1
|
Mattos-Graner RO, Klein MI, Alves LA. The complement system as a key modulator of the oral microbiome in health and disease. Crit Rev Microbiol 2024; 50:138-167. [PMID: 36622855 DOI: 10.1080/1040841x.2022.2163614] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/10/2023]
Abstract
In this review, we address the interplay between the complement system and host microbiomes in health and disease, focussing on oral bacteria known to contribute to homeostasis or to promote dysbiosis associated with dental caries and periodontal diseases. Host proteins modulating complement activities in the oral environment and expression profiles of complement proteins in oral tissues were described. In addition, we highlight a sub-set of bacterial proteins involved in complement evasion and/or dysregulation previously characterized in pathogenic species (or strains), but further conserved among prototypical commensal species of the oral microbiome. Potential roles of these proteins in host-microbiome homeostasis and in the emergence of commensal strain lineages with increased virulence were also addressed. Finally, we provide examples of how commensal bacteria might exploit the complement system in competitive or cooperative interactions within the complex microbial communities of oral biofilms. These issues highlight the need for studies investigating the effects of the complement system on bacterial behaviour and competitiveness during their complex interactions within oral and extra-oral host sites.
Collapse
Affiliation(s)
- Renata O Mattos-Graner
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Sao Paulo, Brazil
| | - Marlise I Klein
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Sao Paulo, Brazil
| | - Lívia Araújo Alves
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Sao Paulo, Brazil
- School of Dentistry, Cruzeiro do Sul University (UNICSUL), Sao Paulo, Brazil
| |
Collapse
|
2
|
Gan XX, Li YY, Li SJ, Mo SS, Feng JH, Shen F, Cai WS, Lai YQ, Xu B. Significance of DMBT1 in Papillary Thyroid Carcinoma Concurrent With Hashimoto's Thyroiditis. Front Oncol 2021; 11:680873. [PMID: 34422633 PMCID: PMC8372325 DOI: 10.3389/fonc.2021.680873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/10/2021] [Indexed: 12/13/2022] Open
Abstract
Background Papillary thyroid carcinoma (PTC) concurrent with Hashimoto’s thyroiditis (HT) was associated with a better clinical prognosis. This study aimed to investigate a potential mRNA gene that affects the development of PTC, which helps PTC concurrent with HT patients have a better prognosis. Material/Methods PTC data were obtained from The Cancer Genome Atlas (TCGA) database. And the validation data of tissue specimens were collected from Guangzhou First People’s Hospital. The thyroid tissue sections were hybridized with deleted in malignant brain tumor 1 (DMBT1) probes by situ hybridization. Survival rates were analyzed using Kaplan-Meier curves, and the log-rank test was used to compare group survival rates. Prognosis clinicopathological factors were analyzed by Cox regression. Gene Ontology (GO) and Kyoto Gene and Genomic Encyclopedia (KEGG) pathway enrichment analyses were performed using single-sample gene set enrichment analysis (ssGSEA). Finally, the correlation of deletion in DMBT1 expression with overall immune status, tumor purity, and human leukocyte antigen (HLA) gene expression profile was analyzed. Results HT was significantly associated with sex, tumor foci, extrathyroidal extension (ETE), residual tumor, and tumor stage (T stage). Moreover, PTC concurrent with HT had a lower risk of recurrence versus non-HT groups. A total of 136 differentially expressed mRNAs (DEMs) were identified between HT and non-HT groups. Among them, the expression level of DMBT1 in HT groups was statistically higher than that in non-HT groups. A significant association with ETE and recurrence was revealed in the high expression and the low expression of DMBT1. Furthermore, DMBT1 was an independent predictor of survival. The overall immune activity of high expression of DMBT1 was higher than that of the low-expression group. Conclusions The PTC patients with HT had better behavior features and prognosis than those with simple PTC. DMBT1 in PTC-HT patients was a potential possible factor that inhibits tumors. High expression of DMBT1 may improve PTC prognosis by immune-related pathways.
Collapse
Affiliation(s)
- Xiao-Xiong Gan
- Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ya-Yi Li
- Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Si-Jin Li
- Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shi-Sen Mo
- General Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jian-Hua Feng
- Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Fei Shen
- Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wen-Song Cai
- Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ye-Qian Lai
- Department of Thyroid Surgery, Meizhou People's Hospital, Meizhou City, China
| | - Bo Xu
- Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
3
|
Singh P, Banerjee R, Piao S, Costa de Medeiros M, Bellile E, Liu M, Damodaran Puthiya Veettil D, Schmitd LB, Russo N, Danella E, Inglehart RC, Pineault KM, Wellik DM, Wolf G, D’Silva NJ. Squamous cell carcinoma subverts adjacent histologically normal epithelium to promote lateral invasion. J Exp Med 2021; 218:e20200944. [PMID: 33835136 PMCID: PMC8042603 DOI: 10.1084/jem.20200944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 01/04/2021] [Accepted: 02/16/2021] [Indexed: 12/26/2022] Open
Abstract
Recurrent and new tumors, attributed in part to lateral invasion, are frequent in squamous cell carcinomas and lead to poor survival. We identified a mechanism by which cancer subverts adjacent histologically normal epithelium to enable small clusters of cancer cells to burrow undetected under adjacent histologically normal epithelium. We show that suppression of DMBT1 within cancer promotes aggressive invasion and metastasis in vivo and is associated with metastasis in patients. Cancer cells via TGFβ1 and TNFα also suppress DMBT1 in adjacent histologically normal epithelium, thereby subverting it to promote invasion of a small population of tumor cells. The sufficiency of DMBT1 in this process is demonstrated by significantly higher satellite tumor nests in Dmbt1-/- compared with wild-type mice. Moreover, in patients, invasion of small tumor nests under adjacent histologically normal epithelium is associated with increased risk for recurrence and shorter disease-free survival. This study demonstrates a crucial role of adjacent histologically normal epithelium in invasion and its important role in the tumor microenvironment and opens new possibilities for therapeutic strategies that reduce tumor recurrence.
Collapse
Affiliation(s)
- Priyanka Singh
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Rajat Banerjee
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Songlin Piao
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Marcell Costa de Medeiros
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Emily Bellile
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Min Liu
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | | | - Ligia B. Schmitd
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Nickole Russo
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Erika Danella
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Ronald C. Inglehart
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Kyriel M. Pineault
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI
| | - Deneen M. Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI
| | - Greg Wolf
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI
| | - Nisha J. D’Silva
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
- Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
4
|
Jiang Y, Bian Y, Lian N, Wang Y, Xie K, Qin C, Yu Y. iTRAQ-Based Quantitative Proteomic Analysis of Intestines in Murine Polymicrobial Sepsis with Hydrogen Gas Treatment. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4885-4900. [PMID: 33209018 PMCID: PMC7670176 DOI: 10.2147/dddt.s271191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/10/2020] [Indexed: 12/11/2022]
Abstract
Objective Sepsis-associated intestinal injury has a higher morbidity and mortality in patients with sepsis, but there is still no effective treatment. Our research team has proven that inhaling 2% hydrogen gas (H2) can effectively improve sepsis and related organ damage, but the specific molecular mechanism of its role is not clear. In this study, isobaric tags for relative and absolute quantitation (iTRAQ)-based quantitative proteomics analysis was used for studying the effect of H2 on intestinal injury in sepsis. Methods Male C57BL/6J mice were used to prepare a sepsis model by cecal ligation and puncture (CLP). The 7-day survival rates of mice were measured. 4-kd fluorescein isothiocyanate-conjugated Dextran (FITC-dextran) blood concentration measurement, combined with hematoxylin-eosinstain (HE) staining and Western blotting, was used to study the effect of H2 on sepsis-related intestinal damage. iTRAQ-based liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis was used for studying the proteomics associated with H2 for the treatment of intestinal injury. Results H2 can significantly improve the 7-day survival rates of sepsis mice. The load of blood and peritoneal lavage bacteria was increased, and H2 treatment can significantly reduce it. CLP mice had significant intestinal damage, and inhalation of 2% hydrogen could significantly reduce this damage. All 4194 proteins were quantified, of which 199 differentially expressed proteins were associated with the positive effect of H2 on sepsis. Functional enrichment analysis indicated that H2 may reduce intestinal injury in septic mice through the effects of thyroid hormone synthesis and nitrogen metabolism signaling pathway. Western blot showed that H2 was reduced by down-regulating the expressions of deleted in malignant brain tumors 1 protein (DMBT1), insulin receptor substrate 2 (IRS2), N-myc downregulated gene 1 (NDRG1) and serum amyloid A-1 protein (SAA1) intestinal damage in sepsis mice. Conclusion A total of 199 differential proteins were related with H2 in the intestinal protection of sepsis. H2-related differential proteins were notably enriched in the following signaling pathways, including thyroid hormone synthesis signaling pathway, nitrogen metabolism signaling pathways, digestion and absorption signaling pathways (vitamins, proteins and fats). H2 reduced intestinal injury in septic mice by down-regulating the expressions of SAA1, NDRG1, DMBT1 and IRS2.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Yingxue Bian
- Department of Anesthesiology, Tianjin Union Medical Center, Tianjin, People's Republic of China
| | - Naqi Lian
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Yaoqi Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Chao Qin
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| |
Collapse
|
5
|
Reichhardt MP, Loimaranta V, Lea SM, Johnson S. Structures of SALSA/DMBT1 SRCR domains reveal the conserved ligand-binding mechanism of the ancient SRCR fold. Life Sci Alliance 2020; 3:3/4/e201900502. [PMID: 32098784 PMCID: PMC7043408 DOI: 10.26508/lsa.201900502] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 02/14/2020] [Accepted: 02/14/2020] [Indexed: 02/06/2023] Open
Abstract
The structures of SALSA SRCR domains 1 and 8 reveal a cation-dependent mechanism for ligand recognition, contributing to important roles in the immune system and cellular signalling. The cation-binding sites are conserved across all SRCR domains, suggesting conserved functional mechanisms. The scavenger receptor cysteine-rich (SRCR) family of proteins comprises more than 20 membrane-associated and secreted molecules. Characterised by the presence of one or more copies of the ∼110 amino-acid SRCR domain, this class of proteins have widespread functions as antimicrobial molecules, scavenger receptors, and signalling receptors. Despite the high level of structural conservation of SRCR domains, no unifying mechanism for ligand interaction has been described. The SRCR protein SALSA, also known as DMBT1/gp340, is a key player in mucosal immunology. Based on detailed structural data of SALSA SRCR domains 1 and 8, we here reveal a novel universal ligand-binding mechanism for SALSA ligands. The binding interface incorporates a dual cation-binding site, which is highly conserved across the SRCR superfamily. Along with the well-described cation dependency on most SRCR domain–ligand interactions, our data suggest that the binding mechanism described for the SALSA SRCR domains is applicable to all SRCR domains. We thus propose to have identified in SALSA a conserved functional mechanism for the SRCR class of proteins.
