - Seppo Meri
- Immunobiology Research Program, Research Programs Unit, Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki , Helsinki , Finland
| |
Collapse
|
65
|
Reichhardt MP, Jarva H, Lokki AI, Laivuori H, FINNPEC study group, Vuorela P, Loimaranta V, Glasner A, Siwetz M, Huppertz B, Meri S. The Salivary Scavenger and Agglutinin (SALSA) in Healthy and Complicated Pregnancy. PLoS One 2016; 11:e0147867. [PMID: 26828433 PMCID: PMC4734712 DOI: 10.1371/journal.pone.0147867] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 01/08/2016] [Indexed: 11/22/2022] Open
Abstract
Pre-eclampsia is a leading cause of maternal and perinatal morbidity and mortality worldwide. The etiology is not clear, but an immune attack towards components of placenta or fetus has been indicated. This involves activation of the complement system in the placenta. We have previously described the presence of the complement-regulating protein salivary scavenger and agglutinin (SALSA) in amniotic fluid. In this study we investigated the potential role of SALSA in pregnancy by analyzing its presence in amniotic fluid and placental tissue during healthy and complicated pregnancies. SALSA levels in amniotic fluid increased during pregnancy. Before 20 weeks of gestation the levels were slightly higher in patients who later developed pre-eclampsia than in gestation age-matched controls. In the placenta of pre-eclamptic patients syncytial damage is often followed by the formation of fibrinoid structures. SALSA was found clustered into these fibrinoid structures in partial co-localization with complement C1q and fibronectin. In vitro analysis showed direct protein binding of SALSA to fibronectin. SALSA binds also to fibrin/fibrinogen but did not interfere with the blood clotting process in vitro. Thus, in addition to antimicrobial defense and epithelial differentiation, the data presented here suggest that SALSA, together with fibronectin and C1q, may be involved in the containment of injured placental structures into fibrinoids.
Collapse
Affiliation(s)
- Martin Parnov Reichhardt
- Immunobiology Research Program, Research Programs Unit, and Department of Bacteriology & Immunology, Medical Faculty, University of Helsinki, Helsinki, Finland
- * E-mail: (MPR); (SM)
| | - Hanna Jarva
- Immunobiology Research Program, Research Programs Unit, and Department of Bacteriology & Immunology, Medical Faculty, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Laboratory (HUSLAB), Helsinki, Finland
| | - Anna Inkeri Lokki
- Immunobiology Research Program, Research Programs Unit, and Department of Bacteriology & Immunology, Medical Faculty, University of Helsinki, Helsinki, Finland
- Medical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hannele Laivuori
- Medical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | | | - Piia Vuorela
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Obstetrics and Gynecology, Porvoo Hospital, Porvoo, Finland
| | - Vuokko Loimaranta
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | | | - Monika Siwetz
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Berthold Huppertz
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
- Biobank Graz, Organizational Unit of Research Infrastructure, Medical University of Graz, Graz, Austria
| | - Seppo Meri
- Immunobiology Research Program, Research Programs Unit, and Department of Bacteriology & Immunology, Medical Faculty, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Laboratory (HUSLAB), Helsinki, Finland
- * E-mail: (MPR); (SM)
| |
Collapse
|
66
|
Copy number variation of scavenger-receptor cysteine-rich domains within DMBT1 and Crohn's disease. Eur J Hum Genet 2016; 24:1294-300. [PMID: 26813944 PMCID: PMC4851238 DOI: 10.1038/ejhg.2015.280] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 12/08/2015] [Accepted: 12/09/2015] [Indexed: 12/21/2022] Open
Abstract
Previous work has shown that the gene DMBT1, which encodes a large secreted epithelial glycoprotein known as salivary agglutinin, gp340, hensin or muclin, is an innate immune defence protein that binds bacteria. A deletion variant of DMBT1 has been previously associated with Crohn's disease, and a DMBT1−/− knockout mouse has increased levels of colitis induced by dextran sulphate. DMBT1 has a complex copy number variable structure, with two, independent, rapidly mutating copy number variable regions, called CNV1 and CNV2. Because the copy number variable regions are predicted to affect the number of bacteria-binding domains, different alleles may alter host–microbe interactions in the gut. Our aim was to investigate the role of this complex variation in susceptibility to Crohn's disease by assessing the previously reported association. We analysed the association of both copy number variable regions with presence of Crohn's disease, and its severity, on three case–control cohorts. We also reanalysed array comparative genomic hybridisation data (aCGH) from a large case–control cohort study for both copy number variable regions. We found no association with a linear increase in copy number, nor when the CNV1 is regarded as presence or absence of a deletion allele. Taken together, we show that the DMBT1 CNV does not affect susceptibility to Crohn's disease, at least in Northern Europeans.
Collapse
|
67
|
Liu Z, Fan L, Xiao H, Cao C. A multiple covalent crosslinked soft hydrogel for bioseparation. Chem Commun (Camb) 2016; 52:3247-50. [DOI: 10.1039/c5cc09944g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A multiple covalent crosslinked gel of poly(acrylamide–acrylic acid) was synthesized by using acrylic acid and acrylamide both as a monomer and as a covalent crosslinker.
Collapse
Affiliation(s)
- Zhen Liu
- Laboratory of Bioseparation and Analytical Biochemistry
- State Key Laboratory of Microbial Metabolism
- School of Life Science and Biotechnology
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Liuyin Fan
- Laboratory of Bioseparation and Analytical Biochemistry
- State Key Laboratory of Microbial Metabolism
- School of Life Science and Biotechnology
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Hua Xiao
- Laboratory of Bioseparation and Analytical Biochemistry
- State Key Laboratory of Microbial Metabolism
- School of Life Science and Biotechnology
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Chengxi Cao
- Laboratory of Bioseparation and Analytical Biochemistry
- State Key Laboratory of Microbial Metabolism
- School of Life Science and Biotechnology
- Shanghai Jiao Tong University
- Shanghai 200240
| |
Collapse
|
68
|
Gregson AL, Hoji A, Injean P, Poynter ST, Briones C, Palchevskiy V, Weigt SS, Shino MY, Derhovanessian A, Sayah D, Saggar R, Ross D, Ardehali A, Lynch JP, Belperio JA. Altered Exosomal RNA Profiles in Bronchoalveolar Lavage from Lung Transplants with Acute Rejection. Am J Respir Crit Care Med 2015. [PMID: 26308930 DOI: 10.1164/rccm.201503-0558oc]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
RATIONALE The mechanism by which acute allograft rejection leads to chronic rejection remains poorly understood despite its common occurrence. Exosomes, membrane vesicles released from cells within the lung allograft, contain a diverse array of biomolecules that closely reflect the biologic state of the cell and tissue from which they are released. Exosome transcriptomes may provide a better understanding of the rejection process. Furthermore, biomarkers originating from this transcriptome could provide timely and sensitive detection of acute cellular rejection (AR), reducing the incidence of severe AR and chronic lung allograft dysfunction and improving outcomes. OBJECTIVES To provide an in-depth analysis of the bronchoalveolar lavage fluid exosomal shuttle RNA population after lung transplantation and evaluate for differential expression between acute AR and quiescence. METHODS Serial bronchoalveolar lavage specimens were ultracentrifuged to obtain the exosomal pellet for RNA extraction, on which RNA-Seq was performed. MEASUREMENTS AND MAIN RESULTS AR demonstrates an intense inflammatory environment, skewed toward both innate and adaptive immune responses. Novel, potential upstream regulators identified offer potential therapeutic targets. CONCLUSIONS Our findings validate bronchoalveolar lavage fluid exosomal shuttle RNA as a source for understanding the pathophysiology of AR and for biomarker discovery in lung transplantation.
