Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mod Pathol ; 37(2): 100401, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38043787

ABSTRACT

Intrahepatic cholangiocarcinoma (iCCA) is an aggressive cancer composed of large-duct and small-duct types. Understanding the tumor immune microenvironment and its related vascular system is important for developing novel and efficient therapies. We focused on tertiary lymphoid structure (TLS) as a hallmark of antitumor immunity and investigated the clinicopathologic significance of TLSs and the influence of vascular microenvironment on TLS formation in iCCAs. We examined 261 iCCA cases clinicopathologically and analyzed the vascular system using immunohistochemistry. Single-cell (102,685 cells) and bulk RNA (33 iCCA cases) sequencing analyses were performed using data sets downloaded from public databases, and endothelial cell characteristics in iCCA tissues and functional networks related to the tumor microenvironment were bioinformatically examined. High densities of both intratumoral and peritumoral TLSs were significantly associated with prolonged survival only in large-duct-type iCCA. Multivariate analyses showed that peritumoral TLS was a prognostic factor for the large-duct type. TLS-rich iCCA had a significantly higher vein density and tumor-infiltrating T-cell count than TLS-poor iCCA. Both the presence of TLSs and high vein endothelial cells in iCCA tissues were significantly associated with molecular networks representing active immune responses in transcriptomic analysis. Vein density was a prognostic factor in patients with large-duct and small-duct types. This suggests that TLS formation is involved in a microenvironment with high vein density, which represents an antitumor-directed immune microenvironment.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Tertiary Lymphoid Structures , Humans , Prognosis , Tertiary Lymphoid Structures/pathology , Tumor Microenvironment , Endothelial Cells/pathology , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Bile Ducts, Intrahepatic/pathology , Bile Duct Neoplasms/pathology
2.
Histopathology ; 79(5): 872-879, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34288030

ABSTRACT

AIMS: Serous (cystic) neoplasm (SCN) of the pancreas is generally benign, and surgical treatment is recommended in only a limited number of cases. To avoid unnecessary surgery, an accurate diagnosis of SCN is essential. In the present study, we aimed to identify new immunohistochemical markers with which to distinguish SCN from other tumours. METHODS AND RESULTS: We compared the comprehensive gene expression profiles of SCN with those of normal pancreas and pancreatic ductal adenocarcinoma (PDAC). We selected the candidate molecules that were up-regulated in SCN, were minimally expressed or unexpressed in PDAC, and had specific and available antibodies suitable for immunohistochemistry, and then analysed their immunohistochemical expression in various tumours. We selected aquaporin 1 (AQP1), stereocilin (STRC), fibroblast growth factor receptor 3 (FGFR3), and transmembrane protein 255B (TMEM255B), which were diffusely expressed in SCN cells in 79%, 100%, 100% and 100% of SCN cases. AQP1 was not expressed in other tumours, except in 20% of mucinous cystic neoplasms (MCNs) and 19% of PDACs. STRC was rarely expressed in MCNs, neuroendocrine neoplasms (NENs), and PDACs. FGFR3 was expressed in 31% of intraductal papillary mucinous neoplasms (IPMNs), 50% of intraductal oncocytic papillary neoplasms, 40% of NENs, 30% of acinar cell carcinomas, 40% of solid pseudopapillary neoplasms, and 52% of PDACs. TMEM255B was not expressed in the other tumours, except in 50% of MCNs, 80% of gastric-subtype IPMNs, and 29% of PDACs. All antigens were usually expressed in a small proportion of cells when they were positive in tumours other than SCN. CONCLUSIONS: These findings indicate that AQP1 and STRC, and potentially TMEM255B, may act as SCN markers.


Subject(s)
Adenocarcinoma, Mucinous , Biomarkers, Tumor/metabolism , Pancreatic Neoplasms , Adenocarcinoma, Mucinous/diagnosis , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Aquaporin 1/metabolism , Diagnosis, Differential , Gene Expression Profiling , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Pancreas/pathology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms
3.
Oncol Lett ; 15(1): 747-754, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29391893

ABSTRACT

UL16 binding protein 1 (ULBP1) expressed on the tumor cell surface binds to the natural killer group 2 member D (NKG2D) receptor presenting on natural killer (NK), cluster of differentiation (CD)8+ T, and γ δ T cells. However, the roles of ULBP1 and NKG2D expression and associated immune responses in gastric cancer are unclear. The present study investigated the associations between ULBP1 and NKG2D expression and clinical outcomes in patients with gastric cancer. The levels of ULBP1 and NKG2D expression were examined in human gastric cancer cell lines and gastric cancer tissues from 98 patients who underwent surgery from 2004 to 2008. MKN-74 cells expressed ULBP1 with ULBP2, -5, or -6. NKG2D was expressed at a higher level following activation of T cells and NK cells. Among the tissue sections positive for NKG2D expression, 6 patients were positive for CD8 and CD56. In all tissues, NKG2D-expressing cells were typically aCD8+ T cells. Patients with NKG2D expression in tumors exhibited significantly longer overall survival (OS) compared with patients without NKG2D expression in tumors (P=0.0217). The longest OS was observed in patients positive for ULBP1 and NKG2D, whereas the shortest OS was observed in patients negative for ULBP1 and NKG2D. The interaction between ULBP1 and NKG2D may improve OS in patients with gastric cancer, and may have applications in immunotherapy for the induction of adaptive immunity in patients with cancer. Additionally, ULBP1 and NKG2D may be useful as prognostic biomarkers in gastric cancer.

4.
Anticancer Res ; 37(8): 4093-4101, 2017 08.
Article in English | MEDLINE | ID: mdl-28739693

ABSTRACT

BACKGROUND/AIM: We investigated the relationship between the expression of natural killer group 2, member D ligands (NKG2DLs) and the antitumor effects of protein-bound polysaccharide-K (PSK). MATERIALS AND METHODS: PSK was administered to evaluate its effectiveness against tumor growth. The expression of Rae-1 and H60 were analyzed in multiple cell lines. RESULTS: PSK showed the highest antitumor effects in mice implanted with cells expressing neither Rae-1 nor H60. PSK had little antitumor effect in mice implanted with cells expressing both Rae-1 and H60. A correlation between the expression of NKG2DLs and the antitumor effect of PSK was observed. After PSK administration, INF-γ production in CD8+ T cells increased in mice with cells expressing neither Rae-1 nor H60, but did not change in mice implanted with cells expressing both Rae-1 and H60. CONCLUSION: We demonstrated that the expression of NKG2DLs affects tumor immunity and the efficacy of immuno therapy in tumor-bearing mouse model.


Subject(s)
Fungal Proteins/administration & dosage , Minor Histocompatibility Antigens/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , Neoplasms/drug therapy , Nuclear Matrix-Associated Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Polysaccharides/administration & dosage , Animals , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Mice , Minor Histocompatibility Antigens/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Neoplasms/genetics , Neoplasms/pathology , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Xenograft Model Antitumor Assays
5.
Nihon Rinsho ; 75(2): 245-250, 2017 02.
Article in Japanese | MEDLINE | ID: mdl-30562859

ABSTRACT

Recently immune checkpoint inhibitors such as anti-CTLA-4, PD-1 and PD-L1 blocking antibodies have been developed resulting in the dramatic change of standard-of-care in can- cer treatment. Furthermore, in phase 3 study of nivolumab plus ipilimumab for metastatic malignant melanoma, the combination therapy prolonged progression-free survival and im- proved objective response rate, showing the promising activity of the combination therapy with immune checkpoint inhibitors. On the other hand, the frequencies of immune related adverse events increased. Now new immune checkpoint inhibitor is expected to develop so as to substitute for these or to be used with combination. Here we will address the mecha- nism and the function of Lag-3, TIGIT and Tim-3 that are expected as immune checkpoint inhibitors.


