Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Hum Mutat ; 41(12): 2143-2154, 2020 12.
Article in English | MEDLINE | ID: mdl-32935393

ABSTRACT

In cystic fibrosis (CF), the correction of splicing defects represents an interesting therapeutic approach to restore normal CFTR function. In this study, we focused on 10 common mutations/variants 711+3A>G/C, 711+5G>A, TG13T3, TG13T5, TG12T5, 1863C>T, 1898+3A>G, 2789+5G>A, and 3120G>A that induce skipping of the corresponding CFTR exons 5, 10, 13, 16, and 18. To rescue the splicing defects we tested, in a minigene assay, a panel of modified U1 small nuclear RNAs (snRNAs), named Exon Specific U1s (ExSpeU1s), that was engineered to bind to intronic sequences downstream of each defective exon. Using this approach, we show that all 10 splicing mutations analyzed are efficiently corrected by specific ExSpeU1s. Using complementary DNA-splicing competent minigenes, we also show that the ExspeU1-mediated splicing correction at the RNA level recovered the full-length CFTR protein for 1863C>T, 1898+3A>G, 2789+5G>A variants. In addition, detailed mutagenesis experiments performed on exon 13 led us to identify a novel intronic regulatory element involved in the ExSpeU1-mediated splicing rescue. These results provide a common strategy based on modified U1 snRNAs to correct exon skipping in a group of disease-causing CFTR mutations.


Subject(s)
Cystic Fibrosis/genetics , Exons/genetics , Mutation/genetics , RNA, Small Nuclear/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , HeLa Cells , Humans , Introns/genetics , Nucleic Acid Conformation , RNA Splicing/genetics , RNA, Small Nuclear/chemistry
2.
Hum Mutat ; 37(2): 209-15, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26553470

ABSTRACT

Cystic fibrosis (CF), the most common life-threatening genetic disease in Caucasians, is caused by ∼2,000 different mutations in the CF transmembrane conductance regulator (CFTR) gene. A significant fraction of these (∼13%) affect pre-mRNA splicing for which novel therapies have been somewhat neglected. We have previously described the effect of the CFTR splicing mutation c.2657+5G>A in IVS16, showing that it originates transcripts lacking exon 16 as well as wild-type transcripts. Here, we tested an RNA-based antisense oligonucleotide (AON) strategy to correct the aberrant splicing caused by this mutation. Two AONs (AON1/2) complementary to the pre-mRNA IVS16 mutant region were designed and their effect on splicing was assessed at the RNA and protein levels, on intracellular protein localization and function. To this end, we used the 2657+5G>A mutant CFTR minigene stably expressed in HEK293 Flp-In cells that express a single copy of the transgene. RNA data from AON1-treated mutant cells show that exon 16 inclusion was almost completely restored (to 95%), also resulting in increased levels of correctly localized CFTR protein at the plasma membrane (PM) and with increased function. A novel two-color CFTR splicing reporter minigene developed here allowed the quantitative monitoring of splicing by automated microscopy localization of CFTR at the PM. The AON strategy is thus a promising therapeutic approach for the specific correction of alternative splicing.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/diagnosis , Cystic Fibrosis/genetics , Cystic Fibrosis/therapy , Oligonucleotides, Antisense/genetics , RNA Precursors/genetics , Base Sequence , Cystic Fibrosis/pathology , Exons , Genetic Therapy , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Introns , Molecular Sequence Data , Mutation , Oligonucleotides, Antisense/metabolism , RNA Precursors/metabolism , RNA Splicing , Sequence Analysis, DNA
3.
Am J Physiol Endocrinol Metab ; 303(6): E708-19, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22811466

