Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Clin Transl Oncol ; 22(7): 978-988, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31691207

ABSTRACT

Clinical variability is commonly seen in Li-Fraumeni syndrome. Phenotypic heterogeneity is present among different families affected by the same pathogenic variant in TP53 gene and among members of the same family. However, causes of this huge clinical spectrum have not been studied in depth. TP53 type mutation, polymorphic variants in TP53 gene or in TP53-related genes, copy number variations in particular regions, and/or epigenetic deregulation of TP53 expression might be responsible for clinical heterogeneity. In this review, recent advances in the understanding of genetic and epigenetic aspects influencing Li-Fraumeni phenotype are discussed.


Subject(s)
Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/physiopathology , Tumor Suppressor Protein p53/genetics , Anticipation, Genetic , DNA Copy Number Variations , Epigenesis, Genetic , Gene-Environment Interaction , Humans , Mutation , Oxidative Stress , Phenotype , Polymorphism, Genetic , Proto-Oncogene Proteins c-mdm2/genetics , Telomere/metabolism
3.
Cell Death Differ ; 20(7): 898-909, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23538418

ABSTRACT

Mutant p53 proteins not only lose their tumor-suppressor function but some acquire oncogenic gain of function (GOF). The published mutp53 knock-in (KI) alleles (R172H, R270H, R248W) manifest GOF by broader tumor spectrum and more metastasis compared with the p53-null allele, but do not shorten survival. However, whether GOF also occurs with other mutations and whether they are all biologically equal is unknown. To answer this, we created novel humanized mutp53 KI mice harboring the hot spot alleles R248Q and G245S. Intriguingly, their impact was very different. Compared with p53-null mice, R248Q/- mice had accelerated onset of all tumor types and shorter survival, thus unprecedented strong GOF. In contrast, G245S/- mice were similar to null mice in tumor latency and survival. This was associated with a twofold higher T-lymphoma proliferation in R248Q/- mice compared with G245S/- and null mice. Moreover, R248Q/- hematopoietic and mesenchymal stem cells were expanded relative to G245S/- and null mice, the first indication that GOF also acts by perturbing pretumorous progenitor pools. Importantly, these models closely mirror Li-Fraumeni patients who show higher tumor numbers, accelerated onset and shorter tumor-free survival by 10.5 years when harboring codon R248Q mutations as compared with Li-Fraumeni patients with codon G245S mutations or p53 deletions/loss. Conversely, both KI alleles caused a modest broadening of tumor spectrum with enhanced Akt signaling compared with null mice. These models are the first in vivo proof for differential oncogenic strength among p53 GOF alleles, with genotype-phenotype correlations borne out in humans.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology , Adult , Alleles , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/pathology , Li-Fraumeni Syndrome/physiopathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Tumor Suppressor Protein p53/deficiency
4.
Cell ; 148(1-2): 59-71, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22265402

ABSTRACT

Genomic rearrangements are thought to occur progressively during tumor development. Recent findings, however, suggest an alternative mechanism, involving massive chromosome rearrangements in a one-step catastrophic event termed chromothripsis. We report the whole-genome sequencing-based analysis of a Sonic-Hedgehog medulloblastoma (SHH-MB) brain tumor from a patient with a germline TP53 mutation (Li-Fraumeni syndrome), uncovering massive, complex chromosome rearrangements. Integrating TP53 status with microarray and deep sequencing-based DNA rearrangement data in additional patients reveals a striking association between TP53 mutation and chromothripsis in SHH-MBs. Analysis of additional tumor entities substantiates a link between TP53 mutation and chromothripsis, and indicates a context-specific role for p53 in catastrophic DNA rearrangements. Among these, we observed a strong association between somatic TP53 mutations and chromothripsis in acute myeloid leukemia. These findings connect p53 status and chromothripsis in specific tumor types, providing a genetic basis for understanding particularly aggressive subtypes of cancer.


Subject(s)
Brain Neoplasms/genetics , Gene Rearrangement , Medulloblastoma/genetics , Tumor Suppressor Protein p53/genetics , Animals , Child , Chromosome Aberrations , DNA Copy Number Variations , DNA Mutational Analysis , Disease Models, Animal , Humans , Leukemia, Myeloid, Acute/genetics , Li-Fraumeni Syndrome/physiopathology , Mice , Middle Aged
5.
Clin J Oncol Nurs ; 15(1): 79-87, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21278043

ABSTRACT

Li-Fraumeni syndrome (LFS) is an inherited cancer syndrome that affects a small percentage of the population worldwide. LFS is characterized by multiple cancers in affected family members and is devastating to all diagnosed patients and their relatives. A link has been identified between LFS and mutations in the tumor-suppressor gene that encodes for the P53 protein, and much research has been done on the effect of this mutation in tumorigenesis. However, the natural history of the disease has no definitive pathway, and additional research is under way. LFS is rare, can present in many ways, requires complex management, and has tumors that often first present in childhood. Therefore, pediatric nurse practitioners should be aware of LFS as a potential differential diagnosis in patients with multiple tumors, certain rare tumors, or most importantly, a family history notable for multiple early-onset cancers.