Collapse
Affiliation(s)
- Aric L Gregson
- 1 Division of Infectious Diseases, Department of Medicine
| | - Aki Hoji
- 2 Department of Transplantation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Patil Injean
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Steven T Poynter
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Claudia Briones
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Vyacheslav Palchevskiy
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - S Sam Weigt
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Michael Y Shino
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Ariss Derhovanessian
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - David Sayah
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Rajan Saggar
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - David Ross
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - Abbas Ardehali
- 4 Division of Cardiothoracic Surgery, Department of Surgery, University of California, Los Angeles, California; and
| | - Joseph P Lynch
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| | - John A Belperio
- 3 Division of Pulmonary, Critical Care, Allergy, and Immunology, Department of Medicine, and
| |
Collapse
|
69
|
Müller H, Renner M, Helmke BM, Mollenhauer J, Felderhoff-Müser U. Elevated DMBT1 levels in neonatal gastrointestinal diseases. Histochem Cell Biol 2015; 145:227-37. [DOI: 10.1007/s00418-015-1381-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2015] [Indexed: 01/01/2023]
|
70
|
Ray A, Dittel BN. Interrelatedness between dysbiosis in the gut microbiota due to immunodeficiency and disease penetrance of colitis. Immunology 2015. [PMID: 26211540 DOI: 10.1111/imm.12511] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The composition of the microbiome in health and disease has only recently become a major research focus. Although it is clear that an imbalance or dysbiosis in the microbiota is associated with disease, its interrelatedness to disease penetrance is largely unknown. Inflammatory bowel disease (IBD) is an excellent disease in which to explore these questions because of the extensive genetic studies identifying disease susceptibility loci and the ability to easily sample the intestinal microbiota in IBD patients due to the accessibility of stool samples. In addition, mouse models of IBD have contributed to our understanding of the interrelatedness of the gut microbiota and genes associated with IBD. The power of the mouse studies is that multiple colitis models exist that can be used in combination with genetically modified mice that harbour deficiencies in IBD susceptibility genes. Collectively, these studies revealed that bacterial dysbiosis does occur in human IBD and in mouse colitis models. In addition, with an emphasis on immune genes, the mouse studies provided evidence that specific immune regulatory proteins associated with IBD influence the gut microbiota in a manner consistent with disease penetrance. In this review, we will discuss studies in both humans and mice that demonstrate the impact of immunodeficiences in interleukin-10, interleukin-17, nucleotide-binding oligomerization domain (NOD) 2, NOD-like receptor proteins 3 and 6, Toll-like receptor or IgA have on the interrelatedness between the composition of the gut microbiota and disease penetrance of IBD and its mouse models.
Collapse
Affiliation(s)
- Avijit Ray
- BloodCenter of Wisconsin, Blood Research Institute, Milwaukee, WI, USA
| | - Bonnie N Dittel
- BloodCenter of Wisconsin, Blood Research Institute, Milwaukee, WI, USA
| |
Collapse
|
71
|
Mignon-Grasteau S, Narcy A, Rideau N, Chantry-Darmon C, Boscher MY, Sellier N, Chabault M, Konsak-Ilievski B, Le Bihan-Duval E, Gabriel I. Impact of Selection for Digestive Efficiency on Microbiota Composition in the Chicken. PLoS One 2015; 10:e0135488. [PMID: 26267269 PMCID: PMC4534097 DOI: 10.1371/journal.pone.0135488] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/22/2015] [Indexed: 01/23/2023] Open
Abstract
Objectives Feed efficiency and its digestive component, digestive efficiency, are key factors in the environmental impact and economic output of poultry production. The interaction between the host and intestinal microbiota has a crucial role in the determination of the ability of the bird to digest its food and to the birds’ feed efficiency. We therefore investigated the phenotypic and genetic relationships between birds’ efficiency and the composition of the cecal microbiota in a F2 cross between broiler lines divergently selected for their high or low digestive efficiency. Methods Analyses were performed on 144 birds with extreme feed efficiency values at 3 weeks, with feed conversion values of 1.41±0.05 and 2.02±0.04 in the efficient and non-efficient groups, respectively. The total numbers of Lactobacillus, L. salivarius, L. crispatus, C. coccoides, C. leptum and E. coli per gram of cecal content were measured. Results The two groups mainly differed in larger counts of Lactobacillus, L. salivarius and E. coli in less efficient birds. The equilibrium between bacterial groups was also affected, efficient birds showing higher C. leptum, C. coccoides and L. salivarius to E. coli ratios. The heritability of the composition of microbiota was also estimated and L. crispatus, C. leptum, and C. coccoides to E. coli ratios were moderately but significantly heritable (0.16 to 0.24). The coefficient of fecal digestive use of dry matter was genetically and positively correlated with L. crispatus, C. leptum, C. coccoides (0.50 to 0.76) and negatively with E. coli (-0.66). Lipid digestibility was negatively correlated with E. coli (-0.64), and AMEn positively correlated with C. coccoides and with the C. coccoides to Lactobacillus ratio (0.48 to 0.64). We also detected 14 Quantitative Trait Loci (QTL) for microbiota on the host genome, mostly on C. leptum and Lactobacillus. The QTL for C. leptum on GGA6 was close to genome-wide significance. This region mainly includes genes involved in anti-inflammatory responses and in the motility of the gastrointestinal tract.
Collapse
Affiliation(s)
| | - Agnès Narcy
- UR83 Recherches Avicoles, INRA, Nouzilly, France
| | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Deleted in malignant brain tumors 1 (DMBT1) elicits increased VEGF and decreased IL-6 production in type II lung epithelial cells. BMC Pulm Med 2015; 15:32. [PMID: 25885541 PMCID: PMC4426184 DOI: 10.1186/s12890-015-0027-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 03/24/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Deleted in malignant brain tumors 1 (DMBT1) is an innate defence protein expressed in the lungs of preterm infants and adults. Recent studies showed that DMBT1 is important in angiogenesis and can bind to different growth factors including VEGF. We aimed at examining relationships between VEGF and IL-6 levels to DMBT1 expression in the lungs of preterm and term infants and in lung epithelial cells in vitro. METHODS We examined by ELISA VEGF levels in 120 tracheal aspirates of 57 preterm and term infants and tested for correlation with different perinatal factors as well as with DMBT1 levels. To examine the effect of DMBT1 on VEGF and IL-6 expression we compared type II lung epithelial A549 cells stably transfected with a DMBT1 expression plasmid (DMBT1+ cells) to A549 cells stably transfected with an empty expression plasmid (DMBT1- cells). The concentrations of VEGF and IL-6 were determined via ELISA in the supernatant of the unstimulated cells and after stimulation with LPS, TNFα and Phorbol-12-myristate-13-acetate (PMA). RESULTS The VEGF levels in the tracheal aspirates of preterm and term infants were significantly correlated with DMBT1 levels (p = 0.0032), the postnatal age (p = 0.0073) and the presence of neonatal infection/sepsis (p = 0.0002). Unstimulated DMBT1+ A549 cells showed significantly higher VEGF expression (p = 0.0017) than DMBT1- cells. Significantly elevated VEGF levels were also confirmed for DMBT1+ cells after stimulation with TNFα (p = 0.0008), LPS (p = 0.0232) and PMA (p = 0.0025). The IL-6 levels were comparable in DMBT1+ versus DMBT1- cells without stimulation (p = 0.6028), but they were significantly reduced in DMBT1+ cells after stimulation with TNFα (p = 0.0003), LPS (p = 0.0088) and PMA (p = 0.0039). CONCLUSIONS The data indicate that DMBT1 promotes VEGF and suppresses IL-6 production in alveolar tissues, which could point to DMBT1 having a possible role in the transition from inflammation to regeneration and being a potentially useful clinical marker.
Collapse
|
73
|
Salazar KA, Joffe NR, Dinguirard N, Houde P, Castillo MG. Transcriptome analysis of the white body of the squid Euprymna tasmanica with emphasis on immune and hematopoietic gene discovery. PLoS One 2015; 10:e0119949. [PMID: 25775132 PMCID: PMC4361686 DOI: 10.1371/journal.pone.0119949] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 01/27/2015] [Indexed: 02/07/2023] Open
Abstract
In the mutualistic relationship between the squid Euprymna tasmanica and the bioluminescent bacterium Vibrio fischeri, several host factors, including immune-related proteins, are known to interact and respond specifically and exclusively to the presence of the symbiont. In squid and octopus, the white body is considered to be an immune organ mainly due to the fact that blood cells, or hemocytes, are known to be present in high numbers and in different developmental stages. Hence, the white body has been described as the site of hematopoiesis in cephalopods. However, to our knowledge, there are no studies showing any molecular evidence of such functions. In this study, we performed a transcriptomic analysis of white body tissue of the Southern dumpling squid, E. tasmanica. Our primary goal was to gain insights into the functions of this tissue and to test for the presence of gene transcripts associated with hematopoietic and immune processes. Several hematopoiesis genes including CPSF1, GATA 2, TFIID, and FGFR2 were found to be expressed in the white body. In addition, transcripts associated with immune-related signal transduction pathways, such as the toll-like receptor/NF-κβ, and MAPK pathways were also found, as well as other immune genes previously identified in E. tasmanica's sister species, E. scolopes. This study is the first to analyze an immune organ within cephalopods, and to provide gene expression data supporting the white body as a hematopoietic tissue.
