Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
AJNR Am J Neuroradiol ; 40(5): 872-877, 2019 05.
Article in English | MEDLINE | ID: mdl-30948375

ABSTRACT

BACKGROUND AND PURPOSE: Atypical teratoid/rhabdoid tumors are rare, aggressive central nervous system tumors that are predominantly encountered in very young children. Our aim was to determine whether in vivo metabolic profiles correlate with molecular features of central nervous system pediatric atypical teratoid/rhabdoid tumors. MATERIALS AND METHODS: Twenty confirmed patients with atypical teratoid/rhabdoid tumors who underwent MR spectroscopy were included in this study. In vivo metabolite levels of atypical teratoid/rhabdoid tumors were compared with molecular subtypes assessed by achaete-scute homolog 1 expression. Additionally, brain-specific creatine kinase levels were determined in tissue samples. RESULTS: In vivo creatine concentrations were higher in tumors that demonstrated achaete-scute homolog 1 expression compared with those without achaete-scute homolog 1 expression (3.42 ± 1.1 versus 1.8 ± 0.8 IU, P < .01). Additionally, levels of myo-inositol (mI) (9.0 ± 1.5 versus 4.7 ± 3.6 IU, P < .05) were significantly different, whereas lipids approached significance (44 ± 20 versus 80 ± 30 IU, P = .07) in these 2 cohorts. Higher brain-specific creatine kinase levels were observed in the cohort with achaete-scute homolog 1 expression (P < .05). Pearson correlation analysis showed a significant positive correlation of brain-specific creatine kinase with absolute creatine (P < .05) and myo-inositol (P < .05) concentrations. CONCLUSIONS: In vivo MR spectroscopy may predict key molecular features of atypical teratoid/rhabdoid tumors at initial diagnosis, leading to timely patient risk stratification and accelerating the development of targeted therapies.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/metabolism , Magnetic Resonance Spectroscopy/methods , Rhabdoid Tumor/metabolism , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Male , Neuroimaging/methods , Retrospective Studies , Rhabdoid Tumor/diagnostic imaging , Rhabdoid Tumor/pathology , Teratoma/diagnostic imaging , Teratoma/metabolism , Teratoma/pathology
2.
Clin Neuropathol ; 30(5): 235-41, 2011.
Article in English | MEDLINE | ID: mdl-21955927

ABSTRACT

Two unrelated female infants presented at 9 days and 2 months, respectively, with apneic episodes in the former and gaze preference in the latter. MRI revealed enlargement of almost the entire right hemisphere, apparently smooth cortex, simplification of the gyral pattern, and expanded white matter with abnormal signal intensity containing multiple intraparenchymal cysts. Histologic examination of both cases revealed white matter infiltration by a hypocellular lesion composed of uniform, fibrillary astrocytes in a microcystic background. Multilocular tumor cysts were prominent, but Rosenthal fibers and eosinophilic granular bodies were absent. Very rare mitoses were seen in the absence of necrosis or vascular change. There was no convincing cortical infiltration, but the subpial zone was diffusely expanded by a band of astrocytes set in a dense fibrillar feltwork which opened out into numerous cystic spaces. No desmoplastic changes or associated atypical ganglion cells were identified. There was no evidence for a BRAFKIAA1549 fusion or BRAF mutation in one case tested. In conclusion, both lesions are not desmoplastic infantile astrocytoma/ganglioglioma, fibrillary astrocytoma, or typical for pilocytic astrocytoma. Such extreme subpial spread with cysts is most unusual and may suggest a novel variant of infantile astrocytoma.


Subject(s)
Astrocytoma/pathology , Brain Neoplasms/pathology , Cysts/pathology , Astrocytoma/metabolism , Brain Neoplasms/metabolism , Cysts/metabolism , Female , Humans , Immunohistochemistry , Infant, Newborn
3.
Leukemia ; 23(11): 2109-17, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19626050

ABSTRACT

Xenotransplantation of human acute myeloid leukemia (AML) in immunocompromised animals has been critical for defining leukemic stem cells. However, existing immunodeficient strains of mice have short life spans and low levels of AML cell engraftment, hindering long-term evaluation of primary human AML biology. A recent study suggested that NOD/LtSz-scid IL2Rgammac null (NSG) mice have enhanced AML cell engraftment, but this relied on technically challenging neonatal injections. Here, we performed extensive analysis of AML engraftment in adult NSG mice using tail vein injection. Of the 35 AML samples analyzed, 66% showed bone marrow engraftment over 0.1%. Further, 37% showed high levels of engraftment (>10%), with some as high as 95%. A 2-44-fold expansion of AML cells was often seen. Secondary and tertiary recipients showed consistent engraftment, with most showing further AML cell expansion. Engraftment did not correlate with French-American-British subtype or cytogenetic abnormalities. However, samples with FLT3 mutations showed a higher probability of engraftment than FLT3 wild type. Importantly, animals developed organomegaly and a wasting illness consistent with advanced leukemia. We conclude that the NSG xenotransplantation model is a robust model for human AML cell engraftment, which will allow better characterization of AML biology and testing of new therapies.


