Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Cancer Med ; 12(14): 15650-15663, 2023 07.
Article in English | MEDLINE | ID: mdl-37293953

ABSTRACT

INTRODUCTION: B-cell lymphoma/leukemia 11B (BCL11B) is a subunit of SWI/SNF chromatin remodeling complexes and functions in cell cycle regulation and apoptosis upon DNA replication stress and damages via transcription. Many malignancies were reported to exhibit changes in BCL11B gene expression; however, no study has focused on the relationship between BCL11B and hepatocellular carcinoma, which potentially exhibits DNA replication stress and damages upon its oncogenesis. Thus, in this study, we examined the molecular characterization of BCL11B expression in hepatocellular carcinoma. METHODS AND RESULTS: The cumulative progression-free survival and overall survival were significantly longer in the clinical cases of BCL11B-negative hepatocellular carcinoma than BCL11B-positve cases. Microarray and real-time PCR analyses in hepatocellular carcinoma cell lines indicated a correlation between BCL11B and GATA6, a gene reported to be correlated with oncogenic activities and resistance to anthracycline, which is often used for hepatocellular carcinoma chemotherapy. Consequently, BCL11B-overexpressing cell lines exhibited resistance to anthracycline in cell growth assays and the resistance has been evidenced by the increased expression of BCL-xL in cell lines. The results were supported by the analyses of human HCC samples showing the correlation between BCL11B and GATA6 expressions. DISCUSSIONS AND CONCLUSION: Our results indicated that overexpression of BCL11B amplifies GATA6 expression in hepatocellular carcinoma in vitro and in vivo that leads to anti-apoptotic signal activation, and induces resistance to chemotherapy, which influenced the postoperative prognosis.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Transcription Factors/genetics , Prognosis , Anthracyclines , Apoptosis/physiology , Gene Expression Regulation, Neoplastic
2.
Biomedicines ; 10(9)2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36140322

ABSTRACT

An MSM/Ms strain was established using Japanese wild mice, which exhibit resistance to several phenotypes associated with aging, such as obesity, inflammation, and tumorigenesis, compared to common inbred mouse strains. MSM/Ms strain is resistant to age-related hearing loss, and their auditory abilities are sustained for long durations. The age-related hearing loss 3 (ahl3) locus contributes to age-related hearing in MSM/Ms strain. We generated ahl3 congenic strains by transferring a genomic region on chromosome 17 from MSM/Ms mice into C57BL/6J mice. Although C57BL/6J mice develop age-related hearing loss because of the ahl allele of the cadherin 23 gene, the development of middle- to high-frequency hearing loss was significantly delayed in an ahl3 congenic strain. Moreover, the novel age-related hearing loss 10 (ahl10) locus associated with age-related hearing resistance in MSM/Ms strain was mapped to chromosome 12. Although the resistance effects in ahl10 congenic strain were slightly weaker than those in ahl3 congenic strain, slow progression of age-related hearing loss was confirmed in ahl10 congenic strain despite harboring the ahl allele of cadherin 23. These results suggest that causative genes and polymorphisms of the ahl3 and ahl10 loci are important targets for the prevention and treatment of age-related hearing loss.

3.
J Invest Dermatol ; 139(7): 1459-1469, 2019 07.
Article in English | MEDLINE | ID: mdl-30684556

ABSTRACT

Identification of the specific genetic variants responsible for the increased susceptibility to familial or sporadic cancers is important. Using a forward genetics approach to map such loci in a mouse skin cancer model, we previously identified a strong genetic locus, Stmm3, conferring resistance to chemically induced skin papillomas on chromosome 4. Here, we report the cyclin-dependent kinase inhibitor gene Cdkn2a/p19Arf as a major responsible gene for the Stmm3 locus. We provide evidence that the function of Stmm3 is dependent on p53 and that p19ArfMSM confers stronger resistance to papillomas than p16Ink4aMSMin vivo. In addition, we found that genetic polymorphism in p19Arf between a resistant strain, MSM/Ms (Val), and a susceptible strain, FVB/N (Leu), alters the susceptibility to papilloma development, malignant conversion, and the epithelial-mesenchymal transition. Moreover, we demonstrated that the p19ArfMSM allele more efficiently activates the p53 pathway than the p19ArfFVB allele in vitro and in vivo. Furthermore, we found polymorphisms in CDKN2A in the vicinity of a polymorphism in mouse Cdkn2a associated with the risk of human cancers in the Japanese population. Genetic polymorphisms in Cdkn2a and CDKN2A may affect the cancer risk in both mice and humans.


