Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(10)2023 May 16.
Article in English | MEDLINE | ID: mdl-37240163

ABSTRACT

Annexin A7/ANXA7 is a calcium-dependent membrane fusion protein with tumor suppressor gene (TSG) properties, which is located on chromosome 10q21 and is thought to function in the regulation of calcium homeostasis and tumorigenesis. However, whether the molecular mechanisms for tumor suppression are also involved in the calcium- and phospholipid-binding properties of ANXA7 remain to be elucidated. We hypothesized that the 4 C-terminal endonexin-fold repeats in ANXA7 (GX(X)GT), which are contained within each of the 4 annexin repeats with 70 amino acids, are responsible for both calcium- and GTP-dependent membrane fusion and the tumor suppressor function. Here, we identified a dominant-negative triple mutant (DNTM/DN-ANXA7J) that dramatically suppressed the ability of ANXA7 to fuse with artificial membranes while also inhibiting tumor cell proliferation and sensitizing cells to cell death. We also found that the [DNTM]ANA7 mutation altered the membrane fusion rate and the ability to bind calcium and phospholipids. In addition, in prostate cancer cells, our data revealed that variations in phosphatidylserine exposure, membrane permeabilization, and cellular apoptosis were associated with differential IP3 receptor expression and PI3K/AKT/mTOR modulation. In conclusion, we discovered a triple mutant of ANXA7, associated with calcium and phospholipid binding, which leads to the loss of several essential functions of ANXA7 pertinent to tumor protection and highlights the importance of the calcium signaling and membrane fusion functions of ANXA7 for preventing tumorigenesis.


Subject(s)
Phosphatidylinositol 3-Kinases , Prostatic Neoplasms , Male , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Calcium/metabolism , Calcium Signaling , Prostatic Neoplasms/metabolism , Cell Proliferation/genetics , Cell Line, Tumor , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Carcinogenesis
2.
PLoS One ; 13(10): e0205837, 2018.
Article in English | MEDLINE | ID: mdl-30321230

ABSTRACT

BACKGROUND: Annexin A7 (ANXA7) is a member of the multifunctional calcium or phospholipid-binding annexin gene family. While low levels of ANXA7 are associated with aggressive types of cancer, the clinical impact of ANXA7 in prostate cancer remains unclear. Tissue microarrays (TMA) have revealed several new molecular markers in human tumors. Herein, we have identified the prognostic impact of ANXA7 in a prostate cancer using a tissue microarray containing 637 different specimens. METHODS: The patients were diagnosed with prostate cancer and long-term follow-up information on progression (median 5.3 years), tumor-specific and overall survival data (median 5.9 years) were available. Expression of Ki67, Bcl-2, p53, CD-10 (neutral endopeptidase), syndecan-1 (CD-138) and ANXA7 were analyzed by immunohistochemistry. RESULTS: A bimodal distribution of ANXA7 was observed. Tumors expressing either high or no ANXA7 were found to be associated with poor prognosis. However, ANXA7 at an optimal level, in between high and no ANXA7 expression, had a better prognosis. This correlated with low Ki67, Bcl-2, p53 and high syndecan-1 which are known predictors of early recurrence. At Gleason grade 3, ANXA7 is an independent predictor of poor overall survival with a p-value of 0.003. Neoadjuvant hormonal therapy, which is known to be associated with overexpression of Bcl-2 and inhibition of Ki67 LI and CD-10, was found to be associated with under-expression of ANXA7. CONCLUSIONS: The results of this TMA study identified ANXA7 as a new prognostic factor and indicates a bimodal correlation to tumor progression.


Subject(s)
Annexin A7/blood , Prostatic Neoplasms/blood , Tissue Array Analysis/methods , Aged , Aged, 80 and over , Disease Progression , Humans , Kaplan-Meier Estimate , Ki-67 Antigen/metabolism , Male , Middle Aged , Neoplasm Grading , Neprilysin/metabolism , Prognosis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Syndecan-1/metabolism , Time Factors , Treatment Outcome
3.
Anticancer Res ; 38(7): 3831-3842, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29970503

ABSTRACT

BACKGROUND/AIM: Our studies showed that ANXA7 is a novel tumor suppressor gene that is lost in various aggressive forms of prostate cancer. However, little is known about the role of ANXA7 in the anticancer drug treatment towards different cancers. MATERIALS AND METHODS: The expression of ANXA7 was measured in the 60 cancer cell lines of the NCI-60 ADS project and correlated with the enhanced sensitivity to over 30,000 natural and synthetic compounds. RESULTS: Eucalyptol showed a high positive correlation with ANXA7 expression and castration-resistant prostate cancer cell death occurred very effectively in response to the combination of eucalyptol and overexpressed wt-ANXA7 than either agent alone. The synergistic effects of ANXA7 and eucalyptol resulted in concordant changes in gene expression profiles particularly of Ras family members, MDM4, NF-ĸB and VEGF. CONCLUSION: Overexpression of ANXA7 enhances eucalyptol cytotoxicity in prostate cancer cell lines.