ABSTRACT

Abnormal microRNA (miRNA) expression profiles have recently been associated with sporadic pituitary adenomas, suggesting that miRNAs can contribute to tumor formation; miRNAs are small noncoding RNAs that inhibit posttranscriptional expression of target mRNAs by binding to target sequences usually located in the 3'-UTR. In this study, we investigated the role played by miR-107, a miRNA associated with different human cancers, in sporadic pituitary adenomas and its interaction with the pituitary tumor suppressor gene aryl hydrocarbon receptor-interacting protein (AIP). miR-107 expression was evaluated in pituitary adenoma and normal pituitary samples using microRNA screen TLDA (TaqMan Low-Density Array) and RT-qPCR assays. We show that miR-107 expression was significantly upregulated in GH-secreting and nonfunctioning pituitary adenomas. We found that human AIP-3'-UTR is a target of miR-107 since miR-107 inhibited in vitro AIP expression to 53.9 ± 2% of the miRNA control in a luciferase assay and reduced endogenous AIP mRNA expression to 53 ± 22% of the miRNA control in human cells. However, we did not observe a negative correlation between AIP and miR-107 expression in the human tumor samples. Furthermore, we show that miR-107 overexpression inhibited cell proliferation in human neuroblastoma and rat pituitary adenoma cells. In conclusion, miR-107 is overexpressed in pituitary adenomas and may act as a tumor suppressor. We have identified and confirmed AIP as a miR-107 target gene. Expression data in human samples suggest that the expression of AIP and miR-107 could be influenced by a combination of tumorigenic factors as well as compensatory mechanisms stimulated by the tumorigenic process.


Subject(s)
Adenoma/metabolism , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/biosynthesis , MicroRNAs/biosynthesis , Neoplasm Proteins/biosynthesis , Pituitary Neoplasms/metabolism , Up-Regulation , 3' Untranslated Regions , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Profiling , Human Growth Hormone/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/biosynthesis , Mutant Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neuroblastoma/metabolism , Oligonucleotide Array Sequence Analysis , Pituitary Gland/metabolism , Prolactin/metabolism , RNA, Messenger/metabolism , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism
4.
Stress ; 15(5): 554-61, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22217206

ABSTRACT

5' adenosine monophosphate-activated protein kinase (AMPK) plays a prominent role as a metabolic stress sensor. The role of hypothalamic AMPK in response to restraint and surgical stress has not been previously investigated. It has been recently suggested that the renin-angiotensin system, in addition to its role in stress regulation, may play a significant role in regulating metabolic pathways including the regulation of the AMPK system. This study was thus aimed to evaluate the effects of candesartan, an angiotensin II AT1 receptor blocker drug, on hypothalamic AMPK activity under basal conditions and after restraint in conscious rats or after surgical stress under general anesthesia. Male Wistar rats were treated with 5 mg/kg/day candesartan in the drinking water for 2 weeks. The hypothalamic AMPK activity was determined under basal and stress conditions, using a kinase activity assay. Chronic administration of candesartan significantly increased hypothalamic AMPK activity. Hypothalamic AMPK activity was also increased by restraint stress whereas no change was observed during surgical stress under anesthesia. The high levels of hypothalamic AMPK activation observed in candesartan-treated rats were not changed by restraint stress but were reduced to control levels by anesthesia and surgery. In conclusion, chronic candesartan treatment and restraint stress in conscious rats stimulate the hypothalamic AMPK activity, whereas surgical stress under anesthesia inhibits pathways regulating the AMPK activity even in candesartan-treated rats.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Hypothalamus/drug effects , Stress, Physiological , Tetrazoles/pharmacology , Animals , Biphenyl Compounds , Hypothalamus/metabolism , Male , Rats , Rats, Wistar , Renin-Angiotensin System/drug effects
5.
Hum Mutat ; 31(8): 950-60, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20506337

