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










Publication year range
1.
Phys Chem Chem Phys ; 24(17): 9953-9963, 2022 May 04.
Article in English | MEDLINE | ID: mdl-35445226

ABSTRACT

The radiative and multiphonon non-radiative relaxation rates of lanthanide ions are intrinsic parameters to characterize the optical properties, which are the basic data for the theoretical model and numerical simulation of lanthanide upconversion systems. However, there are complex energy transfer processes, such as energy migration, energy transfer upconversion, and cross-relaxation in the lanthanide-doped upconversion materials, so it is difficult to accurately measure the intrinsic radiative and multiphonon relaxation rates. Therefore, a method to determine the relaxation rates of multi-level upconversion systems is proposed based on multi-wavelength excitation and level-by-level parameter calculations in this paper. For a dilute doped multi-level luminescence system excited at low powers, a model based on the measurements of steady-state emission spectra and luminescence decay curves is established through the macroscopic rate equations at multi-wavelength excitation, which can be used for the level-by-level calculation of the multi-level radiative and multiphonon relaxation rates. With the dilute doped ß-NaYF4:Er3+ six-level luminescence system as an example, the measurement method and the model are introduced in detail. Under the experimental conditions of neglecting the energy transfer effect between ions, the materials are excited by five lasers with central wavelengths of 1523 nm, 980 nm, 808 nm, 660 nm, and 520 nm to form five subsystems. The steady-state emission spectra and luminescence decay curves of the luminescence system excited by each wavelength were recorded. The intrinsic relaxation rates including 11 radiative relaxation rates and 4 multiphonon relaxation rates in the ß-NaYF4:Er3+ six-level system were determined based on the established model and method, which experimentally verified the applicability of the method proposed in this paper. This work will provide basic data for the analysis and regulation of the luminescence properties of lanthanide upconversion systems.

2.
Nat Commun ; 13(1): 1006, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35194044

ABSTRACT

During decidualization in rodents, uterine stroma undergoes extensive reprograming into distinct cells, forming the discrete regions defined as the primary decidual zone (PDZ), the secondary decidual zone (SDZ) and the layer of undifferentiated stromal cells respectively. Here we show that uterine deletion of Men1, a member of the histone H3K4 methyltransferase complex, disrupts the terminal differentiation of stroma, resulting in chaotic decidualization and pregnancy failure. Genome-wide epigenetic profile reveals that Men1 binding in chromatin recapitulates H3K4me3 distribution. Further transcriptomic investigation demonstrates that Men1 directly regulates the expression of PTX3, an extra-cellular trap for FGF2 in decidual cells. Decreased Ptx3 upon Men1 ablation leads to aberrant activation of ERK1/2 in the SDZ due to the unrestrained FGF2 signal emanated from undifferentiated stromal cells, which blunt BMP2 induction and decidualization. In brief, our study provides genetic and molecular mechanisms for epigenetic rewiring mediated decidual regionalization by Men1 and sheds new light on pregnancy maintenance.


Subject(s)
C-Reactive Protein/metabolism , Fibroblast Growth Factor 2 , Serum Amyloid P-Component/metabolism , Transcription Factors , Decidua/metabolism , Embryo Implantation , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Pregnancy , Signal Transduction , Stromal Cells , Transcription Factors/metabolism , Uterus/physiology
3.
Endocrinology ; 156(1): 48-57, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25343275

ABSTRACT

The tumor suppressor menin is recognized as a key regulator of ß-cell proliferation. To induce tumorigenesis within the pancreatic ß-cells, floxed alleles of Men1 were selectively ablated using Cre-recombinase driven by the insulin promoter. Despite the ß-cell specificity of the RipCre, glucagon-expressing tumors as well as insulinomas developed in old mutant mice. These glucagon-expressing tumor cells were menin deficient and expressed the mature α-cell-specific transcription factors Brain-specific homeobox POU domain protein 4 (Brn4) and v-maf musculoaponeurotic fibrosarcoma oncogene family, protein B (MafB). Moreover, the inactivation of ß-cell-specific transcription factors was observed in mutant ß-cells. Our work shows that Men1 ablation in the pancreatic ß-cells leads to the inactivation of specific transcription factors, resulting in glucagon-expressing tumor development, which sheds light on the mechanisms of islet tumorigenesis.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Glucagon/metabolism , Glucagonoma/metabolism , Insulin-Secreting Cells/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/genetics , Animals , Female , Gene Deletion , Genotype , Glucagon-Secreting Cells/physiology , Glucagonoma/genetics , Male , Mice , Mice, Knockout , Pancreatic Neoplasms/genetics , Transcription Factors
4.
World J Diabetes ; 5(6): 847-53, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25512786