Subject(s)
Disease Models, Animal , Leukemia, Myeloid, Acute/pathology , Mice, Inbred NOD , Neoplasm Transplantation/methods , Transplantation, Heterologous/methods , Animals , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/physiopathology , Mice , Mice, SCID , Point Mutation , Primary Myelofibrosis/pathology , Receptors, Interleukin-2/genetics , Severity of Illness Index , T-Lymphocytes, Cytotoxic/pathology , fms-Like Tyrosine Kinase 3/genetics
4.
Clin Neuropathol ; 28(1): 1-10, 2009.
Article in English | MEDLINE | ID: mdl-19216214

ABSTRACT

OBJECTIVE: Atypical teratoid/rhabdoid tumors are aggressive neoplasms of the central nervous system occurring mainly in the early childhood and rarely in adults. We described a case of this tumor in an 18-year-old male patient without previous medical history. MATERIAL AND METHODS: The neoplasm was localized in the right frontotemporal area of the brain and was totally excised. The specimen was fixed in formalin and embedded in paraffin. The histological and immunohistochemical features of the neoplasm were assessed, while sequencing analysis as well as interphase fluorescence in situ hybridization (FISH) were performed. RESULTS: Histological and immunohistochemical analysis demonstrated atypical rhabdoid cells strongly and diffusely positive for EMA and Vimentin as well as focally immunoreactive for SMA and GFAP. Additionally, though no abnormalities detected in the coding sequence of the INI1 gene, interphase FISH studies were consistent with a homozygous deletion of the INI1 gene in the majority of examined nuclei. INI1 immunostaining demonstrated diffuse loss of nuclear INI1 expression in tumor cells. Taken together, the results were consistent with a diagnosis of atypical teratoid/rhabdoid tumor (ATRT). CONCLUSIONS: 26 previous cases of ATRT have been reported in adults, thus far. To our knowledge, this is the eighth case of an ATRT reported in an adult patient having genetic confirmation and the first one in which the tumor is, partly, localized in the right temporal area of the brain. This unusual presentation underlines the necessity of considering this devastating neoplasm in the differential diagnosis of malignant brain tumors of young adults.


Subject(s)
Brain Neoplasms/pathology , Rhabdoid Tumor/pathology , Teratoma/pathology , Adolescent , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Male , Rhabdoid Tumor/genetics , Rhabdoid Tumor/metabolism , SMARCB1 Protein , Teratoma/genetics , Teratoma/metabolism , Transcription Factors/genetics
5.
J Med Genet ; 46(1): 68-72, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19124645

ABSTRACT

BACKGROUND: The role of germline and somatic SMARCB1 gene mutations in malignant rhabdoid tumour (MRT) predisposition is well known. Germline SMARCB1 mutations have also recently been identified in a subset of individuals with schwannomatosis. Surprisingly, MRT predisposition and schwannomatosis have never been reported to co-occur in a family. The correlation between genotype and phenotype for mutations in SMARCB1 has not been determined. RESULTS: We have identified a germline 2631 bp duplication that includes exon 6 of SMARCB1 in a unique family with a four generation history of MRT predisposition and schwannomatosis. This duplication segregates with disease in individuals affected with both conditions, linking MRT predisposition and schwannomatosis as components of the same syndrome in this family. CONCLUSION: The unique combination of tumours that result from the duplication described in this report may provide important clues about the mechanisms that influence the phenotype associated with a given SMARCB1 mutation.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Gene Duplication , Neurilemmoma/genetics , Rhabdoid Tumor/genetics , Transcription Factors/genetics , Base Sequence , Exons , Family , Genotype , Germ-Line Mutation , Humans , Molecular Sequence Data , Neurilemmoma/pathology , Pedigree , Phenotype , Rhabdoid Tumor/pathology , SMARCB1 Protein
6.
Hum Mutat ; 29(3): 433-40, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18033723