Subject(s)
Li-Fraumeni Syndrome/nursing , Nurse Practitioners , Nurse's Role , Pediatrics , Child , Genes, Tumor Suppressor , Genes, p53 , Germ-Line Mutation , Humans , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/physiopathology
6.
J Child Neurol ; 24(12): 1526-35, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19955345

ABSTRACT

The majority of tumors of the nervous system are sporadic. However, a subset of patients with tumors and their families are predisposed to developing cancers of the central nervous system and other organs because of a germline mutation. In the last decade, many of the genes responsible for these typically autosomal dominant familial tumor syndromes have been identified. Additionally, our understanding of the mechanisms of carcinogenesis in these syndromes has increased, allowing for more targeted therapies for these patients as well as those with sporadic cancers. Because these patients present a unique set of issues regarding diagnosis and neurooncological management, the most common familial cancer syndromes involving the nervous system are reviewed: neurofibromatosis type 1 and 2; tuberous sclerosis complex; von Hippel Lindau, Li-Fraumeni, Gorlin, and Turcot syndrome.


Subject(s)
Genetic Predisposition to Disease/genetics , Germ-Line Mutation/genetics , Inheritance Patterns/genetics , Neoplasms, Nerve Tissue/genetics , Basal Cell Nevus Syndrome/genetics , Basal Cell Nevus Syndrome/pathology , Basal Cell Nevus Syndrome/physiopathology , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/pathology , Li-Fraumeni Syndrome/physiopathology , Neurofibromatoses/genetics , Neurofibromatoses/pathology , Neurofibromatoses/physiopathology , Tuberous Sclerosis/genetics , Tuberous Sclerosis/pathology , Tuberous Sclerosis/physiopathology , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/pathology , von Hippel-Lindau Disease/physiopathology
7.
Curr Neurol Neurosci Rep ; 7(3): 200-7, 2007 May.
Article in English | MEDLINE | ID: mdl-17488585

ABSTRACT

Hereditary central nervous tumor syndromes are a varied group of conditions that include neurofibromatosis type 1 and 2, tuberous sclerosis, Von Hippel-Lindau disease, and Cowden, Turcot, and Gorlin syndromes. The responsible genes have been identified in most of these disorders. These genes typically act as tumor suppressor genes, maintain normal cellular function and homeostasis, and regulate cell growth and differentiation. Familial central nervous system tumors are mostly inherited as autosomal dominant traits and involve germline mutations. Neoplastic development occurs when a somatic mutation inactivates the second allele. These patients also present unique challenges for their management. This review highlights the clinical manifestations, molecular genetics, pathophysiology, and current treatment options of these disorders with a focus on neuro-oncologic manifestations of the diseases.


Subject(s)
Central Nervous System Neoplasms , Neoplastic Syndromes, Hereditary , Basal Cell Nevus Syndrome/genetics , Basal Cell Nevus Syndrome/pathology , Basal Cell Nevus Syndrome/physiopathology , Basal Cell Nevus Syndrome/therapy , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/physiopathology , Central Nervous System Neoplasms/therapy , Diagnosis, Differential , Hamartoma Syndrome, Multiple/genetics , Hamartoma Syndrome, Multiple/pathology , Hamartoma Syndrome, Multiple/physiopathology , Hamartoma Syndrome, Multiple/therapy , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/pathology , Li-Fraumeni Syndrome/physiopathology , Li-Fraumeni Syndrome/therapy , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Neoplastic Syndromes, Hereditary/physiopathology , Neoplastic Syndromes, Hereditary/therapy , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Neurofibromatosis 1/physiopathology , Neurofibromatosis 1/therapy , Neurofibromatosis 2/genetics , Neurofibromatosis 2/pathology , Neurofibromatosis 2/physiopathology , Neurofibromatosis 2/therapy , Tuberous Sclerosis/genetics , Tuberous Sclerosis/pathology , Tuberous Sclerosis/physiopathology , Tuberous Sclerosis/therapy , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/pathology , von Hippel-Lindau Disease/physiopathology , von Hippel-Lindau Disease/therapy
9.
J Interferon Cytokine Res ; 26(1): 14-26, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16426144