Collapse
Affiliation(s)
| | | | - Susan M Lea
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.,Central Oxford Structural Molecular Imaging Centre, University of Oxford, Oxford, UK
| | - Steven Johnson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
6
|
Sun Z, Xu L. Expression of PDK-1 and DMBT1 in the thyroid carcinoma and its clinicopathological significance. Oncol Lett 2019; 18:2819-2824. [PMID: 31452760 PMCID: PMC6676455 DOI: 10.3892/ol.2019.10639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 05/10/2019] [Indexed: 12/14/2022] Open
Abstract
This study was designed to explore the expression of phosphoinositide-dependent protein kinase 1 (PDK-1), deleted in malignant brain tumors (DMBT1) in the thyroid carcinoma. A total of 87 fresh samples of thyroid carcinoma from surgical resection in The Second People's Hospital of Lianyungang from June 2016 to March 2018 were collected for the ELISA to detect the protein expression of PDK-1 and DMBT1. Then the pathological significance of the expression of PDK-1 and DMBT1 in the thyroid carcinoma and the correlation between them were analyzed, using the ROC curve to study the diagnostic value of each index. The expression of PDK-1 in the thyroid carcinoma tissue was significantly higher than that in the normal thyroid tissue with a statistical difference between them (P<0.05); the expression of DMBT1 in the thyroid carcinoma was statistically significantly lower than that in the normal thyroid tissue (P<0.05); the PDK-1 and DMBT1 expressions were in negative correlation in the thyroid carcinoma (r=−0.889, P<0.001). The AUG, specificity and the sensitivity of the PDK-1 in diagnosing the thyroid carcinoma were 0.862, 86.21% and 78.16%, respectively; the AUG, specificity and the sensitivity of the DMBT1 in diagnosing the thyroid carcinoma were 0.708, 66.67% and 67.82%, respectively; while the AUG, the specificity and the sensitivity of the combination of PDK-1 and DMBT1 in diagnosing the thyroid carcinoma were 0.888, 89.66% and 81.61%. In conclusion, the occurrence and progression of the thyroid carcinoma were related to the high expression of the PDK-1 and the low expression of the DMBT1 in the thyroid carcinoma tissues, the two of which were in connection with factors involving lymph node metastasis, pathological type, neoplasm staging, and clinical staging. Thus, the combined detection of PDK-1 and DMBT1 could be used as an effective index to determine the occurrence of thyroid carcinoma.
Collapse
Affiliation(s)
- Zhichao Sun
- Department of Pathology, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu 222006, P.R. China
| | - Lei Xu
- Department of Pathology, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu 222006, P.R. China
| |
Collapse
|
7
|
Du N, Bao W, Zhang K, Lu X, Crew R, Wang X, Liu G, Wang F. Cytogenetic characterization of the malignant primitive neuroectodermal SK-PN-DW tumor cell line. BMC Cancer 2019; 19:412. [PMID: 31046733 PMCID: PMC6498632 DOI: 10.1186/s12885-019-5625-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 04/18/2019] [Indexed: 11/10/2022] Open
Abstract
Background The SK-PN-DW cell line was established in 1979 and is commercially available. Despite the use of this cell line as an in vitro model for functional and therapeutic studies of malignant primitive neuroectodermal tumor (PNET), there is a lack of complete information about the genetic alterations that are present at the cytogenetic level. Thus, the current study aimed to characterize the cytogenetic profile of this cell line. Methods Routine G-banded chromosome analysis, fluorescence in situ hybridization, and oligonucleotide array comparative genomic hybridization assays were performed to characterize the chromosomal changes in this cell line. Results The G-banded karyotype analysis showed that the number of chromosomes in this cell line ranged between 36 and 41. Importantly, all cells displayed a loss of chromosomes Y, 11, 13, and 18. However, some cells showed an additional loss of chromosome 10. Additionally, the observed structural changes indicated: a) unbalanced translocation between chromosomes 1 and 7; b) translocation between chromosomes 11 and 22 at breakpoints 11q24 and 22q12, which is a classical translocation that is associated with Ewing sarcoma; c) a derivative chromosome due to a whole arm translocation between chromosomes 16 and 17 at likely breakpoints 16p10 and 17q10; and d) possible rearrangement in the short arm of chromosome 18. Moreover, a variable number of double minutes were also observed in each metaphase cell. Furthermore, the microarray assay results not only demonstrated genomic-wide chromosomal imbalance in this cell line and precisely placed chromosomal breakpoints on unbalanced, rearranged chromosomes, but also revealed information about subtle chromosomal changes and the chromosomal origin of double minutes. Finally, the fluorescence in situ hybridization assay confirmed the findings of the routine cytogenetic analysis and microarrays. Conclusion The accurate determination of the cytogenetic profile of the SK-PN-DW cell line is helpful in enabling the research community to utilize this cell line for future identity and comparability studies, in addition to demonstrating the utility of the complete cytogenetic profile, as a public resource.
Collapse
Affiliation(s)
- Na Du
- Department of Infectious Diseases, the First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, 130021, People's Republic of China.,Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Wanguo Bao
- Department of Infectious Diseases, the First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, 130021, People's Republic of China
| | - Kaiyu Zhang
- Department of Infectious Diseases, the First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, 130021, People's Republic of China
| | - Xianglan Lu
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Rebecca Crew
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Xianfu Wang
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Guangming Liu
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.,Department of Gastroenterology, the First Hospital of Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Feng Wang
- Department of Infectious Diseases, the First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, 130021, People's Republic of China.
| |
Collapse
|
8
|
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: 3.6] [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]
|
9
|
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: 1.6] [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
|
10
|
Tuttolomondo M, Casella C, Hansen PL, Polo E, Herda LM, Dawson KA, Ditzel HJ, Mollenhauer J. Human DMBT1-Derived Cell-Penetrating Peptides for Intracellular siRNA Delivery. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 8:264-276. [PMID: 28918028 PMCID: PMC5514624 DOI: 10.1016/j.omtn.2017.06.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 06/27/2017] [Accepted: 06/27/2017] [Indexed: 12/18/2022]
Abstract
Small interfering RNA (siRNA) is a promising molecule for gene therapy, but its therapeutic administration remains problematic. Among the recently proposed vectors, cell-penetrating peptides show great promise in in vivo trials for siRNA delivery. Human protein DMBT1 (deleted in malignant brain tumor 1) is a pattern recognition molecule that interacts with polyanions and recognizes and aggregates bacteria. Taking advantage of these properties, we investigated whether specific synthetic DMBT1-derived peptides could be used to formulate nanoparticles for siRNA administration. Using an electrophoretic mobility shift assay and UV spectra, we identified two DMBT1 peptides that could encapsulate the siRNA with a self- and co-assembly mechanism. The complexes were stable for at least 2 hr in the presence of either fetal bovine serum (FBS) or RNase A, with peptide-dependent time span protection. ζ-potential, circular dichroism, dynamic light scattering, and transmission electron microscopy revealed negatively charged nanoparticles with an average diameter of 10–800 nm, depending on the reaction conditions, and a spherical or rice-shaped morphology, depending on the peptide and β-helix conformation. We successfully transfected human MCF7 cells with fluorescein isothiocyanate (FITC)-DMBT1-peptide-Cy3-siRNA complexes. Finally, DMBT1 peptides encapsulating an siRNA targeting a fluorescent reporter gene showed efficient gene silencing in MCF7-recombinant cells. These results lay the foundation for a new research line to exploit DMBT1-peptide nanocomplexes for therapeutic siRNA delivery.