ABSTRACT

Familial isolated pituitary adenoma (FIPA) is an autosomal dominant condition with variable genetic background and incomplete penetrance. Germline mutations of the aryl hydrocarbon receptor interacting protein (AIP) gene have been reported in 15-40% of FIPA patients. Limited data are available on the functional consequences of the mutations or regarding the regulation of the AIP gene. We describe a large cohort of FIPA families and characterize missense and silent mutations using minigene constructs, luciferase and beta-galactosidase assays, as well as in silico predictions. Patients with AIP mutations had a lower mean age at diagnosis (23.6+/-11.2 years) than AIP mutation-negative patients (40.4+/-14.5 years). A promoter mutation showed reduced in vitro activity corresponding to lower mRNA expression in patient samples. Stimulation of the protein kinase A-pathway positively regulates the AIP promoter. Silent mutations led to abnormal splicing resulting in truncated protein or reduced AIP expression. A two-hybrid assay of protein-protein interaction of all missense variants showed variable disruption of AIP-phosphodiesterase-4A5 binding. In summary, exonic, promoter, splice-site, and large deletion mutations in AIP are implicated in 31% of families in our FIPA cohort. Functional characterization of AIP changes is important to identify the functional impact of gene sequence variants.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Mutation/genetics , Pituitary Neoplasms/genetics , Adult , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Cell Line , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Family , Female , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Molecular Sequence Data , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense/genetics , Pedigree , Pituitary Neoplasms/enzymology , Promoter Regions, Genetic/genetics , RNA Splice Sites/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction
6.
Clin Endocrinol (Oxf) ; 70(2): 259-64, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18710468

ABSTRACT

OBJECTIVE: Germline mutations in the MEN1 gene predispose to the multiple endocrine neoplasia (MEN1) syndrome; however, approximately 10-20% of patients with MEN1 do not have a detectable MEN1 mutation. A rat strain with multiple endocrine tumours, a phenotypic overlap of both MEN1 and MEN2, has been reported to have a homozygous germline p27 (CDKN1B) mutation. Recently, two MEN1 mutation-negative MEN1 syndrome patients have been identified to harbour a germline CDKN1B mutation. The recently identified gene AIP can also cause familial isolated pituitary adenoma, but no other specific tumour is associated with this syndrome. The objective of this study was to evaluate the possible contribution of CDKN1B and AIP germline mutations in a cohort of MEN1 mutation-negative MEN1 syndrome patients. PATIENTS: Eighteen sporadic and three familial cases of MEN1 mutation-negative MEN1 syndrome were studied (18 pituitary adenomas, 12 hyperparathyroidism, 10 neuroendocrine tumours including 2 ACTH-secreting lesions and one adrenal nodular hyperplasia). Clinical data and genomic DNA were analysed for mutations in the CDKN1B and AIP genes. RESULTS: There were no mutations in the coding region or exon/intron junction of the CDKN1B and AIP genes in any patient. Although we have a limited number of patients in our cohort, our data is consistent with others in the literature suggesting that CDKN1B and AIP mutations are extremely rare in MEN1 syndrome. CONCLUSION: Our results suggest that mutations in the CDKN1B and AIP genes are relatively uncommon in MEN1 mutation-negative MEN1 syndrome patients.


Subject(s)
Germ-Line Mutation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Proto-Oncogene Proteins/genetics , Adult , Aged , Cohort Studies , Cyclin-Dependent Kinase Inhibitor p27 , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Serbia , United Kingdom
7.
FASEB J ; 22(6): 1672-83, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18198220

ABSTRACT

Chronic exposure to glucocorticoid hormones, resulting from either drug treatment or Cushing's syndrome, results in insulin resistance, central obesity, and symptoms similar to the metabolic syndrome. We hypothesized that the major metabolic effects of corticosteroids are mediated by changes in the key metabolic enzyme adenosine monophosphate-activated protein kinase (AMPK) activity. Activation of AMPK is known to stimulate appetite in the hypothalamus and stimulate catabolic processes in the periphery. We assessed AMPK activity and the expression of several metabolic enzymes in the hypothalamus, liver, adipose tissue, and heart of a rat glucocorticoid-excess model as well as in in vitro studies using primary human adipose and primary rat hypothalamic cell cultures, and a human hepatoma cell line treated with dexamethasone and metformin. Glucocorticoid treatment inhibited AMPK activity in rat adipose tissue and heart, while stimulating it in the liver and hypothalamus. Similar data were observed in vitro in the primary adipose and hypothalamic cells and in the liver cell line. Metformin, a known AMPK regulator, prevented the corticosteroid-induced effects on AMPK in human adipocytes and rat hypothalamic neurons. Our data suggest that glucocorticoid-induced changes in AMPK constitute a novel mechanism that could explain the increase in appetite, the deposition of lipids in visceral adipose and hepatic tissue, as well as the cardiac changes that are all characteristic of glucocorticoid excess. Our data suggest that metformin treatment could be effective in preventing the metabolic complications of chronic glucocorticoid excess.