Collapse
Affiliation(s)
- Karla A. Salazar
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Nina R. Joffe
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Nathalie Dinguirard
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Peter Houde
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Maria G. Castillo
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
- * E-mail:
| |
Collapse
|
74
|
Buckley KM, Rast JP. Diversity of animal immune receptors and the origins of recognition complexity in the deuterostomes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 49:179-189. [PMID: 25450907 DOI: 10.1016/j.dci.2014.10.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/01/2014] [Accepted: 10/20/2014] [Indexed: 06/04/2023]
Abstract
Invertebrate animals are characterized by extraordinary diversity in terms of body plan, life history and life span. The past impression that invertebrate immune responses are controlled by relatively simple innate systems is increasingly contradicted by genomic analyses that reveal significant evolutionary novelty and complexity. One accessible measure of this complexity is the multiplicity of genes encoding homologs of pattern recognition receptors. These multigene families vary significantly in size, and their sequence character suggests that they vary in function. At the same time, certain aspects of downstream signaling appear to be conserved. Here, we analyze five major classes of immune recognition receptors from newly available animal genome sequences. These include the Toll-like receptors (TLR), Nod-like receptors (NLR), SRCR domain scavenger receptors, peptidoglycan recognition proteins (PGRP), and Gram negative binding proteins (GNBP). We discuss innate immune complexity in the invertebrate deuterostomes, which was first recognized in sea urchins, within the wider context of emerging genomic information across animal phyla.
Collapse
MESH Headings
- Animals
- Biodiversity
- Evolution, Molecular
- Genetic Variation
- Genome/genetics
- Immunity, Innate/genetics
- Immunity, Innate/immunology
- Invertebrates/classification
- Invertebrates/genetics
- Invertebrates/immunology
- Multigene Family/genetics
- Multigene Family/immunology
- Phylogeny
- Receptors, Immunologic/classification
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Pattern Recognition/genetics
- Receptors, Pattern Recognition/immunology
- Receptors, Scavenger/genetics
- Receptors, Scavenger/immunology
- Species Specificity
- Toll-Like Receptors/genetics
- Toll-Like Receptors/immunology
Collapse
Affiliation(s)
- Katherine M Buckley
- Department of Immunology and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Sunnybrook Research Institute, Toronto, ON, Canada.
| | - Jonathan P Rast
- Department of Immunology and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
75
|
Xu AT, Li Y, Zhao D, Shen J, Xu XT, Qiao YQ, Zhu MM, Wang TR, Cui Y, Ai LY, Ran ZH. High suppressor of cytokine signaling-3 expression impairs STAT3-dependent protective effects of interleukin-22 in ulcerative colitis in remission. Inflamm Bowel Dis 2015; 21:241-250. [PMID: 25545374 DOI: 10.1097/mib.0000000000000267] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND High SOCS3 expression in intestinal epithelial cells (IECs) of patients with ulcerative colitis (UC) in remission reflects the shorter time to relapse. We investigated whether high SOCS3 increased risk for relapse through violating STAT3-dependent protective effects of interleukin (IL)-22 during UC remission. METHODS Expression of IL-22 and c-Myc in UC remission mucosa was analyzed by immunohistochemistry. Effects of IL-22 on migration and proliferation of IEC cell lines with enforced SOCS3 expression were assessed with wounding assay and CCK-8 assay, respectively. Influence of STAT3 interference and SOCS3 overexpression on IL-22-regulated expression of antimicrobial peptide and proliferation-related molecules, including DMBT1, c-Myc, Survivin, Bcl-2, and Bcl-xL, were performed with quantitative real-time polymerase chain reaction or Western blot. RESULTS Patients with UC in remission showed significantly more IL-22-positive immune cells, but no difference of epithelial c-Myc levels, in mucosa compared with healthy controls. Overexpression of SOCS3 nearly abolished IL-22-induced activation of STAT3. By inhibiting STAT3 signaling, SOCS3 influenced IL-22-induced expression of DMBT1, c-Myc, Survivin, and Bcl-2 as well as proliferation and migration processes in cultured IEC cell line. CONCLUSIONS SOCS3 overexpression impairs IL-22-mediated epithelial homeostasis and mucosal wound healing, which could be the mechanism for high SOCS3 IEC expression contributed early relapse of mucosal inflammation. Prevention of SOCS3 expression or enhancement of IL-22/STAT3 signaling in IEC seems to be rational therapeutic strategies for UC remission maintenance.
Collapse
Affiliation(s)
- An Tao Xu
- Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Hsu H, Chen C, Nenninger A, Holz L, Baldwin CL, Telfer JC. WC1 is a hybrid γδ TCR coreceptor and pattern recognition receptor for pathogenic bacteria. THE JOURNAL OF IMMUNOLOGY 2015; 194:2280-8. [PMID: 25632007 DOI: 10.4049/jimmunol.1402021] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
WC1 proteins are uniquely expressed on γδ T cells and belong to the scavenger receptor cysteine-rich (SRCR) superfamily. While present in variable, and sometimes high, numbers in the genomes of mammals and birds, in cattle there are 13 distinct genes (WC1-1 to WC1-13). All bovine WC1 proteins can serve as coreceptors for the TCR in a tyrosine phosphorylation dependent manner, and some are required for the γδ T cell response to Leptospira. We hypothesized that individual WC1 receptors encode Ag specificity via coligation of bacteria with the γδ TCR. SRCR domain binding was directly correlated with γδ T cell response, as WC1-3 SRCR domains from Leptospira-responsive cells, but not WC1-4 SRCR domains from Leptospira-nonresponsive cells, bound to multiple serovars of two Leptospira species, L. borgpetersenii, and L. interrogans. Three to five of eleven WC1-3 SRCR domains, but none of the eleven WC1-4 SRCR domains, interacted with Leptospira spp. and Borrelia burgdorferi, but not with Escherichia coli or Staphylococcus aureus. Mutational analysis indicated that the active site for bacterial binding in one of the SRCR domains is composed of amino acids in three discontinuous regions. Recombinant WC1 SRCR domains with the ability to bind leptospires inhibited Leptospira growth. Our data suggest that WC1 gene arrays play a multifaceted role in the γδ T cell response to bacteria, including acting as hybrid pattern recognition receptors and TCR coreceptors, and they may function as antimicrobials.
Collapse
Affiliation(s)
- Haoting Hsu
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003; and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA 01003
| | - Chuang Chen
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003; and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA 01003
| | - Ariel Nenninger
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003; and
| | - Lauren Holz
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003; and
| | - Cynthia L Baldwin
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003; and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA 01003
| | - Janice C Telfer
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003; and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA 01003
| |
Collapse
|
77
|
Lim C, Hammond CJ, Hingley ST, Balin BJ. Chlamydia pneumoniae infection of monocytes in vitro stimulates innate and adaptive immune responses relevant to those in Alzheimer's disease. J Neuroinflammation 2014; 11:217. [PMID: 25540075 PMCID: PMC4295513 DOI: 10.1186/s12974-014-0217-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/07/2014] [Indexed: 11/24/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a progressive neurodegenerative disorder in which infection with Chlamydia pneumoniae (Cpn) has been associated. Cpn is an obligate intracellular respiratory pathogen that may enter the central nervous system (CNS) following infection and trafficking of monocytes through the blood-brain barrier. Following this entry, these cells may secrete pro-inflammatory cytokines and chemokines that have been identified in the AD brain, which have been thought to contribute to AD neurodegeneration. The objectives of this work were: (i) to determine if Cpn infection influences monocyte gene transcript expression at 48 hours post-infection and (ii) to analyze whether pro-inflammatory cytokines are produced and secreted from these cells over 24 to 120 hours post-infection. Methods Gene transcription was analyzed by RT-PCR using an innate and adaptive immunity microarray with 84 genes organized into 5 functional categories: inflammatory response, host defense against bacteria, antibacterial humoral response, septic shock, and cytokines, chemokines and their receptors. Statistical analysis of the results was performed using the Student's t-test. P-values ≤ 0.05 were considered to be significant. ELISA was performed on supernatants from uninfected and Cpn-infected THP1 monocytes followed by statistical analysis with ANOVA. Results When Cpn-infected THP1 human monocytes were compared to control uninfected monocytes at 48 hours post-infection, 17 genes were found to have a significant 4-fold or greater expression, and no gene expression was found to be down-regulated. Furthermore, cytokine secretion (IL-1β, IL-6, IL-8) appears to be maintained for an extended period of infection. Conclusions Utilizing RT-PCR and ELISA techniques, our data demonstrate that Cpn infection of THP1 human monocytes promotes an innate immune response and suggests a potential role in the initiation of inflammation in sporadic/late-onset Alzheimer’s disease.
