Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Biotech Histochem ; 89(7): 518-28, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24799092

ABSTRACT

Unfixed tissue specimens most frequently are stored for long term research uses at either -80° C or in vapor phase liquid nitrogen (VPLN). There is little information concerning the effects such long term storage on tissue RNA or protein available for extraction. Aliquots of 49 specimens were stored for 5-12 years at -80° C or in VPLN. Twelve additional paired specimens were stored for 1 year under identical conditions. RNA was isolated from all tissues and assessed for RNA yield, total RNA integrity and mRNA integrity. Protein stability was analyzed by surface-enhanced or matrix-assisted laser desorption ionization time of flight mass spectrometry (SELDI-TOF-MS, MALDI-TOF-MS) and nano-liquid chromatography electrospray ionization tandem mass spectrometry (nLC-ESI-MS/MS). RNA yield and total RNA integrity showed significantly better results for -80° C storage compared to VPLN storage; the transcripts that were preferentially degraded during VPLN storage were these involved in antigen presentation and processing. No consistent differences were found in the SELDI-TOF-MS, MALDI-TOF-MS or nLC-ESI-MS/MS analyses of specimens stored for more than 8 years at -80° C compared to those stored in VPLN. Long term storage of human research tissues at -80° C provides at least the same quality of RNA and protein as storage in VPLN.


Subject(s)
Freezing , Proteins/chemistry , RNA/chemistry , Tissue Preservation/methods , Cold Temperature , Gene Expression Profiling , Humans , Microarray Analysis , Neoplasms/pathology , Proteomics/methods , RNA, Messenger/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
2.
Am J Pathol ; 159(3): 1121-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549605

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant hereditary disorder characterized by multiple parathyroid, pancreatic, duodenal, and pituitary neuroendocrine tumors. Nonendocrine mesenchymal tumors, such as lipomas, collagenomas, and angiofibromas have also been reported. MEN1-associated neuroendocrine and some mesenchymal tumors have documented MEN1 gene alterations on chromosome 11q13. To test whether the MEN1 gene is involved in the pathogenesis of multiple smooth muscle tumors, we examined the 11q13 loss of heterozygosity (LOH) and clonality patterns in 15 leiomyomata of the esophagus, lung, and uterus from five patients with MEN1. Forty sporadic uterine leiomyomata were also studied for 11q13 LOH. LOH analysis was performed using four polymorphic DNA markers at the MEN1 gene locus; D11S480, PYGM, D11S449, and INT-2. 11q13 LOH was detected in 10 of 12 (83%) MEN1-associated esophageal and uterine smooth muscle tumors. In contrast, LOH at the MEN1 gene locus was demonstrated only in 2 of 40 (5%) sporadic uterine tumors. LOH at 11q13 was not documented in three lung smooth muscle tumors from a single patient with MEN1. Ten tumors from two female patients were additionally assessed for clonality by X-chromosome inactivation analysis. The results demonstrated different clonality patterns in multiple tumors in the same organ in each individual patient. The data indicate that leiomyomata of the esophagus and uterus in MEN1 patients arise as independent clones, develop through MEN1 gene alterations, and are an integral part of MEN1. However, the MEN1 gene is not a significant contributor to the tumorigenesis of sporadic uterine leiomyomata.


Subject(s)
Esophageal Neoplasms/genetics , Leiomyomatosis/genetics , Lung Neoplasms/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Uterine Neoplasms/genetics , Adult , Chromosome Mapping , Chromosomes, Human, Pair 11/genetics , Female , Gene Silencing , Humans , Loss of Heterozygosity , Middle Aged , X Chromosome/genetics
4.
Cancer Res ; 60(22): 6223-6, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11103773

ABSTRACT

Inherited mutations of the RET proto-oncogene are tumorigenic in patients with multiple endocrine neoplasia type 2 (MEN 2). However, it is not understood why only few of the affected cells in the target organs develop into tumors. Genetic analysis of nine pheochromocytomas from five unrelated patients with MEN 2 showed either duplication of the mutant RET allele in trisomy 10 or loss of the wild-type RET allele. Our results suggest a "second hit" causing a dominant effect of the mutant RET allele, through either duplication of the mutant allele or loss of the wild-type allele, as a possible mechanism for pheochromocytoma tumorigenesis in patients with MEN 2.