ABSTRACT

The presence of chromosome-specific low-copy repeats (LCRs) predisposes chromosome 22 to deletions and duplications. The current diagnostic procedure for detecting aberrations at 22q11.2 is chromosomal analysis coupled with fluorescence in situ hybridization (FISH) or PCR-based multiplex ligation dependent probe amplification (MLPA). However, there are copy number variations (CNVs) in 22q11.2 that are only detected by high-resolution platforms such as array comparative genomic hybridization (aCGH). We report on development of a high-definition MLPA (MLPA-HD) 22q11 kit that detects copy number changes at 37 loci on the long arm of chromosome 22. These include the 3-Mb region commonly deleted in DiGeorge/velocardiofacial syndrome (DGS/VCFS), the cat eye syndrome (CES) region, and more distal regions in 22q11 that have recently been shown to be deleted. We have used this MLPA-HD probe set to analyze 363 previously well-characterized samples with a variety of different rearrangements at 22q11 and demonstrate that it can detect copy number alterations with high sensitivity and specificity. In addition to detection of the common recurrent deletions associated with DGS/VCFS, variant and novel chromosome 22 aberrations have been detected. These include duplications within as well as deletions distal to this region. Further, the MLPA-HD detects deletion endpoint differences between patients with the common 3-Mb deletion. The MLPA-HD kit is proposed as a cost effective alternative to the currently available detection methods for individuals with features of the 22q11 aberrations. In patients with the relevant phenotypic characteristics, this MLPA-HD probe set could replace FISH for the clinical diagnosis of 22q11.2 deletions and duplications.


Subject(s)
Chromosomes, Human, Pair 22/genetics , Molecular Probe Techniques , Chromosome Aberrations , Chromosome Deletion , Coloboma/genetics , Craniofacial Abnormalities/genetics , DiGeorge Syndrome/genetics , Gene Dosage , Genetic Variation , Humans , Nucleic Acid Amplification Techniques/methods , Rhabdoid Tumor/genetics
7.
AJNR Am J Neuroradiol ; 27(5): 962-71, 2006 May.
Article in English | MEDLINE | ID: mdl-16687525

ABSTRACT

BACKGROUND AND PURPOSE: Primary atypical teratoid/rhabdoid tumors (AT/RTs) are rare malignant intracranial neoplasms, usually occurring in young children. The objectives of this study were to characterize the MR imaging features and locations of primary intracranial AT/RTs, to determine the frequency of disseminated disease in the central nervous system (CNS) at diagnosis and postoperatively, and to assess patient outcomes. METHODS: The preoperative cranial MR images of 13 patients with AT/RTs were retrospectively reviewed for evaluation of lesion location, size, MR signal intensity and enhancement characteristics, and the presence of disseminated intracranial tumor. Postoperative MR images of the head and spine for 17 patients were reviewed for the presence of locally recurrent or residual tumor and disseminated neoplasm. Imaging data were correlated with patient outcomes. RESULTS: Patients ranged in age from 4 months to 15 years (median age, 2.9 years). Primary AT/RTs were intra-axial in 94% of patients. The single primary extra-axial lesion was located in the cerebellopontine angle cistern. AT/RTs were infratentorial in 47%, supratentorial in 41%, and both infra- and supratentorial in 12%. A germ-line mutation of the hSNF5/INI1 tumor-suppressor gene was responsible for the simultaneous occurrence of an intracranial AT/RT and a malignant renal rhabdoid tumor in a 4-month-old patient. Mean tumor sizes were 3.6 x 3.8 x 3.9 cm. On short TR images, AT/RTs typically had heterogeneous intermediate signal intensity, as well as zones of low (54%), high (8%), or both low and high (31%) signal intensity from cystic and/or necrotic regions, hemorrhage, or both, respectively. On long TR/long TE images, solid portions of AT/RTs typically had heterogeneous intermediate-to-slightly-high signal intensity with additional zones of high (54%) or both high and low signal intensity (38%), secondary to cystic and/or necrotic regions, edema, prior hemorrhage, and/or calcifications. AT/RT had isointense and/or slightly hyperintense signal intensity relative to gray matter on fluid-attenuated inversion-recovery (FLAIR) and long TR/long TE images, and showed restricted diffusion. All except 1 AT/RT showed contrast enhancement. The fraction of tumor volume showing enhancement was greater than two thirds in 58%, between one third and two thirds in 33%, and less than one third in 9%. Disseminated tumor in the leptomeninges was seen with MR imaging in 24% of patients at diagnosis/initial staging and occurred in another 35% from 4 months to 2.8 years (mean, 1.1 years) after surgery and earlier imaging examinations with negative findings. The overall 1-year and 5-year survival probabilities were 71% and 28%, respectively. Patients with MR imaging evidence of disseminated leptomeningeal tumor had a median survival rate of 16 months compared with 149 months for those without disseminated tumor (P < .004, logrank test). CONCLUSION: AT/RTs are typically intra-axial lesions, which can be infra- and/or supratentorial. The unenhanced and enhanced MR imaging features of AT/RT are often variable secondary to cystic/necrotic changes, hemorrhage, and/or calcifications. Poor prognosis is associated with MR imaging evidence of disseminated leptomeningeal tumor.