ABSTRACT

DNA hypermethylation in gene promoters is an epigenetic mechanism regulating gene expression in cellular immortalization, an important step in carcinogenesis. Previously, we studied the genes dysregulated during immortalization using spontaneously immortalized fibroblasts from patients with Li-Fraumeni syndrome (LFS), who carry a germline mutation in the tumor suppressor gene p53. We found that multiple interferon (IFN) signaling pathway genes were regulated by epigenetic silencing. In this study we focused on a key regulator of that pathway, the signal transducer and transcription activator 1 (Stat1) gene. Although Stat1 is downregulated after cellular immortalization and upregulated in immortal MDAH041 cells after 5-aza-2'-deoxycytidine (5-aza-dC) treatment, we detected no methylation of the Stat1 promoter region in these cells before or after immortalization. To analyze the function of Stat1 in immortalization, we expressed Stat1 in immortal MDAH041 cells by stable infection, expecting to induce IFN-regulated genes or cellular senescence or both. However, the overexpression of Stat1 alone was not sufficient to repress the proliferation rate of immortal MDAH041 cells or induce senescence in immortal MDAH041 cells. We concluded that factor(s) additional to Stat1 (whether IFN dependent or not) are required for the immortalization of LFS fibroblasts.


Subject(s)
Cell Transformation, Neoplastic , Cellular Senescence/physiology , Gene Expression Regulation, Neoplastic , Interferons/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction/physiology , Animals , Cell Line , DNA Methylation , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/physiopathology , Promoter Regions, Genetic , STAT1 Transcription Factor/genetics , Sequence Analysis, DNA
10.
Methods Mol Biol ; 222: 117-29, 2003.
Article in English | MEDLINE | ID: mdl-12710683

ABSTRACT

Germline TP53 mutations are responsible for the large majority of classic LFS families, and a smaller proportion of LFL families. In some of the families shown to have no germline TP53 mutation, germline hChk2 mutations have been described. In some cases the functional consequences of the latter have been demonstrated, although there are still relatively few reports of such mutations. Due to the paucity of families currently described with hChk2 mutations, it is not possible to reach any conclusions concerning the phenotypic/clinical differences between the two types of germline mutation. At least one family with a germline hChk2 mutation is a classic LFS family, whereas others are LFL, variant-LFS, or phenotypically suggestive of LFS. However, there is still a significant number of LFS/LFL families for which no underlying genetic determinant has been identified. It will be fascinating to see what genetic defects are responsible, and whether they involve additional components of DNA damage recognition, repair, or cell cycle checkpoint pathways.


Subject(s)
Li-Fraumeni Syndrome/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases , Tumor Suppressor Protein p53/genetics , Checkpoint Kinase 2 , Gene Frequency , Humans , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/physiopathology , Mutation , Tumor Suppressor Protein p53/metabolism
11.
Oncogene ; 16(25): 3291-8, 1998 Jun 25.
Article in English | MEDLINE | ID: mdl-9681828

ABSTRACT

We report an extensive Li-Fraumeni-like family in which there is an unusual spectrum of tumours at relatively late onset. A germline TP53 splice donor mutation in exon 4 is present in all affected family members available for testing. The mutation abolishes correct splicing of intron 4 and techniques of RT-PCR have identified three different aberrant transcripts from the mutant TP53 allele. Using the yeast functional assay to analyse transcripts in cells from a number of family members with the mutant allele, TP53 appears wild-type. Functional studies have been carried out on cells from patients with and without cancer who carry the germline mutation, and on cells from unaffected individuals from the same family who do not carry the mutation. Using a number of functional endpoints known to distinguish between cells carrying mutant or wild-type TP53 alleles, we were unable to discriminate normal (wt/wt) from heterozygous (wt/mut) cells by lymphocyte apoptosis and fibroblast survival following low dose rate ionising radiation exposure. However germline mutation carriers show increased sensitivity to radiation-induced chromosome damage in the G2 phase of the cell cycle, and decreased transient and permanent G1 arrest. These studies demonstrate the importance of fully characterising the effects of TP53 germline mutations, and may explain some of the phenotypic features of this family.


Subject(s)
Alternative Splicing , Germ-Line Mutation/genetics , Li-Fraumeni Syndrome/genetics , Tumor Suppressor Protein p53/genetics , Adult , Apoptosis/genetics , Apoptosis/physiology , Family Health , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Germ-Line Mutation/physiology , Humans , Li-Fraumeni Syndrome/physiopathology , Lymphocytes/cytology , Lymphocytes/metabolism , Male , Pedigree , Point Mutation/genetics , Point Mutation/physiology , Tumor Suppressor Protein p53/physiology , Yeasts/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...