Subject(s)
Immunotherapy , Neoplasms/therapy , Humans , Neoplasms/immunology
6.
Cancer Sci ; 107(11): 1599-1609, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27561105

ABSTRACT

Cancer stem-like cells (CSLCs) in solid tumors are thought to be resistant to conventional chemotherapy or molecular targeting therapy and to contribute to cancer recurrence and metastasis. In this study, we aimed to identify a biomarker of pancreatic CSLCs (P-CSLCs). A P-CSLC-enriched population was generated from pancreatic cancer cell lines using our previously reported method and its protein expression profile was compared with that of parental cells by 2-D electrophoresis and tandem mass spectrometry. The results indicated that a chaperone protein calreticulin (CRT) was significantly upregulated in P-CSLCs compared to parental cells. Flow cytometry analysis indicated that CRT was mostly localized to the surface of P-CSLCs and did not correlate with the levels of CD44v9, another P-CSLC biomarker. Furthermore, the side population in the CRThigh /CD44v9low population was much higher than that in the CRTlow /CD44v9high population. Calreticulin expression was also assessed by immunohistochemistry in pancreatic cancer tissues (n = 80) obtained after radical resection and was found to be associated with patients' clinicopathological features and disease outcomes in the Cox proportional hazard regression model. Multivariate analysis identified CRT as an independent prognostic factor for pancreatic cancer patients, along with age and postoperative therapy. Our results suggest that CRT can serve as a biomarker of P-CSLCs and a prognostic factor associated with poorer survival of pancreatic cancer patients. This novel biomarker can be considered as a therapeutic target for cancer immunotherapy.


Subject(s)
Calreticulin/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , ATP-Binding Cassette Transporters/metabolism , CD47 Antigen/metabolism , Cell Line, Tumor , Humans , Hyaluronan Receptors/metabolism , Kaplan-Meier Estimate , Prognosis , Proportional Hazards Models , Proteomics
7.
J Immunol ; 187(11): 5921-30, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22043012

ABSTRACT

Class II major histocompatibility molecules are the primary susceptibility locus for many autoimmune disorders, including type 1 diabetes. Human DQ8 and I-A(g7), in the NOD mouse model of spontaneous autoimmune diabetes, confers diabetes risk by modulating presentation of specific islet peptides in the thymus and periphery. We used an in silico molecular docking program to screen a large "druglike" chemical library to define small molecules capable of occupying specific structural pockets along the I-A(g7) binding groove, with the objective of influencing presentation to T cells of the autoantigen insulin B chain peptide consisting of amino acids 9-23. In this study we show, using both murine and human cells, that small molecules can enhance or inhibit specific TCR signaling in the presence of cognate target peptides, based upon the structural pocket targeted. The influence of compounds on the TCR response was pocket dependent, with pocket 1 and 6 compounds inhibiting responses and molecules directed at pocket 9 enhancing responses to peptide. At nanomolar concentrations, the inhibitory molecules block the insulin B chain peptide consisting of amino acids 9-23, endogenous insulin, and islet-stimulated T cell responses. Glyphosine, a pocket 9 compound, enhances insulin peptide presentation to T cells at concentrations as low as 10 nM, upregulates IL-10 secretion, and prevents diabetes in NOD mice. These studies present a novel method for identifying small molecules capable of both stimulating and inhibiting T cell responses, with potentially therapeutic applications.


Subject(s)
Antigen Presentation/immunology , Autoantigens/immunology , Diabetes Mellitus, Type 1/immunology , Glycine/analogs & derivatives , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation/immunology , Organophosphorus Compounds/immunology , Alleles , Animals , Autoantigens/genetics , Diabetes Mellitus, Type 1/genetics , Enzyme-Linked Immunosorbent Assay , Glycine/immunology , Glycine/pharmacology , Histocompatibility Antigens Class II/genetics , Humans , Insulin/genetics , Insulin/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Models, Molecular , Organophosphorus Compounds/pharmacology , Peptide Fragments/genetics , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Small Molecule Libraries , Structure-Activity Relationship
8.
Exp Eye Res ; 90(1): 17-25, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19748503

