Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Bioinformatics ; 23(5): 654-5, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17237056

ABSTRACT

UNLABELLED: Primary immunodeficiencies (PIDs) belong to the group of rare diseases. The European Society for Immunodeficiencies (ESID), is establishing an innovative European patient and research database network for continuous long-term documentation of patients, in order to improve the diagnosis, classification, prognosis and therapy of PIDs. The ESID Online Database is a web-based system aimed at data storage, data entry, reporting and the import of pre-existing data sources in an enterprise business-to-business integration (B2B). The online database is based on Java 2 Enterprise System (J2EE) with high-standard security features, which comply with data protection laws and the demands of a modern research platform. AVAILABILITY: The ESID Online Database is accessible via the official website (http://www.esid.org/). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Databases, Factual , Immunologic Deficiency Syndromes/diagnosis , Database Management Systems , Humans , Immunologic Deficiency Syndromes/classification , Immunologic Deficiency Syndromes/therapy , Information Storage and Retrieval , Internet , User-Computer Interface
2.
Clin Exp Immunol ; 147(2): 306-12, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17223972

ABSTRACT

Because primary immunodeficiencies (PID) are rare diseases, transnational studies are essential to maximize the scientific outcome and lead to improved diagnosis and therapy. Immunologists in Europe have united to determine the prevalence of PID in Europe and to establish and evaluate harmonized guidelines for the diagnosis and treatment of PID as well as to improve the awareness of PID in Europe. In order to achieve this aim we have developed an internet-based database for clinical and research data on patients with PID. This database forms the platform for studies of demographics, the development of new diagnostic and therapeutic strategies and the identification of novel disease-associated genes. The database is completely secure, while providing access to researchers via a standard browser using password and encrypted log-in sessions and conforms to all European and national ethics and data protection guidelines. So far 2386 patients have been documented by 35 documenting centres in 20 countries. Common variable immunodeficiency (CVID) is the most common entity, accounting for almost 30% of all entries. First statistical analyses on the quality of life of patients show the advantages of immunoglobulin replacement therapy, at the same time revealing a mean diagnostic delay of over 4 years. First studies on specific questions on selected PID are now under way. The platform of this database can be used for any type of medical condition.


Subject(s)
Databases as Topic/organization & administration , Immunologic Deficiency Syndromes/epidemiology , Internet , Adolescent , Adult , Age Distribution , Aged , Biomedical Research , Child , Child, Preschool , Europe/epidemiology , Female , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/therapy , Infant , Infant, Newborn , Male , Middle Aged , Quality of Life , Registries , Sex Distribution , Time Factors
3.
Int J Cancer ; 86(6): 863-9, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10842202

ABSTRACT

In an attempt to obtain suitable in vivo models for optimizing new tumor therapy strategies for intestinal adenocarcinomas, carcinoembryonic antigen (CEA) promoter/SV40 T antigen gene constructs have been used to generate transgenic mice. One transgenic line (L5496), which contains a 424-bp CEA promoter/SV40 T antigen transgene, exclusively developed multi-focal carcinomas in the pyloric region of the stomach in 100% of the offspring. Tumors were already observable in 37-day-old animals as dysplastic cell foci within the mucosal layer. In 50-day-old mice, the tumor mass was mainly restricted to the mucosa with invasive growth into the submucosal tissue. The animals became moribund at 100-130 days of age due to blockage of the pylorus. At this time, the tumor had penetrated into the duodenum and had invaded all tissue layers within the stomach. In contrast to most other stomach tumor models, this one perfectly matches the development of the most common stomach cancers found in humans. Furthermore, after crossing these mice with mice that are transgenic for the human CEA gene, the double transgenic offspring revealed expression of CEA in the resulting tumors. Thus, as well as being a model for studying gastric carcinoma development and prevention, this system should provide a useful preclinical model for CEA-targeted gastric tumor therapy.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Carcinoembryonic Antigen/genetics , Genetic Therapy , Stomach Neoplasms/therapy , Animals , Antigens, Polyomavirus Transforming/physiology , Carcinoembryonic Antigen/physiology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Stomach Neoplasms/etiology
4.
Blood ; 91(2): 663-72, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9427723

ABSTRACT

The nonspecific cross-reacting antigen-95 (NCA-95/CD66b), is a member of the human carcinoembryonic antigen (CEA) family encoded by the CGM6 gene that is exclusively expressed in neutrophils and eosinophils. No murine counter-part is known to exist. We have analyzed a cosmid containing the complete CGM6 gene. The coding sequence is contained within six exons spanning a 16.5 kb region. The main transcriptional start site was mapped to a tight cluster between nucleotides -95 and -101 relative to the translational start site. As with other members of the CEA gene family, no typical TATA or CAAT-box sequences were found in the CGM6 gene. Transgenic mice were established with the cosmid insert. CD66b expression is first seen in the fetal liver on day 12.5 of mouse embryonic development, and it first appears in the bone marrow at day 17.5. Northern blot analysis showed that CD66b transcripts are confined to the bone marrow of adult mice, whereas immunohistochemistry also showed CD66b-positive granulocytes in the spleen, thymus, and lungs. FACScan analyses of bone marrow and spleen cells showed CD66b expression to be exclusive to granulocytes. Thus, all the elements necessary for regulating granulocyte-specific expression are present within this cosmid clone. These mice could provide a model for transplantation and for inflammation studies using CD66b as a granulocyte-specific marker.