Collapse
Affiliation(s)
- Martina Tuttolomondo
- Lundbeckfonden Center of Excellence NanoCAN, University of Southern Denmark, 5000 Odense C, Denmark; Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark.
| | - Cinzia Casella
- Lundbeckfonden Center of Excellence NanoCAN, University of Southern Denmark, 5000 Odense C, Denmark; Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Pernille Lund Hansen
- Lundbeckfonden Center of Excellence NanoCAN, University of Southern Denmark, 5000 Odense C, Denmark; Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Ester Polo
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Luciana M Herda
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kenneth A Dawson
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Henrik J Ditzel
- Lundbeckfonden Center of Excellence NanoCAN, University of Southern Denmark, 5000 Odense C, Denmark; Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark; Department of Oncology, Odense University Hospital, 5000 Odense C, Denmark.
| | - Jan Mollenhauer
- Lundbeckfonden Center of Excellence NanoCAN, University of Southern Denmark, 5000 Odense C, Denmark; Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| |
Collapse
|
11
|
The scavenging capacity of DMBT1 is impaired by germline deletions. Immunogenetics 2017; 69:401-407. [PMID: 28364129 PMCID: PMC5435793 DOI: 10.1007/s00251-017-0982-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/27/2017] [Indexed: 12/31/2022]
Abstract
The Scavenger Receptor Cysteine-Rich (SRCR) proteins are an archaic group of proteins characterized by the presence of multiple SRCR domains. They are membrane-bound or secreted proteins, which are generally related to host defense systems in animals. Deleted in Malignant Brain Tumors 1 (DMBT1) is a SRCR protein which is secreted in mucosal fluids and involved in host defense by pathogen binding by its SRCR domains. Genetic polymorphism within DMBT1 leads to DMBT1-alleles giving rise to polypeptides with interindividually different numbers of SRCR domains, ranging from 8 SRCR domains (encoded by 6 kb DMBT1 variant) to 13 SRCR domains (encoded by the 8 kb DMBT1 variant). In the present study, we have investigated whether reduction from 13 to 8 amino-terminal SRCR domains leads to reduction of bacterial binding. The 6 kb variant bound ~20–45% less bacteria compared to the 8 kb variant. These results support the hypothesis that genetic variation in DMBT1 may influence microbial defense.
Collapse
|
12
|
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: 2.9] [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
|
13
|
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: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2015] [Indexed: 01/01/2023]
|
14
|
Lin YL, Pavlidis P, Karakoc E, Ajay J, Gokcumen O. The evolution and functional impact of human deletion variants shared with archaic hominin genomes. Mol Biol Evol 2015; 32:1008-19. [PMID: 25556237 PMCID: PMC4379406 DOI: 10.1093/molbev/msu405] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Allele sharing between modern and archaic hominin genomes has been variously interpreted to have originated from ancestral genetic structure or through non-African introgression from archaic hominins. However, evolution of polymorphic human deletions that are shared with archaic hominin genomes has yet to be studied. We identified 427 polymorphic human deletions that are shared with archaic hominin genomes, approximately 87% of which originated before the Human–Neandertal divergence (ancient) and only approximately 9% of which have been introgressed from Neandertals (introgressed). Recurrence, incomplete lineage sorting between human and chimp lineages, and hominid-specific insertions constitute the remaining approximately 4% of allele sharing between humans and archaic hominins. We observed that ancient deletions correspond to more than 13% of all common (>5% allele frequency) deletion variation among modern humans. Our analyses indicate that the genomic landscapes of both ancient and introgressed deletion variants were primarily shaped by purifying selection, eliminating large and exonic variants. We found 17 exonic deletions that are shared with archaic hominin genomes, including those leading to three fusion transcripts. The affected genes are involved in metabolism of external and internal compounds, growth and sperm formation, as well as susceptibility to psoriasis and Crohn’s disease. Our analyses suggest that these “exonic” deletion variants have evolved through different adaptive forces, including balancing and population-specific positive selection. Our findings reveal that genomic structural variants that are shared between humans and archaic hominin genomes are common among modern humans and can influence biomedically and evolutionarily important phenotypes.
Collapse
Affiliation(s)
- Yen-Lung Lin
- Department of Biological Sciences, State University of New York at Buffalo, NY, US
| | - Pavlos Pavlidis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation of Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Emre Karakoc
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Jerry Ajay
- Department of Computer Science and Engineering, State University of New York at Buffalo, NY, US
| | - Omer Gokcumen
- Department of Biological Sciences, State University of New York at Buffalo, NY, US
| |
Collapse
|
15
|
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: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
16
|
Casella C, Tuttolomondo M, Høilund-Carlsen PF, Mollenhauer J. Natural pattern recognition mechanisms at epithelial barriers and potential use in nanomedicine. EUROPEAN JOURNAL OF NANOMEDICINE 2014. [DOI: 10.1515/ejnm-2014-0020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
17
|
Ambruosi B, Accogli G, Douet C, Canepa S, Pascal G, Monget P, Moros C, Holmskov U, Mollenhauer J, Robbe-Masselot C, Vidal O, Desantis S, Goudet G. Deleted in malignant brain tumor 1 is secreted in the oviduct and involved in the mechanism of fertilization in equine and porcine species. Reproduction 2013; 146:119-33. [DOI: 10.1530/rep-13-0007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oviductal environment affects preparation of gametes for fertilization, fertilization itself, and subsequent embryonic development. The aim of this study was to evaluate the effect of oviductal fluid and the possible involvement of deleted in malignant brain tumor 1 (DMBT1) on IVF in porcine and equine species that represent divergent IVF models. We first performed IVF after pre-incubation of oocytes with or without oviductal fluid supplemented or not with antibodies directed against DMBT1. We showed that oviductal fluid induces an increase in the monospermic fertilization rate and that this effect is canceled by the addition of antibodies, in both porcine and equine species. Moreover, pre-incubation of oocytes with recombinant DMBT1 induces an increase in the monospermic fertilization rate in the pig, confirming an involvement of DMBT1 in the fertilization process. The presence of DMBT1 in the oviduct at different stages of the estrus cycle was shown by western blot and confirmed by immunohistochemical analysis of ampulla and isthmus regions. The presence of DMBT1 in cumulus–oocyte complexes was shown by western blot analysis, and the localization of DMBT1 in the zona pellucida and cytoplasm of equine and porcine oocytes was observed using immunofluorescence analysis and confocal microscopy. Moreover, we showed an interaction between DMBT1 and porcine spermatozoa using surface plasmon resonance studies. Finally, a bioinformatic and phylogenetic analysis allowed us to identify the DMBT1 protein as well as a DMBT1-like protein in several mammals. Our results strongly suggest an important role of DMBT1 in the process of fertilization.
Collapse
|
18
|
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: 41] [Impact Index Per Article: 3.7] [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
|
19
|
DMBT1Homozygous Deletion in Diffuse Astrocytomas Is Associated With Unfavorable Clinical Outcome. J Neuropathol Exp Neurol 2012; 71:702-7. [DOI: 10.1097/nen.0b013e31825f2e5d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
20
|
Dodurga Y, Avci CB, Yilmaz S, Dogan ZO, Kesen Z, Tataroglu C, Satiroglu-Tufan NL, Bushra T, Gunduz C. Evaluation of deleted in malignant brain tumors 1 (DMBT1) gene expression in bladder carcinoma cases: preliminary study. Biomarkers 2011; 16:610-5. [PMID: 21999583 DOI: 10.3109/1354750x.2011.620627] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
This study was undertaken to evaluate the expression of DMBT1 in bladder cancer and its correlation with clinico-pathological parameters analyzed in bladder carcinoma patients. We investigated DMBT1 in 56 paraffin embedded specimens of transitional cell carcinoma of the urinary bladder. We assessed DMBT1 gene expression at mRNA level by RT-PCR. Our results show 100% expression of DMBT1 in bladder carcinoma samples. Due to this preliminary results; gene expression was compared to tumor grade, and a significant difference was detected between grade 1 and 3 (p = 0.028). The down-regulation of DMBT1 gene expression in carcinomas suggests the possible role in bladder cancer.
Collapse
Affiliation(s)
- Yavuz Dodurga
- Department of Medical Biology, Pamukkale University School of Medicine, Denizli, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
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: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [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
|
22
|
Fukui H, Sekikawa A, Tanaka H, Fujimori Y, Katake Y, Fujii S, Ichikawa K, Tomita S, Imura J, Chiba T, Fujimori T. DMBT1 is a novel gene induced by IL-22 in ulcerative colitis. Inflamm Bowel Dis 2011; 17:1177-88. [PMID: 20824812 DOI: 10.1002/ibd.21473] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2010] [Accepted: 07/30/2010] [Indexed: 01/09/2023]
Abstract
BACKGROUND Interleukin (IL)-22 is a recently identified cytokine that is suggested to play pivotal roles in various inflammatory diseases. Although the IL-22 receptor 1 (IL-22R1) is restrictively expressed in epithelial cells in the colon, the role of IL-22 in colonic diseases still remains unclear. In this study microarray analyses revealed that deleted in malignant brain tumors 1 (DMBT1) is a novel upregulated gene in IL-22-stimulated colon cancer cells. Therefore, we investigated the involvement of DMBT1 and IL-22 in ulcerative colitis (UC) tissues and examined the mechanism regulating the expression of DMBT1 in response to IL-22 stimulation. METHODS Changes of gene expression in IL-22-stimulated SW403 cells were investigated by microarray analyses. The effects of IL-22 on DMBT1 expression were examined in SW403 cells using a small interfering RNA (si)RNA for STAT3 or inhibitors for MEK, PI3K, and nuclear factor kappa B (NF-κB). The element responsible for IL-22-induced DMBT1 promoter activation was determined by a promoter deletion and electrophoretic mobility shift assay (EMSA). Expression of IL-22, IL-22R1, and DMBT1 in UC tissues was analyzed by real-time reverse-transcription polymerase chain reaction (RT-PCR) and immunohistochemistry. RESULTS IL-22 treatment enhanced the expression of DMBT1 through STAT3 tyrosine phosphorylation and NF-κB activation in colon cancer cells. The IL-22-responsive element was located between -187 and -179 in the DMBT1 promoter region. In the UC mucosa the levels of DMBT1 and IL-22 mRNA expression were significantly enhanced and positively correlated, the numbers of IL-22-positive lymphocytes were increased, and the expression of IL-22R1 and DMBT1 was enhanced in the inflamed epithelium. CONCLUSIONS The IL-22/DMBT1 axis may play a pivotal role in the pathophysiology of UC.