Collapse
Affiliation(s)
- Charles Lim
- Department of Bio-Medical Sciences, Center for Chronic Disorders of Aging, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA, 19131, USA.
| | - Christine J Hammond
- Department of Bio-Medical Sciences, Center for Chronic Disorders of Aging, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA, 19131, USA.
| | - Susan T Hingley
- Department of Bio-Medical Sciences, Center for Chronic Disorders of Aging, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA, 19131, USA.
| | - Brian J Balin
- Department of Bio-Medical Sciences, Center for Chronic Disorders of Aging, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA, 19131, USA.
| |
Collapse
|
78
|
Reichhardt MP, Jarva H, de Been M, Rodriguez JM, Jimenez Quintana E, Loimaranta V, Meindert de Vos W, Meri S. The Salivary Scavenger and Agglutinin in Early Life: Diverse Roles in Amniotic Fluid and in the Infant Intestine. THE JOURNAL OF IMMUNOLOGY 2014; 193:5240-8. [DOI: 10.4049/jimmunol.1401631] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
79
|
Abstract
BACKGROUND Many aspects of autoimmune disease are not well understood, including the specificities of autoimmune targets, and patterns of co-morbidity and cross-heritability across diseases. Prior work has provided evidence that somatic mutation caused by gene conversion and deletion at segmentally duplicated loci is relevant to several diseases. Simple tandem repeat (STR) sequence is highly mutable, both somatically and in the germ-line, and somatic STR mutations are observed under inflammation. RESULTS Protein-coding genes spanning STRs having markers of mutability, including germ-line variability, high total length, repeat count and/or repeat similarity, are evaluated in the context of autoimmunity. For the initiation of autoimmune disease, antigens whose autoantibodies are the first observed in a disease, termed primary autoantigens, are informative. Three primary autoantigens, thyroid peroxidase (TPO), phogrin (PTPRN2) and filaggrin (FLG), include STRs that are among the eleven longest STRs spanned by protein-coding genes. This association of primary autoantigens with long STR sequence is highly significant (p<3.0x10(-7)). Long STRs occur within twenty genes that are associated with sixteen common autoimmune diseases and atherosclerosis. The repeat within the TTC34 gene is an outlier in terms of length and a link with systemic lupus erythematosus is proposed. CONCLUSIONS The results support the hypothesis that many autoimmune diseases are triggered by immune responses to proteins whose DNA sequence mutates somatically in a coherent, consistent fashion. Other autoimmune diseases may be caused by coherent somatic mutations in immune cells. The coherent somatic mutation hypothesis has the potential to be a comprehensive explanation for the initiation of many autoimmune diseases.
Collapse
Affiliation(s)
- Kenneth Andrew Ross
- Department of Computer Science, Columbia University, New York, New York, United States of America
| |
Collapse
|
80
|
Miró-Julià C, Escoda-Ferran C, Carrasco E, Moeller JB, Vadekaer DF, Gao X, Paragas N, Oliver J, Holmskov U, Al-Awqati Q, Lozano F. Expression of the innate defense receptor S5D-SRCRB in the urogenital tract. ACTA ACUST UNITED AC 2014; 83:273-85. [DOI: 10.1111/tan.12330] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 01/28/2014] [Accepted: 02/11/2014] [Indexed: 12/19/2022]
Affiliation(s)
- C. Miró-Julià
- Grup d'Immunoreceptors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); Centre Esther Koplowitz; Barcelona Spain
| | - C. Escoda-Ferran
- Grup d'Immunoreceptors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); Centre Esther Koplowitz; Barcelona Spain
| | - E. Carrasco
- Grup d'Immunoreceptors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); Centre Esther Koplowitz; Barcelona Spain
| | - J. B. Moeller
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
| | - D. F. Vadekaer
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
| | - X. Gao
- Department of Medicine, College of Physicians and Surgeons; Columbia University; New York NY USA
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons; Columbia University; New York NY USA
| | - N. Paragas
- Department of Medicine, College of Physicians and Surgeons; Columbia University; New York NY USA
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons; Columbia University; New York NY USA
| | - J. Oliver
- Department of Medicine, College of Physicians and Surgeons; Columbia University; New York NY USA
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons; Columbia University; New York NY USA
| | - U. Holmskov
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
| | - Q. Al-Awqati
- Department of Medicine, College of Physicians and Surgeons; Columbia University; New York NY USA
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons; Columbia University; New York NY USA
| | - F. Lozano
- Grup d'Immunoreceptors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); Centre Esther Koplowitz; Barcelona Spain
- Servei d'Immunologia, Centre de Diagnòstic Biomèdic; Hospital Clínic de Barcelona; Barcelona Spain
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina; Universitat de Barcelona; Barcelona Spain
| |
Collapse
|
81
|
Lipinski S, Rosenstiel P. Debug Your Bugs - How NLRs Shape Intestinal Host-Microbe Interactions. Front Immunol 2013; 4:479. [PMID: 24409180 PMCID: PMC3873519 DOI: 10.3389/fimmu.2013.00479] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 12/09/2013] [Indexed: 12/14/2022] Open
Abstract
The host's ability to discriminate friend and foe and to establish a precise homeostasis with its associated microbiota is crucial for its survival and fitness. Among the mediators of intestinal host-microbe interactions, NOD-like receptor (NLR) proteins take center stage. They are present in the epithelial lining and innate immune cells that constantly monitor microbial activities at the intestinal barrier. Dysfunctional NLRs predispose to intestinal inflammation as well as sensitization to extra-intestinal immune-mediated diseases and are linked to the alteration of microbial communities. Here, we review advances in our understanding of their reciprocal relationship in the regulation of intestinal homeostasis and implications for intestinal health.
Collapse
Affiliation(s)
- Simone Lipinski
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| |
Collapse
|
82
|
Diegelmann J, Czamara D, Le Bras E, Zimmermann E, Olszak T, Bedynek A, Göke B, Franke A, Glas J, Brand S. Intestinal DMBT1 expression is modulated by Crohn's disease-associated IL23R variants and by a DMBT1 variant which influences binding of the transcription factors CREB1 and ATF-2. PLoS One 2013; 8:e77773. [PMID: 24223725 PMCID: PMC3818382 DOI: 10.1371/journal.pone.0077773] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 09/09/2013] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES DMBT is an antibacterial pattern recognition and scavenger receptor. In this study, we analyzed the role of DMBT1 single nucleotide polymorphisms (SNPs) regarding inflammatory bowel disease (IBD) susceptibility and examined their functional impact on transcription factor binding and downstream gene expression. METHODS Seven SNPs in the DMBT1 gene region were analyzed in 2073 individuals including 818 Crohn's disease (CD) patients and 972 healthy controls in two independent case-control panels. Comprehensive epistasis analyses for the known CD susceptibility genes NOD2, IL23R and IL27 were performed. The influence of IL23R variants on DMBT1 expression was analyzed. Functional analysis included siRNA transfection, quantitative PCR, western blot, electrophoretic mobility shift and luciferase assays. RESULTS IL-22 induces DMBT1 protein expression in intestinal epithelial cells dependent on STAT3, ATF-2 and CREB1. IL-22 expression-modulating, CD risk-associated IL23R variants influence DMBT1 expression in CD patients and DMBT1 levels are increased in the inflamed intestinal mucosa of CD patients. Several DMBT1 SNPs were associated with CD susceptibility. SNP rs2981804 was most strongly associated with CD in the combined panel (p = 3.0 × 10(-7), OR 1.42; 95% CI 1.24-1.63). All haplotype groups tested showed highly significant associations with CD (including omnibus P-values as low as 6.1 × 10(-18)). The most strongly CD risk-associated, non-coding DMBT1 SNP rs2981804 modifies the DNA binding sites for the transcription factors CREB1 and ATF-2 and the respective genomic region comprising rs2981804 is able to act as a transcriptional regulator in vitro. Intestinal DMBT1 expression is decreased in CD patients carrying the rs2981804 CD risk allele. CONCLUSION We identified novel associations of DMBT1 variants with CD susceptibility and discovered a novel functional role of rs2981804 in regulating DMBT1 expression. Our data suggest an important role of DMBT1 in CD pathogenesis.