Subject(s)
Annexin A7/genetics , Cyclohexanols/pharmacology , Drug Resistance, Neoplasm/genetics , Monoterpenes/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Cell Line, Tumor , Cell Proliferation , Cyclohexanols/therapeutic use , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Eucalyptol , Gene Expression Profiling , Humans , Male , Monoterpenes/therapeutic use , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology
4.
Methods Mol Biol ; 1513: 23-35, 2017.
Article in English | MEDLINE | ID: mdl-27807828

ABSTRACT

Chromosomal abnormalities, including homozygous deletions and loss of heterozygosity at 10q, are commonly observed in most human tumors, including prostate, breast, and kidney cancers. The ANXA7-GTPase is a tumor suppressor, which is frequently inactivated by genomic alterations at 10q21. In the last few years, considerable amounts of data have accumulated describing inactivation of ANXA7-GTPase in a variety of human malignancies and demonstrating the tumor suppressor potential of ANXA7-GTPase. ANXA7-GTPase contains a calcium binding domain that classifies it as a member of the annexin family. The cancer-specific expression of ANXA7-GTPase, coupled with its importance in regulating cell death, cell motility, and invasion, makes it a useful diagnostic marker of cancer and a potential target for cancer treatment. Recently, emerging evidence suggests that ANXA7-GTPase is a critical factor associated with the metastatic state of several cancers and can be used as a risk biomarker for HER2 negative breast cancer patients. Cross talk between ANXA7, PTEN, and EGFR leads to constitutive activation of PI3K-AKT signaling, a central pathway of tumor cell survival and proliferation. This review focuses on the recent progress in understanding the tumor suppressor functions of ANXA7-GTPase emphasizing the role of this gene in Ca2+ metabolism, and exploring opportunities for function as an example of a calcium binding GTPase acting as a tumor suppressor and opportunities for ANXA7-GTPase gene cancer therapy.


Subject(s)
Annexin A7/genetics , Breast Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Kidney Neoplasms/therapy , Prostatic Neoplasms/therapy , Annexin A7/agonists , Annexin A7/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium/metabolism , Chromosomes, Human, Pair 10 , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Genetic Vectors/therapeutic use , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Loss of Heterozygosity , Male , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction
5.
J Proteomics Bioinform ; 8(2): 23-30, 2015 Feb.
Article in English | MEDLINE | ID: mdl-27087761

ABSTRACT

Tocopherol succinate (TS) has been shown to protect mice against acute radiation syndrome, however, its exact mechanism of action and its possible use in humans has not yet been evaluated. Our approach has been to test the radioprotectant properties of TS on CD34-positive stem cells from healthy volunteers. We hypothesize that a radioproteomics strategy can identify a drug-dependent, personalized proteomics signature for radioprotection. To directly test the radioproteomics hypothesis, we treated human CD34-positive stem cells with 20 µM TS for 24 h, and then exposed the cells to 2 Gy of cobalt-60 gamma-radiation. We isolated protein from all cultures and used a high throughput Antibody Microarray (AbMA) platform to measure concentrations of 725 low abundance proteins. As an in vivo control, we also tested mouse CD34-positive stem cells using the same preemptive TS paradigm on progenitor colony forming units. TS pretreatment of in vitro or in vivo CD34-positive stem cells rescued radiation-induced loss of colony-forming potential of progenitors. We identified 50 of 725 proteins that could be preemptively rescued from radiation-induced reduction by pretreatment with TS. Ingenuity Pathway Analysis (IPA) reveals that the modified proteins fall into categories dominated by epigenetic regulation, DNA repair, and inflammation. Our results suggest that radioproteomics can be used to develop personalized medicine for radioprotection using protein signatures from primary CD34-positive progenitors derived from the patient or victim prior to radiation exposure. The protective effect of TS may be due to its ability to preemptively activate epigenetic mechanisms relevant to radioprotection and to preemptively activate the programs for DNA repair and inflammation leading to cell survival.