Subject(s)
Adrenal Gland Neoplasms/genetics , Chromosomes, Human, Pair 10 , Drosophila Proteins , Loss of Heterozygosity , Multiple Endocrine Neoplasia Type 2a/genetics , Pheochromocytoma/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Trisomy , Alleles , DNA, Neoplasm/blood , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Germ-Line Mutation , Humans , In Situ Hybridization, Fluorescence , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret
5.
Am J Pathol ; 157(5): 1615-21, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11073821

ABSTRACT

Microcystic adenoma and cysts of the pancreas occur sporadically or as a part of von Hippel-Lindau (VHL) disease. The pathology of pancreatic cystic disease in VHL patients has not been well characterized. Furthermore, it is presently unknown whether the alteration of the VHL gene is responsible for the development of the entire spectrum of pancreatic serous cystic lesions. We performed a histopathological analysis of 21 cysts and 98 microcystic adenomas in nine VHL patients with a known germline mutation. In addition, PCR-amplified DNA from 27 pancreatic cystic lesions in three informative patients was studied for allelic deletions with polymorphic markers spanning the VHL gene locus. In all patients, pancreatic lesions were multiple: 21 benign serous cysts, 63 microscopic microcystic adenomas (size <0.4 cm), and 35 macroscopic microcystic adenomas (size >0.5 cm). The average number of lesions per patient was 2.1 benign cysts (range, 0-8), 7.7 (1-37) microscopic microcystic adenomas, and 3 (0-21) macroscopic microcystic adenomas. All lesions showed similar histology and contained prominent fibrous stroma, clear and/or amphophilic, glycogen-rich epithelial cells, endothelial and smooth muscle cells. VHL deletions were detected in all types of pancreatic cystic lesions. The presence of VHL gene allelic deletions in the spectrum of multifocal pancreatic cystic lesions provides direct molecular evidence of their neoplastic nature and integral association with VHL disease. The histopathological and molecular data establish a serous cyst-microcystic adenoma continuum in the development of pancreatic cystic neoplasia in VHL disease.


Subject(s)
Adenoma/pathology , Cysts/pathology , Pancreatic Diseases/pathology , Pancreatic Neoplasms/pathology , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/pathology , Adenoma/genetics , Adult , Aged , Cysts/genetics , DNA, Neoplasm/genetics , Female , Humans , Loss of Heterozygosity , Male , Middle Aged , Molecular Biology/methods , Pancreatic Diseases/genetics , Pancreatic Neoplasms/genetics
6.
J Clin Endocrinol Metab ; 85(11): 4146-56, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11095446

ABSTRACT

The p16INK4a/CDKN2A gene (p16INK4a) is frequently altered by homozygous deletion, mutation, or methylation in many nonendocrine tumors, and these alterations may be predictive of recurrence, tumor growth, or aggressiveness. Whether this is true of neuroendocrine tumors such as gastrinomas is unclear. To address this question we analyzed the gastrinomas from 44 patients for p16INK4a gene mutations and correlated the results to the tumor's biological behavior, growth pattern, and aggressiveness. No gastrinomas had mutations of exon 1 or exon 2 of the p16INK4a gene, although polymorphisms were found in 54%. No homozygous deletions were found. In 52% of the gastrinomas, hypermethylation of a 5'-CpG island of the p16INK4a gene promoter was found. To assess the growth behavior of the gastrinomas, all patients were assessed yearly with at least three conventional imaging studies (computed tomography scan, magnetic resonance imaging, and ultrasound), and since 1994 have been assessed with radionuclide scanning using [111In-diethylenetriamine pentaacetic acid,DPhe1]octreotide. The mean follow-up was 5.1+/-0.4 yr (range, 1.2-11.7). The presence or absence of methylation of the p16INK4a gene did not correlate with clinical characteristics of the gastrinoma, biological behavior (gastrin release and basal or maximal acid output), the presence or absence of known prognostic factors (tumor size, gastrinoma location, lymph node metastases, liver metastases, and curability), or growth pattern of the gastrinoma postresection. These results indicate that methylation of the p16INK4a gene is the most common gene alteration described to date in gastrinomas. Furthermore, because it is independent of disease stage it is probably an early event in the pathogenesis and because it is independent of the primary gastrinoma location, which is now thought to have different origins, methylation of the p16INK4a gene is probably a central process in the molecular pathogenesis of these tumors.


Subject(s)
Carrier Proteins/genetics , Duodenal Neoplasms/genetics , Gastrinoma/genetics , Genes, Tumor Suppressor , Mutation , Pancreatic Neoplasms/genetics , Polymorphism, Genetic , Adolescent , Adult , Cyclin-Dependent Kinase Inhibitor p16 , DNA Methylation , Duodenal Neoplasms/pathology , Duodenal Neoplasms/surgery , Exons , Female , Follow-Up Studies , Gastrinoma/diagnostic imaging , Gastrinoma/pathology , Gastrinoma/surgery , Gastrins/blood , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasms, Unknown Primary/genetics , Neoplasms, Unknown Primary/pathology , Neoplasms, Unknown Primary/surgery , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Radionuclide Imaging , Time Factors , Tumor Cells, Cultured
7.
Cancer Res ; 60(19): 5536-42, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11034099