Collapse
Affiliation(s)
- Julia Diegelmann
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University (LMU), Munich, Germany
- Department of Preventive Dentistry and Periodontology, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Darina Czamara
- Max-Planck-Institute for Psychiatry, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Emmanuelle Le Bras
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Eva Zimmermann
- Department of Preventive Dentistry and Periodontology, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Torsten Olszak
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University (LMU), Munich, Germany
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Andrea Bedynek
- Department of Clinical Chemistry, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Burkhard Göke
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - Jürgen Glas
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University (LMU), Munich, Germany
- Department of Preventive Dentistry and Periodontology, Ludwig-Maximilians-University (LMU), Munich, Germany
- Department of Human Genetics, Rheinisch-Westfälische Technische Hochschule (RWTH), Aachen, Germany
| | - Stephan Brand
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University (LMU), Munich, Germany
| |
Collapse
|
83
|
Barber C, Weissman D, Barnhart K, Dalvi M, Abrams WR, Malamud D. An electrochemiluminescence assay for gp340 (DMBT1). Anal Biochem 2013; 440:78-80. [PMID: 23727557 DOI: 10.1016/j.ab.2013.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 05/17/2013] [Accepted: 05/21/2013] [Indexed: 11/18/2022]
Abstract
Gp340 is a member of the scavenger receptor cysteine-rich family of innate immune molecules and also functions as a tumor suppressor. This study describes a picogram-level assay using electrochemiluminescence technology on the MesoScale Discovery platform. Antibodies were evaluated and the best pair was used to assay whole-mouth stimulated saliva and cervical/vaginal lavage. The assay was tested using specimens obtained from healthy volunteers to determine if gp340 concentration in saliva correlates with levels in vaginal lavage fluid. Interestingly, no correlation was determined between gp340 content in these two fluids.
Collapse
Affiliation(s)
- Cheryl Barber
- Department of Basic Sciences, College of Dentistry, New York University, New York, NY 10010, USA
| | | | | | | | | | | |
Collapse
|
84
|
Chu Y, Li J, Wu X, Hua Z, Wu Z. Identification of human immunodeficiency virus type 1 (HIV-1) gp120-binding sites on scavenger receptor cysteine rich 1 (SRCR1) domain of gp340. J Biomed Sci 2013; 20:44. [PMID: 23815775 PMCID: PMC3721995 DOI: 10.1186/1423-0127-20-44] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/26/2013] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND gp340, a member of scavenger receptor cysteine rich family encoded by Deleted in Malignant Brain Tumors 1 (DMBT1), is an important component in innate immune defense. The first scavenger receptor cysteine rich domain (SRCR1) of gp340 has been shown to inhibit HIV-1 infection through binding to the N-terminal flank of the V3 loop of HIV-1 gp120. RESULTS Through homology modeling and docking analysis of SRCR1 to a gp120-CD4-X5 antibody complex, we identified three loop regions containing polar or acidic residues that directly interacted with gp120. To confirm the docking prediction, a series of over-lapping peptides covering the SRCR1 sequence were synthesized and analyzed by gp120-peptide binding assay. Five peptides coincide with three loop regions showed the relative high binding index. An alanine substitution scan revealed that Asp34, Asp35, Asn96 and Glu101 in two peptides with the highest binding index are the critical residues in SRCR1 interaction with gp120. CONCLUSION We pinpointed the vital gp120-binding regions in SRCR1 and narrowed down the amino acids which play critical roles in contacting with gp120.
Collapse
Affiliation(s)
- Ying Chu
- The Center for Public Health Research, School of Medicine, Nanjing University, Nanjing 210093, Jiangsu Province, P. R. China
| | - Jiahuang Li
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, Jiangsu Province, P. R. China
| | - Xilin Wu
- The Center for Public Health Research, School of Medicine, Nanjing University, Nanjing 210093, Jiangsu Province, P. R. China
- Present address: AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam Hong Kong SAR, Hong Kong, People’s Republic of China
| | - Zichun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, Jiangsu Province, P. R. China
| | - Zhiwei Wu
- The Center for Public Health Research, School of Medicine, Nanjing University, Nanjing 210093, Jiangsu Province, P. R. China
- State Key Lab of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210093, Jiangsu Province, P. R. China
- Nanjing University, Meng Minwei 2104, 22 Hankou Road, Nanjing, Jiangsu 210093, P. R. China
| |
Collapse
|
85
|
Abstract
Mucins--large, highly glycosylated proteins--are important for the luminal protection of the gastrointestinal tract. Enterocytes have their apical surface covered by transmembrane mucins and goblet cells produce the secreted gel-forming mucins that form mucus. The small intestine has a single unattached mucus layer, which in cystic fibrosis becomes attached, accounting for the intestinal manifestations of this disease. The stomach and colon have two layers of mucus; the inner layer is attached and the outer layer is less dense and unattached. In the colon, the outer mucus layer is the habitat for commensal bacteria. The inner mucus layer is impervious to bacteria and is renewed every hour by surface goblet cells. The crypt goblet cells have the ability to restitute the mucus layer by secretion, for example after an ischaemic challenge. Proteases of certain parasites and some bacteria can cleave mucins and dissolve the mucus as part of their pathogenicity. The inner mucus layer can, however, also become penetrable to bacteria by several other mechanisms, including aberrations in the immune system. When bacteria reach the epithelial surface, the immune system is activated and inflammation is triggered. This mechanism might occur in some types of ulcerative colitis.
Collapse
|
86
|
Madsen J, Sorensen GL, Nielsen O, Tornøe I, Thim L, Fenger C, Mollenhauer J, Holmskov U. A variant form of the human deleted in malignant brain tumor 1 (DMBT1) gene shows increased expression in inflammatory bowel diseases and interacts with dimeric trefoil factor 3 (TFF3). PLoS One 2013; 8:e64441. [PMID: 23691218 PMCID: PMC3654909 DOI: 10.1371/journal.pone.0064441] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 04/15/2013] [Indexed: 12/27/2022] Open
Abstract
The protein deleted in malignant brain tumors (DMBT1) and the trefoil factor (TFF) proteins have all been proposed to have roles in epithelial cell growth and cell differentiation and shown to be up regulated in inflammatory bowel diseases. A panel of monoclonal antibodies was raised against human DMBT1gp340. Analysis of lung washings and colon tissue extracts by Western blotting in the unreduced state, two antibodies (Hyb213-1 and Hyb213-6) reacted with a double band of 290 kDa in lung lavage. Hyb213-6, in addition, reacted against a double band of 270 kDa in colon extract while Hyb213-1 showed no reaction. Hyb213-6 showed strong cytoplasmic staining in epithelial cells of both the small and large intestine whereas no staining was seen with Hyb213-1. The number of DMBT1gp340 positive epithelial cells, stained with Hyb213-6, was significantly up regulated in inflammatory colon tissue sections from patients with ulcerative colitis (p<0.0001) and Crohn’s disease (p = 0.006) compared to normal colon tissue. Immunohistochemical analysis of trefoil factor TFF1, 2 and 3 showed that TFF1 and 3 localized to goblet cells in both normal colon tissue and in tissue from patients with ulcerative colitis or Crohn’s disease. No staining for TFF2 was seen in goblet cells in normal colon tissue whereas the majority of tissue sections in ulcerative colitis and Crohn’s disease showed sparse and scattered TFF2 positive goblet cells. DMBT1 and TFF proteins did therefore not co-localize in the same cells but localized in adjacent cells in the colon. The interaction between DMBT1gp340 and trefoil TFFs proteins was investigated using an ELISA assay. DMBT1gp340 bound to solid-phase bound recombinant dimeric TFF3 in a calcium dependent manner (p<0.0001) but did not bind to recombinant forms of monomeric TFF3, TFF2 or glycosylated TFF2. This implies a role for DMBT1 and TFF3 together in inflammatory bowel disease.
Collapse
Affiliation(s)
- Jens Madsen
- Sir Henry Wellcome Laboratories, Department of Child Health, Clinical and Experimental Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Cardiac amyloidosis induces up-regulation of Deleted in Malignant Brain Tumors 1 (DMBT1). Cardiovasc Pathol 2013; 22:195-202. [DOI: 10.1016/j.carpath.2012.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 10/26/2012] [Accepted: 10/28/2012] [Indexed: 01/20/2023] Open
|
88
|
Abstract
PURPOSE OF REVIEW Recent advances in molecular techniques have enabled a deep view into the structure and function of the host's immune system and the stably associated commensal intestinal flora. This review outlines selected aspects of the interplay of innate immune recognition and effectors that shape the ecological niches for the intestinal microbiota. RECENT FINDINGS Several studies have demonstrated a pivotal role of innate immune receptor pathways (NOD-like receptors and Toll-like receptors) for the maintenance of microbial communities in the gut. Genetic deficiencies in these pathways have been associated with increased susceptibility to inflammation that in animal models can be transmitted via direct contact or by stool transplantation in the absence of abundant pathogens. SUMMARY The genetic architecture of the human host shapes the diversity and function of its stably associated intestinal microflora. Innate immune receptors such as NOD2 or the inflammasome component NOD-like receptor, pyrin-domain containing 6 play a major role in licensing the microbiota under physiological conditions. Understanding the symbiotic interplay in the intestinal tract should help develop procedures and therapeutic interventions aiming at the identification and restoration of disturbed microbiota states. Indeed, these states may be the missing trigger factor for the manifestation of a multitude of civilization disorders including inflammatory bowel disease and gastrointestinal cancer.