6.
Biomed Res Int ; 2014: 193635, 2014.
Article in English | MEDLINE | ID: mdl-24864229

ABSTRACT

Tumor suppressor function of the calcium/phospholipid-binding Annexin-A7 (ANXA7) has been shown in Anxa7-deficient mice and validated in human cancers. In the androgen-resistant prostate cancer cells, ANXA7 and p53 showed similar cytotoxicity levels. However, in the androgen-sensitive LNCaP, ANXA7 greatly exceeded the p53-induced cytotoxicity. We hypothesized that the p53 underperformance in LNCaP could be due to the involvement of p53-responsive SGK1 and FOXO3A. In this study, we show that p53 failed to match programmed cell death (PCD) and G1-arrest that were induced by ANXA7 in LNCaP. WT-ANXA7 preserved total FOXO3A expression with no hyperphosphorylation that could enable FOXO3A nuclear translocation and proapoptotic transcription. In contrast, in the p53-transfected LNCaP cells with maintained cell proliferation, the phosphorylated (but not total) FOXO3A fraction was increased implying a predominantly cytoplasmic localization and, subsequently, a lack of FOXO3A proapoptotic transcription. In addition, p53 reduced the expression of aberrant SGK1 protein form in LNCaP. Using Ingenuity Pathway Analysis and p53-signature genes, we elucidated the role of distinct SGK1/FOXO3A-associated regulation in p53 versus ANXA7 responses and proposed that aberrant SGK1 could affect reciprocal SGK1-FOXO3A-Akt regulation. Thus, the failure of the cell growth regulator p53 versus the phospholipid-binding ANXA7 could be potentially attributed to its diverse effects on SGK1-FOXO3A-Akt pathway in the PTEN-deficient LNCaP.


Subject(s)
Annexin A7/metabolism , Forkhead Transcription Factors/metabolism , Immediate-Early Proteins/genetics , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Androgens/pharmacology , Animals , Base Sequence , Cell Count , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Forkhead Box Protein O3 , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immediate-Early Proteins/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Molecular Sequence Data , Molecular Weight , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/drug effects , Transfection
7.
Cell Physiol Biochem ; 29(5-6): 697-704, 2012.
Article in English | MEDLINE | ID: mdl-22613970

ABSTRACT

Insulin secretion from the pancreatic ß-cell is controlled by changes in membrane potential and intracellular Ca(2+). The contribution of intracellular Ca(2+) stores to this process is poorly understood. We have previously shown that ß-cells of mice lacking one copy of the Annexin 7 gene (Anx7(+/-)) express reduced levels of IP(3) receptors and defects in IP(3)-dependent Ca(2+) signaling. To further elucidate the effect of the Anx7(+/-) mutation on signaling related to intracellular Ca(2+) stores in the ß-cell, we measured the effects of Ca(2+) mobilizing agents on electrical activity, intracellular Ca(2+) and insulin secretion in control and mutant ß-cells. We found that the muscarinic agonist carbachol and the ryanodine receptor agonists caffeine and 4-chloro-m-cresol had more potent depolarizing effects on Anx7(+/-) ß-cells compared to controls. Accordingly, glucose-induced insulin secretion was augmented to a greater extent by caffeine in mutant islets. Surprisingly, ryanodine receptor-mediated Ca(2+) mobilization was not affected by the Anx7(+/-) mutation, suggesting that the mechanism underlying the observed differences in electrical and secretory responsiveness does not involve intracellular Ca(2+) stores. Our results provide evidence that both IP3 receptors and ryanodine receptors play important roles in regulating ß-cell membrane potential and insulin secretion, and that the Anx7(+/-) mutation is associated with alterations in the signaling pathways related to these receptors.


