Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Clin Endocrinol Metab ; 94(6): 2085-91, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19293268

ABSTRACT

BACKGROUND: The "complex of myxomas, spotty skin pigmentation, and endocrine overactivity," or "Carney complex" (CNC), is caused by inactivating mutations of the regulatory subunit type 1A of the cAMP-dependent protein kinase (PRKAR1A) gene and as yet unknown defect(s) in other gene(s). Delineation of a genotype-phenotype correlation for CNC patients is essential for understanding PRKAR1A function and providing counseling and preventive care. METHODS: A transatlantic consortium studied the molecular genotype and clinical phenotype of 353 patients (221 females and 132 males, age 34 +/- 19 yr) who carried a germline PRKAR1A mutation or were diagnosed with CNC and/or primary pigmented nodular adrenocortical disease. RESULTS: A total of 258 patients (73%) carried 80 different PRKAR1A mutations; 114 (62%) of the index cases had a PRKAR1A mutation. Most PRKAR1A mutations (82%) led to lack of detectable mutant protein (nonexpressed mutations) because of nonsense mRNA mediated decay. Patients with a PRKAR1A mutation were more likely to have pigmented skin lesions, myxomas, and thyroid and gonadal tumors; they also presented earlier with these tumors. Primary pigmented nodular adrenocortical disease occurred earlier, was more frequent in females, and was the only manifestation of CNC with a gender predilection. Mutations located in exons were more often associated with acromegaly, myxomas, lentigines, and schwannomas, whereas the frequent c.491-492delTG mutation was commonly associated with lentigines, cardiac myxomas, and thyroid tumors. Overall, nonexpressed PRKAR1A mutations were associated with less severe disease. CONCLUSION: CNC is genetically and clinically heterogeneous. Certain tumors are more frequent, with specific mutations providing some genotype-phenotype correlation for PRKAR1A mutations.


Subject(s)
Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Mutation , Adolescent , Adrenal Cortex Diseases/complications , Adrenal Cortex Diseases/genetics , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , DNA Mutational Analysis , Female , Genotype , Humans , Infant , Male , Middle Aged , Mutation/physiology , Myxoma/complications , Myxoma/genetics , Phenotype , Young Adult
2.
Hormones (Athens) ; 7(3): 251-4, 2008.
Article in English | MEDLINE | ID: mdl-18694864

ABSTRACT

OBJECTIVE: We recently reported the association of the Sp1 site polymorphism of the COL1A1 gene with lumbar disk disease (LDD). In the present study we searched for a different polymorphism of the COL1A1 gene (which is usually not in linkage disequilibrium with the Sp1 site) in subjects with LDD. DESIGN: Blood was collected from 24 Greek army recruits, aged 29+/-7.6 years, with LDD, and 66 healthy men, aged 26+/-4.38 years, matched for body mass index (BMI) and age, with normal BMD and with no history of trauma or fractures, who served as controls. DNA was extracted and the COL1A1 gene was sequenced. Of the control subjects, 12 were army recruits and 54 were selected from the general population. RESULTS: The four base-pair insertion polymorphism in the COL1A1 gene analyzed by polymerase chain reaction amplification of DNA produces two different fragments (alleles A1 and A2): 14 patients (58.3%) were homozygous for A2A2, versus 35 controls (53%), while 3 patients (12.5%) were A1A1, and 8 of the control subjects (12%) had this genotype. There were no statistically significant differences in the presence of the two alleles of this polymorphism between patients with LDD and control subjects. CONCLUSIONS: A four base-pair insertion polymorphism of the COL1A1 gene is not associated with the presence of LDD in young males, unlike the Sp1 site polymorphism of the same gene. These data reinforce the association between LDD and the functional polymorphisms of the Sp1 site by showing that other polymorphic sites of the of the COL1A1 gene in the same population of patients are not linked to the disease.


Subject(s)
Collagen Type I/genetics , Intervertebral Disc/metabolism , Lumbar Vertebrae/metabolism , Polymorphism, Genetic , Spinal Diseases/genetics , Adult , Case-Control Studies , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , DNA Mutational Analysis , Gene Frequency , Genetic Predisposition to Disease , Greece , Homozygote , Humans , Male , Military Personnel , Polymerase Chain Reaction , Sp1 Transcription Factor/metabolism , Spinal Diseases/metabolism
3.
Eur J Hum Genet ; 16(1): 79-88, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17667967

ABSTRACT

Gastrointestinal stromal tumors (GISTs) may be caused by germline mutations of the KIT and platelet-derived growth factor receptor-alpha (PDGFRA) genes and treated by Imatinib mesylate (STI571) or other protein tyrosine kinase inhibitors. However, not all GISTs harbor these genetic defects and several do not respond to STI571 suggesting that other molecular mechanisms may be implicated in GIST pathogenesis. In a subset of patients with GISTs, the lesions are associated with paragangliomas; the condition is familial and transmitted as an autosomal-dominant trait. We investigated 11 patients with the dyad of 'paraganglioma and gastric stromal sarcoma'; in eight (from seven unrelated families), the GISTs were caused by germline mutations of the genes encoding subunits B, C, or D (the SDHB, SDHC and SDHD genes, respectively). In this report, we present the molecular effects of these mutations on these genes and the clinical information on the patients. We conclude that succinate dehydrogenase deficiency may be the cause of a subgroup of GISTs and this offers a therapeutic target for GISTs that may not respond to STI571 and its analogs.


Subject(s)
Gastrointestinal Stromal Tumors/enzymology , Gastrointestinal Stromal Tumors/genetics , Germ-Line Mutation , Iron-Sulfur Proteins/genetics , Membrane Proteins/genetics , Neoplastic Syndromes, Hereditary/enzymology , Neoplastic Syndromes, Hereditary/genetics , Paraganglioma/enzymology , Paraganglioma/genetics , Succinate Dehydrogenase/genetics , Adolescent , Adult , Alleles , Antineoplastic Agents/therapeutic use , Base Sequence , Benzamides , Child , DNA Primers/genetics , DNA, Neoplasm/genetics , Female , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Genes, Dominant , Heterozygote , Humans , Imatinib Mesylate , Loss of Heterozygosity , Male , Neoplastic Syndromes, Hereditary/drug therapy , Neoplastic Syndromes, Hereditary/pathology , Paraganglioma/drug therapy , Paraganglioma/pathology , Piperazines/therapeutic use , Pyrimidines/therapeutic use
4.
Breast Cancer Res Treat ; 107(3): 431-41, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17453335

ABSTRACT

127 Greek breast/ovarian cancer families were screened for germline BRCA1/2 mutations by dHPLC followed by direct sequencing. Our results indicated 16 and 5 breast/ovarian cancer families bearing deleterious mutations in the BRCA1 and BRCA2 genes, respectively. Two novel BRCA2 germline mutations (G4X and 3783del10) are reported here for the first time. Subsequent compilation of our present findings with previously reported mutation data reveals that in a total of 287 Greek breast/ovarian cancer families, 46 and 13 carry a deleterious mutation in BRCA1 and BRCA2, respectively. It should be noted that two BRCA1 mutations, 5382insC and G1738R, both located in exon 20, account for 46% of the families found to carry a mutation. Based on our mutation analysis results, we propose here a hierarchical, cost-effective BRCA1/2 mutation screening protocol for individuals of Greek ethnic origin. The suggested protocol can impact on the clinical management of breast-ovarian cancer families on a national healthcare system level.


Subject(s)
Breast Neoplasms/genetics , Genes, BRCA1 , Genes, BRCA2 , Mutation , Ovarian Neoplasms/genetics , Cost-Benefit Analysis , Female , Greece , Humans
5.
Am J Surg Pathol ; 31(11): 1764-75, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18059235

ABSTRACT

Pigmented epithelioid melanocytoma (PEM) is a recently described entity comprising most cases previously described as "animal-type melanoma" and epithelioid blue nevus (EBN) occurring in patients with the multiple neoplasia syndrome Carney complex (CNC). Mutations of the protein kinase A regulatory subunit type 1alpha (R1alpha) (coded by the PRKAR1A gene) are found in more than half of CNC patients. In this study, we investigated whether PEM and EBN are related at the molecular level, and whether changes in the PRKAR1A gene status and the expression of the R1alpha protein may be involved in the pathogenesis of PEM and other melanocytic lesions. Histologic analysis of hematoxylin and eosin-stained sections and immunohistochemistry (IHC) with R1alpha antibody were performed on 34 sporadic PEMs, 8 CNC-associated PEMs from patients with known PRKAR1A mutations, 297 benign and malignant melanocytic tumors (127 conventional sections of 10 compound nevi, 10 Spitz nevi, 5 deep-penetrating nevi, 5 blue nevi, 6 cellular blue nevi, 2 malignant blue nevi, 3 lentigo maligna, and 86 melanomas of various types); in addition, 170 tissue microarray sections consisting of 35 benign nevi, 60 primary melanomas, and 75 metastatic melanomas, and 5 equine dermal melanomas, were examined. Histologic diagnoses were based on preexisting pathologic reports and were confirmed for this study. DNA studies [loss of heterozygosity (LOH) for the 17q22-24 locus and the PRKAR1A gene sequencing] were performed on 60 melanomas and 7 PEMs. IHC showed that R1alpha was expressed in all but one core from tissue microarrays (169/170), and in all 127 melanocytic lesions evaluated in conventional sections. By contrast, R1alpha was not expressed in the 8 EBN from patients with CNC and PRKAR1A mutations. Expression of R1alpha was lost in 28 of 34 PEMs (82%). R1alpha was expressed in the 5 equine melanomas studied. DNA studies correlated with IHC findings: there were no PRKAR1A mutations in any of the melanomas studied and the rate of LOH for 17q22-24 was less than 7%; 5 of the 7 PEMs showed extensive 17q22-24 LOH but no PRKAR1A mutations. The results support the concept that PEM is a distinct melanocytic tumor occurring in a sporadic setting and in the context of CNC. They also suggest that PEM differs from melanomas in equine melanotic disease, further arguing that the term animal-type melanoma may be a misnomer for this group of lesions. Loss of expression of R1alpha offers a useful diagnostic test that helps to distinguish PEM from lesions that mimic it histologically.


Subject(s)
Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/analysis , Horse Diseases/enzymology , Melanocytes/enzymology , Melanoma/enzymology , Neoplasms, Multiple Primary/enzymology , Nevus, Blue/enzymology , Nevus, Epithelioid and Spindle Cell/enzymology , Skin Neoplasms/enzymology , Animals , Chromosomes, Human, Pair 17 , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Diagnosis, Differential , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Horse Diseases/genetics , Horse Diseases/pathology , Horses , Humans , Immunohistochemistry , Loss of Heterozygosity , Melanocytes/pathology , Melanoma/pathology , Melanoma/veterinary , Mutation , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/pathology , Nevus, Blue/classification , Nevus, Blue/genetics , Nevus, Blue/pathology , Nevus, Epithelioid and Spindle Cell/classification , Nevus, Epithelioid and Spindle Cell/genetics , Nevus, Epithelioid and Spindle Cell/pathology , Skin Neoplasms/classification , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/veterinary , Terminology as Topic , Tissue Array Analysis
6.
Hormones (Athens) ; 6(2): 152-6, 2007.
Article in English | MEDLINE | ID: mdl-17704047

ABSTRACT

Multiple endocrine neoplasia type 2A (MEN2A) is a syndrome of familial neoplasias characterized by medullary thyroid carcinoma (MTC), pheochromocytoma and hyperplasia of the parathyroid glands. RET protooncogene mutations are responsible for MEN 2A. Mutations in exons 10 or 11 have been identified in more than 96% of patients with MEN 2A. We herein report for the first time a patient with MEN 2A harboring a mutation (Gly(533)Cys) in exon 8. A 66-year old male patient was referred to our department for bilateral adrenal nodules. The patient's family history was remarkable in that his mother had pheochromocytoma. Biochemical evaluation and findings of the magnetic resonance imaging of the adrenals were compatible with the diagnosis of bilateral pheochromocytomas. The patient underwent laparoscopic bilateral adrenalectomy and histological examination confirmed the preoperative diagnosis of pheochromocytoma. Absence of phenotypic characteristics of VHL or NF1 and elevated calcitonin levels both basal and post pentagastrin stimulation, raised the possibility of MEN 2A syndrome. Total thyroidectomy was performed and histological examination showed the presence of MTC. Direct sequencing of exon 8 from the patient's genomic DNA revealed the mutation c.1,597G-->T (Gly533Cys). Although this missense point mutation has been associated with familial MTC (FMTC), to the best of our knowledge mutations in exon 8 have not previously been identified in patients with MEN 2A. In conclusion, in patients with clinical suspicion of MEN 2A syndrome, analysis of RET exon 8 should be considered when the routine evaluation of MEN 2A-associated mutations is negative. Furthermore, patients with FMTC and exon 8 mutations should also be screened for pheochromocytoma.


Subject(s)
Exons , Multiple Endocrine Neoplasia Type 2a/genetics , Mutation , Proto-Oncogene Proteins c-ret/genetics , Adrenal Gland Neoplasms/diagnosis , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Adrenal Glands/pathology , Aged , Base Sequence , Cysteine , Glycine , Guanine , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Male , Molecular Sequence Data , Multiple Endocrine Neoplasia Type 2a/diagnosis , Pedigree , Pheochromocytoma/diagnosis , Pheochromocytoma/genetics , Pheochromocytoma/metabolism , Thymine
7.
J Clin Endocrinol Metab ; 92(9): 3728-32, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17566086

ABSTRACT

CONTEXT: Gastrointestinal stromal tumors (GISTs) may be caused by somatic or germline mutations of the KIT and PDGFRA genes, but most GISTs associated with neuroendocrine tumors (NETs) are not, suggesting that other molecular pathways are implicated in their pathogenesis. OBJECTIVE: In the course of investigating NETs and GIST genetics, we encountered a patient who had a unique combination of multiple fibrous polyps and lipomas of the small intestine and several gastric GISTs. DESIGN: The study included the clinical description of a unique patient, DNA sequencing of germline and tumor DNA, and comparative genomic hybridization (CGH) and allelic marker analysis of tumor DNA. RESULTS: The patient was found to carry a germline PDGFRA mutation (V561D) in the heterozygote state; it has only been seen rarely before and only in the somatic state in sporadic GISTs. CGH identified losses of chromosomal regions 1p33-36, 9q12-24, 11q13, and 16q; loss of the 14q region that is commonly lost in NETs and GISTs was shown by DNA marker analysis. These changes are likely to point to secondary and tertiary genetic hits involved in the formation of these rare tumors. CONCLUSIONS: Multiple GISTs and other tumors may be caused by germline PDGFRA gene mutations; the V561D mutation can occur in the germline state and lead to a syndrome that should not be confused with other genetic conditions associated with a predisposition to NETs and other tumors. A number of chromosomal loci are likely to be involved in the PDGFRA V561D-dependent tumorigenesis, as shown by CGH and other DNA analyses.


Subject(s)
Gastrointestinal Stromal Tumors/genetics , Germ-Line Mutation , Neoplasms, Multiple Primary/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Aspartic Acid/genetics , Base Sequence , DNA Mutational Analysis , Female , Humans , Middle Aged , Valine/genetics
8.
J Clin Endocrinol Metab ; 92(8): 2938-43, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17535989

ABSTRACT

CONTEXT: Carney triad (CT) describes the association of paragangliomas (PGLs) with gastrointestinal stromal tumors (GISTs) and pulmonary chondromas. Inactivating mutations of the mitochondrial complex II succinate dehydrogenase (SDH) enzyme subunits SDHB, SDHC, and SDHD are found in PGLs, gain-of-function mutations of c-kit (KIT), and platelet-derived growth factor receptor A (PDGFRA) in GISTs. OBJECTIVE: Our objective was to investigate the possibility that patients with CT and/or their tumors may harbor mutations of the SDHB, SDHC, SDHD, KIT, and PDGFRA genes and identify any other genetic alterations in CT tumors. DESIGN: Three males and 34 females with CT were studied retrospectively. We sequenced the stated genes and performed comparative genomic hybridization on a total of 41 tumors. RESULTS: No patient had coding sequence mutations of the investigated genes. Comparative genomic hybridization revealed a number of DNA copy number changes: losses dominated among benign lesions, there were an equal number of gains and losses in malignant lesions, and the average number of alterations in malignant tumors was higher compared with benign lesions. The most frequent and greatest contiguous change was 1q12-q21 deletion, a region that harbors the SDHC gene. Another frequent change was loss of 1p. Allelic losses of 1p and 1q were confirmed by fluorescent in situ hybridization and loss-of-heterozygosity studies. CONCLUSIONS: We conclude that CT is not due to SDH-inactivating or KIT- and PDGFRA-activating mutations. GISTs and PGLs in CT are associated with chromosome 1 and other changes that appear to participate in tumor progression and point to their common genetic cause.


Subject(s)
Chromosomes, Human, Pair 1/genetics , Gastrointestinal Neoplasms/genetics , Gastrointestinal Stromal Tumors/genetics , Germ-Line Mutation/genetics , Lung Neoplasms/genetics , Paraganglioma/genetics , Adolescent , Adult , Child , DNA, Neoplasm/genetics , Female , Gene Dosage , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Loss of Heterozygosity , Male , Nucleic Acid Hybridization , Retrospective Studies , Succinate Dehydrogenase/genetics , Tumor Cells, Cultured
9.
Clin Endocrinol (Oxf) ; 64(5): 561-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16649977

ABSTRACT

OBJECTIVE: Familial medullary thyroid carcinoma (FMTC) is caused by germ-line mutations in the RET proto-oncogene. These mutations concern mainly cysteine residues in exons 10 and 11, whereas noncysteine mutations in exons 13-16 are rare. Mutations in other exons have been reported only in isolated families. In this study we have analysed the RET gene in two FMTC families negative for mutations in the above exons. DESIGN: We have analysed exons 7-19 and 21 in one index patient from each family using DNA sequencing. PATIENTS: Twenty-eight subjects from both families were clinically assessed and subsequently molecularly analysed for the presence of RET gene mutations. RESULTS: We have found the mutation c.1597G-->T (Gly533Cys) in two Greek families with FMTC. The mutation was detected in all seven MTC patients of both families as well as in 13 asymptomatic relatives in the heterozygote state, although one of the patients was also a homozygote due to consanguinity. The mutation shows a wide clinical heterogeneity, as there are carrier patients with age of diagnosis ranging from 23 to 88 years. CONCLUSIONS: It is likely that this mutation causes FMTC, as no other mutation was found in the RET gene, the mutation co-segregates with FMTC, and family members without the mutation are clinically unaffected. As the same point mutation was previously found in a large Brazilian family, it may be present in other populations as well. Therefore, exon 8 of RET should be screened in FMTC families with no identified common RET mutations.


Subject(s)
Carcinoma, Medullary/genetics , Point Mutation , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogenes , Thyroid Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Consanguinity , DNA Mutational Analysis , Exons , Female , Genetic Testing , Greece , Heterozygote , Humans , Male , Middle Aged , Pedigree , Proto-Oncogene Mas
10.
Am J Surg Pathol ; 30(1): 42-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16330941

ABSTRACT

Functioning paraganglioma and gastrointestinal stromal tumor (GIST) are uncommon tumors that occur mostly in a sporadic and isolated form, occasionally as components of multiple neoplasia syndromes, either separately or together. Separately, they occur in several inherited syndromes including multiple endocrine neoplasia 2, and the GIST, lentigines, and mast cell tumor syndrome. Together, they are variably prominent components of three syndromes: the familial paraganglioma and gastric GIST syndrome, neurofibromatosis type 1, and the Carney triad. The two former conditions are inherited as autosomal dominant traits; the latter does not appear to be inherited and affects young women predominantly. This article reports the nonfamilial occurrence of functioning paraganglioma and GIST of the jejunum in 3 women, 1 young (22 years) at initial presentation. The occurrences were unexpected because of the infrequency of the tumors. The neoplasms, respectively, did not show germline SDHA, SDHB, SDHC, and SDHD, and KIT mutations associated with familial paraganglioma and familial GIST. The paraganglioma-jejunal GIST combination may be the harbinger of a rare genetic syndrome, a variant of the Carney triad or the paraganglioma-gastric stromal sarcoma syndrome, or be coincidental.