Collapse
|
89
|
Staphylococcus aureus SasA is responsible for binding to the salivary agglutinin gp340, derived from human saliva. Infect Immun 2013; 81:1870-9. [PMID: 23439307 DOI: 10.1128/iai.00011-13] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen that can colonize the nasal cavity, skin, intestine, and oral cavity as a commensal bacterium. gp340, also known as DMBT1 (deleted in malignant brain tumors 1), is associated with epithelial differentiation and innate immunity. In the oral cavity, gp340 induces salivary aggregation with several oral bacteria and promotes bacterial adhesion to tissues such as the teeth and mucosa. S. aureus is often isolated from the oral cavity, but the mechanism underlying its persistence in the oral cavity remains unclear. In this study, we investigated the interaction between S. aureus and gp340 and found that S. aureus interacts with saliva- and gp340-coated resin. We then identified the S. aureus factor(s) responsible for binding to gp340. The cell surface protein SasA, which is rich in basic amino acids (BR domain) at the N terminus, was responsible for binding to gp340. Inactivation of the sasA gene resulted in a significant decrease in S. aureus binding to gp340-coated resin. Also, recombinant SasA protein (rSasA) showed binding affinity to gp340, which was inhibited by the addition of N-acetylneuraminic acid. Surface plasmon resonance analysis showed that rSasA significantly bound to the NeuAcα(2-3)Galβ(1-4)GlcNAc structure. These results indicate that SasA is responsible for binding to gp340 via the N-acetylneuraminic acid moiety.
Collapse
|
90
|
Baik J, Hong S, Choi S, Jeon J, Park OJ, Cho K, Seo DG, Kum KY, Yun CH, Han S. Alpha-amylase is a human salivary protein with affinity to lipopolysaccharide ofAggregatibacter actinomycetemcomitans. Mol Oral Microbiol 2012. [DOI: 10.1111/omi.12011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- J.E. Baik
- Department of Oral Microbiology and Immunology; Dental Research Institute and BK21 Program; School of Dentistry; Seoul National University; Seoul; Korea
| | - S.W. Hong
- Department of Oral Microbiology and Immunology; Dental Research Institute and BK21 Program; School of Dentistry; Seoul National University; Seoul; Korea
| | - S. Choi
- Department of Oral Microbiology and Immunology; Dental Research Institute and BK21 Program; School of Dentistry; Seoul National University; Seoul; Korea
| | - J.H. Jeon
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences; Seoul National University; Seoul; Korea
| | - O.-J. Park
- Department of Oral Microbiology and Immunology; Dental Research Institute and BK21 Program; School of Dentistry; Seoul National University; Seoul; Korea
| | - K. Cho
- Division of Mass Spectrometry Research; Korea Basic Science Institute; Ochang; Korea
| | - D.-G. Seo
- Department of Conservative Dentistry and Dental Research Institute; School of Dentistry; Seoul National University; Seoul; Korea
| | - K.-Y. Kum
- Department of Conservative Dentistry and Dental Research Institute; School of Dentistry; Seoul National University; Seoul; Korea
| | - C.-H. Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences; Seoul National University; Seoul; Korea
| | - S.H. Han
- Department of Oral Microbiology and Immunology; Dental Research Institute and BK21 Program; School of Dentistry; Seoul National University; Seoul; Korea
| |
Collapse
|
91
|
Bueno SM, Riquelme S, Riedel CA, Kalergis AM. Mechanisms used by virulent Salmonella to impair dendritic cell function and evade adaptive immunity. Immunology 2012; 137:28-36. [PMID: 22703384 DOI: 10.1111/j.1365-2567.2012.03614.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Innate and adaptive immunity are inter-related by dendritic cells (DCs), which directly recognize bacteria through the binding of pathogen-associated molecular patterns (PAMPs) to specialized receptors on their surface. After capturing and degrading bacteria, DCs present their antigens as small peptides bound to MHC molecules and prime naive bacteria-specific T cells. In response to PAMP recognition DCs undergo maturation, which is a phenotypic change that increases their immunogenicity and promotes the activation of naive T cells. As a result, a specific immune response that targets bacteria-derived antigens is initiated. Therefore, the characterization of DC-bacteria interactions is important to understand the mechanisms used by virulent bacteria to avoid adaptive immunity. Furthermore, any impairment of DC function might contribute to bacterial survival and dissemination inside the host. An example of a bacterial pathogen capable of interfering with DC function is Salmonella enterica serovar Typhimurium (S. Typhimurium). Virulent strains of this bacterium are able to differentially modulate the entrance to DCs, avoid lysosomal degradation and prevent antigen presentation on MHC molecules. These features of virulent S. Typhimurium are controlled by virulence proteins, which are encoded by pathogenicity islands. Modulation of DC functions by these gene products is supported by several studies showing that pathogenesis might depend on this attribute of virulent S. Typhimurium. Here we discuss some of the recent data reported by the literature showing that several virulence proteins from Salmonella are required to modulate DC function and the activation of host adaptive immunity.
Collapse
Affiliation(s)
- Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genetica Molecular y Microbiologia, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | | | | | |
Collapse
|
92
|
High DMBT1 concentrations in breast milk correlate with increased risk of infection in preterm and term neonates. BMC Pediatr 2012; 12:157. [PMID: 23034003 PMCID: PMC3518203 DOI: 10.1186/1471-2431-12-157] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 09/26/2012] [Indexed: 12/13/2022] Open
Abstract
Background Human milk contains immune molecules involved in the protection of newborns against infections. We analyzed the concentration of Deleted in Malignant Brain Tumors 1 (DMBT1), a protein with functions in innate immunity, in breast milk. Methods DMBT1 was detected in breast milk by Western blotting and its concentration was quantified by ELISA in 95 breast milk samples collected from mothers of preterm and term neonates during the first four weeks after delivery. Possible effects of maternal or neonatal parameters were analyzed by different statistical tests. Results The mean DMBT1 concentration (± standard error of the mean) in the tested milk samples was 2.48 ± 0.26 μg/mL (range: 0.112 μg/mL to 17.984 μg/mL) and represented 0.0087% of the total protein content. The comparison between the newborns with infection and the newborns without infection revealed significantly higher DMBT1 concentrations in breast milk in the group with infection (6.72 ± 2.53 μg/mL versus 2.20 ± 0.35 μg/mL (P = 0.031)). Neither maternal nor neonatal parameters showed a correlation with the milk DMBT1 levels. Conclusions DMBT1 is a component of breast milk after birth and is up-regulated in the breast milk from mothers with newborns suffering from neonatal infection. Thus, breast milk DMBT1 may be part of the innate immunity similar to secretory IgA.
Collapse
|
93
|
Ibarra Sierra E, Díaz Chávez J, Cortés-Malagón EM, Uribe-Figueroa L, Hidalgo-Miranda A, Lambert PF, Gariglio P. Differential gene expression between skin and cervix induced by the E7 oncoprotein in a transgenic mouse model. Virology 2012; 433:337-45. [PMID: 22980503 DOI: 10.1016/j.virol.2012.08.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 06/20/2012] [Accepted: 08/17/2012] [Indexed: 10/27/2022]
Abstract
HPV16 E7 oncoprotein expression in K14E7 transgenic mice induces cervical cancer after 6 months of treatment with the co-carcinogen 17β-estradiol. In untreated mice, E7 also induces skin tumors late in life albeit at low penetrance. These findings indicate that E7 alters cellular functions in cervix and skin so as to predispose these organs to tumorigenesis. Using microarrays, we determined the global genes expression profile in cervical and skin tissue of young adult K14E7 transgenic mice without estrogen treatment. In these tissues, the E7 oncoprotein altered the transcriptional pattern of genes involved in several biological processes including signal transduction, transport, metabolic process, cell adhesion, apoptosis, cell differentiation, immune response and inflammatory response. Among the E7-dysregulated genes were ones not previously known to be involved in cervical neoplasia including DMBT1, GLI1 and 17βHSD2 in cervix, as well as MMP2, 12, 14, 19 and 27 in skin.