ABSTRACT

Evidence is mounting that not only microangiopathy, but also neurodegenerative events occur in the retinas of humans and rodents with early diabetes. Diverse pathologies are known to alter the amount and/or location of glial expression of the water-selective channels aquaporins (AQPs) 1 and 4. However, the temporal relationships among glial activation, the altered expression of the AQP proteins and neuronal death in the retinas of diabetic animals remains to be investigated. Male spontaneously diabetic Torii (SDT) rats reportedly develop diabetes by 40 weeks of age at the latest and manifest proliferative diabetic retinopathy at 50 weeks or later. This study compared temporal changes in neuroretinal apoptosis, glial fibrillary acidic protein (GFAP) expression and the expression of AQPs 1 and 4 between SDT rat retinas and age-matched Sprague-Dawley (SD) rat retinas. Cell death was detected by terminal deoxynucleotidyl transferase-mediated deoxy-uridine triphosphate nick end-labeling on retinal flatmounts and activated caspase 3 immunofluorescence of retinal cryosections. The expression of GFAP and AQPs 1 and 4 was assessed by immunohistochemistry of cryosections and retinal flatmounts. Diabetes started to develop around 15 weeks in SDT rats. Apoptotic cells in the ganglion cell layer and the inner nuclear layer were significantly more numerous in 40-week-old SDT rat retinas than in either age-matched SD rat retinas or 10-week-old SDT rats. GFAP immunoreactivity was confined to the nerve fiber layer both in SD and SDT rats at 10 weeks, whereas it spanned the whole retina in SDT rats, but not in SD rats, at 40 weeks. AQP1 was expressed in the outer retina, whereas AQP4 was expressed in the perivascular and end feet of Müller cells and astrocytes in the inner retina in the control SD rats and the SDT rats at 10 weeks. The perivascular AQPs shifted from AQP4 to AQP1 in 40-week-old SDT rats that exhibited marked hyperglycemia. Thus, the development of diabetes increases neuroretinal apoptosis, and this coincides with an altered expression pattern of GFAP and water-selective channels AQPs 1 and 4 in SDT rats.


Subject(s)
Apoptosis , Aquaporin 1/metabolism , Aquaporin 4/metabolism , Diabetic Retinopathy/metabolism , Glial Fibrillary Acidic Protein/metabolism , Retinal Neurons/pathology , Animals , Blood Glucose/analysis , Body Weight , Caspase 3/metabolism , Diabetic Retinopathy/pathology , Disease Models, Animal , Fluorescent Antibody Technique, Indirect , Glycated Hemoglobin/analysis , In Situ Nick-End Labeling , Male , Microscopy, Fluorescence , Rats , Rats, Sprague-Dawley , Time Factors
9.
Physiol Genomics ; 35(1): 96-105, 2008 Sep 17.
Article in English | MEDLINE | ID: mdl-18612083

ABSTRACT

The pathogenesis of inflammation and fibrosis in the pancreatic islets in diabetes is largely unknown. Spontaneously diabetic Torii (SDT) rats exhibit inflammation and fibrosis in and around the islets during the development of the disease. We investigated genetic factors for diabetes, islet inflammation, and fibrosis in the SDT rat. We produced F1 and F2 rats by intercross between SDT and F344 rats, examined the onset of diabetes, glucose tolerance, and histology of the pancreas, and performed genetic analysis of these traits. We then established a congenic strain carrying the SDT allele at the strongest diabetogenic locus on the F344 genetic background and characterized glucose tolerance and histology of the pancreas. F1 rats showed glucose intolerance and inflammatory changes mainly in the islets. Genetic analysis of diabetes identified a major locus on chromosome 3, designated Dmsdt1, at which a dominantly acting SDT allele was involved. Quantitative trait locus (QTL) analysis of glucose tolerance revealed, in addition to Dmsdt1 [logarithm of odds (LOD) 5.3 near D3Mit12], three other loci, designated Dmsdt2 (LOD 4.2 at D8Rat46), Dmsdt3 (LOD 3.8 near D13Arb5), and Dmsdt4 (LOD 5.8 at D14Arb18). Analysis of a congenic strain for Dmsdt1 indicates that the dominantly acting SDT allele induces islet inflammation and fibrosis. Thus we have found a major locus on chromosome 3 for islet inflammation and fibrosis in the SDT rat. Identification of the genes responsible should provide insight into the pathogenesis of diabetes.


Subject(s)
Diabetes Mellitus/genetics , Islets of Langerhans/pathology , Pancreatitis/genetics , Quantitative Trait Loci , Alleles , Animals , Chromosomes, Mammalian/genetics , Diabetes Mellitus/pathology , Disease Models, Animal , Female , Fibrosis , Male , Pancreatitis/pathology , Phenotype , Rats , Rats, Inbred F344 , Rats, Inbred Strains
10.
Comp Med ; 58(6): 560-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19149413

ABSTRACT

The Komeda miniature rat Ishikawa (KMI) is a spontaneous animal model of dwarfism caused by a mutation in Prkg2, which encodes cGMP-dependent protein kinase type II (cGKII). This strain has been maintained as a segregating inbred strain for the mutated allele mri. In this study, we characterized the phenotype of the KMI strain, particularly growth traits, craniofacial measurements, and organ weights. The homozygous mutant (mri/mri) animals were approximately 70% to 80% of the size of normal, heterozygous (mri/+) animals in regard to body length, weight, and naso-occipital length of the calvarium, and the retroperitoneal fat of mri/mri rats was reduced greatly. In addition, among progeny of the (BNxKMI-mri/mri)F1xKMI-mri/mri backcross, animals with the KMI phenotype (mri/mri) were easily distinguished from those showing the wild-type phenotype (mri/+) by using growth traits such as body length and weight. Genetic analysis revealed that all of the backcrossed progeny exhibiting the KMI phenotype were homozygous for the KMI allele in the 1.2-cM region between D14Rat5 and D14Rat80 on chromosome 14, suggesting strongly that mri acts in a completely recessive manner. The KMI strain is the first and only rat model with a confirmed mutation in Prkg2 and is a valuable model for studying dwarfism and longitudinal growth traits in humans and for functional studies of cGKII.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/genetics , Dwarfism/genetics , Mutation , Animals , Body Weight/genetics , Chromosome Mapping , Cyclic GMP-Dependent Protein Kinase Type II , Dwarfism/enzymology , Dwarfism/pathology , Female , Male , Organ Size/genetics , Phenotype , Rats , Rats, Inbred BN , Rats, Mutant Strains
11.
Diabetes ; 55(7): 1930-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16804060

ABSTRACT

The ATP-sensitive K(+) channel (K(ATP) channel) in pancreatic beta-cells is a critical regulator in insulin secretion. We previously reported that transgenic mice expressing a dominant-negative form (Kir6.2G132S) of Kir6.2, a subunit of the K(ATP) channel, specifically in beta-cells develop severe hyperglycemia in adults (8 weeks of age). In this study, we conducted a long-term investigation of the phenotype of these transgenic mice. Surprisingly, hyperglycemia was spontaneously improved with concomitant improvement of pancreatic insulin content in the transgenic mice at >25 weeks of age. Insulin-positive cells and pancreatic duodenal homeobox 1 (PDX1)-positive cells both were clearly increased in the older compared with the younger transgenic mice. Interestingly, cells labeled with the lectin Dolichos biflorus agglutinin (DBA), a potential indicator of uncommitted pancreatic epithelial/ductal cells, were detected in the islets of the transgenic mice but not in those of wild-type mice. In addition, a subset of the DBA-labeled cells was positive for PDX1, insulin, glucagon, somatostatin, or pancreatic polypeptide. Moreover, some of the DBA-labeled cells were also positive for a proliferating cell marker. These results show that the Kir6.2G132S transgenic mouse is a useful model for studying beta-cell regeneration and that DBA-labeled cells participate in the process.


Subject(s)
Hyperglycemia/prevention & control , Islets of Langerhans/physiology , Potassium Channels, Inwardly Rectifying/physiology , Amino Acid Substitution , Animals , Apoptosis , Blood Glucose/metabolism , Glucose Tolerance Test , Glycine , In Situ Nick-End Labeling , Insulin/blood , Mice , Mice, Transgenic , Potassium Channels, Inwardly Rectifying/genetics , Serine
12.
Exp Anim ; 52(4): 295-301, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14562605