Collapse
Affiliation(s)
- Hirokazu Fukui
- Department of Surgical and Molecular Pathology, Dokkyo University School of Medicine, Tochigi, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Du J, Guan M, Fan J, Jiang H. Loss of DMBT1 Expression in Human Prostate Cancer and Its Correlation with Clinical Progressive Features. Urology 2011; 77:509.e9-13. [DOI: 10.1016/j.urology.2010.09.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2010] [Revised: 08/22/2010] [Accepted: 09/03/2010] [Indexed: 01/15/2023]
|
24
|
Deleted in malignant brain tumors-1 protein (DMBT1): a pattern recognition receptor with multiple binding sites. Int J Mol Sci 2010; 11:5212-33. [PMID: 21614203 PMCID: PMC3100851 DOI: 10.3390/ijms1112521] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 12/09/2010] [Accepted: 12/09/2010] [Indexed: 12/22/2022] Open
Abstract
Deleted in Malignant Brain Tumors-1 protein (DMBT1), salivary agglutinin (DMBT1(SAG)), and lung glycoprotein-340 (DMBT1(GP340)) are three names for glycoproteins encoded by the same DMBT1 gene. All these proteins belong to the scavenger receptor cysteine-rich (SRCR) superfamily of proteins: a superfamily of secreted or membrane-bound proteins with SRCR domains that are highly conserved down to sponges, the most ancient metazoa. In addition to SRCR domains, all DMBT1s contain two CUB domains and one zona pellucida domain. The SRCR domains play a role in the function of DMBT1s, which is the binding of a broad range of pathogens including cariogenic streptococci, Helicobacter pylori and HIV. Mucosal defense proteins like IgA, surfactant proteins and lactoferrin also bind to DMBT1s through their SRCR domains. The binding motif on the SRCR domains comprises an 11-mer peptide in which a few amino acids are essential for binding (GRVEVLYRGSW). Adjacent to each individual SRCR domain are glycosylation domains, where the attached carbohydrate chains play a role in the binding of influenza A virus and Helicobacter pylori. The composition of the carbohydrate chains is not only donor specific, but also varies between different organs. These data demonstrate a role for DMBT1s as pattern recognition molecules containing various peptide and carbohydrate binding motifs.
Collapse
|
25
|
Rossez Y, Coddeville B, Elass E, Quinchon JF, Vidal O, Corfield AP, Gosset P, Lacroix JM, Michalski JC, Robbe-Masselot C. Interaction between DMBT1 and galectin 3 is modulated by the structure of the oligosaccharides carried by DMBT1. Biochimie 2010; 93:593-603. [PMID: 21167898 DOI: 10.1016/j.biochi.2010.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 12/06/2010] [Indexed: 11/29/2022]
Abstract
DMBT1 (deleted in malignant brain tumor 1), a human mucin-like glycoprotein, belonging to the scavenger receptor cysteine-rich (SRCR) superfamily, is mainly secreted from mucosal epithelia. It has been shown previously that interaction of hensin, the rabbit ortholog of DMBT1, with galectin 3, a β-galactoside-binding lectin, induces a terminal differentiation of epithelial cells. In this paper, we have used surface plasmon resonance (SPR), to analyse the binding of galectin 3 to two purified samples of human DMBT1:recombinant DMBT1 produced in CHO cells and DMBT1 isolated from intestinal tissues. Characterization of their glycosylation profile by nano-ESI-Q-TOF tandem mass spectrometry showed significant differences in O-glycans between the two DMBT1 samples. Results obtained by SPR demonstrated that the oligosaccharide side chains of DMBT1 are recognized by the carbohydrate-recognition domain (CRD) of galectin 3 and modification in the pattern of oligosaccharides modulates the binding parameters of DMBT1 with galectin 3. Moreover, using immunohistochemistry on paraffin-embedded colonic tissue sections, we could show a co-localisation of DMBT1 and galectin 3 in human intestine, suggesting a potential physiological interaction.
Collapse
|
26
|
Madsen J, Mollenhauer J, Holmskov U. Review: Gp-340/DMBT1 in mucosal innate immunity. Innate Immun 2010; 16:160-7. [PMID: 20418254 DOI: 10.1177/1753425910368447] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Deleted in Malignant Brain Tumour 1 (DMBT1) is a gene that encodes alternatively spliced proteins involved in mucosal innate immunity. It also encodes a glycoprotein with a molecular mass of 340 kDa, and is referred to as gp-340 (DMBT1(gp340)) and salivary agglutinin (DMBT1(SAG)). DMBT1(gp340) is secreted into broncho-alveolar surface lining fluid whereas DMBT(SAG) is present in the saliva. The two molecules were shown to be identical and both interact with and agglutinate several Gram-negative and Gram-positive bacteria including Streptococcus mutans, a bacterium responsible for caries in the oral cavity. DMBT1(gp340) interacts with surfactant proteins A and D (SP-D). DMBT1(gp340) and SP-D can individually and together interact and agglutinate influenza A virus. DMBT1(gp340) also binds to HIV-1 and facilitates transcytosis of the virus into epithelial cells. DMBT1 binds to a variety of other host proteins, including serum and secretory IgA, C1q, lactoferrin, MUC5B and trefoil factor 2 (TFF2), all molecules with involvement in innate immunity and/or wound-healing processes. Recent generation of Dmbt1-deficient mice has provided the research field of DMBT1 with a model that allows research to progress from in vitro studies to in vivo functional studies of the multifunctional proteins encoded by the DMBT1 gene.
Collapse
Affiliation(s)
- Jens Madsen
- University of Southampton, Southampton General Hospital, UK.
| | | | | |
Collapse
|
27
|
Tchatchou S, Riedel A, Lyer S, Schmutzhard J, Strobel-Freidekind O, Gronert-Sum S, Mietag C, D'Amato M, Schlehe B, Hemminki K, Sutter C, Ditsch N, Blackburn A, Hill LZ, Jerry DJ, Bugert P, Weber BHF, Niederacher D, Arnold N, Varon-Mateeva R, Wappenschmidt B, Schmutzler RK, Engel C, Meindl A, Bartram CR, Mollenhauer J, Burwinkel B. Identification of a DMBT1 polymorphism associated with increased breast cancer risk and decreased promoter activity. Hum Mutat 2010; 31:60-6. [PMID: 19830809 DOI: 10.1002/humu.21134] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
According to present estimations, the unfavorable combination of alleles with low penetrance but high prevalence in the population might account for the major part of hereditary breast cancer risk. Deleted in Malignant Brain Tumors 1 (DMBT1) has been proposed as a tumor suppressor for breast cancer and other cancer types. Genomewide mapping in mice further identified Dmbt1 as a potential modulator of breast cancer risk. Here, we report the association of two frequent and linked single-nucleotide polymorphisms (SNPs) with increased breast cancer risk in women above the age of 60 years: DMBT1 c.-93C>T, rs2981745, located in the DMBT1 promoter; and DMBT1 c.124A>C, p.Thr42Pro, rs11523871(odds ratio [OR]=1.66, 95% confidence interval [CI]=1.21-2.29, P=0.0017; and OR=1.66; 95% CI=1.21-2.28, P=0.0016, respectively), based on 1,195 BRCA1/2 mutation-negative German breast cancer families and 1,466 unrelated German controls. Promoter studies in breast cancer cells demonstrate that the risk-increasing DMBT1 -93T allele displays significantly decreased promoter activity compared to the DMBT1 -93C allele, resulting in a loss of promoter activity. The data suggest that DMBT1 polymorphisms in the 5'-region are associated with increased breast cancer risk. In accordance with previous results, these data link decreased DMBT1 levels to breast cancer risk.
Collapse
Affiliation(s)
- Sandrine Tchatchou
- Helmholtz-University Group Molecular Epidemiology, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Molecular cloning and analysis of SSc5D, a new member of the scavenger receptor cysteine-rich superfamily. Mol Immunol 2009; 46:2585-96. [DOI: 10.1016/j.molimm.2009.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 05/11/2009] [Indexed: 11/18/2022]
|
29
|
Warburton PE, Hasson D, Guillem F, Lescale C, Jin X, Abrusan G. Analysis of the largest tandemly repeated DNA families in the human genome. BMC Genomics 2008; 9:533. [PMID: 18992157 PMCID: PMC2588610 DOI: 10.1186/1471-2164-9-533] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Accepted: 11/07/2008] [Indexed: 01/26/2023] Open
Abstract
Background Tandemly Repeated DNA represents a large portion of the human genome, and accounts for a significant amount of copy number variation. Here we present a genome wide analysis of the largest tandem repeats found in the human genome sequence. Results Using Tandem Repeats Finder (TRF), tandem repeat arrays greater than 10 kb in total size were identified, and classified into simple sequence e.g. GAATG, classical satellites e.g. alpha satellite DNA, and locus specific VNTR arrays. Analysis of these large sequenced regions revealed that several "simple sequence" arrays actually showed complex domain and/or higher order repeat organization. Using additional methods, we further identified a total of 96 additional arrays with tandem repeat units greater than 2 kb (the detection limit of TRF), 53 of which contained genes or repeated exons. The overall size of an array of tandem 12 kb repeats which spanned a gap on chromosome 8 was found to be 600 kb to 1.7 Mbp in size, representing one of the largest non-centromeric arrays characterized. Several novel megasatellite tandem DNA families were observed that are characterized by repeating patterns of interspersed transposable elements that have expanded presumably by unequal crossing over. One of these families is found on 11 different chromosomes in >25 arrays, and represents one of the largest most widespread megasatellite DNA families. Conclusion This study represents the most comprehensive genome wide analysis of large tandem repeats in the human genome, and will serve as an important resource towards understanding the organization and copy number variation of these complex DNA families.