Collapse
Affiliation(s)
- E Ibarra Sierra
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados, México DF, Mexico
| | | | | | | | | | | | | |
Collapse
|
94
|
Adrianto I, Lin CP, Hale JJ, Levin AM, Datta I, Parker R, Adler A, Kelly JA, Kaufman KM, Lessard CJ, Moser KL, Kimberly RP, Harley JB, Iannuzzi MC, Rybicki BA, Montgomery CG. Genome-wide association study of African and European Americans implicates multiple shared and ethnic specific loci in sarcoidosis susceptibility. PLoS One 2012; 7:e43907. [PMID: 22952805 PMCID: PMC3428296 DOI: 10.1371/journal.pone.0043907] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 07/27/2012] [Indexed: 12/21/2022] Open
Abstract
Sarcoidosis is a systemic inflammatory disease characterized by the formation of granulomas in affected organs. Genome-wide association studies (GWASs) of this disease have been conducted only in European population. We present the first sarcoidosis GWAS in African Americans (AAs, 818 cases and 1,088 related controls) followed by replication in independent sets of AAs (455 cases and 557 controls) and European Americans (EAs, 442 cases and 2,284 controls). We evaluated >6 million SNPs either genotyped using the Illumina Omni1-Quad array or imputed from the 1000 Genomes Project data. We identified a novel sarcoidosis-associated locus, NOTCH4, that reached genome-wide significance in the combined AA samples (rs715299, P(AA-meta) = 6.51 × 10(-10)) and demonstrated the independence of this locus from others in the MHC region in the same sample. We replicated previous European GWAS associations within HLA-DRA, HLA-DRB5, HLA-DRB1, BTNL2, and ANXA11 in both our AA and EA datasets. We also confirmed significant associations to the previously reported HLA-C and HLA-B regions in the EA but not AA samples. We further identified suggestive associations with several other genes previously reported in lung or inflammatory diseases.
Collapse
Affiliation(s)
- Indra Adrianto
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Chee Paul Lin
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Jessica J. Hale
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Albert M. Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Indrani Datta
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Ryan Parker
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Adam Adler
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Jennifer A. Kelly
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Kenneth M. Kaufman
- Division of Rheumatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- The United States Department of Veterans Affairs Medical Center, Cincinnati, Ohio, United States of America
| | - Christopher J. Lessard
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Kathy L. Moser
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Robert P. Kimberly
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - John B. Harley
- Division of Rheumatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- The United States Department of Veterans Affairs Medical Center, Cincinnati, Ohio, United States of America
| | - Michael C. Iannuzzi
- Department of Medicine, SUNY Upstate Medical University, Syracuse, New York, United States of America
| | - Benjamin A. Rybicki
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Courtney G. Montgomery
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
95
|
IFN-γ renders human intestinal epithelial cells responsive to lipopolysaccharide of Vibrio cholerae by down-regulation of DMBT1. Comp Immunol Microbiol Infect Dis 2012; 35:345-54. [PMID: 22437006 DOI: 10.1016/j.cimid.2012.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 02/18/2012] [Accepted: 02/24/2012] [Indexed: 11/23/2022]
Abstract
Although intestinal epithelial cells (IECs) are continuously exposed to high densities of enteric bacteria, they are not highly responsive to microbe-associated molecular patterns (MAMPs). However, inflammatory cytokines such as interferon-γ (IFN-γ) are potentially capable of priming IECs to enhance responsiveness to MAMPs. In this study, we observed that heat-killed Vibrio cholerae (HKVC) and its lipopolysaccharide (LPS) poorly induced IL-8 production in a human IEC line, HT-29. However, both HKVC and the LPS showed a substantial induction of IL-8 production in IFN-γ-primed HT-29 cells. LPS-induced IL-8 production was proportional to the IFN-γ-priming period and LPS could not induce IL-8 production in the presence of polymyxin B. Moreover, LPS-induced IL-8 production in the IFN-γ-primed HT-29 cells was mediated through signaling pathways requiring p38 kinase and ERK, but not the JNK/SAPK pathway. Since deleted in malignant brain tumor 1 (DMBT1) is known to interact with and antagonize the action of LPS, we hypothesized that IFN-γ enhanced the responsiveness to LPS in HT-29 through down-regulation of DMBT1. We found that IFN-γ indeed attenuated DMBT1 expression at both the mRNA and protein levels in HT-29 cells. Conversely, when the cells were transfected with small interfering RNA to specifically silence DMBT1, IL-8 expression was augmented even in the absence of IFN-γ and the augmentation was further enhanced by treatment with V. cholerae LPS. Since IFN-γ is known to increase IFN-β expression in the IECs, we examined if IFN-β functioned similar to IFN-γ. Although IFN-β alone was able to induce IL-8 expression, it failed to render HT-29 cells responsive to V. cholerae LPS. In conclusion, our study suggests that IFN-γ primes IECs to become responsive to V. cholerae and its LPS by suppressing the expression of DMBT1.
Collapse
|
96
|
Kaemmerer E, Schneider U, Klaus C, Plum P, Reinartz A, Adolf M, Renner M, Wolfs TGAM, Kramer BW, Wagner N, Mollenhauer J, Gassler N. Increased levels of deleted in malignant brain tumours 1 (DMBT1) in active bacteria-related appendicitis. Histopathology 2012; 60:561-569. [PMID: 22296301 DOI: 10.1111/j.1365-2559.2011.04159.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2023]
Abstract
AIMS Deleted in malignant brain tumours 1 (DMBT1; gp340) is a secreted glycoprotein which is found in the surface lining epithelia of human small and large intestine. DMBT1 is suggested to play a role in enterocyte differentiation and surface protection from intestinal bacteria. The aim of this study was to elucidate DMBT1 expression in bacteria-related active intestinal inflammation such as appendicitis. METHODS AND RESULTS mRNA and protein levels of DMBT1 were analysed in surgical resections of 50 appendices (active inflammation: n = 25). In non-actively inflamed appendices, inter-individual differences in basal DMBT1 levels of enterocytes and some non-epithelial cells were found. In active appendicitis, enterocytic DMBT1 mRNA expression was increased approximately fivefold, which was paralleled by a corresponding increase of cytoplasmic and secreted DMBT1 protein levels. Increased DMBT1 expression was predominant in enterocytes adjacent to erosive lesions or ulcers. CONCLUSIONS Our data demonstrate that bacteria-related active inflammation results in a sharp increase of DMBT1 levels in enterocytes. These findings substantiate the view that DMBT1 is of functional relevance for host defence and modulation of the course of intestinal bacteria-related inflammatory responses.
Collapse
Affiliation(s)
- Elke Kaemmerer
- Department of Pediatrics Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Presley LL, Ye J, Li X, LeBlanc J, Zhang Z, Ruegger PM, Allard J, McGovern D, Ippoliti A, Roth B, Cui X, Jeske DR, Elashoff D, Goodglick L, Braun J, Borneman J. Host-microbe relationships in inflammatory bowel disease detected by bacterial and metaproteomic analysis of the mucosal-luminal interface. Inflamm Bowel Dis 2012; 18:409-17. [PMID: 21698720 PMCID: PMC3179764 DOI: 10.1002/ibd.21793] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 05/16/2011] [Indexed: 12/16/2022]
Abstract
BACKGROUND Host-microbe interactions at the intestinal mucosal-luminal interface (MLI) are critical factors in the biology of inflammatory bowel disease (IBD). METHODS To address this issue, we performed a series of investigations integrating analysis of the bacteria and metaproteome at the MLI of Crohn's disease, ulcerative colitis, and healthy human subjects. After quantifying these variables in mucosal specimens from a first sample set, we searched for bacteria exhibiting strong correlations with host proteins. This assessment identified a small subset of bacterial phylotypes possessing this host interaction property. Using a second and independent sample set, we tested the association of disease state with levels of these 14 "host interaction" bacterial phylotypes. RESULTS A high frequency of these bacteria (35%) significantly differentiated human subjects by disease type. Analysis of the MLI metaproteomes also yielded disease classification with exceptional confidence levels. Examination of the relationships between the bacteria and proteins, using regularized canonical correlation analysis (RCCA), sorted most subjects by disease type, supporting the concept that host-microbe interactions are involved in the biology underlying IBD. Moreover, this correlation analysis identified bacteria and proteins that were undetected by standard means-based methods such as analysis of variance, and identified associations of specific bacterial phylotypes with particular protein features of the innate immune response, some of which have been documented in model systems. CONCLUSIONS These findings suggest that computational mining of mucosa-associated bacteria for host interaction provides an unsupervised strategy to uncover networks of bacterial taxa and host processes relevant to normal and disease states. (Inflamm Bowel Dis 2012;).