ABSTRACT

The Komeda diabetes-prone (KDP) rat is a spontaneous animal model of human autoimmune type 1 diabetes. By positional cloning of the non-MHC major susceptibility locus lddm/kdp1, we recently identified a nonsense mutation in Cblb and also found that lymphocytes of KDP rats infiltrate into various tissues, indicating autoimmunity. The maintenance and production of KDP rats has been a critical problem owing to the poor reproductive ability of diabetic animals. To solve the problem, we here established the KDP rat as a segregating inbred strain. We first identified animals that were heterozygous at the lddm/kdp1 region in a breeding colony of KDP rats. The heterozygous region spans at least from D11Yok1 to Cblb on rat chromosome 11. By mating between the heterozygous rats, we obtained homozygotes, heterozygotes and wild-types with the expected ratio of 1:2:1 and found that only the homozygotes developed diabetes, suggesting that these genotypes represent those of lddm/kdp1. We then tried to maintain KDP rats by mating between the heterozygotes, which resulted in a segregating inbred strain. Within 210 d of age, about 80% of lddm/kdp1 homozygotes developed diabetes with severe insulitis, while neither heterozygotes nor wild-types developed diabetes. The phenotypic characteristics of the homozygotes are the same as those of progeny of diabetic parents in the original KDP rats. The segregating inbred KDP rat strain described here would serve as a useful animal model for autoimmune diseases, including type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Disease Models, Animal , Rats, Inbred Strains/genetics , Animals , Autoimmune Diseases/genetics , Codon, Nonsense , Rats
13.
Brain Res Mol Brain Res ; 112(1-2): 1-7, 2003 Apr 10.
Article in English | MEDLINE | ID: mdl-12670697

ABSTRACT

Reelin (Reln) is an extracellular matrix protein secreted from distinct neuronal populations and controls neural cell positioning during brain development. Alterations of human RELN have been reported in two pedigrees with an autosomal recessive lissencephaly. Although several alleles of the mouse reeler mutation were identified as disruptions of Reln, there is no other animal model with a confirmed mutation in Reln. We recently established the Komeda Zucker creeping (KZC) rat strain with an autosomal recessive mutation creeping (cre), showing a reeler-like phenotype. We also found that creeping was located in the genomic segment on rat chromosome 4 containing Reln and that the expression level of Reln mRNA was markedly reduced in cre/cre homozygous mutant animals. Here we report positional candidate cloning of creeping, and identify a nucleotide insertion mutation in Reln. This mutation leads to a translational frameshift and results in truncation of the predicted protein in the fourth reelin-specific repeat, removing the C-terminal region required for secretion and function of the protein. We conclude that the mutation detected here is causative and is probably a null allele. The KZC rat is the first rat model with a confirmed Reln mutation and would therefore contribute to the understanding of the Reln function.


Subject(s)
Brain/abnormalities , Cell Adhesion Molecules, Neuronal/deficiency , Extracellular Matrix Proteins/deficiency , Movement Disorders/genetics , Mutation/genetics , Neurons/metabolism , Animals , Base Sequence/genetics , Brain/metabolism , Brain/pathology , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/isolation & purification , Chromosome Mapping , Cloning, Molecular , Codon, Nonsense/genetics , DNA Mutational Analysis , DNA, Complementary/analysis , DNA, Complementary/genetics , Disease Models, Animal , Exons/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/isolation & purification , Genotype , Molecular Sequence Data , Movement Disorders/metabolism , Nerve Tissue Proteins , Neurons/pathology , Phenotype , Rats , Rats, Inbred Strains , Reelin Protein , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Serine Endopeptidases
14.
Biochem Biophys Res Commun ; 304(1): 196-206, 2003 Apr 25.
Article in English | MEDLINE | ID: mdl-12705906

ABSTRACT

The Spontaneously Diabetic Torii (SDT) rat has recently been established as a new rat model of nonobese type 2 diabetes. In this study, we characterized diabetic features in SDT rats, and performed quantitative trait locus (QTL) analysis for glucose intolerance using 319 male (BNxSDT)xSDT backcrosses. Male SDT rats exhibited glucose intolerance at 20 weeks, and spontaneously developed diabetes with the incidence of 100% at 38 weeks, and glucose intolerance is well associated with the development of diabetes. The QTL analysis identified three highly significant QTLs (Gisdt1, Gisdt2, and Gisdt3) for glucose intolerance on rat chromosomes 1, 2, and X, respectively. The SDT allele for these QTLs significantly exacerbated glucose intolerance. Furthermore, synergistic interactions among these QTLs were detected. These findings indicate that diabetic features in SDT rats are inherited as polygenic traits and that SDT rats would provide insights into genetics of human type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Rats, Inbred Strains , Age of Onset , Animals , Blood Glucose/analysis , Body Weight/genetics , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Genotype , Glucose Intolerance/blood , Glucose Intolerance/genetics , Male , Phenotype , Quantitative Trait Loci , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...