Subject(s)
Cushing Syndrome/metabolism , Glucocorticoids/pharmacology , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Adipose Tissue/enzymology , Adipose Tissue/metabolism , Animals , Cells, Cultured , Cushing Syndrome/enzymology , Dexamethasone/pharmacology , Humans , Hypothalamus/enzymology , Hypothalamus/metabolism , Liver/enzymology , Liver/metabolism , Metabolism/drug effects , Metformin/pharmacology , Myocardium/enzymology , Myocardium/metabolism , Organ Specificity , Rats
8.
J Clin Endocrinol Metab ; 93(6): 2390-401, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18381572

ABSTRACT

CONTEXT: Mutations have been identified in the aryl hydrocarbon receptor-interacting protein (AIP) gene in familial isolated pituitary adenomas (FIPA). It is not clear, however, how this molecular chaperone is involved in tumorigenesis. OBJECTIVE: AIP sequence changes and expression were studied in FIPA and sporadic adenomas. The function of normal and mutated AIP molecules was studied on cell proliferation and protein-protein interaction. Cellular and ultrastructural AIP localization was determined in pituitary cells. PATIENTS: Twenty-six FIPA kindreds and 85 sporadic pituitary adenoma patients were included in the study. RESULTS: Nine families harbored AIP mutations. Overexpression of wild-type AIP in TIG3 and HEK293 human fibroblast and GH3 pituitary cell lines dramatically reduced cell proliferation, whereas mutant AIP lost this ability. All the mutations led to a disruption of the protein-protein interaction between AIP and phosphodiesterase-4A5. In normal pituitary, AIP colocalizes exclusively with GH and prolactin, and it is found in association with the secretory vesicle, as shown by double-immunofluorescence and electron microscopy staining. In sporadic pituitary adenomas, however, AIP is expressed in all tumor types. In addition, whereas AIP is expressed in the secretory vesicle in GH-secreting tumors, similar to normal GH-secreting cells, in lactotroph, corticotroph, and nonfunctioning adenomas, it is localized to the cytoplasm and not in the secretory vesicles. CONCLUSIONS: Our functional evaluation of AIP mutations is consistent with a tumor-suppressor role for AIP and its involvement in familial acromegaly. The abnormal expression and subcellular localization of AIP in sporadic pituitary adenomas indicate deranged regulation of this protein during tumorigenesis.


Subject(s)
Adenoma/genetics , Pituitary Neoplasms/genetics , Proteins/physiology , Acromegaly/genetics , Acromegaly/metabolism , Adenoma/metabolism , Adolescent , Adult , Aged , Cell Proliferation , Child , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Female , Gene Expression Regulation, Neoplastic , Genetic Testing , Human Growth Hormone/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Male , Middle Aged , Pituitary Neoplasms/metabolism , Protein Binding , Proteins/genetics , Proteins/metabolism , Transfection , Tumor Cells, Cultured
9.
Eur J Hum Genet ; 22(6): 784-91, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24129438