Collapse
Affiliation(s)
- Peter E Warburton
- Deptartment of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Cannon G, Yi Y, Ni H, Stoddard E, Scales DA, Van Ryk DI, Chaiken I, Malamud D, Weissman D. HIV envelope binding by macrophage-expressed gp340 promotes HIV-1 infection. THE JOURNAL OF IMMUNOLOGY 2008; 181:2065-70. [PMID: 18641344 DOI: 10.4049/jimmunol.181.3.2065] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The scavenger receptor cysteine-rich protein gp340 functions as part of the host innate immune defense system at mucosal surfaces. In the genital tract, its expression by cervical and vaginal epithelial cells promotes HIV trans-infection and may play a role in sexual transmission. Gp340 is an alternatively spliced product of the deleted in malignant brain tumors 1 (DMBT1) gene. In addition to its innate immune system activity, DMBT1 demonstrates instability in multiple types of cancer and plays a role in epithelial cell differentiation. We demonstrate that monocyte-derived macrophages express gp340 and that HIV-1 infection is decreased when envelope cannot bind it. Inhibition of infection occurred at the level of fusion of M-, T-, and dual-tropic envelopes. Additional HIV-1 envelope binding molecules, such as dendritic cell-specific ICAM-3-grabbing nonintegrin (DC-SIGN), mannose-binding lectin, and heparan sulfate, enhance the efficiency of infection of the cells that express them by increasing the local concentration of infectious virus. Our data suggest that gp340, which is expressed by macrophages in vivo, may function to enhance infection in much the same manner. Its expression on tissue macrophages and epithelial cells suggests important new opportunities for HIV-1 pathogenesis investigation and therapy.
Collapse
Affiliation(s)
- Georgetta Cannon
- Division of Infectious Diseases, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ligtenberg AJM, Veerman ECI, Nieuw Amerongen AV, Mollenhauer J. Salivary agglutinin/glycoprotein-340/DMBT1: a single molecule with variable composition and with different functions in infection, inflammation and cancer. Biol Chem 2008; 388:1275-89. [PMID: 18020944 DOI: 10.1515/bc.2007.158] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Salivary agglutinin (SAG), lung glycoprotein-340 (gp-340) and Deleted in Malignant Brain Tumours 1 (DMBT1) are three names for identical proteins encoded by the dmbt1 gene. DMBT1/SAG/gp-340 belongs to the scavenger receptor cysteine-rich (SRCR) superfamily of proteins, a superfamily of secreted or membrane-bound proteins with SRCR domains that are highly conserved down to sponges, the most ancient metazoa. On the one hand, DMBT1 may represent an innate defence factor acting as a pattern recognition molecule. It interacts with a broad range of pathogens, including cariogenic streptococci and Helicobacter pylori, influenza viruses and HIV, but also with mucosal defence proteins, such as IgA, surfactant proteins and MUC5B. Stimulation of alveolar macrophage migration, suppression of neutrophil oxidative burst and activation of the complement cascade point further to an important role in the regulation of inflammatory responses. On the other hand, DMBT1 has been demonstrated to play a role in epithelial and stem cell differentiation. Inactivation of the gene coding for this protein may lead to disturbed differentiation, possibly resulting in tumour formation. These data strongly point to a role for DMBT1 as a molecule linking innate immune processes with regenerative processes.
Collapse
Affiliation(s)
- Antoon J M Ligtenberg
- Department of Oral Biochemistry, Academic Centre for Dentistry, Free University, van de Boechorststraat 7, NL-1081 BT Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
32
|
Renner M, Bergmann G, Krebs I, End C, Lyer S, Hilberg F, Helmke B, Gassler N, Autschbach F, Bikker F, Strobel-Freidekind O, Gronert-Sum S, Benner A, Blaich S, Wittig R, Hudler M, Ligtenberg AJ, Madsen J, Holmskov U, Annese V, Latiano A, Schirmacher P, Amerongen AVN, D'Amato M, Kioschis P, Hafner M, Poustka A, Mollenhauer J. DMBT1 confers mucosal protection in vivo and a deletion variant is associated with Crohn's disease. Gastroenterology 2007; 133:1499-509. [PMID: 17983803 DOI: 10.1053/j.gastro.2007.08.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2007] [Accepted: 07/19/2007] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS Impaired mucosal defense plays an important role in the pathogenesis of Crohn's disease (CD), one of the main subtypes of inflammatory bowel disease (IBD). Deleted in malignant brain tumors 1 (DMBT1) is a secreted scavenger receptor cysteine-rich protein with predominant expression in the intestine and has been proposed to exert possible functions in regenerative processes and pathogen defense. Here, we aimed at analyzing the role of DMBT1 in IBD. METHODS We studied DMBT1 expression in IBD and normal tissues by quantitative reverse transcription-polymerase chain reaction, immunohistochemistry, and mRNA in situ hybridization. Genetic polymorphisms within DMBT1 were analyzed in an Italian IBD case-control sample. Dmbt1(-/-) mice were generated, characterized, and analyzed for their susceptibility to dextran sulfate sodium-induced colitis. RESULTS DMBT1 levels correlate with disease activity in inflamed IBD tissues. A highly significant fraction of the patients with IBD displayed up-regulation of DMBT1 specifically in the intestinal epithelial surface cells and Paneth cells. A deletion allele of DMBT1 with a reduced number of scavenger receptor cysteine-rich domain coding exons is associated with an increased risk of CD (P = .00056; odds ratio, 1.75) but not for ulcerative colitis. Dmbt1(-/-) mice display enhanced susceptibility to dextran sulfate sodium-induced colitis and elevated Tnf, Il6, and Nod2 expression levels during inflammation. CONCLUSIONS DMBT1 may play a role in intestinal mucosal protection and prevention of inflammation. Impaired DMBT1 function may contribute to the pathogenesis of CD.
Collapse
Affiliation(s)
- Marcus Renner
- Division of Molecular Genome Analysis, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Mollenhauer J, End C, Renner M, Lyer S, Poustka A. DMBT1 as an archetypal link between infection, inflammation, and cancer. ACTA ACUST UNITED AC 2007. [DOI: 10.1016/s0213-9626(07)70089-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
34
|
Rosenstiel P, Sina C, End C, Renner M, Lyer S, Till A, Hellmig S, Nikolaus S, Fölsch UR, Helmke B, Autschbach F, Schirmacher P, Kioschis P, Hafner M, Poustka A, Mollenhauer J, Schreiber S. Regulation of DMBT1 via NOD2 and TLR4 in intestinal epithelial cells modulates bacterial recognition and invasion. THE JOURNAL OF IMMUNOLOGY 2007; 178:8203-11. [PMID: 17548659 DOI: 10.4049/jimmunol.178.12.8203] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mucosal epithelial cell layers are constantly exposed to a complex resident microflora. 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 pathogen binding. This report describes the regulation and function of DMBT1 in intestinal epithelial cells, which form the primary immunological barrier for invading pathogens. We report that intestinal epithelial cells up-regulate DMBT1 upon proinflammatory stimuli (e.g., TNF-alpha, LPS). We demonstrate that DMBT1 is a target gene for the intracellular pathogen receptor NOD2 via NF-kappaB activation. DMBT1 is strongly up-regulated in the inflamed intestinal mucosa of Crohn's disease patients with wild-type, but not with mutant NOD2. We show that DMBT1 inhibits cytoinvasion of Salmonella enterica and LPS- and muramyl dipeptide-induced NF-kappaB activation and cytokine secretion in vitro. Thus, DMBT1 may play an important role in the first line of mucosal defense conferring immune exclusion of bacterial cell wall components. Dysregulated intestinal DMBT1 expression due to mutations in the NOD2/CARD15 gene may be part of the complex pathophysiology of barrier dysfunction in Crohn's disease.
Collapse
Affiliation(s)
- Philip Rosenstiel
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Campus Kiel, Schittenhelmstrache 12, Kiel, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Haase B, Humphray SJ, Lyer S, Renner M, Poustka A, Mollenhauer J, Leeb T. Molecular characterization of the porcine deleted in malignant brain tumors 1 gene (DMBT1). Gene 2006; 376:184-91. [PMID: 16624504 DOI: 10.1016/j.gene.2006.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Revised: 02/28/2006] [Accepted: 03/01/2006] [Indexed: 11/21/2022]
Abstract
The human gene deleted in malignant brain tumors 1 (DMBT1) is considered to play a role in tumorigenesis and pathogen defense. It encodes a protein with multiple scavenger receptor cysteine-rich (SRCR) domains, which are involved in recognition and binding of a broad spectrum of bacterial pathogens. The SRCR domains are encoded by highly homologous repetitive exons, whose number in humans may vary from 8 to 13 due to genetic polymorphism. Here, we characterized the porcine DMBT1 gene on the mRNA and genomic level. We assembled a 4.5 kb porcine DMBT1 cDNA sequence from RT-PCR amplified seminal vesicle RNA. The porcine DMBT1 cDNA contains an open reading frame of 4050 nt. The transcript gives rise to a putative polypeptide of 1349 amino acids with a calculated mass of 147.9 kDa. Compared to human DMBT1, it contains only four N-terminal SRCR domains. Northern blotting revealed transcripts of approximately 4.7 kb in size in the tissues analyzed. Analysis of ESTs suggested the existence of secreted and transmembrane variants. The porcine DMBT1 gene spans about 54 kb on chromosome 14q28-q29. In contrast to the characterized cDNA, the genomic BAC clone only contained 3 exons coding for N-terminal SRCR domains. In different mammalian DMBT1 orthologs large interspecific differences in the number of SRCR exons and utilization of the transmembrane exon exist. Our data suggest that the porcine DMBT1 gene may share with the human DMBT1 gene additional intraspecific variations in the number of SRCR-coding exons.