Subject(s)
Chromosomes, Human, Pair 4/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Genotype , Papilloma/genetics , Skin Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Alleles , Animals , Carcinogenesis/genetics , Disease Models, Animal , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Mice , Mice, Knockout , Papilloma/chemically induced , Polymorphism, Single Nucleotide , Skin Neoplasms/chemically induced , Tumor Suppressor Protein p53/genetics
4.
Sci Rep ; 7(1): 11208, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28894263

ABSTRACT

Using a forward genetics approach to map loci in a mouse skin cancer model, we previously identified a genetic locus, Skin tumour modifier of MSM 1 (Stmm1) on chromosome 7, conferring strong tumour resistance. Sub-congenic mapping localized Parathyroid hormone (Pth) in Stmm1b. Here, we report that serum intact-PTH (iPTH) and a genetic polymorphism in Pth are important for skin tumour resistance. We identified higher iPTH levels in sera from cancer-resistant MSM/Ms mice compared with susceptible FVB/NJ mice. Therefore, we performed skin carcinogenesis experiments with MSM-BAC transgenic mice (Pth MSM-Tg) and Pth knockout heterozygous mice (Pth +/-). As a result, the higher amounts of iPTH in sera conferred stronger resistance to skin tumours. Furthermore, we found that the coding SNP (rs51104087, Val28Met) localizes in the mouse Pro-PTH encoding region, which is linked to processing efficacy and increased PTH secretion. Finally, we report that PTH increases intracellular calcium in keratinocytes and promotes their terminal differentiation. Taken together, our data suggest that Pth is one of the genes responsible for Stmm1, and serum iPTH could serve as a prevention marker of skin cancer and a target for new therapies.


Subject(s)
Calcium-Regulating Hormones and Agents/genetics , Calcium-Regulating Hormones and Agents/metabolism , Genetic Predisposition to Disease , Parathyroid Hormone/genetics , Parathyroid Hormone/metabolism , Skin Neoplasms/epidemiology , Skin Neoplasms/genetics , Animals , Disease Models, Animal , Mice , Mice, Knockout , Mice, Transgenic , Polymorphism, Single Nucleotide
5.
Nat Commun ; 8(1): 702, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28951542

ABSTRACT

T-lineage committed precursor thymocytes are screened by a fate-determination process mediated via T cell receptor (TCR) signals for differentiation into distinct lineages. However, it remains unclear whether any antecedent event is required to couple TCR signals with the transcriptional program governing lineage decisions. Here we show that Bcl11b, known as a T-lineage commitment factor, is essential for proper expression of ThPOK and Runx3, central regulators for the CD4-helper/CD8-cytotoxic lineage choice. Loss of Bcl11b results in random expression of these factors and, thereby, lineage scrambling that is disconnected from TCR restriction by MHC. Initial Thpok repression by Bcl11b prior to the pre-selection stage is independent of a known silencer for Thpok, and requires the last zinc-finger motif in Bcl11b protein, which by contrast is dispensable for T-lineage commitment. Collectively, our findings shed new light on the function of Bcl11b in priming lineage-specifying genes to integrate TCR signals into subsequent transcriptional regulatory mechanisms.CD4 and CD8 T cells develop in the thymus with their transcription programs controlled by ThPOK and Runx3, respectively. Here the authors show that a pre-commitment event modulated by the transcription factor, Bcl11b, is required for the proper expression of ThPOK and Runx3 and correct CD4/CD8 lineage commitment.


Subject(s)
Cell Differentiation/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Repressor Proteins/genetics , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Helper-Inducer/cytology , Thymocytes/cytology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Lineage , Gene Expression Regulation , Mice , Receptors, Antigen, T-Cell/genetics
6.
Biosci Biotechnol Biochem ; 81(5): 922-930, 2017 May.
Article in English | MEDLINE | ID: mdl-28067590

ABSTRACT

BCL11B is a zinc finger-type transcription factor that regulates the development of the white adipose tissue (WAT), skin, central nervous system, and immune system. BCL11B is required for proper adipocyte differentiation, and BCL11B-/- embryos at E19.5 have very low amounts of the subcutaneous WAT. Here, we demonstrated that BCL11B+/- mice have lower body weight than BCL11B+/+ mice, whereas the expression of adipogenic marker genes in the WAT was comparable between BCL11B+/+ and BCL11B+/- mice. Histological analysis indicated that BCL11B+/- mice fed a high-fat diet have much smaller white adipocytes and lipid droplets in the WAT and liver, respectively. In addition, BCL11B+/- mice had increased energy consumption under both standard and high-fat diets. Thus, this study identifies BCL11B as a regulator of energy metabolism, and it is unlikely that BCL11B functions in the WAT contribute to energy metabolism in BCL11B+/- mice.