Subject(s)
Annexin A7/physiology , Calcium/metabolism , Islets of Langerhans/physiopathology , Mutation , Animals , Annexin A7/genetics , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Membrane Potentials , Mice , Mice, Knockout , Ryanodine Receptor Calcium Release Channel/drug effects
8.
Cancer Biomark ; 7(1): 17-23, 2010.
Article in English | MEDLINE | ID: mdl-21045261

ABSTRACT

Evaluation of gene expression profiles in CWR22 prostate tumor xenografts in nude mice revealed overexpression of the Cytochrome b561, a transmembrane electron transport protein abundant in neuroendocrine vesicles, in the castration recurrent prostate cancers (CRPC). Four fold higher levels of the Cytochrome b561 was present in highly metastatic and androgen refractory LNCaP/C4-2 prostate cancer cells in comparison to androgen responsive and non-metastatic LNCaP cells. In LNCaP cells, Cytochrome b561 expression was induced by the synthetic androgen, R1881. Of note, Cytochrome b561 expression pattern correlated with known androgen regulated genes in epithelial transcriptome of primary prostate tumors. Taken together, these novel findings suggest that the expression of Cytochrome b561, is androgen regulated in the context of CaP cells and its increased expression in CRPC reflects increased androgen receptor signaling in tumor cells. These observations warrant further evaluation of functions and biomarker potential of Cytochrome b561 in CRPC.


Subject(s)
Cytochrome b Group/biosynthesis , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Humans , Male , Metribolone/pharmacology , Mice , Mice, Nude , Neoplasm Recurrence, Local , Neoplasm Transplantation , Orchiectomy , Prostatic Neoplasms/pathology , Testosterone Congeners/pharmacology
9.
Int J Cancer ; 125(11): 2528-39, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19610065

ABSTRACT

The tumor suppressor role of annexin-A7 (ANXA7) was previously demonstrated by cancer susceptibility in Anxa7(+/-)-mice and by ANXA7 loss in human cancers, especially in hormone-resistant prostate tumors. To gain mechanistic insights into ANXA7 tumor suppression, we undertook an in vitro study in which we compared wild-type (WT)-ANXA7 and dominant-negative (DN)-ANXA7 effects to a conventional tumor suppressor p53 in prostate cancer cells with different androgen sensitivity. Unlike p53 (which caused cell growth arrest and apoptosis to a noticeable extent in benign PrEC), WT-ANXA7 demonstrated profound cytotoxicityin androgen-sensitive LNCaP as well as in the androgen-resistant DU145 and PC3 prostate cancer cells, but not in PrEC. In androgen-sensitive LNCaP, WT-ANXA7 decreased low-molecular-weight (LMW) AR protein forms and maintained higher retinoblastoma 1 (RB1)/phospho-RB1 ratio. In contrast, DN-ANXA7 (which lacks phosphatidylserine liposome aggregation properties) increased LMW-AR forms and hyperphosphorylated RB1 that was consistent with the lack of DN-ANXA7 cytotoxicity. According to the microarray-based Ingenuity Pathways Analysis, a major WT-ANXA7 effect in androgen-sensitive LNCaP constituted of upregulation of the RB1-binding transcription factor E2F1 along with its downstream proapoptotic targets such as ASK1 and ASPP2. These results suggested a reversal of the RBdependent repression of the proapoptotic E2F-mediated transcription. However, DN-ANXA7 increased RB1/2 (but not E2F1) expression and induced the proliferation-promoting ERK5, thereby maintaining the RB-dependent repression of E2F-mediated apoptosis in LNcaP. On the other hand, in androgen-resistant cells, WT-ANXA7 tumor suppressor effects involved PTEN and NFkB pathways. Thus, ANXA7 revived the RB-associated cell survival control and overcame androgen resistance and dysfunctional status of major tumor suppressors commonly mutated in prostate cancer. Published 2009 UICC.


Subject(s)
Annexin A7/pharmacology , Drug Resistance, Neoplasm , Neoplasms, Hormone-Dependent/pathology , Prostate/drug effects , Prostatic Neoplasms/pathology , Retinoblastoma Protein/metabolism , Tumor Suppressor Proteins/pharmacology , Adenoviridae/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , DNA, Neoplasm/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Profiling , Genetic Vectors , Humans , Male , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Transfection , Tumor Cells, Cultured
10.
Cancer Lett ; 210(2): 239-44, 2004 Jul 16.
Article in English | MEDLINE | ID: mdl-15183540