Collapse
Affiliation(s)
- Bianca Haase
- Institute of Genetics, Vetsuisse Faculty, University of Berne, Bremgartenstrasse 109a, 3001 Berne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
36
|
End C, Lyer S, Renner M, Stahl C, Ditzer J, Holloschi A, Kuhn HM, Flammann HT, Poustka A, Hafner M, Mollenhauer J, Kioschis P. Generation of a vector system facilitating cloning of DMBT1 variants and recombinant expression of functional full-length DMBT1. Protein Expr Purif 2005; 41:275-86. [PMID: 15866713 DOI: 10.1016/j.pep.2005.02.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 01/26/2005] [Indexed: 10/25/2022]
Abstract
Deleted in malignant brain tumours 1 (DMBT1) codes for a approximately 340kDa glycoprotein with highly repetitive scavenger receptor cysteine-rich (SRCR) domains. DMBT1 was implicated in cancer, defence against viral and bacterial infections, and differentiation of epithelial cells. Recombinant expression and purification of DMBT1 is an essential step for systematic standardized functional research and towards the evaluation of its therapeutical potential. So far, DMBT1 is obtained from natural sources such as bronchioalveolar lavage or saliva, resulting in time consuming sample collection, low yields, and protein preparations which may substantially vary due to differential processing and genetic polymorphism, all of which impedes functional research on DMBT1. Cloning of DMBT1 cDNAs is hampered because of the size and the 13 highly homologous SRCR exons. In this study, we report on the setup of a vector system that facilitates cloning of DMBT1 variants. We demonstrate applicability of the vector system by expression of the largest DMBT1 variant in a tetracycline-inducible mammalian expression system using the Chinese hamster ovary cell line. Yields up to 30 mg rDMBT1 per litre of cell culture supernatant could be achieved with an optimized production procedure. By harnessing the specific bacteria-binding property of DMBT1 we established an affinity purification procedure which allows the isolation of more than 3 mg rDMBT1 with a purity of about 95%. Although the glycosylation moieties of rDMBT1 are different from DMBT1(SAG) isolated from saliva, we demonstrate that rDMBT1 is functionally active in aggregating Gram-positive and Gram-negative bacteria and binding to C1q and lactoferrin, which represent two known endogenous DMBT1 ligands.
Collapse
Affiliation(s)
- Caroline End
- Institute of Molecular Biology and Cell Culture Technology, University of Applied Sciences Mannheim, Windeckstrasse 98, 68163 Mannheim, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Schwartz GJ, Al-Awqati Q. Role of hensin in mediating the adaptation of the cortical collecting duct to metabolic acidosis. Curr Opin Nephrol Hypertens 2005; 14:383-8. [PMID: 15931009 DOI: 10.1097/01.mnh.0000172727.82993.aa] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW The cortical collecting duct is able to secrete HCO3-, a state that can be converted to acid secretion during metabolic acidosis. Bicarbonate secretion in this segment is mediated by beta-intercalated cells whereas alpha-intercalated cells perform acid secretion. During metabolic acidosis, the number of beta-intercalated cells is reduced while that of alpha-intercalated cells increases without a change in the total number of intercalated cells, suggesting conversion of one cell type to another. Using an immortalized intercalated cell line we found that this adaptation is mediated by an extracellular protein named hensin. Hensin is secreted as a monomer which is then polymerized in the extracellular environment by a complex process requiring at least three other proteins. RECENT FINDINGS We describe that a cyclophilin, via its cis/trans prolyl isomerase activity, is required for this polymerization. This may explain the distal renal tubular acidosis observed with cyclosporin A therapy. In addition, galectin-3 is needed to aggregate the protein. Finally, we recently found that activation of integrins is also necessary for the development of the hensin fiber. Hensin is expressed in all epithelia and deletion of its gene is embryonic lethal at an early stage when the first columnar epithelia develop. SUMMARY These studies suggest that the response of intercalated cells to metabolic acidosis uses a pathway that is involved in terminal differentiation of columnar epithelia.
Collapse
Affiliation(s)
- George J Schwartz
- Departments of Pediatrics and Medicine, University of Rochester School of Medicine, Rochester, New York 14642, USA.
| | | |
Collapse
|
38
|
Ace CI, Okulicz WC. The 5' Flanking Region of the Rhesus Monkey H3 (DMBT1) Gene Contains Putative Progesterone Response Elements. DNA Cell Biol 2005; 24:345-9. [PMID: 15941386 DOI: 10.1089/dna.2005.24.345] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
DMBT1 (deleted in malignant brain tumors) encodes a large scavenger receptor cysteine rich (SRCR) protein with proposed tumor suppressor properties due to its frequent deletion or lack of expression in a variety of different tumors including endometrial cancers. The gene is alternatively spliced to produce a number of related proteins with suspected functions in mucosal inflammation and epithelial regeneration. Expression of DMBT1 has been demonstrated in a wide variety of cell types, mostly of epithelial origin, including tissues of the respiratory system, the alimentary system, brain, and reproductive system. We have previously identified a Rhesus monkey cDNA clone H3 (homologous to human DMBT1) as a progesterone-induced gene in Rhesus monkey endometrium during the secretory phase of the menstrual cycle. As an initial step in understanding the molecular mechanisms of H3 (DMBT1) regulation we have cloned and sequenced 1.5 kb of the 5'-flanking region expected to contain promoter sequences of the Rhesus monkey gene and identified six putative progesterone receptor binding sites in the 5'-upstream region.
Collapse
Affiliation(s)
- Christopher I Ace
- Department of Physiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
39
|
Grys TE, Siegel MB, Lathem WW, Welch RA. The StcE protease contributes to intimate adherence of enterohemorrhagic Escherichia coli O157:H7 to host cells. Infect Immun 2005; 73:1295-303. [PMID: 15731026 PMCID: PMC1064933 DOI: 10.1128/iai.73.3.1295-1303.2005] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is a diarrheal pathogen that causes attaching and effacing (A/E) lesions on intestinal epithelial cells. Strains of the O157 serogroup carry the large virulence plasmid pO157, which encodes the etp type II secretion system that secretes the genetically linked zinc metalloprotease StcE. The Ler regulator controls expression of many genes involved in A/E lesion formation, as well as StcE, suggesting StcE may be important at a similar time during colonization. Our laboratory has previously demonstrated that StcE cleaves C1-esterase inhibitor, a regulator of multiple inflammation pathways. Here we report two new substrates for StcE, mucin 7 and glycoprotein 340, and that purified StcE reduces the viscosity of human saliva. We tested the hypothesis that StcE contributes to intimate adherence of EHEC to host cells by cleavage of glycoproteins from the cell surface. The fluorescent actin stain (FAS) test was used to observe the intimate adherence represented by fluorescently stained bacteria colocalized with regions of bundled actin formed on HEp-2 cells. An E. coli O157:H7 strain with a stcE gene deletion was not affected in its ability to generally adhere to HEp-2 cells, but it did score threefold lower on the FAS test than wild-type or complemented strains. Addition of exogenous recombinant StcE increased intimate adherence of the mutant to wild-type levels. Thus, StcE may help block host clearance of E. coli O157:H7 by destruction of some classes of glycoproteins, and it contributes to intimate adherence of E. coli O157:H7 to the HEp-2 cell surface.
Collapse
Affiliation(s)
- Thomas E Grys
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 1300 University Ave., Room 481 MSC, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
40
|
Wiltshire RN, Herndon JE, Lloyd A, Friedman HS, Bigner DD, Bigner SH, McLendon RE. Comparative genomic hybridization analysis of astrocytomas: prognostic and diagnostic implications. J Mol Diagn 2005; 6:166-79. [PMID: 15269292 PMCID: PMC1867634 DOI: 10.1016/s1525-1578(10)60507-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Astrocytoma is comprised of a group of common intracranial neoplasms that are classified into four grades based on the World Health Organization histological criteria and patient survival. To date, histological grade, patient age, and clinical performance, as reflected in the Karnofsky score, are the most reliable prognostic predictors. Recently, there has been a significant effort to identify additional prognostic markers using objective molecular genetic techniques. We believe that the identification of such markers will characterize new chromosomal loci important in astrocytoma progression and aid clinical diagnosis and prognosis. To this end, our laboratory used comparative genomic hybridization to identify DNA sequence copy number changes in 102 astrocytomas. Novel losses of 19p loci were detected in low-grade pilocytic astrocytomas and losses of loci on 9p, 10, and 22 along with gains on 7, 19, and 20 were detected in a significant proportion of high-grade astrocytomas. The Cox proportional hazards statistical modeling showed that the presence of +7q and -10q comparative genomic hybridization alterations significantly increased a patient's risk of dying, independent of histological grade. This investigation demonstrates the efficacy of comparative genomic hybridization for identifying tumor suppressor and oncogene loci in different astrocytic grades. The cumulative effect of these loci is an important consideration in their diagnostic and prognostic implications.
Collapse
Affiliation(s)
- Rodney N Wiltshire
- Duke University Medical Center, Department of Pathology, Box 3712, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Bikker FJ, Ligtenberg AJM, End C, Renner M, Blaich S, Lyer S, Wittig R, van't Hof W, Veerman ECI, Nazmi K, de Blieck-Hogervorst JMA, Kioschis P, Nieuw Amerongen AV, Poustka A, Mollenhauer J. Bacteria Binding by DMBT1/SAG/gp-340 Is Confined to the VEVLXXXXW Motif in Its Scavenger Receptor Cysteine-rich Domains. J Biol Chem 2004; 279:47699-703. [PMID: 15355985 DOI: 10.1074/jbc.m406095200] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The scavenger receptor cysteine-rich (SRCR) proteins form an archaic group of metazoan proteins characterized by the presence of SRCR domains. These proteins are classified in group A and B based on the number of conserved cysteine residues in their SRCR domains, i.e. six for group A and eight for group B. The protein DMBT1 (deleted in malignant brain tumors 1), which is identical to salivary agglutinin and lung gp-340, belongs to the group B SRCR proteins and is considered to be involved in tumor suppression and host defense by pathogen binding. In a previous study we used nonoverlapping synthetic peptides covering the SRCR consensus sequence to identify a 16-amino acid bacteria-binding protein loop (peptide SRCRP2; QGRVEVLYRGSWGTVC) within the SRCR domains. In this study, using overlapping peptides, we pinpointed the minimal bacteria-binding site on SRCRP2, and thus DMBT1, to an 11-amino acid motif (DMBT1 pathogen-binding site 1 or DMBT1pbs1; GRVEVLYRGSW). An alanine substitution scan revealed that VEVL and Trp are critical residues in this motif. Bacteria binding by DMBT1pbs1 was different from the bacteria binding by the macrophage receptor MARCO in which an RXR motif was critical. In addition, the homologous consensus sequences of a number of SRCR proteins were synthesized and tested for bacteria binding. Only consensus sequences of DMBT1 orthologues bound bacteria by this motif.
Collapse
Affiliation(s)
- Floris J Bikker
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit en Universiteit van Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hermans KG, van Alewijk DC, Veltman JA, van Weerden W, van Kessel AG, Trapman J. Loss of a small region around the PTEN locus is a major chromosome 10 alteration in prostate cancer xenografts and cell lines. Genes Chromosomes Cancer 2004; 39:171-84. [PMID: 14732919 DOI: 10.1002/gcc.10311] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We examined 11 prostate cancer xenografts and 4 cell lines for chromosome 10 alterations. Conventional comparative genomic hybridization (CGH) and array-based CGH revealed a pattern of loss of distal 10p, gain of proximal 10p and 10q, and loss of distal 10q. In addition, array CGH identified 2 high-level amplifications in the cell line PC3, homozygous deletions of PTEN in PC3 and in the xenografts PCEW, PC133, and PC324, and small single- or double-copy deletions around PTEN in PCEW, PC82, PC324, PC346, and LNCaP. Allelotype analysis confirmed all 10p losses, 5 of 6 large 10q losses, the homozygous deletions, and the small regions of one copy loss. MXI1, DMBT1, and KLF6 were excluded as important tumor-suppressor genes. The sizes of homozygous deletions around PTEN ranged from 1.2 Mbp (PC133) to <30 kbp (PTEN exon 5 in PC295). The regions of small single- or double-copy loss around PTEN were all less than 4.5 Mbp. The loss of 1 or 2 copies of PTEN was always accompanied by loss of the distal flanking gene FLJ11218 and, in most cases, by loss of the proximal flanking genes MINPP1, PAPSS2, and FLJ14600. Furthermore, differential expression was detected for FLJ11218 and PAPSS2. Complete deletion or inactivating mutation of PAPSS2 was found in at least 3 samples. In addition to 4 homozygous deletions, 1 missense mutation was detected in FLJ11218. In conclusion, our data provide evidence that loss of a small region around PTEN is the major chromosome 10 alteration in prostate cancer xenografts and cell lines. In some of the samples, PTEN inactivation was accompanied by loss of 1 MINPP1 allele, loss of 1 copy, mutation, or low expression of PAPSS2, and most frequently by loss of 1 or 2 copies or low expression of FLJ11218.
Collapse
Affiliation(s)
- Karin G Hermans
- Department of Pathology, Josephine Nefkens Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
43
|
Suzuki T, Maruno M, Wada K, Kagawa N, Fujimoto Y, Hashimoto N, Izumoto S, Yoshimine T. Genetic analysis of human glioblastomas using a genomic microarray system. Brain Tumor Pathol 2004; 21:27-34. [PMID: 15696966 DOI: 10.1007/bf02482174] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Genomic microarray systems can simultaneously provide substantial genetic and chromosomal information in a relatively short time. We have analyzed genomic DNA from frozen sections of 30 cases of primary glioblastomas by GenoSensor Array 300 in order to characterize gene amplifications, gene deletions, and chromosomal information in the whole genome. Genes that were frequently amplified included RFC2/CYLN2 (63.3%), EGFR (53.3%), IL6 (53.3%), ABCB1 (MDR1) (36.7%), and PDGFRA (26.7%). Genes that were frequently deleted included (56.7%), FGFR2 (66.7%), MTAP (60.0%), DMBT1 CDKN2A (p16)/MTAP (50.0%), PIK3CA (43.3%), and EGR2 (43.3%), but deletion of RB1 or TP53 was rarely detected. Chromosomal gains were observed frequently for 7q (33.3%), 7p (20.0%), and 17q (13.3%). Loss of the 10q was frequently detected in 13 of 30 cases (46.7%). Loss of the entire chromosome 10 was seen in 9 of 30 cases (30.0%), and was often accompanied by EGFR amplification (7 cases, 77.8%). The GenoSensor Array 300 proved to be useful for identification of genome-wide molecular changes in glioblastomas. The obtained microarray profile can also yield valuable insight into the molecular events underlying carcinogenesis of brain tumors and may provide clues about clinical correlations, including response to treatment.
Collapse
Affiliation(s)
- Tsuyoshi Suzuki
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Muñoz J, Lázcoz P, Inda MM, Nistal M, Pestaña A, Encío IJ, Castresana JS. Homozygous deletion and expression of PTEN and DMBT1 in human primary neuroblastoma and cell lines. Int J Cancer 2004; 109:673-9. [PMID: 14999773 DOI: 10.1002/ijc.20055] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Neuroblastoma is the most common pediatric solid tumor. Although many allelic imbalances have been described, a bona fide tumor suppressor gene for this disease has not been found yet. In our study, we analyzed 2 genes, PTEN and DMBT1, mapping 10q23.31 and 10q25.3-26.1, respectively, which have been found frequently altered in other kinds of neoplasms. We screened both genes for homozygous deletions in 45 primary neuroblastic tumors and 12 neuroblastoma cell lines. Expression of these genes in cell lines was assessed by RT-PCR analysis. We could detect 2 of 41 (5%) primary tumors harboring PTEN homozygous deletions. Three of 41 (7%) primary tumors and 2 of 12 cell lines presented homozygous losses at the g14 STS on the DMBT1 locus. All cell lines analyzed expressed PTEN, but lack of DMBT1 mRNA expression was detected in 2 of them. We tried to see whether epigenetic mechanisms, such as aberrant promoter hypermethylation, had any role in DMBT1 silencing. The 2 cell lines lacking DMBT1 expression were treated with 5-aza-2'-deoxycytidine; DMBT1 expression was restored in only one of them (MC-IXC). From our work, we can conclude that PTEN and DMBT1 seem to contribute to the development of a small fraction of neuroblastomas, and that promoter hypermethylation might have a role in DMBT1 gene silencing.
Collapse
Affiliation(s)
- Jorge Muñoz
- Departamento de Genética, Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
45
|
Mollenhauer J, Helmke B, Medina D, Bergmann G, Gassler N, Müller H, Lyer S, Diedrichs L, Renner M, Wittig R, Blaich S, Hamann U, Madsen J, Holmskov U, Bikker F, Ligtenberg A, Carlén A, Olsson J, Otto HF, O'Malley B, Poustka A. Carcinogen inducibility in vivo and down-regulation of DMBT1 during breast carcinogenesis. Genes Chromosomes Cancer 2003; 39:185-94. [PMID: 14732920 DOI: 10.1002/gcc.10309] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Deleted in malignant brain tumors 1 (DMBT1) has been proposed as a candidate tumor suppressor for brain and epithelial cancer. Initial studies suggested loss of expression rather than mutation as the predominant mode of DMBT1 inactivation. However, in situ studies in lung cancer demonstrated highly sophisticated changes of DMBT1 expression and localization, pointing to a chronological order of events. Here we report on the investigation of DMBT1 in breast cancer in order to test whether these principles might also be attributable to other tumor types. Comprehensive mutational analyses did not uncover unambiguous inactivating DMBT1 mutations in breast cancer. Expression analyses in the human and mouse mammary glands pointed to the necessity of DMBT1 induction. While age-dependent and hormonal effects could be ruled out, 9 of 10 mice showed induction of Dmbt1 expression after administration of the carcinogen 7,12-dimethybenz(alpha)anthracene prior to the onset of tumorigenesis or other histopathological changes. DMBT1 displayed significant up-regulation in human tumor-flanking tissues compared to in normal breast tissues (P < 0.05). However, the breast tumor cells displayed a switch from lumenal secretion to secretion to the extracellular matrix and a significant down-regulation compared to that in matched normal flanking tissues (P < 0.01). We concluded that loss of expression also is the predominant mode of DMBT1 inactivation in breast cancer. The dynamic behavior of DMBT1 in lung carcinoma is fully reflected in breast cancer, which suggests that this behavior might be common to tumor types arising from monolayered epithelia.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Agglutinins
- Animals
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Calcium-Binding Proteins
- Carcinogens/metabolism
- Cell Line, Tumor
- DNA Mutational Analysis/methods
- DNA-Binding Proteins
- Down-Regulation/genetics
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Genes, Tumor Suppressor
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Mammary Glands, Human/drug effects
- Mammary Glands, Human/pathology
- Mammary Glands, Human/physiology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Middle Aged
- Mutation/drug effects
- Mutation/genetics
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Tumor Suppressor Proteins
Collapse
Affiliation(s)
- Jan Mollenhauer
- Department of Molecular Genome Analysis, Deutsches Krebsforschungszentrum, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mollenhauer J, Deichmann M, Helmke B, Müller H, Kollender G, Holmskov U, Ligtenberg T, Krebs I, Wiemann S, Bantel-Schaal U, Madsen J, Bikker F, Klauck SM, Otto HF, Moldenhauer G, Poustka A. Frequent downregulation of DMBT1 and galectin-3 in epithelial skin cancer. Int J Cancer 2003; 105:149-57. [PMID: 12673672 DOI: 10.1002/ijc.11072] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
DMBT1 and galectin-3 are potential interacting proteins with presumably complex roles in tumorigenesis. While at present a variety of mechanisms are discussed for DMBT1 and its participation in cancer, galectin-3 is commonly known to exert tumor-promoting effects. However, in vitro studies in a rodent system have suggested that DMBT1/galectin-3 interaction in the ECM triggers epithelial differentiation, which would point to tumor-suppressive properties. To improve the understanding of DMBT1/galectin-3 action in cancer, we carried out studies in skin cancer of different origins. Mutational analyses of DMBT1 identified a missense mutation in 1 of 13 melanoma cell lines. It led to an exchange of an evolutionary conserved proline residue for serine and located within the second CUB domain of DMBT1. Immunohistochemical analyses demonstrated absence of DMBT1/galectin-3 expression from melanocytes but induction of DMBT1 expression in 1 of 8 nevi and 1 of 11 melanomas and of galectin-3 expression in 3 of 8 nevi and 4 of 8 melanomas. These data suggest that DMBT1 and galectin-3 are unlikely to act as classical tumor suppressors in melanomas. DMBT1 and galectin-3 appear to be secreted to the ECM by epithelial cells within the epidermis and the hair follicle. Compared to the flanking normal epidermis, skin tumors of epithelial origin frequently displayed downregulation of DMBT1 (18 of 19 cases) and galectin-3 (12 of 12 cases). Thus, loss of DMBT1/galectin-3 expression may play a role in the genesis of epithelial skin cancer. This would support the view that galectin-3 can exert tumor-suppressive effects in certain scenarios, and DMBT1/galectin-3-mediated differentiation represents a candidate mechanism for this effect.
Collapse
MESH Headings
- Agglutinins
- Calcium-Binding Proteins
- Carcinoma, Basal Cell/genetics
- Carcinoma, Basal Cell/metabolism
- Carcinoma, Basal Cell/pathology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Case-Control Studies
- Chromosomes, Human, Pair 10/genetics
- DNA Mutational Analysis
- DNA Primers/chemistry
- DNA-Binding Proteins
- Down-Regulation
- Galectin 3/genetics
- Galectin 3/metabolism
- Gene Expression Regulation, Neoplastic
- Genes, Tumor Suppressor
- Humans
- Immunoenzyme Techniques
- In Vitro Techniques
- Loss of Heterozygosity
- Melanocytes/metabolism
- Melanocytes/pathology
- Melanoma/genetics
- Melanoma/metabolism
- Melanoma/pathology
- Microsatellite Repeats
- Polymerase Chain Reaction
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Skin Neoplasms/genetics
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Tumor Cells, Cultured
- Tumor Suppressor Proteins
Collapse
Affiliation(s)
- Jan Mollenhauer
- Department of Molecular Genome Analysis, Deutsches Krebsforschungszentrum, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kang W, Reid KBM. DMBT1, a regulator of mucosal homeostasis through the linking of mucosal defense and regeneration? FEBS Lett 2003; 540:21-5. [PMID: 12681477 DOI: 10.1016/s0014-5793(03)00217-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
DMBT1 (deleted in malignant brain tumor 1), which encodes a large scavenger receptor cysteine rich (SRCR) B protein, has been proposed to be a tumor suppressor gene, due to the high frequency of its homozygous deletion and the lack of expression in a variety of cancers. However, studies on its physiological functions and its relationship with tumorigenesis are still at an initial stage. Two mucosal defense-related molecules, gp-340 and salivary agglutinin, have been identified to be alternatively spliced products of DMBT1, which suggests that DMBT1 is a pattern recognition receptor in innate immunity. Meanwhile, results from immunohistochemical staining and studies at the cellular level, began to associate DMBT1 with a proliferation to differentiation switching process in gastrointestinal epithelial cells. Together with its up-regulation in inflammation, these findings suggest that DMBT1 might be a local regulator of homeostasis, possibly through linking mucosal inflammation to the modulation of epithelial regeneration, and whose abnormality is a frequent cause of malignancy.
Collapse
Affiliation(s)
- Weiqun Kang
- MRC Immunochemistry Unit, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | | |
Collapse
|
48
|
Mollenhauer J, Helmke B, Müller H, Kollender G, Krebs I, Wiemann S, Holmskov U, Madsen J, Otto HF, Poustka A. An integrative model on the role of DMBT1 in epithelial cancer. CANCER DETECTION AND PREVENTION 2003; 26:266-74. [PMID: 12430631 DOI: 10.1016/s0361-090x(02)00094-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The gene, deleted in malignant brain tumors 1 (DMBT1), has been proposed to play a role in brain and epithelial cancer, but shows unusual features for a classical tumor suppressor gene. We have proposed that its presumptive dual function in protection and differentiation is of importance to understand its role in cancer. To gain insights into its role in tumorigenesis, we conducted a comprehensive study on DMBT1 mutations, expression and location. Twenty-one out of 44 tumors showed variable numbers of tandem repeats (VNTRs) due to genetic polymorphism of DMBT1, whereas 11 out of 44 tumors displayed presumable mutations. However, none of the alterations would be predicted to lead to a complete inactivation of the gene. DMBT1 is mucin-like and shows tissue-specific expression and secretion, pointing to a function in the protection of monolayered epithelia and to an additional function in the differentiation of multilayered epithelia. The expression patterns in carcinomas arising from the respective structures support this view. Accepting this functional dualism gives rise to an initial model on the role of DMBT1 in epithelial cancer.
Collapse
|
49
|
Abstract
All epithelia form sheets of cells connected by tight and adherent junctions and exhibit polarized distribution of membrane proteins and lipids. During their development, epithelia progress from this 'generic' phenotype to terminally differentiated states characterized by the development of apical structures such as microvilli, apical endocytosis and regulated exocytosis as well as characteristic cell shapes. We have identified an extracellular matrix protein, hensin, which when polymerized into a fiber induces the terminal differentiation of renal cells. Hensin is expressed in most epithelia where it exists in tissue-specific alternately spliced forms. Many epithelial tumors have deletions in the human ortholog of hensin. We propose that hensin mediates terminal differentiation of these epithelia.
Collapse
Affiliation(s)
- Qais Al-Awqati
- Department of Medicine and Physiology, College of Physicians and Surgeons of Columbia University, 630 W. 168th St., New York, NY 10032, USA
| | | | | |
Collapse
|
50
|
Sanson M, Leuraud P, Aguirre-Cruz L, He J, Marie Y, Cartalat-Carel S, Mokhtari K, Duffau H, Delattre JY, Hoang-Xuan K. Analysis of loss of chromosome 10q, DMBT1 homozygous deletions, and PTEN mutations in oligodendrogliomas. J Neurosurg 2002; 97:1397-401. [PMID: 12507139 DOI: 10.3171/jns.2002.97.6.1397] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Chromosomal deletions of 10q and candidate genes such as PTEN and DMBT1 have been thoroughly investigated in glioblastomas but few data specifically address oligodendrogliomas. METHODS In this study, 39 pure oligodendrogliomas were investigated for loss of heterozygosity (LOH) on 10q, PTEN mutations, and DMBT1 homozygous deletions. The LOH on 10q was found in 19 (48%) of 39 oligodendrogliomas and was closely related to anaplasia (p = 0.02), shorter time to progression (p = 0.0005), and poorer survival (p = 0.035). The DMBT1 homozygous deletions were found in 10 (26%) of 39 oligodendrogliomas but only one PTEN mutation was detected. The LOH on 10q is a strong predictor of survival and could be a valuable prognostic marker in oligodendrogliomas. CONCLUSIONS Frequent inactivation of DMBT1 contrasting with rare mutations of PTEN may indicate that DMBT1 is preferentially involved in oligodendrogliomas. Nevertheless, the absence of a correlation with survival makes the role of DMBT1 in tumorigenesis still questionable and warrants further investigation.
Collapse
Affiliation(s)
- Marc Sanson
- INSERM U495, Laboratoire de Biologie des Interactions Neurones-Glie, Groupe Hospitalier Pitié-Salpêtrière and Université Pierre et Marie Curie, Paris, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|