Collapse
Affiliation(s)
- Laura L. Presley
- Department of Plant Pathology and Microbiology, University of California, Riverside, CA, 92521, USA
| | - Jingxiao Ye
- Department of Plant Pathology and Microbiology, University of California, Riverside, CA, 92521, USA
| | - Xiaoxiao Li
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - James LeBlanc
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Zhanpan Zhang
- Department of Statistics, University of California, Riverside, CA, 92521, USA
| | - Paul M. Ruegger
- Department of Plant Pathology and Microbiology, University of California, Riverside, CA, 92521, USA
| | - Jeff Allard
- Department of Medicine, Division of Digestive Disease, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Dermot McGovern
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Andrew Ippoliti
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Bennett Roth
- Department of Medicine, Division of Digestive Disease, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Xinping Cui
- Department of Statistics, University of California, Riverside, CA, 92521, USA
| | - Daniel R. Jeske
- Department of Statistics, University of California, Riverside, CA, 92521, USA
| | - David Elashoff
- Department of Human Genetics and Department of Biostatistics, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Lee Goodglick
- Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Jonathan Braun
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA, Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA,Corresponding Authors: James Borneman, Department of Plant Pathology and Microbiology, University of California, Riverside, CA, 92521, USA, Phone: 951-827-3584, Fax: 951-827-4294, . Jonathan Braun, Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA, Phone: 310-825-0650, Fax: 310-0825-5674,
| | - James Borneman
- Department of Plant Pathology and Microbiology, University of California, Riverside, CA, 92521, USA,Corresponding Authors: James Borneman, Department of Plant Pathology and Microbiology, University of California, Riverside, CA, 92521, USA, Phone: 951-827-3584, Fax: 951-827-4294, . Jonathan Braun, Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA, Phone: 310-825-0650, Fax: 310-0825-5674,
| |
Collapse
|
98
|
Abstract
OBJECTIVES: Interleukin-23 (IL-23) has emerged as a new therapeutic target for the treatment of inflammatory bowel disease (IBD). As biomarkers of disease state and treatment efficacy are becoming increasingly important in drug development, we sought to identify efficacy biomarkers for anti-IL-23 therapy in Crohn's disease (CD). METHODS: Candidate IL-23 biomarkers, downstream of IL-23 signaling, were identified using shotgun proteomic analysis of feces and colon lavages obtained from a short-term mouse IBD model (anti-CD40 Rag2−/−) treated preventively with monoclonal antibodies (mAbs) to the IL-23 receptor (IL-23R). The biomarkers were then measured in an IBD T-cell transfer model treated therapeutically with a mAb to IL-23 (p19), confirming their association with IBD. To assess the clinical relevance of these markers, we assessed their concentrations in clinical serum, colon tissue, and feces from CD patients. RESULTS: We identified 57 proteins up or downregulated in diseased animals that returned to control values when the mice were treated with mAbs to IL-23R. Among those, S100A8, S100A9, regenerating protein 3β (REG), REG3γ, lipocalin 2 (LCN2), deleted in malignant tumor 1 (DMBT1), and macrophage migration inhibitory factor (MIF) mRNA levels correlated with disease score and dose titration of mAbs to IL-23R or IL-23(p19). All biomarkers, except DMBT1, were also downregulated after therapeutic administration of mAbs to IL-23(p19) in a T-cell transfer IBD mouse model. In sera from CD patients, we confirmed a significant upregulation of S100A8/A9 (43%), MIF (138%), pancreatitis-associated protein (PAP, human homolog of REG3β/γ 49%), LCN2 (520%), and CCL20 (1280%), compared with control samples, as well as a significant upregulation of S100A8/A9 (887%), PAP (401%), and LCN2 (783%) in human feces from CD patients compared with normal controls. CONCLUSIONS: These studies identify multiple protein biomarkers downstream of IL-23 that could be valuable tools to assess the efficacy of this new therapeutic agent.
Collapse
|
99
|
Martínez VG, Moestrup SK, Holmskov U, Mollenhauer J, Lozano F. The conserved scavenger receptor cysteine-rich superfamily in therapy and diagnosis. Pharmacol Rev 2011; 63:967-1000. [PMID: 21880988 DOI: 10.1124/pr.111.004523] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The scavenger receptor cysteine-rich (SRCR) superfamily of soluble or membrane-bound protein receptors is characterized by the presence of one or several repeats of an ancient and highly conserved protein module, the SRCR domain. This superfamily (SRCR-SF) has been in constant and progressive expansion, now up to more than 30 members. The study of these members is attracting growing interest, which parallels that in innate immunity. No unifying function has been described to date for the SRCR domains, this being the result of the limited knowledge still available on the physiology of most members of the SRCR-SF, but also of the sequence versatility of the SRCR domains. Indeed, involvement of SRCR-SF members in quite different functions, such as pathogen recognition, modulation of the immune response, epithelial homeostasis, stem cell biology, and tumor development, have all been described. This has brought to us new information, unveiling the possibility that targeting or supplementing SRCR-SF proteins could result in diagnostic and/or therapeutic benefit for a number of physiologic and pathologic states. Recent research has provided structural and functional insight into these proteins, facilitating the development of means to modulate the activity of SRCR-SF members. Indeed, some of these approaches are already in use, paving the way for a more comprehensive use of SRCR-SF members in the clinic. The present review will illustrate some available evidence on the potential of well known and new members of the SRCR-SF in this regard.
Collapse
Affiliation(s)
- Vanesa Gabriela Martínez
- Center Esther Koplowitz, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | | | | | | |
Collapse
|
100
|
Diegelmann J, Olszak T, Göke B, Blumberg RS, Brand S. A novel role for interleukin-27 (IL-27) as mediator of intestinal epithelial barrier protection mediated via differential signal transducer and activator of transcription (STAT) protein signaling and induction of antibacterial and anti-inflammatory proteins. J Biol Chem 2011; 287:286-298. [PMID: 22069308 DOI: 10.1074/jbc.m111.294355] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The role of the Th17 cell inhibiting cytokine IL-27 in the pathogenesis of inflammatory bowel disease is contradictory. Its effects on the intestinal barrier have so far not been investigated, which was the aim of this study. We show that intestinal epithelial cells (IEC) express both IL-27 receptor subunits IL-27RA and gp130. The IL-27 receptor expression is up-regulated in intestinal inflammation and during bacterial infection. IL-27 activates ERK and p38 MAPKs as well as Akt, STAT1, STAT3, and STAT6 in IEC. IL-27 significantly enhances cell proliferation and IEC restitution. These functions of IL-27 are dependent on the activation of STAT3 and STAT6 signaling pathways. As analyzed by microarray, IL-27 modulates the expression of 428 target genes in IEC (316 up and 112 down; p<0.05). IL-27 as well as its main target genes are up-regulated in colonic tissue and IEC isolated from mice with dextran sulfate sodium (DSS)-induced colitis. The IL-27-induced expression of the anti-bacterial gene deleted in malignant brain tumor 1 (DMBT1) is mediated by p38 and STAT3 signaling, whereas the activation of the anti-inflammatory and anti-bacterial gene indoleamine 2,3-dioxygenase (IDO1) is dependent on STAT1 signal transduction. IL-27-induced indoleamine 2,3-dioxygenase enzymatic activity leads to growth inhibition of intestinal bacteria by causing local tryptophan depletion. For the first time, we characterize IL-27 as a mediator of intestinal epithelial barrier protection mediated via transcriptional activation of anti-inflammatory and antibacterial target genes.
Collapse
Affiliation(s)
- Julia Diegelmann
- Department of Medicine II-Grosshadern, Ludwig-Maximilians-University, Munich, 81377, Germany; Department of Preventive Dentistry and Periodontology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Torsten Olszak
- Department of Medicine II-Grosshadern, Ludwig-Maximilians-University, Munich, 81377, Germany; Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Burkhard Göke
- Department of Medicine II-Grosshadern, Ludwig-Maximilians-University, Munich, 81377, Germany
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Stephan Brand
- Department of Medicine II-Grosshadern, Ludwig-Maximilians-University, Munich, 81377, Germany.
| |
Collapse
|