ABSTRACT

Loss of heterozygosity (LOH) in the 10q21 region that harbors the tumor suppressor gene ANX7-GTPase gene have been found in 35% of prostate tumors. Therefore, the rationale for this study is that this gene could also be implicated in breast pathogenesis as well. We investigated allelic losses in microsatellites of the 10q21 region, and their correlations with ANX7 status, estrogen receptor (ER) status, progesterone receptor (PR) status, Ki-67 status and pathological phenotype in 30 breast carcinomas with matched control specimens. The LOH analysis was performed by amplifying DNA by PCR, using four markers of the 10q21 region (AFMa299ya5, AFM220xe5, AFM 063xc5, AFM200wf4). LOH in at least one marker of the 10q21 region (AFM220xe5 marker close to ANX7) was found in 66% of the first set of informative tumors containing 10 pairs of specimens. Subsequent comparison between 20 carcinomas using AFM220xe5, with and without LOH in terms of pathological parameters showed significant associations with differences in age (P = 0.04) ER (P = 0.05) Ki-67 (P = 0.04) and PR (P = 0.01) a trend toward significance was found for tumor size (P = 0.06) and histological grade III (P = 0.06). These results suggest that the ANX7 gene, or other genes of the 10q21 region, could be functionally related to breast cancer, probably influencing the hormone receptor expression associated with poor prognosis during development.


Subject(s)
Annexin A7/genetics , Breast Neoplasms/genetics , Carcinoma/genetics , Chromosomes, Human, Pair 10/genetics , GTP Phosphohydrolases/genetics , Loss of Heterozygosity , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Breast Neoplasms/pathology , Carcinoma/pathology , Case-Control Studies , Female , Gene Amplification , Humans , Ki-67 Antigen/analysis , Microsatellite Repeats , Phenotype , Polymerase Chain Reaction
11.
Proc Natl Acad Sci U S A ; 100(24): 14287-92, 2003 Nov 25.
Article in English | MEDLINE | ID: mdl-14608035

ABSTRACT

Annexin 7 (ANX7) acts as a tumor suppressor gene in prostate cancer, where loss of heterozygosity and reduction of ANX7 protein expression is associated with aggressive metastatic tumors. To investigate the mechanism by which this gene controls tumor development, we have developed an Anx7(+/-) knockout mouse. As hypothesized, the Anx7(+/-) mouse has a cancer-prone phenotype. The emerging tumors express low levels of Anx7 protein. Nonetheless, the wild-type Anx7 allele is detectable in laser-capture microdissection-derived tumor tissue cells. Genome array analysis of hepatocellular carcinoma tissue indicates that the Anx7(+/-) genotype is accompanied by profound reductions of expression of several other tumor suppressor genes, DNA repair genes, and apoptosis-related genes. In situ analysis by tissue imprinting from chromosomes in the primary tumor and spectral karyotyping analysis of derived cell lines identify chromosomal instability and clonal chromosomal aberrations. Furthermore, whereas 23% of the mutant mice develop spontaneous neoplasms, all mice exhibit growth anomalies, including gender-specific gigantism and organomegaly. We conclude that haploinsufficiency of Anx7 expression appears to drive disease progression to cancer because of genomic instability through a discrete signaling pathway involving other tumor suppressor genes, DNA-repair genes, and apoptosis-related genes.


Subject(s)
Annexin A7/genetics , Genes, Tumor Suppressor , Animals , Cell Division , Chromosomal Instability , Dictyostelium/cytology , Dictyostelium/genetics , Female , Gene Dosage , Gene Expression , Genes, Protozoan , Genotype , Gigantism/genetics , Heterozygote , Humans , Karyotyping , Liver Neoplasms, Experimental/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Oligonucleotide Array Sequence Analysis , Phenotype , Prostatic Neoplasms/genetics , Signal Transduction
12.
Ann N Y Acad Sci ; 971: 53-60, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12438089

ABSTRACT

The Anx7 gene codes for a Ca(2+)/GTPase with calcium channel and membrane fusion properties that has been proposed to regulate exocytotic secretion in chromaffin and other cell types. We have previously reported that the homozygous Anx7 (+/-) knockout mouse has an embryonically lethal phenotype. However, the viable heterozygous Anx7 (+/-) mouse displays a complex phenotype that includes adrenal gland hypertrophy, chromaffin cell hyperplasia, and defective IP(3) receptor (IP(3)R) expression. To search for a molecular basis for this phenotype, we have used cDNA microarray technology and have challenged control and mutant mice with fed or fasting conditions. We report that in the absence of the Anx7/IP(3)R signaling system, the cells in the adrenal gland are unable to discriminate between the fed and fasted states, in vivo. In control chromaffin cells, fasting is accompanied by an increased expression of structural genes for chromaffin cell contents, including chromogranin A and B, and DbetaH. There are also genes whose expression is specifically reduced. However, the Anx7 (+/-) mutation results in sustained expression of these nutritionally sensitive genes. We hypothesize that the calcium signaling defect due to the missing IP(3)R may be responsible for the global effects of the mutation on nutritionally sensitive genes. We further hypothesize that the tonically elevated expression of chromogranin A, a reportedly master control "switch" for dense core granule formation, may contribute to the process driving glandular hypertrophy and chromaffin cell hyperplasia in the Anx7 (+/-) mutant mouse.