ABSTRACT

Gastrinomas are rare gastrin-secreting endocrine tumors that usually arise in the duodenum or pancreas and, if untreated, can cause severe peptic ulcers or metastatic disease. Although most tumors are sporadic they are especially common in patients with multiple endocrine neoplasia type 1 (MEN1), and most studies of these tumors have focused on the role of the MEN1 gene. Although the gene is commonly altered in sporadic tumors, this finding is not universal, and it is highly likely that other genetic defects play a significant role. In the present study, an in-depth analysis of the DNA of eight tumors was carried out in an effort to localize these areas. The experiments consisted of an analysis of 400 microsatellite marker loci distributed evenly throughout the human genome, and the results were confirmed with comparative genomic hybridization. Whereas deletions encompassing the MEN1 gene were seen in two tumors, the most striking result was multiple large rearrangements on chromosome 1 in two of the tumors with hepatic metastases. In several instances, an individual tumor had abnormalities of every informative maker on a given chromosome, presumably as a result of aneuploidy affecting that chromosome. Such defects were only seen in the four large or aggressive tumors, and the total number of chromosomes affected in a tumor ranged from 1 to a high of 13 in a patient who had an unusually aggressive tumor This tumor also showed microsatellite instability, and this is the first report of such a defect in gastrinomas. This study implicates chromosome 1 defects, aneuploidy, and perhaps mismatch repair defects as importan features of gastrinomas; deletions involving the MEN1 gene were con firmed, but the rest of the genome was free of large deletions or amplifications.


Subject(s)
Duodenal Neoplasms/genetics , Gastrinoma/genetics , Pancreatic Neoplasms/genetics , Allelic Imbalance , Aneuploidy , Chromosome Aberrations , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 5 , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Duodenal Neoplasms/pathology , Genome, Human , Humans , Lymphatic Metastasis , Microsatellite Repeats , Nucleic Acid Hybridization , Pancreatic Neoplasms/pathology , Polymerase Chain Reaction
8.
J Nucl Med ; 41(10): 1646-56, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11037994

ABSTRACT

UNLABELLED: Gastric carcinoids are of increasing clinical concern because they may develop in hypergastrinemic states, especially with the increased chronic use of potent acid suppressants that can cause hypergastrinemia. However, gastric carcinoids are difficult to diagnose. Somatostatin receptor scintigraphy (SRS) has a high sensitivity and specificity for localizing carcinoids in other locations. The purpose of this study was to determine whether SRS could localize gastric carcinoids. METHODS: Two groups of patients with Zollinger-Ellison syndrome (ZES) with hypergastrinemia, each having a different increased risk of developing gastric carcinoids, were studied. One hundred sixty-two consecutive patients with ZES were studied prospectively, with 39 having multiple endocrine neoplasia, type 1 (MEN-1) (high increased risk), and 123 not having MEN-1 (low increased risk). Patients were admitted to the hospital initially and then yearly, undergoing SRS with SPECT, upper gastrointestinal endoscopy, and Jumbo Cup biopsies of any gastric abnormalities, as well as random biopsies of the gastric body. Tumor localization studies were also performed. Both the results of the routine SRS interpretation and the results of a masked review, with particular attention to the stomach of high risk MEN-1 patients, were correlated with the gastric biopsy results. RESULTS: Gastric SRS localization was positive in 19 (12%) of 162 patients. Sixteen patients had a gastric carcinoid, and 12 of these patients had SRS localization. The sensitivity of SRS in localizing a gastric carcinoid was 75%, with a specificity of 95%. Positive and negative predictive values were 63% and 97%, respectively. CONCLUSION: SRS is a noninvasive method that can identify patients with gastric carcinoids with a reasonable sensitivity and a high specificity. SRS should prove useful in the treatment of patients with hypergastrinemic states that have an increased incidence of gastric carcinoids. In patients with MEN-1, one must realize that localization in the upper abdomen on SRS may be caused by a gastric carcinoid and not a pancreatic endocrine tumor.


Subject(s)
Carcinoid Tumor/diagnostic imaging , Receptors, Somatostatin/metabolism , Stomach Neoplasms/diagnostic imaging , Tomography, Emission-Computed, Single-Photon , Carcinoid Tumor/metabolism , Case-Control Studies , Female , Humans , Indium Radioisotopes , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/diagnostic imaging , Octreotide/analogs & derivatives , Pentetic Acid/analogs & derivatives , Predictive Value of Tests , Prospective Studies , Radiopharmaceuticals , Sensitivity and Specificity , Stomach Neoplasms/metabolism , Zollinger-Ellison Syndrome/diagnostic imaging
9.
Am J Clin Pathol ; 114(3): 419-25, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10989643

ABSTRACT

To investigate the optimum number of biopsy specimens to be obtained for enterochromaffin-like (ECL) cell monitoring in hypergastrinemic patients and ECL cell regional variations potentially influencing the results, qualitative ECL cell changes were assessed in 149 patients with Zollinger-Ellison syndrome using jumbo biopsy specimens and a systematic sampling procedure of 4 areas each from the lesser or greater curvature of the gastric body. Of 1,176 specimens examined, 1,101 were adequate. The correlation was excellent between different sites within the greater or lesser curvature. In contrast, a normal ECL cell pattern was more frequent in the lesser curvature, whereas linear hyperplasia was more frequent in the greater curvature. Dysplastic lesions and carcinoid tumors in endoscopically unremarkable mucosa were detected in 3.4% and 1.2% of biopsy specimens, respectively, and were equally distributed between the lesser and greater curvature. Their chances of being diagnosed were related to the number of specimens examined. Extensive sampling of both the lesser and greater curvature is recommended for early diagnosis of dysplastic and/or carcinoid lesions in patients at risk. In contrast, limited sampling in the greater curvature seems to be adequate in patients with no risk for carcinoid development.