Subject(s)
Cell Adhesion Molecules , Granulocytes/metabolism , Membrane Glycoproteins/genetics , Animals , Antigens, CD , Antigens, Neoplasm/genetics , Base Sequence , Exons/genetics , GPI-Linked Proteins , Gene Expression , Humans , Immunohistochemistry , Membrane Glycoproteins/biosynthesis , Mice , Mice, Transgenic , Molecular Sequence Data
5.
Int J Cancer ; 72(1): 197-202, 1997 Jul 03.
Article in English | MEDLINE | ID: mdl-9212243

ABSTRACT

Carcinoembryonic antigen (CEA) is a tumor marker for the most common forms of adenocarcinomas. We have previously described C57BL/6 mice transgenic for the complete human CEA gene. Compared with humans, these mice reveal a conserved spatiotemporal CEA expression pattern. To establish animal models for CEA-targeted tumor immunotherapy, we have crossed CEA transgenic mice with mice that are genetically predisposed to tumor development. These immunocompetent animals should allow optimization of immunotherapy strategies for maximal destruction of tumor tissues with minimal damage to CEA-expressing normal tissues. To develop a breast tumor model, CEA transgenic mice were cross-bred with mice transgenic for the rat neu protooncogene controlled by the mouse mammary tumor virus long terminal repeat. Female offspring developed poorly differentiated breast tumors, none of which, however, expressed CEA. As a model for colorectal tumors, mice bearing a mutation in the Apc gene (Min mice) and the CEA transgene developed multiple intestinal adenomas with strong CEA expression in all tumor cells. CEA expression had no significant effect on tumor growth. Occasional, well-differentiated breast adenocarcinomas in female offspring expressed CEA focally in tumor cells lining pseudolumina. Cross-breeding Apc(Min/+) mice with neu transgenic mice did not reveal a synergistic effect on the kinetics of breast tumor formation. Finally, CEA transgenic mice crossbred with mice transgenic for the SV40 large T antigen regulated by the surfactant protein-C promoter, developed multiple lung adenocarcinomas that revealed a mosaic CEA expression pattern.


Subject(s)
Adenocarcinoma/genetics , Carcinoembryonic Antigen/genetics , Disease Models, Animal , Mice, Transgenic , Adenocarcinoma/metabolism , Animals , Carcinoembryonic Antigen/metabolism , Crosses, Genetic , Female , Genes, erbB-2 , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Lung Neoplasms/genetics , Male , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred C57BL , Mutation , Simian virus 40/genetics
6.
J Invest Dermatol ; 106(1): 64-9, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8592083

ABSTRACT

The carcinoembryonic antigen (CEA) family comprises a group of glycoproteins including the classical CEA, nonspecific cross-reacting antigens (NCA), and biliary glycoprotein (BGP). CEA glycoproteins have been identified in many glandular and mucosal tissues. In view of their putative role in cell adhesion, protein sorting, and signal transduction, CEA glycoproteins are thought to be involved in embryogenesis, architectual integrity, and secretory mechanisms of glandular epithelia. Since there are few data available on the expression of CEA-like proteins in human skin, the aim of this study was to immunohistochemically specify and localize the CEA glycoproteins in cutaneous adult and fetal glands using a panel of well-characterized antibodies. The secretory parts of eccrine sweat glands expressed CEA, NCA-90, and BGP, whereas apocrine glands remained unreactive for CEA glycoproteins. The ductal epithelia of both eccrine and apocrine glands contained CEA and NCA-90. Sebaceous glands were stained for BGP only. Electron microscopy of sweat glands showed CEA glycoprotein expression in cytoplasmic organelles and on microvilli lining the ductal surface. In sebaceous glands, BGP were demonstrated in small vesicles and along the cell membranes of differentiating sebocytes. Fetal development of cutaneous glands was associated with early expression of CEA glycoproteins. Additionally, mice transgenic for human CEA were shown to express CEA in sweat glands. The overall distribution of CEA glycoproteins in cutaneous glands was consistent with that in epithelia of other glandular tissues.