Subject(s)
Adrenal Glands/metabolism , Annexin A7/genetics , Annexin A7/physiology , Food Deprivation , Adrenal Medulla/metabolism , Animal Nutritional Physiological Phenomena , Animals , Annexin A7/metabolism , Calcium Channels/metabolism , Chromaffin Cells/metabolism , Chromogranin A , Chromogranins/metabolism , DNA, Complementary/metabolism , Databases as Topic , Hypertrophy , Inositol 1,4,5-Trisphosphate Receptors , Mice , Mice, Knockout , Mutation , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction
13.
Mol Med ; 8(12): 781-97, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12606813

ABSTRACT

BACKGROUND: Gene expression in islets of Langerhans is profoundly sensitive to glucose and other nutrients. Islets of Langerhans in the Anx7(+/-) knockout mouse exhibit a profound reduction in ITPR3 protein expression, defective intracellular calcium signaling, and defective insulin secretion. Additional data presented here also show that mRNA for ITPR3 is virtually undetectable in isolated Anx7(+/-) islets. IP3Receptor type 3 (ITPR3) expression in islets of Langerhans is closely regulated by secretory stimuli, and it has been suggested that the level of the ITPR3 expression controls the ability of the islets to respond to nutritional signals. We report that although control islets respond to glucose in vitro by a transient increment in ITPR3 mRNA, the islets from the Anx7(+/-) mouse remain low. We therefore hypothesized that the Anx7/IP3 Receptor(3)/Ca(2+) signaling pathway plays a role in beta cell responses to glucose, and that in the absence of the Anx7/ITPR3 signaling system, the islets would be unable to discriminate between fed or fasted states in vivo. MATERIALS AND METHODS: To test this hypothesis, we subjected Anx7(+/-) and control mice to either food and water ad libidum or to an overnight fast with access to water only. We then isolated the respective islets and compared nutrient-dependent changes in global gene expression under the four conditions using genome-based microarray technology. RESULTS: Anx7 protein expression in these islets is only about 50% of control levels in normal littermate controls, and IPTR3 message and protein are virtually zero. cDNA microarray analyses show that in control animals gene expression is significantly affected by the fasting state. Many of the affected genes have historical relevance to development and differentiation of islets. These include preproglucagon, APOJ, cadherin2, phosphoglucoisomerase, oncostatin M, PAX6, HGF, and cytokeratin 18. However, there are also many other nutritionally sensitive genes in control islets that are principally associated with cell division and DNA repair. The latter genes have not specifically been associated with islet physiology in the past. By contrast, Anx7(+/-) mouse islets exhibit a greatly reduced ability to discriminate genomically between fed and fasted states for all classes of identified genes. Many of the validated genes are specific to islets in comparison to liver tissue examined. Real-time quantitative RT-PCR analysis of islets from Anx7 heterozygous mice and littermate controls revealed remarkable down-regulation in PTEN, Glut-2, PDX-1, IGF-1, and Neuro D1 expression, but not in liver. CONCLUSIONS: We conclude that reduced gene dosage in the Anx7(+/-) islet, with concomitant loss of ITPR3 expression and consequent defects in Ca(2+) signaling, may substantially contribute to the mechanism of the loss of genomic discrimination, in vivo, between the fed and fasted states. We believe that the requirement for complete Anx7 gene dosage and IPTR3 expression in islets of Langerhans will prove to be of fundamental importance for understanding the mechanism of nutritional sensing in health and disease.


Subject(s)
Annexin A7/metabolism , Gene Expression Regulation/physiology , Islets of Langerhans/metabolism , Signal Transduction/physiology , Algorithms , Animals , Annexin A7/genetics , Blood Glucose/metabolism , Calcium Channels/biosynthesis , Calcium Channels/genetics , Calcium Channels/metabolism , Inositol 1,4,5-Trisphosphate Receptors , Insulin/blood , Insulin/metabolism , Mice , Mice, Knockout , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...