Subject(s)
Enterochromaffin Cells/pathology , Gastric Mucosa/pathology , Zollinger-Ellison Syndrome/pathology , Biopsy/methods , Carcinoid Tumor/pathology , Cell Count , Enterochromaffin Cells/chemistry , Gastric Mucosa/metabolism , Humans , Hyperplasia , Immunoenzyme Techniques , Zollinger-Ellison Syndrome/metabolism
10.
Cancer Res ; 60(1): 60-3, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10646853

ABSTRACT

Gastrinomas are neuroendocrine neoplasms that occur sporadically and in patients with multiple endocrine neoplasia type 1 (MEN1). In MEN1, multiple gastrinomas have been shown to arise by independent clonal events (Debelenko, et al., Cancer Res., 57: 2238-2243, 1997). The purpose of the present study was to analyze clonality in 20 sporadic gastrinomas from eight patients in whom the tumor was present in at least two separate sites. A combination of methods was used to assess clonality, including MEN1 gene mutation analysis, loss of heterozygosity analysis of the MEN1 locus, and analysis of X-chromosome inactivation at the human androgen receptor locus (human androgen receptor analysis). In three patients, a somantic MEN1 gene mutation was detected in the tumor. Identical mutations were found in other tumors at different sites within the same patients. Human androgen receptor analysis in three informative patients and loss of heterozygosity analysis in five patients revealed identical clonal patterns in the tumors from multiple sites in each patient. We conclude that sporadic gastrinomas at multiple sites are monoclonal and that MEN1 gene alterations in gastrinomas occur before the development of tumor metastases.


Subject(s)
DNA, Neoplasm/genetics , Gastrinoma/genetics , Loss of Heterozygosity , Multiple Endocrine Neoplasia Type 1/genetics , Adult , DNA Mutational Analysis , Dosage Compensation, Genetic , Female , Genetic Markers , Humans , Lymph Nodes , Male , Middle Aged
11.
J Urol ; 163(2): 431-3, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10647647

ABSTRACT

PURPOSE: We describe the earliest renal lesions associated with hereditary papillary renal cancer and estimate the prevalence of microscopic papillary renal tumors. MATERIALS AND METHODS: Grossly normal tissue was obtained from 12 kidneys during renal surgery in 9 patients with hereditary papillary renal cancer. Tissue was examined microscopically and findings were compared to those previously reported to be associated with von Hippel-Lindau disease and sporadic renal cell carcinoma. RESULTS: A total of 92 microscopic papillary renal cell carcinoma lesions were identified on 46 of 88 slides (53%). No other lesions were identified. All tumors were solid and displayed the basophilic papillary histology characteristic of hereditary papillary renal cancer. Extrapolation of the data predicted the prevalence of 1,100 to 3,400 microscopic papillary tumors in a single kidney in a patient with hereditary papillary renal cancer. CONCLUSIONS: The basophilic papillary histology characteristic of clinically apparent renal tumors in patients with hereditary papillary renal cancer also characterizes the multiple microscopic lesions seen in the kidneys. These findings suggest that the earliest renal tumor in patients with an activating hereditary mutation of the met gene is papillary basophilic renal cancer. The large number of microscopic tumors in patients with hereditary papillary renal cancer was comparable to or greater than that seen in those with von Hippel-Lindau disease.


Subject(s)
Carcinoma, Papillary/epidemiology , Carcinoma, Papillary/pathology , Kidney Neoplasms/epidemiology , Kidney Neoplasms/pathology , Adult , Aged , Carcinoma, Papillary/genetics , Female , Humans , Kidney Neoplasms/genetics , Male , Middle Aged , Prevalence
12.
J Clin Endocrinol Metab ; 85(1): 116-23, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10634374

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) gene mutations are reported in some gastrinomas occurring in patients without MEN1 as well as in some other pancreatic endocrine tumors (PETs). In some inherited syndromes phenotype-genotype correlations exist for disease severity, location, or other manifestations. The purpose of the present study was to correlate mutations of the MEN1 gene in a large cohort of patients with sporadic gastrinomas to disease activity, tumor location, extent, and growth pattern. DNA was extracted from frozen gastrinomas from 51 patients and screened by dideoxyfinger-printing (ddF) for abnormalities in the 9 coding exons and adjacent splice junctions of the MEN1 gene. Tumor DNA exhibiting abnormal ddF patterns was sequenced for mutations. The findings were correlated with clinical manifestations of the disease, primary tumor site, disease extent, and tumor growth postoperatively. Tumor growth was determined by serial imaging studies. Sixteen different MEN1 gene mutations in the 51 sporadic gastrinomas (31%) were identified (11 truncating, 4 missense, and 1 in-frame deletion). Nine of the 16 mutations were located in exon 2 compared to 7 of 16 in the remaining 8 coding exons (P = 0.005 on a per nucleotide basis). Primary pancreatic or lymph node gastrinomas with a mutation had only exon 2 mutations, whereas duodenal tumors uncommonly harbored exon 2 mutations (P = 0.011). Similarly, small primary tumors (<1 cm) more frequently contained a nonexon 2 mutation (P = 0.02). There was no difference between patients with or without a mutation with respect to clinical characteristics, primary tumor site, disease extent, or proportion of patients disease free after surgery. Postoperative tumor growth tended to be more aggressive in patients with a mutation (P = 0.09). No correlation in the rate of disease-free status or postoperative tumor growth in patients with active disease to the location of the mutation was seen. These results demonstrate that the MEN1 gene is mutated in 31% of sporadic gastrinomas, and mutations are clustered between amino acids 66-166, which differs from patients with familial MEN1, in whom mutations occur throughout the gene. The presence of an MEN1 gene mutation does not correlate with clinical characteristics of patients with gastrinomas, gastrinoma extent, or growth pattern; however, the location of the mutation differed with gastrinoma location. These data suggest that mutations in the MEN1 gene are important in a proportion of sporadic gastrinomas, but the presence or absence of these mutations will not identify the clinically important subgroups with different growth patterns.


Subject(s)
Gastrinoma/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Mutation/genetics , Pancreatic Neoplasms/genetics , Base Sequence , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Exons/genetics , Female , Genotype , Humans , Male , Middle Aged , Molecular Sequence Data , Phenotype , Polymerase Chain Reaction , Zollinger-Ellison Syndrome/genetics
13.
J Clin Endocrinol Metab ; 85(12): 4776-80, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11134142

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN 1) is associated with parathyroid, enteropancreatic, pituitary, and other tumors. The MEN1 gene, a tumor suppressor, is located on chromosome 11. Affected individuals inherit a mutated MEN1 allele, and tumorigenesis in specific tissues follows inactivation of the remaining MEN1 allele. MEN 1-associated endocrine tumors usually become clinically evident in late adolescence or young adulthood, as high levels of PTH, gastrin, or PRL. Because each of these tumors can usually be controlled with medications and/or surgery, MEN 1 has been regarded mainly as a treatable endocrinopathy of adults. Unlike in MEN 2, early testing of children in MEN 1 families is not recommended. We report a 2.3-cm pituitary macroadenoma in a 5-yr-old boy with familial MEN 1. He presented with growth acceleration, acromegaloid features, and hyperprolactinemia. We tested systematically to see whether his pituitary tumor had causes similar to or different from a typical MEN 1 tumor. Germ line DNA of the propositus and his affected relatives revealed a heterozygous point mutation in the MEN1 gene, which leads to a His139Asp (H139D) amino acid substitution. The patient had no other detectable germ-line mutations on either MEN1 allele. DNA sequencing and fluorescent in situ hybridization with a MEN1 genomic DNA sequence probe each demonstrated one copy of the MEN1 gene to be deleted in the pituitary tumor and not in normal DNA, proving MEN1 "second hit" as a tumor cause. Gsalpha mutation, common in nonhereditary GH-producing tumors, was not detected in this tumor. We conclude that this pituitary macroadenoma showed molecular genetic features of a typical MEN 1-associated tumor. This patient represents the earliest presentation of any morbid endocrine tumor in MEN 1. A better understanding of early onset MEN 1 disease is needed to formulate recommendations for early MEN 1 genetic testing.


Subject(s)
Adenoma/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Pituitary Neoplasms/genetics , Adenoma/pathology , Alleles , Child, Preschool , DNA/genetics , DNA/isolation & purification , DNA Mutational Analysis , Gene Deletion , Humans , In Situ Hybridization, Fluorescence , Male , Multiple Endocrine Neoplasia Type 1/pathology , Pituitary Neoplasms/pathology , Point Mutation/genetics
14.
Am J Surg Pathol ; 23(12): 1527-31, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10584706

ABSTRACT

Mucolipidosis type IV (ML-IV) is an autosomal recessive lysosomal storage disease that causes severe neurologic abnormalities. The brain disease is characterized by pigmented cytoplasmic granules in neurons and accumulation of lamellated membrane structures in lysosomes. The gastrointestinal disease in ML-IV was not previously recognized. Clinical examination of 20 patients with ML-IV (age range, 2-23 years) at the National Institutes of Health showed hypergastrinemia and constitutive achlorhydria. Endoscopic biopsy specimens from the gastric fundus, body, and antrum and from the duodenum of four such patients (ages 4, 6, 7, and 22 years) were evaluated histologically and by electron microscopy. Histologically, all gastric fundus and body biopsy specimens showed parietal cells in normal numbers. However, a striking cytoplasmic vacuolization of parietal cells was seen on hematoxylin and eosin stain. Electron microscopy showed the parietal cells to be markedly distended by large lysosomes containing lamellar, concentric, and cystic membranous inclusions. Additionally, chronic atrophic gastritis and enterochromaffin-like (ECL) cell hyperplasia were observed. Foveolar and chief cells in stomach and duodenum biopsy specimens were normal. We conclude that the cytoplasmic lysosomal inclusions in gastric parietal cells is a unique histologic feature of gastric biopsy in ML-IV.


Subject(s)
Achlorhydria/etiology , Gastrins/blood , Gastritis, Atrophic/etiology , Inclusion Bodies/ultrastructure , Lysosomes/ultrastructure , Mucolipidoses/complications , Parietal Cells, Gastric/ultrastructure , Achlorhydria/blood , Achlorhydria/pathology , Adult , Biopsy , Child , Child, Preschool , Enterochromaffin-like Cells/pathology , Female , Gastric Fundus/pathology , Gastritis, Atrophic/blood , Gastritis, Atrophic/pathology , Gastroscopy , Humans , Hyperplasia , Male , Mucolipidoses/blood , Mucolipidoses/pathology
15.
Recent Prog Horm Res ; 54: 397-438; discussion 438-9, 1999.
Article in English | MEDLINE | ID: mdl-10548885

ABSTRACT

MEN1 is a syndrome of parathyroid adenomas, gastrinomas, prolactinomas, and other endocrine tumors. Collagenomas and facial angiofibromas are newly recognized but common skin expressions. Many tumors in MEN1 are benign; however, many entero-pancreatic neuroendocrine tumors and foregut carcinoid tumors are malignant. MEN1 is thus the expression of a cancer gene but without available prevention or cure for malignancy. Hereditary (as compared to sporadic) endocrine tumors show early onset age and multiplicity, because each cell of the body has "one hit" by inheritance. Multiple neoplasia syndromes with endocrine tumor(s) all include nonendocrine components; their known defective genes seem mainly to disturb cell accumulation. Hereditary neoplasia/hyperplasia of one endocrine tissue reflects a defect that is tissue selective and directed at cell secretion. Though the hereditary endocrine neoplasias are rare, most of their identified genes also contribute to common sporadic endocrine neoplasms. Hereditary tumors may be caused by activation of an oncogene (e.g., RET) or, more often, by inactivation of a tumor suppressor gene (e.g., P53, MEN1). Recently, MEN1 was identified by positional cloning. This strategy included narrowing the gene candidate interval, identifying many or all genes in that interval, and testing the newly identified candidate genes for mutation in MEN1 cases. MEN1 was identified because it showed mutation in 14 of 15 MEN1 cases. NIH testing showed germline MEN1 mutations in 47 of 50 MEN1 index cases and in seven of eight cases with sporadic MEN1. Despite proven capacity to find germline MEN1 mutation, NIH testing found no MEN1 mutation among five families with isolated hyperparathyroidism, suggesting that this often arises from mutation of other gene(s). Analogous studies in Japan found that familial isolated pituitary tumors also did not show MEN1 germline mutation. MEN1 mutation testing can now be considered for cases of MEN1 and its phenocopies and for asymptomatic members of families with known MEN1 mutation. Germline MEN1 testing does not have the urgency of RET testing in MEN2a and 2b, as MEN1 testing does not commonly lead to an important intervention. Somatic MEN1 mutation was found in sporadic tumors: parathyroid adenoma (21%), gastrinoma (33%), insulinoma (17%), and bronchial carcinoid (36%). For each of these, MEN1 was the known gene most frequently mutated. MEN1 has a widely expressed mRNA that encodes a protein (menin) of 610 amino acids. The protein sequence is not informative about domains or functions. The protein was mainly nuclear. Menin binds to JunD, an AP-1 transcription factor, inhibiting JunD's activation of transcription. Most of the germline and somatic MEN1 mutations predict truncation of menin, a likely destructive change. Inactivating MEN1 mutations in germline and in sporadic neoplasms support prior predictions that MEN1 is a tumor suppressor gene. Germline MEN1 mutation underlies all or most cases of MEN1 (familial or sporadic). Somatic MEN1 mutation is the most common gene mutation in many sporadic endocrine tumor types.


Subject(s)
Multiple Endocrine Neoplasia Type 1/physiopathology , Amino Acid Sequence , Hormones/metabolism , Humans , Molecular Sequence Data , Multiple Endocrine Neoplasia Type 1/epidemiology , Multiple Endocrine Neoplasia Type 1/therapy , Pedigree , Prevalence , Secretory Rate
16.
J Urol ; 162(4): 1246-58, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10492174

ABSTRACT

PURPOSE: We characterize the genetic abnormalities associated with pathological subtypes of renal tumors, which may help diagnosis or prognostication. MATERIALS AND METHODS: A comprehensive literature review of genetic abnormalities associated with different renal tumor subtypes was performed. RESULTS: Studies of sporadic and hereditary forms suggest that abnormalities in the von Hippel-Lindau and met genes are the earliest changes in conventional (clear cell) and papillary basophilic renal cancers, respectively. Renal oncocytoma and chromophobe carcinoma have common genetic abnormalities, suggesting a relationship. A similar finding has been observed between papillary adenoma and papillary basophilic renal cancer. CONCLUSIONS: These findings suggest that molecular diagnostic testing will help determine histopathological diagnosis, identify tumor types with similar genetic abnormalities suggesting a common origin and indicate potential prognostic markers for future study.


Subject(s)
Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Humans , Kidney Neoplasms/classification
17.
Mod Pathol ; 12(9): 919-24, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10496602

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is characterized by the development of endocrine tumors of the parathyroid and pituitary glands, pancreas, and duodenum. Less frequently occurring tumors associated with MEN1 include non-endocrine tumors such as lipomas and angiofibromas. An increased incidence of thyroid neoplasms, leiomyomas, adrenal cortical hyperplasia, hepatic focal nodular hyperplasia, and renal angiomyolipoma has been noted in the MEN1 population. The pathogenesis of non-neuroendocrine tumors in MEN1 is unknown. We report a complex clinical course and a detailed morphologic and genetic analysis of a series of tumors that developed in a patient with MEN1. All tumors were microdissected and analyzed for loss of heterozygosity of the MEN1 gene. A germline mutation of the MEN1 gene was detected, and deletions of the MEN1 gene were consistently detected in multiple neuroendocrine tumors involving the parathyroid glands and the pancreas and a hepatic neuroendocrine tumor metastasis, as predicted by Knudson's "two hit" hypothesis. Two hits of the MEN1 gene were also detected in esophageal leiomyoma tissue, suggesting that tumorigenesis was directly related to the patient's underlying MEN1. In contrast, follicular thyroid adenoma, papillary thyroid carcinoma, hepatic focal nodular hyperplasia, and adrenal cortical hyperplasia consistently showed retained heterozygosity of the MEN1 gene with flanking markers and an intragenic marker. Therefore, these tumors appear to develop along pathogenetic pathways that are different from classical MEN1-associated tumors.


Subject(s)
Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/pathology , Proto-Oncogene Proteins , Adult , Chromosomes, Human, Pair 11/genetics , DNA, Neoplasm/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Humans , Leiomyoma/genetics , Leiomyoma/pathology , Loss of Heterozygosity , Microsatellite Repeats , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Parathyroid Neoplasms/genetics , Parathyroid Neoplasms/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
18.
Am J Pathol ; 155(2): 517-26, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10433944

ABSTRACT

Germline mutations of c-met oncogene at 7q31 have been detected in patients with hereditary papillary renal cell carcinoma. In addition, c-met mutations were shown to play a role in 13% of patients with papillary renal cell carcinoma and no family history of renal tumors. The histopathology of papillary renal cell carcinoma with c-met mutations has not been previously described. We analyzed the histopathology of 103 bilateral archival papillary renal cell carcinomas and 4 metastases in 29 patients from 6 hereditary papillary renal cell carcinoma families with germline c-met mutations and 6 papillary renal cell carcinomas with c-met mutations from 5 patients with no family history of renal tumors. Twenty-five sporadic renal tumors with prominent papillary architecture and without somatic c-met mutations were evaluated for comparison. All papillary renal cell carcinomas with c-met mutations were 75 to 100% papillary/tubulopapillary in architecture and showed chromophil basophilic, papillary renal cell carcinoma type 1 histology. Fuhrman nuclear grade 1-2 was seen in tumors from 23 patients, and nuclear grade 3 was observed focally in 8 patients. Seventeen patients had multiple papillary adenomas and microscopic papillary lesions in the surrounding renal parenchyma. Clear cells with intracytoplasmic lipid and glycogen were focally present in tumors of 94% papillary renal cell carcinoma patients. Clear cells of papillary renal cell carcinoma had small basophilic nuclei, and clear cell areas lacked a fine vascular network characteristic of conventional (clear) cell renal cell carcinoma. We conclude that papillary renal cell carcinoma patients with c-met mutations develop multiple, bilateral, papillary macroscopic and microscopic renal lesions. Renal tumors with c-met genotype show a distinctive papillary renal cell carcinoma type 1 phenotype and are genetically and histologically different from renal tumors seen in other hereditary renal syndromes and most sporadic renal tumors with papillary architecture. Although all hereditary and sporadic papillary renal cell carcinomas with c-met mutations share papillary renal cell carcinoma type 1 histology, not all type 1 sporadic papillary renal cell carcinomas harbor c-met mutations.


Subject(s)
Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Germ-Line Mutation , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Proto-Oncogene Proteins c-met/genetics , Adenoma/genetics , Adenoma/pathology , Adult , Aged , Chromosomes, Human, Pair 7 , Family Health , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Pedigree , Phenotype , Proto-Oncogene Proteins c-met/metabolism , Trisomy
19.
Bone ; 25(1): 119-22, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10423035

ABSTRACT

Multiple endocrine neoplasia type 1 (MENI) is a promising model to understand endocrine and other tumors. Its most common endocrine expressions are tumors of parathyroids, entero-pancreatic neuro-endocrine tissue, and anterior pituitary. Recently, collagenomas and multiple angiofibromas of the dermis also have been recognized as very common. MEN1 can be characterized from different perspectives: (a) as a hormone (parathyroid hormone, gastrin, prolactin, etc.) excess syndrome with excellent therapeutic options; (b) as a syndrome with sometimes lethal outcomes from malignancy of entero-pancreatic neuro-endocrine or foregut carcinoid tissues; or (c) as a disorder than can give insight about cell regulation in the endocrine, the dermal, and perhaps other tissue systems. The MEN1 gene was identified recently by positional cloning, a comprehensive strategy of narrowing the candidate interval and evaluating all or most genes in that interval. This discovery has opened new approaches to basic and clinical issues. Germline MEN1 mutations have been identified in most MEN1 families. Germline MENI mutations were generally not found in families with isolated hyperparathyroidism or with isolated pituitary tumor. Thus, studies with the MENI gene helped establish that mutation of other gene(s) is likely causative of these two MEN1 phenocopies. MEN1 proved to be the gene most frequent L4 mutated in common-variety, nonhereditary parathyroid tumor, gastrinoma, insulinoma, or bronchial carcinoid. For example, in common-variety parathyroid tumors, mutation of several other genes (such as cyclin D1 and P53) has been found, but much less frequently than MEN1 mutation. The majority of germline and somatic MEN1 mutations predicted truncation of the encoded protein (menin). Such inactivating mutations strongly supported prior predictions that MEN1 is a tumor suppressor gene insofar as stepwise mutational inactivation of both copies can release a cell from normal growth suppression. Menin is principally a nuclear protein; menin interacts with junD. Future studies, such as discovery of menin's metabolic pathway, could lead to new opportunities in cell biology and in tumor therapy.


Subject(s)
Endocrine Gland Neoplasms/genetics , Gene Expression Regulation, Neoplastic/physiology , Multiple Endocrine Neoplasia Type 1/genetics , Proto-Oncogene Proteins , Genotype , Germ-Line Mutation , Humans , Neoplasm Proteins/physiology , Pedigree , Phenotype
20.
Arch Ophthalmol ; 117(5): 625-30, 1999 May.
Article in English | MEDLINE | ID: mdl-10326959

ABSTRACT

OBJECTIVE: Retinal angioma frequently occurs in von Hippel-Lindau (VHL) disease. However, VHL gene alterations have not been documented in retinal angiomas. METHODS: Using tissue microdissection and polymerase chain reaction amplification, we have analyzed 7 retinal angiomas associated with VHL disease for loss of heterozygosity of the VHL gene. In addition, vascular endothelial growth factor expression was evaluated in these tumors by immunohistochemistry and in situ hybridization. RESULTS: All 6 informative retinal angiomas showed loss of heterozygosity of the VHL gene. Loss of heterozygosity was detected in vacuolated "stromal" cells, but not in vascular cells or reactive glial tissue. Vascular endothelial growth factor protein and messenger RNA were also present in vacuolated "stromal" cells. CONCLUSIONS: These findings suggest that vacuolated "stromal" cells represent the true neoplastic component in retinal angioma. These cells express vascular endothelial growth factor and therefore may be responsible for abundant neovascularization of retinal angioma.


Subject(s)
Endothelial Growth Factors/genetics , Gene Deletion , Gene Expression , Hemangioma, Capillary/genetics , Ligases , Lymphokines/genetics , Proteins/genetics , Retinal Neoplasms/genetics , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , von Hippel-Lindau Disease/genetics , Adult , Endothelial Growth Factors/metabolism , Hemangioma, Capillary/metabolism , Hemangioma, Capillary/pathology , Humans , Immunoenzyme Techniques , In Situ Hybridization , Loss of Heterozygosity , Lymphokines/metabolism , Male , Polymerase Chain Reaction , Proteins/metabolism , Retinal Neoplasms/metabolism , Retinal Neoplasms/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Von Hippel-Lindau Tumor Suppressor Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...