Subject(s)
Brain Neoplasms/diagnosis , Magnetic Resonance Imaging , Rhabdoid Tumor/diagnosis , Teratoma/diagnosis , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male , Retrospective Studies
8.
Mol Ther ; 4(6): 586-92, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11735343

ABSTRACT

A potential consequence of systemic administration of viral vectors is the inadvertent introduction of foreign DNA into recipient germ cells. To evaluate the safety of in vivo recombinant adeno-associated virus (rAAV) mediated gene transfer approaches for hemophilia B, we explored the risk of germline transmission of vector sequences following intramuscular (IM) injection of rAAV in four species of male animals (mouse, rat, rabbit and dog). In vector biodistribution studies in mice and rats, there is a dose-dependent increase in the likelihood that vector sequences can be detected in gonadal DNA using a sensitive PCR technique. However, in dogs DNA extracted from semen is negative for vector sequences. To address this discrepancy, studies were done in rabbits, and both semen and testicular DNAs were analyzed for the presence of vector sequences. These studies showed that no AAV vector sequences were detected in DNA extracted from rabbit semen samples collected at time points ranging from 7 to 90 days following IM injection of 1 x 10(13) vector genomes rAAV (vg) per kg. In contrast, DNA extracted from gonadal tissue was positive for vector sequences, but the positive signals diminished in number and strength with time. By FISH analysis, AAV signals were localized to the testis basement membrane and the interstitial space; no intracellular signal was observed. We observed similar findings following hepatic artery administration of rAAV in rats and dogs, suggesting that our findings are independent of the route of administration of vector. Attempts to transduce isolated murine spermatogonia directly with AAV-lacZ were unsuccessful. In clinical studies human subjects injected IM with an AAV vector at doses up to 2 x 10(12) vg/kg have shown no evidence of vector sequences in semen. Together, these studies suggest that rAAV introduced into skeletal muscle or the hepatic artery does not transduce male germ cells efficiently. We conclude that the risk of inadvertent germline transmission of vector sequences following IM or hepatic artery injection of AAV-2 vectors is extremely low.


Subject(s)
Dependovirus/genetics , Hemophilia B/genetics , Muscle, Skeletal/metabolism , Spermatozoa/virology , Animals , DNA Primers/chemistry , DNA, Viral/analysis , Dogs , Factor IX/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Hemophilia B/pathology , Hemophilia B/therapy , In Situ Hybridization, Fluorescence , Injections, Intramuscular , Male , Mice , Polymerase Chain Reaction , Rabbits , Rats , Recombinant Proteins/genetics , Semen/virology , Testis/virology
9.
Hum Pathol ; 32(8): 884-6, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11521235

ABSTRACT

We report a primary uterine sarcoma with classic histologic, immunohistochemical, and ultrastructural features of a malignant extrarenal rhabdoid tumor (MERT). It arose in a 71-year-old woman who presented with postmenopausal bleeding, ascites, and a right pelvic mass. Malignant cells with rhabdoid morphology were identified by cytologic examination of the peritoneal fluid. Exploratory laparotomy revealed a 10-cm right adnexal mass and disseminated peritoneal tumor. Pathologic study showed diffuse expansion of the endometrial stroma by rhabdoid-like cells with transmural infiltration of the myometrium and extensive involvement of uterine serosa and right ovary by tumor. Neoplastic cells were immunoreactive for vimentin, cytokeratin, and epithelial membrane antigen, and cytoplasmic whorls of intermediate filaments were observed by electron microscopy. Fluorescence in situ hybridization (FISH) studies with chromosome 22-specific probes showed no loss of the INI1 gene, and no coding sequence mutation was identified.


Subject(s)
Ascitic Fluid/diagnosis , DNA-Binding Proteins/genetics , Rhabdoid Tumor/diagnosis , Sarcoma/diagnosis , Uterine Neoplasms/diagnosis , Aged , Antigens, Neoplasm/analysis , Biomarkers, Tumor/analysis , Chromosomal Proteins, Non-Histone , Cytoplasm/ultrastructure , DNA-Binding Proteins/analysis , Diagnosis, Differential , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Intermediate Filaments/ultrastructure , Mutation , Rhabdoid Tumor/chemistry , Rhabdoid Tumor/genetics , SMARCB1 Protein , Sarcoma/chemistry , Sarcoma/genetics , Transcription Factors , Uterine Neoplasms/chemistry , Uterine Neoplasms/genetics
10.
Med Pediatr Oncol ; 36(1): 37-41, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11464901