ABSTRACT

Pancreatic insulin-secreting ß-cells are essential regulators of glucose metabolism. New strategies are currently being investigated to create insulin-producing ß cells to replace deficient ß cells, including the differentiation of either stem or progenitor cells, and the newly uncovered transdifferentiation of mature non-ß islet cell types. However, in order to correctly drive any cell to adopt a new ß-cell fate, a better understanding of the in vivo mechanisms involved in the plasticity and biology of islet cells is urgently required. Here, we review the recent studies reporting the phenomenon of transdifferentiation of α cells into ß cells by focusing on the major candidates and contexts revealed to be involved in adult ß-cell regeneration through this process. The possible underlying mechanisms of transdifferentiation and the interactions between several key factors involved in the process are also addressed. We propose that it is of importance to further study the molecular and cellular mechanisms underlying α- to ß-cell transdifferentiation, in order to make ß-cell regeneration from α cells a relevant and realizable strategy for developing cell-replacement therapy.

5.
Nat Commun ; 5: 5809, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25517963

ABSTRACT

Inactivating MEN1 mutations are the most common genetic defects present in sporadic and inherited pancreatic neuroendocrine tumours (PNETs). The lack of interventional therapies prompts us to explore the therapeutic approach of targeting ß-catenin signalling in MEN1-mutant PNETs. Here we show the MEN1-encoded scaffold protein menin regulates phosphorylation of ß-catenin. ß-catenin signalling is activated in MEN1-mutant human and mouse PNETs. Conditional knockout of ß-catenin suppresses the tumorigenesis and growth of Men1-deficient PNETs, and significantly prolongs the survival time in mice. Suppression of ß-catenin signalling by genetic ablation or a molecular antagonist inhibits the expression of proproliferative genes in menin-null PNETs and potently improves hyperinsulinemia and hypoglycemia in mice. Blockade of ß-catenin has no adverse effect on physiological function of pancreatic ß-cells. Our data demonstrate that ß-catenin signalling is an effective therapeutic target for MEN1-mutant PNETs. Our findings may contribute to individualized and combined medication treatment for PNETs.


Subject(s)
Carcinogenesis/metabolism , Gene Expression Regulation, Neoplastic , Multiple Endocrine Neoplasia Type 1/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/genetics , Signal Transduction , beta Catenin/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Gene Deletion , Humans , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Hypoglycemia/genetics , Hypoglycemia/metabolism , Hypoglycemia/pathology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/mortality , Multiple Endocrine Neoplasia Type 1/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Phosphorylation , Precision Medicine , Proto-Oncogene Proteins/deficiency , Survival Analysis , beta Catenin/deficiency
6.
Ann Endocrinol (Paris) ; 75(3): 133-40, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24997771

ABSTRACT

CONTEXT: Multiple Endocrine Neoplasia Type 1 (MEN1) is an autosomal dominant inherited syndrome, related to mutations in the MEN1 gene. Controversial data suggest that the nonsynonymous p.Ala541Thr variant, usually considered as a non-pathogenic polymorphism, may be associated with an increased risk of MEN1-related lesions in carriers. OBJECTIVE: The aim of this study was to evaluate the pathogenic influence of the p.Ala541Thr variant on clinical and functional outcomes. PATIENTS AND METHODS: We analysed a series of 55 index patients carrying the p.Ala541Thr variant. Their clinical profile was compared to that of 117 MEN1 patients. The biological impact of the p.Ala541Thr variant on cell growth was additionally investigated on menin-deficient Leydig cell tumour (LCT)10 cells generated from Men1+/Men1- heterozygous knock-out mice, and compared with wild type (WT). RESULTS: The mean age at first appearance of endocrine lesions was similar in both p.Ala541Thr carriers and MEN1 patients, but no p.Ala541Thr patient had more than one cardinal MEN1 lesion at initial diagnosis. A second MEN1 lesion was diagnosed in 13% of MEN1 patients and in 7% of p.Ala541Thr carriers in the year following preliminary diagnosis. Functional studies on LCT10 cells showed that overexpression of the p.Ala541Thr variant did not inhibit cell growth, which is in direct contrast to results obtained from investigation of WT menin protein. CONCLUSION: Taken together, these data raise the question of a potential pathogenicity of the p.Ala541Thr missense variant of menin that commonly occurs within the general population. Additional studies are required to investigate whether it may be involved in a low-penetrance MEN1 phenotype.


Subject(s)
Multiple Endocrine Neoplasia Type 1/genetics , Mutation , Polymorphism, Genetic/genetics , Proto-Oncogene Proteins/genetics , Adenoma/genetics , Adult , Animals , Cell Line, Tumor , Female , Heterozygote , Humans , Hyperparathyroidism/genetics , Leydig Cell Tumor/genetics , Male , Mice , Mice, Knockout , Middle Aged , Phenotype , Pituitary Neoplasms/genetics , Proto-Oncogene Proteins/deficiency , Transfection
7.
J Hepatol ; 59(6): 1299-306, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23867312

ABSTRACT

BACKGROUND & AIMS: The development and progression of non-alcoholic fatty liver disease are associated with aging, obesity, and type 2 diabetes. Understanding the precise regulatory networks of this process will contribute to novel therapeutic strategies. METHODS: Hepatocyte-specific Men1 knockout mice were generated using Cre/loxP technology. Lipid and glucose metabolic phenotypes and mechanisms were investigated in aging and high-fat diet fed mice. RESULTS: The expression of menin, encoded by multiple endocrine neoplasia 1 (Men1) gene, is reduced in the liver of aging mice. Hepatocyte-specific deletion of Men1 induces liver steatosis in aging mice. Menin deficiency promotes high-fat diet-induced liver steatosis in mice. Menin recruits SIRT1 to control hepatic CD36 expression and triglyceride accumulation through histone deacetylation. CONCLUSIONS: Our work reveals that the adaptor protein menin is critical for the progression of hepatic steatosis during aging and metabolic imbalance.


Subject(s)
Aging/metabolism , Fatty Liver/etiology , Histones/metabolism , Liver/metabolism , Proto-Oncogene Proteins/physiology , Sirtuin 1/physiology , Acetylation , Animals , CD36 Antigens/physiology , Diet, High-Fat , Fatty Liver/metabolism , Glucose/metabolism , Mice , Mice, Inbred C57BL
8.
Mol Cell Biol ; 29(20): 5477-87, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19651895

ABSTRACT

Menin, which is encoded by the multiple endocrine neoplasia type 1 (MEN1) gene, is a tumor suppressor and transcriptional regulator. Menin controls proliferation and apoptosis of cells, especially pancreatic beta cells. We have found that menin contains two functional nuclear export signals and that there is nuclear accumulation of beta-catenin in Men1-null mouse embryonic fibroblasts and insulinoma tissues from beta-cell-specific Men1 knockout mice. It is reported that the deregulation of Wnt/beta-catenin signaling caused by inactivation of tumor suppressors results in abnormal development or tumorigenesis. We further revealed that overexpression of menin reduces beta-catenin nuclear accumulation and its transcriptional activity. Menin is able to directly interact with beta-catenin and carry beta-catenin out of the nucleus via nuclear-cytoplasmic shuttling in a CRM1-dependent manner. These results imply that menin may control cell proliferation through suppression of Wnt/beta-catenin signaling.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Proto-Oncogene Proteins/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Active Transport, Cell Nucleus , Animals , HeLa Cells , Humans , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Signal Transduction/genetics , Signal Transduction/physiology
9.
J Pharm Biomed Anal ; 49(4): 1097-100, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19268518

ABSTRACT

In order to evaluate the differences and similarities between the liposoluble constituents in Cynomorium songaricum populations, stem liposoluble constituents in five populations of C. songaricum collected from three different geographic regions and four different hosts were obtained by solvent extraction and analyzed by GC-MS. Cluster analysis of the percentage composition of 80 compounds showed differences in chemical composition which were related to the geographic origin rather than the host. Hexadecanoic acid was the most abundant compound in the essential oils of C. songaricum from hosts Nitraria sibirica and Nitraria tanguticum. Whereas (Z)-9-octadecenoic acid was accumulated in the oils of C. songaricum from Zygophyllum xanthoxylum and Peganum harmala. Four of the five populations had characteristic components, which were specific to each population.


Subject(s)
Cynomorium/chemistry , Lipids/chemistry , Plant Stems/chemistry , Cluster Analysis , Gas Chromatography-Mass Spectrometry , Indicators and Reagents , Oils, Volatile/analysis , Solvents
10.
J Endocrinol ; 199(2): 287-98, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18772165

ABSTRACT

Mutations of the multiple endocrine neoplasia type 1 (MEN1) gene predispose patients to MEN1 that affects mainly endocrine tissues, suggesting important physiological functions of the gene in adult endocrine cells. Homozygous disruption of Men1 in mice causes embryonic lethality, whereas the eventual involvement of the gene in embryonic development of the endocrine cells remains unknown. Here, we show that homozygous Men1 knockout mice demonstrate a reduced number of glucagon-positive cells in the E12.5 pancreatic bud associated with apoptosis, whereas the exocrine pancreas development in these mice is not affected. Our data suggest that menin is involved in the survival of the early pancreatic endocrine cells during the first developmental transition. Furthermore, chimerism assay revealed that menin has an autonomous and specific effect on the development of islet cells. In addition, using pancreatic bud culture mimicking the differentiation of alpha- and beta-cells during the second transition, we show that loss of menin leads to the failure of endocrine cell development, altered pancreatic structure and a markedly decreased number of cells expressing neurogenin 3, indicating that menin is also required at this stage of the endocrine pancreas development. Taken together, our results suggest that menin plays an indispensable role in the development of the pancreatic endocrine cells.


Subject(s)
Endocrine Cells/cytology , Endocrine Cells/metabolism , Pancreas/metabolism , Proto-Oncogene Proteins/physiology , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Fluorescent Antibody Technique , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , In Vitro Techniques , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Pancreas/cytology , Pancreas/embryology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Endocr Relat Cancer ; 14(4): 1073-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18045958

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is an inherited tumour syndrome characterized by the development of tumours of the parathyroid, anterior pituitary and pancreatic islets, etc. Heterozygous germ line mutations of MEN1 gene are responsible for the onset of MEN1. We investigated the probands and 31 family members from eight unrelated Chinese families associated with MEN1 and identified four novel mutations, namely 373_374ins18, 822delT, 259delT and 1092delC, as well as three previously reported mutations, such as 357_360delCTGT, 427_428delTA and R108X (CGA>TGA) of MEN1 gene. Furthermore, we detected a loss of heterozygosity (LOH) at chromosome 11q in the removed tumours, including gastrinoma, insulinoma and parathyroid adenoma from two probands of MEN1 families. RT-PCR and direct sequencing showed that mutant MEN1 transcripts remained in the MEN1-associated endocrine tumours, whereas normal menin proteins could not be detected in those tumours by either immunohistochemistry or immunoblotting. In conclusion, MEN1 heterozygous mutations are associated with LOH and menin absence, which are present in MEN1-associated endocrine tumours.


Subject(s)
Multiple Endocrine Neoplasia Type 1/genetics , Mutation , Proto-Oncogene Proteins/genetics , Adolescent , Adult , Aged , Amino Acid Sequence , China , DNA Mutational Analysis , DNA Transposable Elements , Family , Female , Humans , Loss of Heterozygosity , Male , Middle Aged , Peptide Fragments , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion
12.
J Biol Chem ; 279(23): 24477-84, 2004 Jun 04.
Article in English | MEDLINE | ID: mdl-15054094

ABSTRACT

Menin, the product of the tumor suppressor gene MEN1, is widely expressed in mammalian endocrine and non-endocrine tissues, including intestine. Its known abundant expression in several types of cells with high proliferative capacity led us to investigate the physiological function of the protein menin in intestinal epithelium, one of the most rapidly growing epithelia. Here we showed that the Men1 gene is mainly expressed in the crypt compartment of the proximal small intestine and that its expression was increased during fasting in vivo, both suggesting a role of menin in the control of cell growth. Indeed, specific reduction of menin expression by transfected antisense cDNA in the rat duodenal crypt-like cell line, IEC-17, increased cell proliferation. The latter is correlated to a loss of cell-cycle arrest in G(1) phase by resting cells and an overexpression of cyclin D1 and cyclin-dependent kinase (Cdk)-4. Furthermore, these cells lost the inhibition of proliferation induced by transforming growth factor-beta1, associated with a decrease of transforming growth factor-beta type II receptor expression. As a result of deregulated proliferation, antisense menin transfected IEC-17 cells became tumorigenic as shown in vitro as well as in vivo in immunosuppressed animals. These results indicate that menin contributes to proliferation control in intestinal epithelial cells. The present study reveals an unknown physiological function for menin in intestine that may be important in the regulation of epithelial homeostasis.


Subject(s)
Epithelial Cells/metabolism , Intestines/cytology , Proto-Oncogene Proteins/biosynthesis , Agar/metabolism , Animals , Blotting, Western , Cell Cycle , Cell Division , Cell Line , Cell Separation , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , Cytoskeletal Proteins/metabolism , DNA, Complementary/metabolism , Down-Regulation , Fasting , Flow Cytometry , G1 Phase , Heterozygote , Immunohistochemistry , Immunosuppression Therapy , In Situ Hybridization , Intestine, Small/metabolism , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Plasmids/metabolism , Protein Serine-Threonine Kinases , RNA, Messenger/metabolism , Rats , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Time Factors , Trans-Activators/metabolism , Transfection , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , beta Catenin
13.
Mol Endocrinol ; 17(9): 1880-92, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12819299

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is a hereditary syndrome characterized by the occurrence of multiple endocrine tumors of the parathyroid, pancreas, and anterior pituitary in patients. To study tumorigenesis related to the MEN1 syndrome, we have generated Men1 knockout mice using the gene targeting approach. Heterozygous Men1 mutant mice developed the same range of major endocrine tumors as is seen in MEN1 patients, affecting the parathyroid, pancreatic islets, pituitary and adrenal glands, as well as the thyroid, and exhibiting multistage tumor progression with metastatic potential. In particular, extrapancreatic gastrinoma, pancreatic glucagonoma, and mixed hormone-producing tumors in islets were observed. In addition, there was a high incidence of gonadal tumors of endocrine origin, i.e. Leydig cell tumors, and ovary sex-cord stromal cell tumors in heterozygous Men1 mutant mice. Hormonal disturbance, such as abnormal PTH and insulin levels, was also observed in these mice. These tumors were associated with loss of heterozygosity of the wild-type Men1 allele, suggesting that menin is involved in suppressing the development of these endocrine tumors. All of these features are reminiscent of MEN1 symptoms in humans and establish heterozygous Men1 mutant mice as a suitable model for this disease.


Subject(s)
Disease Models, Animal , Multiple Endocrine Neoplasia Type 1/genetics , Proto-Oncogene Proteins/genetics , Animals , Endocrine System/pathology , Genes, Dominant , Heterozygote , Mice , Multiple Endocrine Neoplasia Type 1/metabolism , Mutation , Proto-Oncogene Proteins/metabolism
14.
Mech Dev ; 120(5): 549-60, 2003 May.
Article in English | MEDLINE | ID: mdl-12782272

ABSTRACT

Patients suffering from multiple endocrine neoplasia type 1 (MEN1) are predisposed to multiple endocrine tumors. The MEN1 gene product, menin, is expressed in many embryonic, as well as adult tissues, and interacts with several proteins in vitro and in vivo. However, the biological function of menin remains largely unknown. Here we show that disruption of the Men1 gene in mice causes embryonic lethality at E11.5-E13.5. The Men1 null mutant embryos appeared smaller in size, frequently with body haemorrhages and oedemas, and a substantial proportion of them showed disclosure of the neural tube. Histological analysis revealed an abnormal development of the nervous system and heart hypotrophy in some Men1 null embryos. Furthermore, Men1 null livers generally displayed an altered organization of the epithelial and hematopoietic compartments associated with enhanced apoptosis. Chimerism analysis of embryos generated by injection of Men1 null ES cells, showed that cells lacking menin do not seem to have a general cell-autonomous defect. However, primary Men1 null embryonic fibroblasts entered senescence earlier than their wild-type counterparts. Despite normal proliferation ability, Men1 null ES cells exhibited a deficiency to form embryoid bodies, suggesting an impaired differentiation capacity in these cells. The present study demonstrates that menin plays an important role in the embryonic development of multiple organs in addition to its proposed role in tumor suppression.


Subject(s)
Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Alleles , Animals , Apoptosis , Blotting, Western , Cell Differentiation , Cell Division , Cellular Senescence , Fibroblasts/metabolism , Genetic Vectors , Genotype , Heart/embryology , Hypertrophy/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Liver/cytology , Liver/embryology , Liver/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Models, Genetic , Mutation , Myocardium/metabolism , Neural Crest/embryology , Proto-Oncogene Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...