Subject(s)
Gastrointestinal Stromal Tumors/pathology , Jejunal Neoplasms/pathology , Jejunal Neoplasms/physiopathology , Neoplasms, Multiple Primary/pathology , Paraganglioma/pathology , Adult , Aged , Female , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/physiopathology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Jejunal Neoplasms/genetics , Middle Aged , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/physiopathology , Paraganglioma/genetics , Paraganglioma/physiopathology , Polymerase Chain Reaction , Pregnancy
11.
Cancer Res ; 63(17): 5308-19, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14500362

ABSTRACT

Germ-line protein kinase A (PKA) regulatory-subunit type-Ialpha (RIalpha; PRKAR1A)-inactivating mutations and loss-of-heterozygosity (LOH) of its 17q22-24 locus have been found in Cushing syndrome (CS) caused by primary pigmented nodular adrenocortical disease (PPNAD). We examined whether somatic 17q22-24, PRKAR1A, or PKA changes are present in 44 sporadic adrenocortical tumors (29 adenomas and 15 cancers); 26 of these tumors were responsible for CS. A probe containing the PRKAR1A gene-mapped by fluorescent in situ hybridization to 17q22-24-and corresponding microsatellite markers were used to study allelic losses; PRKAR1A was sequenced in all samples. 17q22-24 losses were seen in 23 and 53% of adenomas and cancers, respectively. In three tumors, somatic, PRKAR1A-inactivating mutations were identified: (a) a nonsense mutation in exon 6 (A751G); (b) a splicing mutation (9IVS-1G/A); and (c) a transition (1050T>C) followed by a 22-bp deletion, also in exon 9; all predicted premature RIalpha protein terminations. Quantitative message and protein studies showed RIalpha down-regulation in tumors with genetic changes; their cortisol secretion pattern was similar to that of PPNAD, and they had higher PKA activity by enzymatic studies. We conclude that somatic allelic losses of the 17q22-24 region, PRKAR1A-inactivating mutations or down-regulation, and corresponding PKA activity changes are present in at least some sporadic adrenocortical tumors, especially those with a PPNAD-like clinical presentation of CS.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Adenoma/genetics , Chromosomes, Human, Pair 17/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Adrenal Cortex Neoplasms/enzymology , Adrenocortical Adenoma/enzymology , Adult , Aged , Alleles , Blotting, Western , Chromosome Mapping , Cushing Syndrome/complications , Cushing Syndrome/enzymology , Cushing Syndrome/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Immunohistochemistry , Loss of Heterozygosity , Male , Middle Aged , Mutation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
12.
FASEB J ; 17(9): 1189-91, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12692076

ABSTRACT

A mouse protein that interacts with the peripheral-type benzodiazepine receptor (PBR) and cAMP-dependent protein kinase A (PKA) regulatory subunit RIalpha (PRKAR1A), named PBR and PKA-associated protein 7 (PAP7), was identified and shown to be involved in hormone-induced steroid biosynthesis. We report the identification of the human PAP7 gene, its expression pattern, genomic structure, and chromosomal mapping to 1q32-1q41. Human PAP7 is a 60-kDa protein highly homologous to the rodent protein. PAP7 is widely present in human tissues and highly expressed in seminal vesicles, pituitary, thyroid, pancreas, renal cortex, enteric epithelium, muscles, myocardium and in steroidogenic tissues, including the gonads and adrenal cortex. These tissues are also targets of Carney complex (CNC), a multiple neoplasia syndrome caused by germline inactivating PRKAR1A mutations (PRKAR1A-mut) and associated with primary pigmented nodular adrenocortical disease (PPNAD) and increased steroid synthesis. PAP7 and PRKAR1A expression were studied in PPNAD and in lymphoblasts from patients bearing PRKAR1A-mut. Like PRKAR1A, PAP7 was decreased in CNC lymphocytes and PPNAD nodules, but not in the surrounding cortex. These studies showed that, like in the mouse, human PAP7 is highly expressed in steroidogenic tissues, where it follows the pattern of PRKAR1A expression, suggesting that it participates in PRKAR1A-mediated tumorigenesis and hypercortisolism.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Adaptor Proteins, Signal Transducing , Adrenal Cortex Diseases/metabolism , Animals , Base Sequence , Cell Line , Cholesterol/metabolism , Chromosome Mapping , Cloning, Molecular , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Cyclic AMP-Dependent Protein Kinases/genetics , Humans , Male , Membrane Proteins , Mice , Mitochondria/metabolism , Models, Biological , Molecular Sequence Data , Mutation , RNA, Messenger/biosynthesis , Receptors, GABA-A/biosynthesis , Receptors, GABA-A/physiology , Steroids/biosynthesis , Testis/metabolism , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...