ABSTRACT

BACKGROUND: Several lines of evidence es tablish that chromosome band 1p36 is frequently deleted in neuroblastoma primary tumors and cell lines, suggesting that a tumor suppressor gene within this region is involved in the development of this tumor. PROCEDURE: We analyzed the status of 1p36 in primary neuroblastomas and cell lines to define the region of consistent rearrangement. RESULTS: Loss of heterozygosity (LOH) studies of primary neuro blastomas identified allelic loss in 135 of 503 tumors (27%), with the smallest region of overlap (SRO) defined distal to D15214 (1p36.3). No homozygous deletions were detected at 120 loci mapping to 1p36.1-p36.3 in a panel of 46 neuroblastoma cell lines. A recently identified patient with neuroblastoma was found to have a constitutional deletion within 1p36.2-p36.3, and this deletion, when combined with the LOH results, defined a smaller SRO of one megabase within 1p36.3. We constructed a comprehensive integrated map of chromosome 1 containing 11,000 markers and large-insert clones, a high-resolution radiation hybrid (RH) map of 1p36, and a P1-artificial chromosome (PAC) contig spanning the SRO, to further characterize the region of interest. Over 768 kb (75%) of the SRO has been sequenced to completion. Further analysis of distal 1p identified 113 transcripts localizing to 1p36, 21 of which were mapped within the SRO. CONCLUSION: This analysis will identify suitable positional candidate transcripts for mutational screening and subsequent identification of the 1p36.3 neuroblastoma suppressor gene.


Subject(s)
Chromosomes, Human, Pair 1/genetics , Neuroblastoma/genetics , Alleles , Chromosome Deletion , Chromosome Mapping , Chromosomes, Human, Pair 1/ultrastructure , Female , Genes, Tumor Suppressor , Genotype , Humans , In Situ Hybridization, Fluorescence , Infant , Loss of Heterozygosity , Microsatellite Repeats , Neuroblastoma/mortality , Neuroblastoma/pathology , Transcription, Genetic
11.
Clin Cancer Res ; 6(7): 2759-63, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10914721

ABSTRACT

Germ-line and somatic mutations of the hSNF5/INI1 gene have been reported in atypical teratoid/rhabdoid tumors (AT/RTs) of the brain, consistent with its role as a tumor suppressor gene. In the present study, we determined the frequency of deletions and mutations of INI1 in 52 children whose original diagnosis was medulloblastoma (MB) or primitive neuroectodermal tumor (PNET) of the central nervous system. Mutations were detected in DNA isolated from four tumors, all from children less than 3 years of age at diagnosis. Two of the four were reviewed and reclassified as atypical teratoid tumor, whereas there was insufficient material to establish this diagnosis in the two remaining cases. The relatively low frequency of mutations, even in a large series of infants, suggests that loss of sequences from chromosome 22 and/or mutations of INI1 do not account for the poor prognosis of children with MB or PNET who are less than 3 years of age at diagnosis. Nevertheless, chromosome 22 deletion and INI1-mutation analysis of infants with MB/PNET should be considered for all children who are less than 1 year of age. Detection of these mutations suggests that the child has an AT/RT, rather than a MB/PNET, a finding with important prognostic value.


Subject(s)
Brain Neoplasms/genetics , Chromosomes, Human, Pair 22 , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Medulloblastoma/genetics , Mutation , Neuroectodermal Tumors, Primitive/genetics , Adolescent , Adult , Amino Acid Sequence , Base Sequence , Brain Neoplasms/surgery , Child , Child, Preschool , Chromosomal Proteins, Non-Histone , Chromosome Mapping , DNA-Binding Proteins/chemistry , Frameshift Mutation , Humans , Infant , Infant, Newborn , Karyotyping , Loss of Heterozygosity , Medulloblastoma/surgery , Monosomy , Neuroectodermal Tumors, Primitive/surgery , SMARCB1 Protein , Sequence Deletion , Transcription Factors
12.
J Clin Oncol ; 18(5): 1027-35, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10694553

ABSTRACT

PURPOSE: To identify biologic prognostic factors in childhood primitive neuroectodermal tumors (PNET), including medulloblastoma, that accurately define patient groups with sufficiently good prognosis to permit a reduction in treatment intensity. PATIENTS AND METHODS: We determined expression levels of the neurotrophin receptor TrkC mRNA in formalin-fixed tumor samples from 87 well characterized PNET patients using in situ hybridization. Comparison of TrkC mRNA expression levels with clinical and other laboratory variables was performed using univariate and multivariate Cox regression analysis. RESULTS: High TrkC mRNA expression was found to be associated more with higher 5-year cumulative survival rate than was low TrkC mRNA expression (89% v 46%, respectively). When compared with established clinical prognostic factors and laboratory variables of potential prognostic significance, TrkC mRNA expression, by univariate analysis, was found to be the single most powerful predictor of outcome (hazards ratio, 4.81; P <.00005), exceeding all clinical prognostic factors. In multivariate analysis, the hazards ratio remained significant (P <.00005). CONCLUSION: High TrkC mRNA expression in PNET is a powerful independent predictor of favorable clinical outcome. Assessment of TrkC mRNA levels may aid in treatment planning for patients with PNETs and should be incorporated prospectively into PNET clinical trials.