Subject(s)
Adipogenesis/genetics , Body Weight/genetics , Energy Metabolism/genetics , Heterozygote , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Cell Differentiation/genetics , Diet, High-Fat/adverse effects , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/chemically induced , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Repressor Proteins/deficiency , Tumor Suppressor Proteins/deficiency
7.
Sci Rep ; 6: 32750, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27586877

ABSTRACT

The differentiation of preadipocytes into adipocytes is controlled by several transcription factors, including peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα), which are known as master regulators of adipogenesis. BCL11B is a zinc finger-type transcription factor that regulates the development of the skin and central nervous and immune systems. Here, we found that BCL11B was expressed in the white adipose tissue (WAT), particularly the subcutaneous WAT and that BCL11B(-/-) mice had a reduced amount of subcutaneous WAT. During adipogenesis, BCL11B expression transiently increased in 3T3-L1 preadipocytes and mouse embryonic fibroblasts (MEFs). The ability for adipogenesis was reduced in BCL11B knockdown 3T3-L1 cells and BCL11B(-/-) MEFs, whereas the ability for osteoblastogenesis was unaffected in BCL11B(-/-) MEFs. Luciferase reporter gene assays revealed that BCL11B stimulated C/EBPß activity. Furthermore, the expression of downstream genes of the Wnt/ß-catenin signaling pathway was not suppressed in BCL11B(-/-) MEFs during adipogenesis. Thus, this study identifies BCL11B as a novel regulator of adipogenesis, which works, at least in part, by stimulating C/EBPß activity and suppressing the Wnt/ß-catenin signaling pathway.


Subject(s)
Adipogenesis , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Repressor Proteins/metabolism , Subcutaneous Fat/physiology , Tumor Suppressor Proteins/metabolism , Adipocytes/physiology , Animals , Cells, Cultured , Fibroblasts/physiology , Gene Expression Profiling , Gene Expression Regulation , Mice , Mice, Knockout , Repressor Proteins/deficiency , Tumor Suppressor Proteins/deficiency , Wnt Signaling Pathway
8.
Dev Biol ; 416(1): 98-110, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27287879

ABSTRACT

Molecular mechanisms underlying the development and morphogenesis of oral epithelia, comprising the gustatory and nongustatory epithelium, remain unclear. Here, we show that Bcl11b, a zinc finger transcription factor, plays an important role in the development of lingual papillae, especially filiform papillae. In both gustatory and nongustatory epithelium, Bcl11b was expressed in keratin 14-positive epithelial basal cells, which differentiate into keratinocytes and/or taste cells. Loss of Bcl11b function resulted in abnormal morphology of the gustatory papillae: flattened fungiform papillae, shorter trench wall in the foliate and circumvallate papillae, and ectopic invagination in more than half of circumvallate papillae. However, Bcl11b loss caused no effect on differentiation of taste receptor cells. In nongustatory epithelium, the impact of Bcl11b deficiency was much more striking, resulting in a smooth surface on the tongue tip and hypoplastic filiform papillae in the dorsal lingual epithelium. Immunohistochemical analyses revealed that a keratinocyte differentiation marker, Tchh expression was severely decreased in the Bcl11b(-/-) filiform papillae. In addition, expression of Pax9, required for morphogenesis of filiform papillae and its downstream target genes, hard keratins, almost disappeared in the tongue tip and was decreased in the dorsal tongue of Bcl11b(-/-) mice. Gene expression analyses demonstrated a delayed onset of expression of epithelial differentiation complex genes, which disturbed barrier formation in the mutant tongue. These results indicate that Bcl11b regulates the differentiation of keratinocytes in the tongue and identify Bcl11b as an essential factor for the lingual papilla morphogenesis.


Subject(s)
Repressor Proteins/physiology , Tongue/embryology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Differentiation , Keratinocytes/cytology , Mice , Morphogenesis/genetics , Mouth Mucosa/cytology , Mouth Mucosa/embryology , Repressor Proteins/genetics , Taste Buds/embryology , Tongue/ultrastructure , Tumor Suppressor Proteins/genetics
9.
Hum Mol Genet ; 25(10): 2045-2059, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26936824

ABSTRACT

Most clinical reports have suggested that patients with congenital profound hearing loss have recessive mutations in deafness genes, whereas dominant alleles are associated with progressive hearing loss (PHL). Jackson shaker (Ush1gjs) is a mouse model of recessive deafness that exhibits congenital profound deafness caused by the homozygous mutation of Ush1g/Sans on chromosome 11. We found that C57BL/6J-Ush1gjs/+ heterozygous mice exhibited early-onset PHL (ePHL) accompanied by progressive degeneration of stereocilia in the cochlear outer hair cells. Interestingly, ePHL did not develop in mutant mice with the C3H/HeN background, thus suggesting that other genetic factors are required for ePHL development. Therefore, we performed classical genetic analyses and found that the occurrence of ePHL in Ush1gjs/+ mice was associated with an interval in chromosome 10 that contains the cadherin 23 gene (Cdh23), which is also responsible for human deafness. To confirm this mutation effect, we generated C57BL/6J-Ush1gjs/+, Cdh23c.753A/G double-heterozygous mice by using the CRISPR/Cas9-mediated Cdh23c.753A>G knock-in method. The Cdh23c.753A/G mice harbored a one-base substitution (A for G), and the homozygous A allele caused moderate hearing loss with aging. Analyses revealed the complete recovery of ePHL and stereocilia degeneration in C57BL/6J-Ush1gjs/+ mice. These results clearly show that the development of ePHL requires at least two mutant alleles of the Ush1g and Cdh23 genes. Our results also suggest that because the SANS and CDH23 proteins form a complex in the stereocilia, the interaction between these proteins may play key roles in the maintenance of stereocilia and the prevention of ePHL.


Subject(s)
Cadherins/genetics , Hearing Loss/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Alleles , Amino Acid Sequence/genetics , Animals , Chromosomes, Human, Pair 10/genetics , Disease Models, Animal , Hair Cells, Auditory, Outer/pathology , Hearing Loss/pathology , Heterozygote , Homozygote , Humans , Mice , Stereocilia/pathology
10.
Carcinogenesis ; 36(6): 622-31, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25827435

ABSTRACT

SWI/SNF chromatin remodeling complexes constitute a highly related family of multi-subunit complexes to modulate transcription, and SWI/SNF subunit genes are collectively mutated in 20% of all human cancers. Bcl11b is a SWI/SNF subunit and acts as a haploinsufficient tumor suppressor in leukemia/lymphomas. Here, we show expression of Bcl11b in intestinal crypt cells and promotion of intestinal tumorigenesis by Bcl11b attenuation in Apc (min/+) mice. Of importance, mutations or allelic loss of BCL11B was detected in one-third of human colon cancers. We also show that attenuated Bcl11b activity in the crypt base columnar (CBC) cells expressing the Lgr5 stem cell marker enhanced regeneration of intestinal epithelial cells after the radiation-induced injury. Interestingly, BCL11B introduction in human cell lines downregulated transcription of ß-catenin target genes, whereas Bcl11b attenuation in Lgr5(+) CBCs increased expression of ß-catenin targets including c-Myc and cyclin D1. Together, our results argue that Bcl11b impairment promotes tumor development in mouse and human intestine at least in part through deregulation of ß-catenin pathway.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomal Proteins, Non-Histone/genetics , Colonic Neoplasms/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , beta Catenin/metabolism , Adenoma/classification , Adenoma/genetics , Animals , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/classification , Cyclin D1/biosynthesis , HCT116 Cells , HEK293 Cells , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-myc/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Repressor Proteins/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/biosynthesis , beta Catenin/genetics
11.
Int Immunol ; 27(4): 205-15, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25422283

ABSTRACT

If Bcl11b activity is compromised, CD4(+)CD8(+) double-positive (DP) thymocytes produce a greatly increased fraction of innate CD8(+) single-positive (SP) cells highly producing IFN-γ, which are also increased in mice deficient of genes such as Itk, Id3 and NF-κB1 that affect TCR signaling. Of interest, the increase in the former two is due to the bystander effect of IL-4 that is secreted by promyelocytic leukemia zinc finger-expressing NKT and γδT cells whereas the increase in the latter is cell intrinsic. Bcl11b zinc-finger proteins play key roles in T cell development and T cell-mediated immune response likely through TCR signaling. We examined thymocytes at and after the DP stage in Bcl11b (F/S826G) CD4cre, Bcl11b (F/+) CD4cre and Bcl11b (+/S826G) mice, carrying the allele that substituted serine for glycine at the position of 826. Here we show that Bcl11b impairment leads to an increase in the population of TCRαß(high)CD44(high)CD122(high) innate CD8SP thymocytes, together with two different developmental abnormalities: impaired positive and negative selection accompanying a reduction in the number of CD8SP cells, and developmental arrest of NKT cells at multiple steps. The innate CD8SP thymocytes express Eomes and secrete IFN-γ after stimulation with PMA and ionomycin, and in this case their increase is not due to a bystander effect of IL-4 but cell intrinsic. Those results indicate that Bcl11b regulates development of different thymocyte subsets at multiple stages and prevents an excess of innate CD8SP thymocytes.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Hyaluronan Receptors/metabolism , Inhibitor of Differentiation Proteins/genetics , Interferon-gamma/biosynthesis , Interleukin-2 Receptor beta Subunit/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , Natural Killer T-Cells/immunology , Protein-Tyrosine Kinases/genetics , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/immunology , Signal Transduction/immunology
12.
BMC Gastroenterol ; 14: 160, 2014 Sep 13.
Article in English | MEDLINE | ID: mdl-25218883

ABSTRACT

BACKGROUND: Intrahepatic cholestasis of pregnancy (ICP) is a cholestasis condition caused by elevated levels of serum bile acids that mainly occurs in the third trimester of pregnancy. Maternal symptoms include pruritus; elevation of transaminases, biliary enzymes, and bilirubin levels; and abnormal liver function tests. Fetal symptoms include spontaneous preterm labor, fetal distress, and intrauterine death. It is more prevalent in the Caucasians and is rarely found in Asian countries, including Japan. The etiology of ICP has been reported as involving various factors such as, environmental factors, hormone balance, and genetic components. The genetic factors include single-nucleotide polymorphisms (SNPs) in the genes of canalicular transporters, including ABCB4 and ABCB11. It has also been reported that the combination of these SNPs induces severe cholestasis and liver dysfunction. CASE PRESENTATION: Here, we report for the first time a 24-year Japanese case of severe ICP diagnosed by typical symptoms, serum biochemical analysis, and treated with the administration of ursodeoxycholic acid which improved cholestasis and liver injury and prevented fetal death. The sequence analysis showed SNPs reported their association with ICP in the ABCB11 (rs2287622, V444A) and ABCB4 (rs1202283, N168N) loci. CONCLUSION: The risk of ICP has been reported to be population-specific, and it is rare in the Japanese population. Our case was successfully treated with ursodeoxycholic acid and the genetic sequence analysis has supported the diagnosis. Because genetic variation in ABCB4 and ABCB11 has also been reported in the Japanese population, we need to be aware of potential ICP cases in pregnant Japanese women although further studies are necessary.


Subject(s)
Cholagogues and Choleretics/therapeutic use , Cholestasis, Intrahepatic/drug therapy , Pregnancy Complications/drug therapy , Ursodeoxycholic Acid/therapeutic use , Asian People , Female , Humans , Pregnancy , Treatment Outcome , Young Adult
13.
Exp Anim ; 63(3): 339-48, 2014.
Article in English | MEDLINE | ID: mdl-25077764

ABSTRACT

Genome-wide association studies have revealed that many low-penetrance cancer susceptibility loci are located throughout the genome; however, a very limited number of genes have been identified so far. Using a forward genetics approach to map such loci in a mouse skin cancer model, we previously identified strong genetic loci conferring resistance to chemically induced skin papillomas on chromosome 4 and 7 with a large number of [(FVB/N × MSM/Ms) F1 × FVB/N] backcross mice. In this report, we describe a combination of congenic mapping and allele-specific alteration analysis of the loci on chromosome 4. We used linkage analysis and a congenic mouse strain, FVB.MSM-Stmm3 to refine the location of Stmm3 (Skin tumor modifier of MSM 3) locus within a physical interval of about 34 Mb on distal chromosome 4. In addition, we used patterns of allele-specific imbalances in tumors from N2 and N10 congenic mice to narrow down further the region of Stmm3 locus to a physical distance of about 25 Mb. Furthermore, immunohistochemical analysis showed papillomas from congenic mice had less proliferative activity. These results suggest that Stmm3 responsible genes may have an influence on papilloma formation in the two-stage skin carcinogenesis by regulating papilloma growth rather than development.


Subject(s)
Chromosomes, Mammalian/genetics , Genetic Loci/genetics , Genetic Predisposition to Disease/genetics , Papilloma/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Animals , Chromosome Mapping , Genome-Wide Association Study , Humans , Mice, Congenic , Mice, Inbred Strains , Neoplasm Staging , Papilloma/chemically induced , Papilloma/pathology , Skin Neoplasms/pathology
14.
PLoS One ; 9(7): e102111, 2014.
Article in English | MEDLINE | ID: mdl-25013928

ABSTRACT

Previous studies have shown that Meis1 plays an important role in blood development and vascular homeostasis, and can induce blood cancers, such as leukemia. However, its role in epithelia remains largely unknown. Here, we uncover two roles for Meis1 in the epidermis: as a critical regulator of epidermal homeostasis in normal tissues and as a proto-oncogenic factor in neoplastic tissues. In normal epidermis, we show that Meis1 is predominantly expressed in the bulge region of the hair follicles where multipotent adult stem cells reside, and that the number of these stem cells is reduced when Meis1 is deleted in the epidermal tissue of mice. Mice with epidermal deletion of Meis1 developed significantly fewer DMBA/TPA-induced benign and malignant tumors compared with wild-type mice, suggesting that Meis1 plays a role in both tumor development and malignant progression. This is consistent with the observation that Meis1 expression increases as tumors progress from benign papillomas to malignant carcinomas. Interestingly, we found that Meis1 localization was altered to neoplasia development. Instead of being localized to the stem cell region, Meis1 is localized to more differentiated cells in tumor tissues. These findings suggest that, during the transformation from normal to neoplastic tissues, a functional switch occurs in Meis1.


Subject(s)
Carcinoma/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Neoplasm Proteins/genetics , Papilloma/genetics , Skin Neoplasms/genetics , 9,10-Dimethyl-1,2-benzanthracene , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Carcinogens , Carcinoma/chemically induced , Carcinoma/metabolism , Carcinoma/pathology , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Progression , Hair Follicle/drug effects , Hair Follicle/metabolism , Hair Follicle/pathology , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Transgenic , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/pathology , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/deficiency , Papilloma/chemically induced , Papilloma/metabolism , Papilloma/pathology , Signal Transduction , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/analogs & derivatives
15.
PLoS One ; 9(5): e97201, 2014.
Article in English | MEDLINE | ID: mdl-24844776

ABSTRACT

Genome-wide association studies have revealed that many low-penetrance cancer susceptibility loci are located throughout the genome; however, a very limited number of genes have been identified so far. Using a forward genetics approach to map such loci in a mouse skin cancer model, we previously identified strong genetic loci conferring resistance to early-stage chemically induced skin papillomas on chromosome 7 with a large number of [(FVB/N×MSM/Ms)×FVB/N] F1 backcross mice. In this report, we describe a combination of congenic mapping and allele-specific alteration analysis of the loci on chromosome 7. We used linkage analysis and congenic mouse strains to refine the location of Stmm1 (Skin tumor modifier of MSM 1) locus within a genetic interval of about 3 cM on proximal chromosome 7. In addition, we used patterns of allele-specific imbalances in tumors from F1 backcross and N10 congenic mice to narrow down further the region of Stmm1 locus to a physical distance of about 5.4 Mb. To gain the insight into the function of Stmm1 locus, we carried out a long term BrdU labelling experiments with congenic mice containing Stmm1 locus. Interestingly, we observed a decrease of BrdU-LRCs (Label Retaining Cells) in a congenic strain heterozygous or homozygous for MSM allele of Stmm1. These results suggest that Stmm1 responsible genes may have an influence on papillomagenesis in the two-stage skin carcinogenesis by regulating epidermal quiescent stem cells.


Subject(s)
Alleles , Cell Transformation, Neoplastic/genetics , Chromosome Mapping , Chromosomes, Mammalian/genetics , Genetic Loci , Papilloma/genetics , Skin Neoplasms/genetics , Animals , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/pathology , Genetic Linkage , Mice , Papilloma/chemically induced , Papilloma/pathology , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology
16.
J Immunol ; 192(5): 2210-8, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24489104

ABSTRACT

γδ T cells develop at the double-negative (DN) 2 and DN3 stages and acquire functions to produce IL-17 and IFN-γ in fetal thymus. However, the relationship between differentiation stages and their functions was unclear. In this study, we found that, although IFN-γ-producing and IL-17-producing γδ T cells developed from DN2 cells, only IFN-γ-producing γδ T cells developed from DN3 cells, indicating the direct generation of IL-17-producing γδ T cells from the DN2 stage, not through the DN3 stage. Single-cell analysis revealed that DN2 cells contained heterogeneous γδ T cell precursors with or without an ability to develop IL-17 producers. Inactivation of B cell leukemia/lymphoma 11b, a zinc finger transcription factor responsible for transition from early to late stages of DN2 cells, completely abrogated the development of IL-17-producing γδ T cells, although a unique subset of IFN-γ-producing γδ T cells expressing a high level of promyelocytic leukemia zinc finger was able to develop. Thus, our results reveal that γδ T cells are functionally differentiated to IFN-γ and IL-17 producers at different developmental stages in fetal thymus.


Subject(s)
Interferon-gamma/immunology , Interleukin-17/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Thymus Gland/embryology , Animals , Interferon-gamma/genetics , Interleukin-17/genetics , Mice , Mice, Mutant Strains , Receptors, Antigen, T-Cell, gamma-delta/genetics , Repressor Proteins/genetics , Repressor Proteins/immunology , T-Lymphocytes/cytology , Thymus Gland/immunology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
17.
Exp Anim ; 62(4): 333-46, 2013.
Article in English | MEDLINE | ID: mdl-24172198

ABSTRACT

The waltzer (v) mouse mutant harbors a mutation in Cadherin 23 (Cdh23) and is a model for Usher syndrome type 1D, which is characterized by congenital deafness, vestibular dysfunction, and prepubertal onset of progressive retinitis pigmentosa. In mice, functionally null Cdh23 mutations affect stereociliary morphogenesis and the polarity of both cochlear and vestibular hair cells. In contrast, the murine Cdh23(ahl) allele, which harbors a hypomorphic mutation, causes an increase in susceptibility to age-related hearing loss in many inbred strains. We produced congenic mice by crossing mice carrying the v niigata (Cdh23(v-ngt)) null allele with mice carrying the hypomorphic Cdh23(ahl) allele on the C57BL/6J background, and we then analyzed the animals' balance and hearing phenotypes. Although the Cdh23(v-ngt/ahl) compound heterozygous mice exhibited normal vestibular function, their hearing ability was abnormal: the mice exhibited higher thresholds of auditory brainstem response (ABR) and rapid age-dependent elevation of ABR thresholds compared with Cdh23(ahl/ahl) homozygous mice. We found that the stereocilia developed normally but were progressively disrupted in Cdh23(v-ngt/ahl) mice. In hair cells, CDH23 localizes to the tip links of stereocilia, which are thought to gate the mechanoelectrical transduction channels in hair cells. We hypothesize that the reduction of Cdh23 gene dosage in Cdh23(v-ngt/ahl) mice leads to the degeneration of stereocilia, which consequently reduces tip link tension. These findings indicate that CDH23 plays an important role in the maintenance of tip links during the aging process.


Subject(s)
Alleles , Cadherins/genetics , Cadherins/physiology , Hearing Loss/genetics , Heterozygote , Mutation , Aging/genetics , Aging/pathology , Animals , Cadherins/metabolism , Disease Progression , Gene Dosage , Hearing Loss/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nerve Degeneration/genetics , Stereocilia/metabolism , Stereocilia/pathology
18.
Mech Dev ; 130(9-10): 482-92, 2013.
Article in English | MEDLINE | ID: mdl-23727454

ABSTRACT

Rodent incisors maintain the ability to grow continuously and their labial dentin is covered with enamel. Bcl11b zinc-finger transcription factor is expressed in ameloblast progenitors in mouse incisors and its absence in Bcl11b(KO/KO) mice results in a defect in embryonic tooth development. However, the role of Bcl11b in incisor maintenance in adult tissue was not studied because of death at birth in Bcl11b(KO/KO) mice. Here, we examined compound heterozygous Bcl11b(S826G/KO) mice, one allele of which has an amino acid substitution of serine at position 826 for glycine, that exhibited hypoplastic maxillary incisors with lower concentrations of minerals at the enamel and the dentin, accompanying the maxillary bone hypoplasia. Histological examinations revealed hypoplasia of the labial cervical loop in incisors, shortening of the ameloblast progenitor region, and impairment in differentiation and proliferation of ameloblast-lineage cells. Interestingly, however, juvenile mice at 5days after birth did not show marked change in these phenotypes. These results suggest that attenuated Bcl11b activity impairs ameloblast progenitors and incisor maintenance. The number of BrdU label-retaining cells, putative stem cells, was lower in Bcl11b(S826G/KO) incisors, which suggests the incisor hypoplasia may be in part a result of the decreased number of stem cells. Interestingly, the level of Shh and FGF3 expressions, which are assumed to play key roles in the development and maintenance of ameloblasts and odontoblasts, was not decreased, though the expressed areas were more restricted in ameloblast progenitor and mesenchyme regions of Bcl11b(S826G/KO) incisors, respectively. Those data suggest that the incisor maintenance by Bcl11b is not directly related to the FGF epithelial-mesenchymal signaling loop including Shh but is intrinsic to ameloblast progenitors and possibly stem cells.


Subject(s)
Ameloblasts/metabolism , Gene Expression Regulation, Developmental , Incisor/metabolism , Maxilla/metabolism , Repressor Proteins/genetics , Stem Cells/metabolism , Tumor Suppressor Proteins/genetics , Age Factors , Ameloblasts/cytology , Amino Acid Substitution , Animals , Animals, Newborn , Cell Count , Cell Differentiation , Fibroblast Growth Factor 3/genetics , Fibroblast Growth Factor 3/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Heterozygote , Incisor/cytology , Incisor/growth & development , Male , Maxilla/cytology , Maxilla/growth & development , Mice , Mice, Knockout , Repressor Proteins/deficiency , Signal Transduction , Stem Cells/cytology , Transcription, Genetic , Tumor Suppressor Proteins/deficiency
19.
Cancer Sci ; 104(8): 1009-16, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23663453

ABSTRACT

Bcl11b is a haploinsufficient tumor suppressor, mutations or deletion of which has been found in 10-16% of T-cell acute lymphoblastic leukemias. Bcl11b(KO) (/+) heterozygous mice are susceptible to thymic lymphomas, a model of T-cell acute lymphoblastic leukemia, when γ-irradiated, and irradiated Bcl11b(KO) (/+) mice generate clonally expanding or premalignant thymocytes before thymic lymphoma development. Cells with radiation-induced DNA damages are assumed to be the cells of origin in tumors; however, which thymocyte is the tumor cell origin remains obscure. In this study we generated Bcl11b(flox/+) ;Lck-Cre and Bcl11b(flox/+) ;CD4-Cre mice; in the former, loss of one Bcl11b allele occurs in thymocytes at the immature CD4(-) CD8(-) stage, whereas in the latter the loss occurs in the more differentiated CD4(+) CD8(+) double-positive stage. We examined clonal expansion and differentiation of thymocytes in mice 60 days after 3 Gy γ-irradiation. Half (9/18) of the thymuses in the Bcl11b(flox/+) ;Lck-Cre group showed limited rearrangement sites at the T-cell receptor-ß (TCRß) locus, indicating clonal cell expansion, but none in the Bcl11b(flox/+) ;CD4-Cre group did. This indicates that the origin of the premalignant thymocytes is not in double-positive cells but immature thymocytes. Interestingly, those premalignant thymocytes underwent rearrangement at various different sites of the TCRα locus and the majority showed a higher expression of TCRß and CD8, and more differentiated phenotypes. This suggests the existence of a subpopulation of immature cells within the premalignant cells that is capable of proliferating and continuously producing differentiated thymocytes.


Subject(s)
Alleles , Neoplasms, Radiation-Induced/genetics , Precancerous Conditions/genetics , Repressor Proteins/genetics , Thymocytes/pathology , Thymocytes/radiation effects , Thymus Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/radiation effects , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/radiation effects , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Differentiation/genetics , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Gamma Rays/adverse effects , Mice , Mice, Inbred C57BL , Neoplasms, Radiation-Induced/metabolism , Neoplasms, Radiation-Induced/pathology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Thymocytes/metabolism , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology
20.
EMBO J ; 32(8): 1183-94, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23481257

ABSTRACT

CD4(+) helper and CD8(+) cytotoxic T cells differentiate from common precursors in the thymus after T-cell receptor (TCR)-mediated selection. Commitment to the helper lineage depends on persistent TCR signals and expression of the ThPOK transcription factor, whereas a ThPOK cis-regulatory element, ThPOK silencer, represses Thpok gene expression during commitment to the cytotoxic lineage. Here, we show that silencer-mediated alterations of chromatin structures in cytotoxic-lineage thymocytes establish a repressive state that is epigenetically inherited in peripheral CD8(+) T cells even after removal of the silencer. When silencer activity is enhanced in helper-lineage cells, by increasing its copy number, a similar heritable Thpok silencing occurs. Epigenetic locking of the Thpok locus may therefore be an independent event from commitment to the cytotoxic lineage. These findings imply that long-lasting TCR signals are needed to establish stable Thpok expression activity to commit to helper T-cell fate and that full commitment to the helper lineage requires persistent reversal of silencer activity during a particular time window.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , Mice , Mice, Transgenic , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...