Subject(s)
Aging/metabolism , Carcinoembryonic Antigen/chemistry , Glycoproteins/physiology , Sebaceous Glands/chemistry , Skin/chemistry , Sweat Glands/chemistry , Adult , Animals , Fetus/metabolism , Humans , Mice , Mice, Transgenic , Sebaceous Glands/ultrastructure , Skin/embryology , Skin/ultrastructure , Sweat Glands/ultrastructure
7.
Tumour Biol ; 16(1): 56-61, 1995.
Article in English | MEDLINE | ID: mdl-7863223

ABSTRACT

The tumor marker carcinoembryonic antigen (CEA) is a 180-kD glycoprotein expressed on epithelial cells of the gastrointestinal tract and respiratory organs. It is a member of a large family belonging to the immunoglobulin superfamily. In order to develop a mouse model to study tumor immunotherapy using CEA as a target antigen, we have established mice which are transgenic for the CEA gene. The human CEA transgene maintains its spatiotemporal expression pattern in transgenic mice thus providing a suitable model in which to optimize different immunotherapy approaches, with the advantage that negative side effects due to the presence of CEA in normal tissue can be analyzed.


Subject(s)
Carcinoembryonic Antigen/immunology , Neoplasms, Experimental/therapy , Animals , Biomarkers, Tumor , Disease Models, Animal , Gene Expression , Mice , Mice, Nude , Mice, Transgenic
8.
Cancer Res ; 54(15): 4169-76, 1994 Aug 01.
Article in English | MEDLINE | ID: mdl-8033149

ABSTRACT

The tumor marker carcinoembryonic antigen (CEA) is predominantly expressed in epithelial cells along the gastrointestinal tract and in a variety of adenocarcinomas. As a basis for investigating its in vivo regulation and for establishing an animal model for tumor immunotherapy, transgenic mice were generated with a 33-kilobase cosmid clone insert containing the complete human CEA gene and flanking sequences. CEA was found in the tongue, esophagus, stomach, small intestine, cecum, colon, and trachea and at low levels in the lung, testis, and uterus of adult mice of independent transgenic strains. CEA was first detected at day 10.5 of embryonic development (embryonic day 10.5) in primary trophoblast giant cells and was found in the developing gut, urethra, trachea, lung, and nucleus pulposus of the vertebral column from embryonic day 14.5 onwards. From embryonic day 16.5 CEA was also visible in the nasal mucosa and tongue. Because this spatiotemporal expression pattern correlates well with that known for humans, it follows that the transferred genomic region contains all of the regulatory elements required for the correct expression of CEA. Furthermore, although mice apparently lack an endogenous CEA gene, the entire repertoire of transcription factors necessary for correct expression of the CEA transgene is conserved between mice and humans. After tumor induction, these immunocompetent mice will serve as a model for optimizing various forms of immunotherapy, using CEA as a target antigen.


Subject(s)
Carcinoembryonic Antigen/analysis , Carcinoembryonic Antigen/genetics , Mice, Transgenic/genetics , Mice, Transgenic/immunology , Animals , Colon/chemistry , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , RNA, Messenger/genetics
9.
Genomics ; 12(4): 761-72, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1572649

ABSTRACT

A long-range physical map of the carcinoembryonic antigen (CEA) gene family cluster, which is located on the long arm of chromosome 19, has been constructed. This was achieved by hybridization analysis of large DNA fragments separated by pulse-field gel electrophoresis and of DNA from human/rodent somatic cell hybrids, as well as the assembly of ordered sets of cosmids for this gene region into contigs. The different approaches yielded very similar results and indicate that the entire gene family is contained within a region located at position 19q13.1-q13.2 between the CYP2A and the D19S15/D19S8 markers. The physical linkage of nine genes belonging to the CEA subgroup and their location with respect to the pregnancy-specific glycoprotein (PSG) subgroup genes have been determined, and the latter are located closer to the telomere. From large groups of ordered cosmid clones, the identity of all known CEA subgroup genes has been confirmed either by hybridization using gene-specific probes or by DNA sequencing. These studies have identified a new member of the CEA subgroup (CGM8), which probably represents a pseudogene due to the existence of two stop codons, one in the leader and one in the N-terminal domain exons. The gene order and orientation, which were determined by hybridization with probes from the 5' and 3' regions of the genes, are as follows: cen/3'-CGM7-5'/3'-CGM2-5'/5'-CEA-3'/5'-NCA-3'/5'-CGM1- 3'/3'-BGP-5'/3'- CGM9-5'/3'-CGM6-5'/5'-CGM8-3'/PSGcluster/qter.


Subject(s)
Carcinoembryonic Antigen/genetics , Multigene Family , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 19 , Cosmids , DNA/genetics , DNA Probes , Electrophoresis, Gel, Pulsed-Field , Genetic Linkage , Humans , Hybrid Cells , Molecular Sequence Data , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...