Subject(s)
Biomarkers, Tumor/biosynthesis , Brain Neoplasms/metabolism , Neuroectodermal Tumors, Primitive/metabolism , Receptor, trkC/biosynthesis , Adolescent , Adult , Age Factors , Antigens, Differentiation/analysis , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Child , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 17 , Female , Humans , Immunohistochemistry , Infant , Male , Neuroectodermal Tumors, Primitive/diagnosis , Neuroectodermal Tumors, Primitive/genetics , Neuroectodermal Tumors, Primitive/mortality , Prognosis , RNA, Messenger/biosynthesis , Sex Factors , Survival Analysis
13.
Genes Chromosomes Cancer ; 28(1): 31-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10738300

ABSTRACT

We describe a four-month-old child who presented with an atypical teratoid/rhabdoid tumor of the brain and subsequently developed a renal rhabdoid tumor. Distinct histologic features, immunophenotypic profiles, and deletions of chromosome 22 were supportive of two primary tumors. An identical mutation in exon 7 of the INI1 rhabdoid tumor suppressor gene was identified in both tumors, as well as in normal kidney tissue. We propose that this germline INI1 mutation predisposed the child to the development of both malignancies. These findings lend support to the hypothesis that rhabdoid tumors in all sites have a common genetic etiology.


Subject(s)
Central Nervous System Neoplasms/genetics , DNA-Binding Proteins/genetics , Germ-Line Mutation/genetics , Kidney Neoplasms/genetics , Rhabdoid Tumor/genetics , Teratoma/genetics , Central Nervous System Neoplasms/pathology , Chromosomal Proteins, Non-Histone , DNA Mutational Analysis , Fatal Outcome , Humans , Immunohistochemistry , Infant , Karyotyping , Kidney Neoplasms/pathology , Loss of Heterozygosity , Male , Rhabdoid Tumor/pathology , SMARCB1 Protein , Teratoma/pathology , Transcription Factors
14.
Gene ; 241(1): 133-41, 2000 Jan 04.
Article in English | MEDLINE | ID: mdl-10607907

ABSTRACT

We employed exon trapping and large-scale genomic sequence analysis of two bacterial artificial chromosome clones to isolate genes from the region between the IGLC and BCR in chromosome 22q11.2. At the time these studies were initiated, one previously identified gene, GNAZ, was known to map to this region. Two genes, RTDR1 and RAB36, were cloned from this portion of 22q11, which is heterozygously or homozygously deleted in pediatric rhabdoid tumors of the brain, kidney and soft tissues. RTDR1 is a novel gene with a slight homology to a yeast vacuolar protein. RAB36 is a member of the Rab family of proteins. A series of primary rhabdoid tumors with chromosome 22q11 deletions were screened for mutations in the coding sequences of RTDR1, GNAZ and RAB36, but did not demonstrate any disease-specific alterations. Recently, INI1, which maps to the distal portion of the deletion region in 22q11, was identified as the candidate rhabdoid tumor suppressor gene. Further studies of RTDR1 and RAB36 are required to determine whether their absence contributes to the progression of rhabdoid tumors. Alternatively, these genes may be candidates for other diseases that map to human chromosome 22.


Subject(s)
Chromosomes, Human, Pair 22 , Gene Deletion , Rhabdoid Tumor/genetics , rab GTP-Binding Proteins/genetics , Amino Acid Sequence , Base Sequence , Contig Mapping , Exons , Gene Expression , Humans , Models, Genetic , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
15.
Genes Chromosomes Cancer ; 27(1): 44-51, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10564585

ABSTRACT

Inactivation of the PTCH tumor suppressor gene occurs in a subset of sporadic medulloblastomas, suggesting that alterations in the PTCH pathway may be important in the development of this tumor. In order to address the frequency of genetic alterations affecting genes in this pathway, we used a combination of loss of heterozygosity (LOH) analysis, single-stranded conformational polymorphism (SSCP) analysis, and direct sequencing of DNA samples from sporadic primitive neuroectodermal tumors (PNETs). To identify alterations in the PTCH gene, we performed LOH analysis on 37 tumor DNA samples. Of those with matched constitutional DNA samples, one demonstrated LOH. Of those without matched constitutional DNA, six were homozygous with all markers. All exons of the PTCH gene were sequenced in these seven tumors, and three mutations were found. To identify alterations in the SHH and SMO genes, we analyzed all exons of both genes in 24 tumors with SSCP and sequenced any exons that showed aberrant band patterns. No mutations were found in either SHH or SMO in any tumor. We also identified the following genes as candidate tumor suppressors based on their roles in controlling hh/ptc signaling in Drosophila: EN-1 and EN-2, deletion of which results in a lack of cerebellar development in mice; SMAD family members 1-7, and protein kinase A subunits RIalpha, RIbeta, RIIbeta, Calpha, and Cbeta. Each of these genes was investigated in a panel of 24 matched constitutional and tumor DNA samples. Our search revealed no mutations in any of these genes. Thus, PTCH is the only gene in this complex pathway that is mutated with notable frequency in PNET. Genes Chromosomes Cancer 27:44-51, 2000.


Subject(s)
Cerebellar Neoplasms/genetics , Drosophila Proteins , Genes, Tumor Suppressor , Medulloblastoma/genetics , Membrane Proteins/genetics , Proteins/genetics , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled , Trans-Activators , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 18/genetics , Chromosomes, Human, Pair 7/genetics , DNA, Neoplasm/genetics , Exons/genetics , Genetic Markers , Hedgehog Proteins , Humans , Loss of Heterozygosity , Microsatellite Repeats , Patched Receptors , Patched-1 Receptor , Polymorphism, Single-Stranded Conformational , Smoothened Receptor
16.
J Cell Sci ; 112 Pt 18: 3039-47, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10462520

ABSTRACT

Basonuclin is a zinc finger protein mainly expressed in keratinocytes of the basal layer of epidermis and the outer root sheath of hair follicles. It is also found in abundance in the germ cells of testis and ovary. In cultured keratinocytes, basonuclin is associated with chromatin in all phases of the cell cycle, including mitosis. By immunocytochemical methods, we demonstrate here that in mitosis basonuclin is associated with the short arms of the acrocentric chromosomes and with other loci on many metaphase chromosomes of human keratinocytes. Using the evolutionarily highly conserved N-terminal pair of zinc fingers in an electrophoresis mobility shift assay, we demonstrate that the DNA target sequences of basonuclin on the acrocentric chromosomes are likely to be within the promoter region of the 45S rRNA gene transcription unit. DNase I footprinting shows that basonuclin zinc fingers interact with the upstream control element of this promoter, which is necessary for the high level of transcription of the rRNA genes. This result suggests that basonuclin may be a tissue-specific transcription factor for the ribosomal RNA genes.


Subject(s)
Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Keratinocytes/metabolism , Proteins/metabolism , RNA, Ribosomal/genetics , Base Sequence , Binding Sites/genetics , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , DNA Footprinting , DNA Primers/genetics , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , DNA-Binding Proteins , Deoxyribonuclease I , Female , Humans , In Situ Hybridization, Fluorescence , Male , Mitosis , Phosphoproteins , Promoter Regions, Genetic , Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Fingers/genetics
17.
Am J Surg Pathol ; 23(3): 249-56, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10078913

ABSTRACT

The clinical, pathologic, and immunohistochemical features of a widely disseminated tumor with rhabdoid phenotype are described in nine infants < or = 3 months of age. Five neonates had tumor evident at birth, two of which had placental metastases. The average survival following diagnosis was < 6 weeks. None of the infants had an apparent primary tumor in either the kidney or brain. In four cases, the dominant mass involved the head and neck region, and in two cases, the primary mass was paraspinal. The histologic features were those of a high-grade, round cell neoplasm with abundant cytoplasm and containing cells with cytoplasmic filamentous inclusions. Immunohistochemical studies revealed polyphenotypic antigen expression. Genetic information was available from eight of nine cases. Karyotype analysis revealed abnormalities of chromosome band 22q11-12 in three of six tumors. Fluorescence in situ hybridization studies or molecular studies demonstrated 22q11.2 deletions in all five cases with available frozen tissue, two of which had translocations involving 22q by karyotype analysis. The similar clinical and pathologic findings in these rapidly fatal tumors in infants and the demonstration of abnormalities of chromosome 22q11 in a majority of the cases supports their histogenetic and nosologic relationship to the family of malignant rhabdoid tumors that typically occur in young children in several anatomic sites, including kidney, soft tissues, liver, and brain. Like neuroblastoma and rhabdomyosarcoma, malignant rhabdoid tumor can appear as disseminated disease at birth or shortly thereafter.


Subject(s)
Chromosomes, Human, Pair 22/genetics , Rhabdoid Tumor/congenital , Rhabdoid Tumor/genetics , Cytoskeleton/ultrastructure , Female , Gene Deletion , Gestational Age , Head and Neck Neoplasms/congenital , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/ultrastructure , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Karyotyping , Male , Prognosis , Rhabdoid Tumor/pathology , Rhabdoid Tumor/ultrastructure
18.
Cancer Res ; 59(1): 74-9, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9892189

ABSTRACT

We examined 18 atypical teratoid and rhabdoid tumors of the brain and 7 renal and 4 extrarenal rhabdoid tumors for mutations in the candidate rhabdoid tumor suppressor gene, INI1. Fifteen tumors had homozygous deletions of one or more exons of the INI1 gene, and the other 14 tumors demonstrated mutations. Germ-line mutations of INI1 were identified in four children, one with an atypical teratoid tumor of the brain and three with renal rhabdoid tumors. These studies suggest that INI1 is a tumor suppressor gene involved in rhabdoid tumors of the brain, kidney, and other extrarenal sites.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Germ-Line Mutation , Rhabdoid Tumor/genetics , Teratoma/genetics , Child , Child, Preschool , Chromosomal Proteins, Non-Histone , Female , Humans , Infant , Infant, Newborn , Male , Mutation , SMARCB1 Protein , Transcription Factors
19.
Neuro Oncol ; 1(2): 139-51, 1999 04.
Article in English | MEDLINE | ID: mdl-11550309

ABSTRACT

Considerable progress has been made toward improving survival for children with brain tumors, and yet there is still relatively little known regarding the molecular genetic events that contribute to tumor initiation or progression. Nonrandom patterns of chromosomal deletions in several types of childhood brain tumors suggest that the loss or inactivation of tumor suppressor genes are critical events in tumorigenesis. Deletions of chromosomal regions 10q, 11 and 17p, and example, are frequent events in medulloblastoma, whereas loss of a region within 22q11.2, which contains the INI1 gene, is involved in the development of atypical teratoid and rhabdoid tumors. A review of the cytogenetic and molecular genetic changes identified to date in childhood brain tumors will be presented.


Subject(s)
Brain Neoplasms/genetics , Astrocytoma/genetics , Child , Child, Preschool , Choroid Plexus Neoplasms/genetics , Chromosome Aberrations , Chromosomes, Human/genetics , Chromosomes, Human/ultrastructure , Ependymoma/genetics , Gene Amplification , Germinoma/genetics , Humans , Infant , Karyotyping , Medulloblastoma/genetics , Monosomy , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplastic Syndromes, Hereditary/genetics , Neuroectodermal Tumors, Primitive/genetics , Nucleic Acid Hybridization , Rhabdoid Tumor/genetics
20.
Cytogenet Cell Genet ; 81(1): 60-4, 1998.
Article in English | MEDLINE | ID: mdl-9691177

ABSTRACT

Several human malignancies frequently exhibit deletions or rearrangements of the distal short arm of chromosome 1 (1p36), and a number of genetic diseases also map to this region. The carbonic anhydrase (CA6) and alpha-enolase (ENO1) genes, previously mapped to 1p36, were physically linked in yeast- and P1-artificial chromosome (YAC and PAC) contigs. PACs from the contig were mapped to 1p36.2 by fluorescence in situ hybridization. The ESTs D1S2068, D1S274E, D1S3275, and stSG4370 were also placed in the same contig. The physical map was integrated with the genetic map of chromosome 1 by assignment of genetic markers D1S160, D1S1615, and D1S503 to the contig. Sequencing of the EST clone representing D1S274E indicated that it was derived from the same transcript as D1S2068E and corresponded to the SLC2A5 (GLUT5) gene, previously assigned to 1p31. Reassignment of SLC2A5 to 1p36.2 was confirmed by somatic cell and radiation hybrid mapping panels and was consistent with previous EST mapping data. Sequencing of the EST clone for D1S274E revealed the presence of intronic sequences, suggesting that the clone was derived from an unprocessed message. The presence of unprocessed and/or alternatively spliced EST clones has potential ramifications for EST-based genomic projects. This information should facilitate the mapping of tumor suppressor and genetic disease loci that have been localized to this region.


Subject(s)
Carbonic Anhydrases/genetics , Chromosomes, Human, Pair 1/genetics , Genes/genetics , Monosaccharide Transport Proteins/genetics , Phosphopyruvate Hydratase/genetics , Chromosome Mapping , Chromosomes, Artificial, Yeast , Gene Library , Genetic Markers/genetics , Glucose Transporter Type 5 , Humans , Hybrid Cells/cytology , Hybrid Cells/metabolism , Sequence Tagged Sites
SELECTION OF CITATIONS
SEARCH DETAIL
...