ABSTRACT

The major purpose of the present study was to quantify correctly spliced CFTR transcripts in human nasal epithelial cells from cystic fibrosis (CF) patients carrying the splicing mutations c.580-1G>T (712-1G>T) and c.2657+5G>A (2789+5G>A) and to assess the applicability of this model in CFTR therapeutic approaches. We performed the relative quantification of CFTR mRNA by reverse transcription quantitative PCR (RT-qPCR) of these splicing mutations in four groups (wild type, CF-F508del controls, CF patients and CF carriers) of individuals. In addition, in vitro assays using minigene constructs were performed to evaluate the effect of a new CF complex allele c.[2657+5G>A; 2562T>G]. Ex vivo qPCR data show that the primary consequence of both mutations at the RNA level is the skipping of their neighboring exon (6 and 16, respectively). The CFTR minigenes results mimicked the ex vivo data, as exon 16 skipping is the main aberrant transcript, and the correctly spliced transcript level was observed in a similar proportion when the c.2657+5G>A mutation is present. In summary, we provide evidence that ex vivo quantitative transcripts analysis using RT/qPCR is a robust technology that could be useful for measuring the efficacy of therapeutic approaches that attempt to achieve an increase in CFTR gene expression.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/genetics , Gene Expression , Mutation , Nasal Mucosa/metabolism , RNA Splicing , Adult , Blotting, Western , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , DNA Mutational Analysis , Female , Genotype , HEK293 Cells , Humans , Male , Nasal Mucosa/cytology , Phenotype , Polymorphism, Single Nucleotide , Reverse Transcriptase Polymerase Chain Reaction
10.
J Clin Endocrinol Metab ; 97(8): E1411-20, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22659247

ABSTRACT

CONTEXT: Somatotroph adenomas harboring aryl hydrocarbon receptor interacting protein (AIP) mutations respond less well to somatostatin analogs, suggesting that the effects of somatostatin analogs may be mediated by AIP. OBJECTIVE: The objective of the investigation was to study the involvement of AIP in the mechanism of effect of somatostatin analogs. DESIGN: In the human study, a 16-wk somatostatin analog pretreatment compared with no pretreatment. In the in vitro cell line study, the effect of somatostatin analog treatment or small interfering RNA (siRNA)/plasmid transfection were studied. SETTING: The study was conducted at a university hospital. PATIENTS: Thirty-nine sporadic and 10 familial acromegaly patients participated in the study. INTERVENTION: Interventions included preoperative lanreotide treatment and pituitary surgery. OUTCOME: For the human study, GH and IGF-I levels, AIP, and somatostatin receptor staining were measured. For the cell line, AIP and ZAC1 (zinc finger regulator of apoptosis and cell cycle arrest) expression, metabolic activity, and clone formation were measured. RESULTS: Lanreotide pretreatment reduced GH and IGF-I levels and tumor volume (all P < 0.0001). AIP immunostaining was stronger in the lanreotide-pretreated group vs. the surgery-only group (P < 0.001). After lanreotide pretreatment, the AIP score correlated to IGF-I changes in females (R = 0.68, P < 0.05). Somatostatin receptor staining was not reduced in samples with AIP mutations. In GH3 cells, 1 nm octreotide increased AIP mRNA and protein (both P < 0.01) and ZAC1 mRNA expression (P < 0.05). Overexpression of wild-type (but not mutant) AIP increased ZAC1 mRNA expression, whereas AIP siRNA knockdown reduced ZAC1 mRNA (both P < 0.05). The siRNA-mediated knockdown of AIP led to an increased metabolic activity and clonogenic ability of GH3 cells compared with cells transfected with a nontargeting control (both P < 0.001). CONCLUSION: These results suggest that AIP may play a role in the mechanism of action of somatostatin analogs via ZAC1 in sporadic somatotroph tumors and may explain their lack of effectiveness in patients with AIP mutations.


Subject(s)
Adenoma/drug therapy , Cell Cycle Proteins/physiology , Growth Hormone-Secreting Pituitary Adenoma/drug therapy , Intracellular Signaling Peptides and Proteins/physiology , Somatostatin/analogs & derivatives , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Cycle Proteins/analysis , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/analysis , Rats , Receptors, Somatostatin/analysis , Signal Transduction , Transcription Factors/analysis , Tumor Suppressor Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL