Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Cancer Discov ; 13(5): 1046-1048, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37139723

ABSTRACT

SUMMARY: In this issue of Cancer Discovery, companion articles from the Prives and Lozano groups describe functional analyses of a common dimeric mutant of p53 found in Li-Fraumeni disease and sporadic cancer: A347D (AD). The authors show that the AD mutant is completely defective for canonical p53 transcriptional function, but interestingly retains some tumor suppressor function, which they show is manifested as "neomorphic" activities in transcription and the control of mitochondrial metabolism. See related article by Gencel-Augusto et al., p. 1230 (7). See related article by Choe et al., p. 1250 (6).


Subject(s)
Li-Fraumeni Syndrome , Humans , Li-Fraumeni Syndrome/metabolism , Tumor Suppressor Protein p53/metabolism , Transcriptional Activation , Cell Death
2.
Cancer Discov ; 13(5): 1250-1273, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37067901

ABSTRACT

Cancer-relevant mutations in the oligomerization domain (OD) of the p53 tumor suppressor protein, unlike those in the DNA binding domain, have not been well elucidated. Here, we characterized the germline OD mutant p53(A347D), which occurs in cancer-prone Li-Fraumeni syndrome (LFS) patients. Unlike wild-type p53, mutant p53(A347D) cannot form tetramers and exists as a hyperstable dimeric protein. Further, p53(A347D) cannot bind or transactivate the majority of canonical p53 target genes. Isogenic cell lines harboring either p53(A347D) or no p53 yield comparable tumorigenic properties, yet p53(A347D) displays remarkable neomorphic activities. Cells bearing p53(A347D) possess a distinct transcriptional profile and undergo metabolic reprogramming. Further, p53(A347D) induces striking mitochondrial network aberration and associates with mitochondria to drive apoptotic cell death upon topoisomerase II inhibition in the absence of transcription. Thus, dimer-forming p53 demonstrates both loss-of-function (LOF) and gain-of-function (GOF) properties compared with the wild-type form of the protein. SIGNIFICANCE: A mutant p53 (A347D), which can only form dimers, is associated with increased cancer susceptibility in LFS individuals. We found that this mutant wields a double-edged sword, driving tumorigenesis through LOF while gaining enhanced apoptogenic activity as a new GOF, thereby yielding a potential vulnerability to select therapeutic approaches. See related commentary by Stieg et al., p. 1046. See related article by Gencel-Augusto et al., p. 1230. This article is highlighted in the In This Issue feature, p. 1027.


Subject(s)
Li-Fraumeni Syndrome , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/pathology , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Apoptosis/genetics , Mitochondria/metabolism
3.
Cancer Discov ; 13(5): 1230-1249, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37067911

ABSTRACT

Cancer-related alterations of the p53 tetramerization domain (TD) abrogate wild-type (WT) p53 function. They result in a protein that preferentially forms monomers or dimers, which are also normal p53 states under basal cellular conditions. However, their physiologic relevance is not well understood. We have established in vivo models for monomeric and dimeric p53, which model Li-Fraumeni syndrome patients with germline p53 TD alterations. p53 monomers are inactive forms of the protein. Unexpectedly, p53 dimers conferred some tumor suppression that is not mediated by canonical WT p53 activities. p53 dimers upregulate the PPAR pathway. These activities are associated with lower prevalence of thymic lymphomas and increased CD8+ T-cell differentiation. Lymphomas derived from dimeric p53 mice show cooperating alterations in the PPAR pathway, further implicating a role for these activities in tumor suppression. Our data reveal novel functions for p53 dimers and support the exploration of PPAR agonists as therapies. SIGNIFICANCE: New mouse models with TP53R342P (monomer) or TP53A347D (dimer) mutations mimic Li-Fraumeni syndrome. Although p53 monomers lack function, p53 dimers conferred noncanonical tumor-suppressive activities. We describe novel activities for p53 dimers facilitated by PPARs and propose these are "basal" p53 activities. See related commentary by Stieg et al., p. 1046. See related article by Choe et al., p. 1250. This article is highlighted in the In This Issue feature, p. 1027.


Subject(s)
Li-Fraumeni Syndrome , Animals , Mice , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/metabolism , Tumor Suppressor Protein p53/metabolism , Transcriptional Activation , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Cell Death
4.
Mod Pathol ; 34(3): 542-548, 2021 03.
Article in English | MEDLINE | ID: mdl-32636452

ABSTRACT

Breast cancer is the most common malignancy in female patients with Li-Fraumeni syndrome (LFS), a rare autosomal dominant hereditary syndrome characterized by germline TP53 mutations. Recent studies have shown that the majority of these tumors are estrogen receptor (ER) positive with frequent HER2 co-expression. However, the morphologic features of these tumors have not been as well studied as other germline-associated breast cancers. We evaluated the pathologic features of 27 invasive and in situ carcinomas from patients with known germline TP53 mutations collected through the Li-Fraumeni Consortium. Overall, 60% of cases were HER2 positive and 44% showed ER co-expression. Most DCIS was high nuclear grade with central necrosis and associated periductal fibrosis and lymphocytic response. Invasive carcinomas were mostly of ductal type (NOS), modified Scarff-Bloom-Richardson (mSBR) high grade, with marked nuclear atypia and high mitotic rate. Prominent tumor infiltrating lymphocytes, syncytial growth pattern, or pushing borders were not seen in these tumors. High p53 IHC expression was seen in tumors from individuals with germline TP53 missense mutations whereas little or no protein expression (<1% nuclear expression, null pattern) was seen in tumors from carriers of non-missense mutations. In this study, we report in detail the morphologic features of invasive and in situ carcinomas in LFS. We found that these tumors share features with cancers harboring somatic TP53 mutations but are distinct from BRCA-associated breast cancers.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Li-Fraumeni Syndrome/pathology , Adult , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Ductal, Breast/genetics , Carcinoma, Intraductal, Noninfiltrating/chemistry , Carcinoma, Intraductal, Noninfiltrating/genetics , Female , Genetic Predisposition to Disease , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/metabolism , Mutation , Neoplasm Invasiveness , Phenotype , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Retrospective Studies , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/genetics
5.
J Vis Exp ; (136)2018 06 13.
Article in English | MEDLINE | ID: mdl-29985349

ABSTRACT

Li-Fraumeni syndrome (LFS) is an autosomal dominant hereditary cancer disorder. Patients with LFS are predisposed to a various type of tumors, including osteosarcoma--one of the most frequent primary non-hematologic malignancies in the childhood and adolescence. Therefore, LFS provides an ideal model to study this malignancy. Taking advantage of iPSC methodologies, LFS-associated osteosarcoma can be successfully modeled by differentiating LFS patient iPSCs to mesenchymal stem cells (MSCs), and then to osteoblasts--the cells of origin for osteosarcomas. These LFS osteoblasts recapitulate oncogenic properties of osteosarcoma, providing an attractive model system for delineating the pathogenesis of osteosarcoma. This manuscript demonstrates a protocol for the generation of iPSCs from LFS patient fibroblasts, differentiation of iPSCs to MSCs, differentiation of MSCs to osteoblasts, and in vivo tumorigenesis using LFS osteoblasts. This iPSC disease model can be extended to identify potential biomarkers or therapeutic targets for LFS-associated osteosarcoma.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Li-Fraumeni Syndrome/genetics , Mesenchymal Stem Cells/metabolism , Osteosarcoma/diagnosis , Adolescent , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/cytology , Li-Fraumeni Syndrome/metabolism , Male , Osteosarcoma/pathology
6.
Trends Mol Med ; 23(8): 737-755, 2017 08.
Article in English | MEDLINE | ID: mdl-28735817

ABSTRACT

Rare hereditary disorders provide unequivocal evidence of the importance of genes in human disease pathogenesis. Familial syndromes that predispose to osteosarcomagenesis are invaluable in understanding the underlying genetics of this malignancy. Recently, patient-derived induced pluripotent stem cells (iPSCs) have been successfully utilized to model Li-Fraumeni syndrome (LFS)-associated bone malignancy, demonstrating that iPSCs can serve as an in vitro disease model to elucidate osteosarcoma etiology. We provide here an overview of osteosarcoma predisposition syndromes and review recently established iPSC disease models for these familial syndromes. Merging molecular information gathered from these models with the current knowledge of osteosarcoma biology will help us to gain a deeper understanding of the pathological mechanisms underlying osteosarcomagenesis and will potentially aid in the development of future patient therapies.


Subject(s)
Bone Neoplasms , Induced Pluripotent Stem Cells , Li-Fraumeni Syndrome , Models, Biological , Osteosarcoma , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/pathology , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology
7.
Cancer Genet ; 211: 9-17, 2017 02.
Article in English | MEDLINE | ID: mdl-28279309

ABSTRACT

Li-Fraumeni syndrome (LFS) is an autosomal dominant disorder where an oncogenic TP53 germline mutation is passed from parent to child. Tumor protein p53 is a key tumor suppressor regulating cell cycle arrest in response to DNA damage. Paradoxically, some mutant TP53 carriers remain unaffected, while their children develop cancer within the first few years of life. To address this paradox, response to UV stress was compared in dermal fibroblasts (dFb) from an affected LFS patient vs. their unaffected carrier parent. UV induction of CDKN1A/p21, a regulatory target of p53, in LFS patient dFb was significantly reduced compared to the unaffected parent. UV exposure also induced significantly greater p53[Ser15]-phosphorylation in LFS patient dFb, a reported property of some mutant p53 variants. Taken together, these results suggested that unaffected parental dFb may express an increased proportion of wild-type vs. mutant p53. Indeed, a significantly increased ratio of wild-type to mutant TP53 allele-specific expression in the unaffected parent dFb was confirmed by RT-PCR-RFLP and RNA-seq analysis. Hence, allele-specific expression of wild-type TP53 may allow an unaffected parent to mount a response to genotoxic stress more characteristic of homozygous wild-type TP53 individuals than their affected offspring, providing protection from the oncogenesis associated with LFS.


Subject(s)
Li-Fraumeni Syndrome/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Alleles , Child , Child, Preschool , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Li-Fraumeni Syndrome/metabolism , Male , Parents
8.
J Clin Invest ; 127(1): 132-136, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27869650

ABSTRACT

Li-Fraumeni syndrome (LFS) is a cancer predisposition disorder caused by germline mutations in TP53 that can lead to increased mitochondrial metabolism in patients. However, the implications of altered mitochondrial function for tumorigenesis in LFS are unclear. Here, we have reported that genetic or pharmacologic disruption of mitochondrial respiration improves cancer-free survival in a mouse model of LFS that expresses mutant p53. Mechanistically, inhibition of mitochondrial function increased autophagy and decreased the aberrant proliferation signaling caused by mutant p53. In a pilot study, LFS patients treated with metformin exhibited decreases in mitochondrial activity concomitant with activation of antiproliferation signaling, thus reproducing the effects of disrupting mitochondrial function observed in LFS mice. These observations indicate that a commonly prescribed diabetic medicine can restrain mitochondrial metabolism and tumorigenesis in an LFS model, supporting its further consideration for cancer prevention in LFS patients.


Subject(s)
Li-Fraumeni Syndrome/prevention & control , Metformin/pharmacology , Mitochondria/metabolism , Neoplasms, Experimental/prevention & control , Oxygen Consumption/drug effects , Adult , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Humans , Jurkat Cells , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/metabolism , Male , Mice , Mice, Mutant Strains , Middle Aged , Mitochondria/genetics , Mitochondria/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Oxygen Consumption/genetics , Pilot Projects , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Lancet Oncol ; 17(9): 1295-305, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27501770

ABSTRACT

BACKGROUND: Carriers of a germline TP53 pathogenic variant have a substantial lifetime risk of developing cancer. In 2011, we did a prospective observational study of members of families who chose to either undergo a comprehensive surveillance protocol for individuals with Li-Fraumeni syndrome or not. We sought to update our assessment of and modify the surveillance protocol, so in this study we report both longer follow-up of these patients and additional patients who underwent surveillance, as well as update the originally presented surveillance protocol. METHODS: A clinical surveillance protocol using physical examination and frequent biochemical and imaging studies (consisting of whole-body MRI, brain MRI, breast MRI, mammography, abdominal and pelvic ultrasound, and colonoscopy) was introduced at three tertiary care centres in Canada and the USA on Jan 1, 2004, for carriers of TP53 pathogenic variants. After confirmation of TP53 mutation, participants either chose to undergo surveillance or chose not to undergo surveillance. Patients could cross over between groups at any time. The primary outcome measure was detection of asymptomatic tumours by surveillance investigations. The secondary outcome measure was 5 year overall survival established from a tumour diagnosed symptomatically (in the non-surveillance group) versus one diagnosed by surveillance. We completed survival analyses using an as-treated approach. FINDINGS: Between Jan 1, 2004, and July 1, 2015, we identified 89 carriers of TP53 pathogenic variants in 39 unrelated families, of whom 40 (45%) agreed to surveillance and 49 (55%) declined surveillance. 19 (21%) patients crossed over from the non-surveillance to the surveillance group, giving a total of 59 (66%) individuals undergoing surveillance for a median of 32 months (IQR 12-87). 40 asymptomatic tumours have been detected in 19 (32%) of 59 patients who underwent surveillance. Two additional cancers were diagnosed between surveillance assessments (false negatives) and two biopsied lesions were non-neoplastic entities on pathological review (false positives). Among the 49 individuals who initially declined surveillance, 61 symptomatic tumours were diagnosed in 43 (88%) patients. 21 (49%) of the 43 individuals not on surveillance who developed cancer were alive compared with 16 (84%) of the 19 individuals undergoing surveillance who developed cancer (p=0·012) after a median follow-up of 46 months (IQR 22-72) for those not on surveillance and 38 months (12-86) for those on surveillance. 5 year overall survival was 88·8% (95% CI 78·7-100) in the surveillance group and 59·6% (47·2-75·2) in the non-surveillance group (p=0·0132). INTERPRETATION: Our findings show that long-term compliance with a comprehensive surveillance protocol for early tumour detection in individuals with pathogenic TP53 variants is feasible and that early tumour detection through surveillance is associated with improved long-term survival. Incorporation of this approach into clinical management of these patients should be considered. FUNDING: Canadian Institutes for Heath Research, Canadian Cancer Society, Terry Fox Research Institute, SickKids Foundation, and Soccer for Hope Foundation.


Subject(s)
Biomarkers, Tumor/metabolism , Germ-Line Mutation/genetics , Li-Fraumeni Syndrome/genetics , Multimodal Imaging/methods , Neoplasms/genetics , Population Surveillance , Tumor Suppressor Protein p53/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Follow-Up Studies , Genetic Predisposition to Disease , Heterozygote , Humans , Infant , Infant, Newborn , Li-Fraumeni Syndrome/diagnostic imaging , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/pathology , Male , Middle Aged , Neoplasm Staging , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Neoplasms/pathology , Prognosis , Prospective Studies , Survival Rate , Young Adult
10.
J Biol Chem ; 291(31): 16011-23, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27467582

ABSTRACT

Li-Fraumeni syndrome (LFS) patients harbor germ line mutations in the TP53 gene and are at increased risk of hormone receptor-positive breast cancers. Recently, elevated levels of aromatase, the rate-limiting enzyme for estrogen biosynthesis, were found in the breast tissue of LFS patients. Although p53 down-regulates aromatase expression, the underlying mechanisms are incompletely understood. In the present study, we found that LFS stromal cells expressed higher levels of Hsp90 ATPase activity and aromatase compared with wild-type stromal cells. Inhibition of Hsp90 ATPase suppressed aromatase expression. Silencing Aha1 (activator of Hsp90 ATPase 1), a co-chaperone of Hsp90 required for its ATPase activity, led to both inhibition of Hsp90 ATPase activity and reduced aromatase expression. In comparison with wild-type stromal cells, increased levels of the Hsp90 client proteins, HIF-1α, and PKM2 were found in LFS stromal cells. A complex comprised of HIF-1α and PKM2 was recruited to the aromatase promoter II in LFS stromal cells. Silencing either HIF-1α or PKM2 suppressed aromatase expression in LFS stromal cells. CP-31398, a p53 rescue compound, suppressed levels of Aha1, Hsp90 ATPase activity, levels of PKM2 and HIF-1α, and aromatase expression in LFS stromal cells. Consistent with these in vitro findings, levels of Hsp90 ATPase activity, Aha1, HIF-1α, PKM2, and aromatase were increased in the mammary glands of p53 null versus wild-type mice. PKM2 and HIF-1α were shown to co-localize in the nucleus of stromal cells of LFS breast tissue. Taken together, our results show that the Aha1-Hsp90-PKM2/HIF-1α axis mediates the induction of aromatase in LFS.


Subject(s)
Adipose Tissue/metabolism , Aromatase/biosynthesis , Breast/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HSP90 Heat-Shock Proteins/metabolism , Li-Fraumeni Syndrome/metabolism , Mammary Glands, Animal/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Thyroid Hormones/metabolism , Adipose Tissue/pathology , Animals , Aromatase/genetics , Breast/pathology , Carrier Proteins/genetics , Cell Line , Female , HSP90 Heat-Shock Proteins/genetics , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/pathology , Mammary Glands, Animal/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neoplasm Proteins/genetics , Stromal Cells/metabolism , Stromal Cells/pathology , Thyroid Hormones/genetics , Thyroid Hormone-Binding Proteins
11.
Proc Natl Acad Sci U S A ; 112(46): E6349-58, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26578795

ABSTRACT

Whereas most mutations in p53 occur in the DNA-binding domain and lead to its functional inactivation, their relevance in the amino-terminal transactivation domain is unclear. We show here that amino-terminal p53 (ATp53) mutations often result in the abrogation of full-length p53 expression, but concomitantly lead to the expression of the amino-terminally truncated p47 isoform. Using genetically modified cancer cells that only express p47, we demonstrate it to be up-regulated in response to various stimuli, and to contribute to cell death, through its ability to selectively activate a group of apoptotic target genes. Target gene selectivity is influenced by K382 acetylation, which depends on the amino terminus, and is required for recruitment of selective cofactors. Consistently, cancers capable of expressing p47 had a better overall survival. Nonetheless, retention of the apoptotic function appears insufficient for tumor suppression, because these mutations are also found in the germ line and lead to Li-Fraumeni syndrome. These data from ATp53 mutations collectively demonstrate that p53's apoptosis proficiency is dispensable for tumor suppression, but could prognosticate better survival.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Li-Fraumeni Syndrome/metabolism , Mutation , Tumor Suppressor Protein p53/biosynthesis , Acetylation , Apoptosis/genetics , Cell Line, Tumor , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/pathology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Structure, Tertiary , Tumor Suppressor Protein p53/genetics
12.
Biomed Res Int ; 2015: 341723, 2015.
Article in English | MEDLINE | ID: mdl-26075229

ABSTRACT

More than one-fifth of ovarian tumors have hereditary susceptibility and, in about 65-85% of these cases, the genetic abnormality is a germline mutation in BRCA genes. Nevertheless, several other suppressor genes and oncogenes have been associated with hereditary ovarian cancers, including the mismatch repair (MMR) genes in Lynch syndrome, the tumor suppressor gene, TP53, in the Li-Fraumeni syndrome, and several other genes involved in the double-strand breaks repair system, such as CHEK2, RAD51, BRIP1, and PALB2. The study of genetic discriminators and deregulated pathways involved in hereditary ovarian syndromes is relevant for the future development of molecular diagnostic strategies and targeted therapeutic approaches. The recent development and implementation of next-generation sequencing technologies have provided the opportunity to simultaneously analyze multiple cancer susceptibility genes, reduce the delay and costs, and optimize the molecular diagnosis of hereditary tumors. Particularly, the identification of mutations in ovarian cancer susceptibility genes in healthy women may result in a more personalized cancer risk management with tailored clinical and radiological surveillance, chemopreventive approaches, and/or prophylactic surgeries. On the other hand, for ovarian cancer patients, the identification of mutations may provide potential targets for biologic agents and guide treatment decision-making.


Subject(s)
BRCA1 Protein , BRCA2 Protein , DNA Mismatch Repair , Genetic Predisposition to Disease , Ovarian Neoplasms , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/therapy , Female , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/pathology , Li-Fraumeni Syndrome/therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy
13.
Proc Natl Acad Sci U S A ; 111(30): 11145-50, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25024203

ABSTRACT

p53(R172H/+) mice inherit a p53 mutation found in Li-Fraumeni syndrome and develop metastatic tumors at much higher frequency than p53(+/-) mice. To explore the mutant p53 metastatic phenotype, we used expression arrays to compare primary osteosarcomas from p53(R172H/+) mice with metastasis to osteosarcomas from p53(+/-) mice lacking metastasis. For this study, 213 genes were differentially expressed with a P value <0.05. Of particular interest, Pla2g16, which encodes a phospholipase that catalyzes phosphatidic acid into lysophosphatidic acid and free fatty acid (both implicated in metastasis), was increased in p53(R172H/+) osteosarcomas. Functional analyses showed that Pla2g16 knockdown decreased migration and invasion in mutant p53-expressing cells, and vice versa: overexpression of Pla2g16 increased the invasion of p53-null cells. Furthermore, Pla2g16 levels were increased upon expression of mutant p53 in both mouse and human osteosarcoma cell lines, indicating that Pla2g16 is a downstream target of the mutant p53 protein. ChIP analysis revealed that several mutant p53 proteins bind the Pla2g16 promoter at E26 transformation-specific (ETS) binding motifs and knockdown of ETS2 suppressed mutant p53 induction of Pla2g16. Thus, our study identifies a phospholipase as a transcriptional target of mutant p53 that is required for metastasis.


Subject(s)
Bone Neoplasms/metabolism , Li-Fraumeni Syndrome/metabolism , Mutation , Osteosarcoma/metabolism , Phospholipases A2, Calcium-Independent/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/pathology , Mice , Mice, Mutant Strains , Neoplasm Invasiveness , Osteosarcoma/genetics , Osteosarcoma/pathology , Phospholipases A2, Calcium-Independent/genetics , Response Elements , Tumor Suppressor Proteins/genetics
14.
Proc Natl Acad Sci U S A ; 111(19): 7006-11, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24778235

ABSTRACT

p53 is a well-known tumor suppressor that is mutated in over 50% of human cancers. These mutations were shown to exhibit gain of oncogenic function compared with the deletion of the gene. Additionally, p53 has fundamental roles in differentiation and development; nevertheless, mutant p53 mice are viable and develop malignant tumors only on adulthood. We set out to reveal the mechanisms by which embryos are protected from mutant p53-induced transformation using ES cells (ESCs) that express a conformational mutant of p53. We found that, despite harboring mutant p53, the ESCs remain pluripotent and benign and have relatively normal karyotype compared with ESCs knocked out for p53. Additionally, using high-content RNA sequencing, we show that p53 is transcriptionally active in response to DNA damage in mutant ESCs and elevates p53 target genes, such as p21 and btg2. We also show that the conformation of mutant p53 protein in ESCs is stabilized to a WT conformation. Through MS-based interactome analyses, we identified a network of proteins, including the CCT complex, USP7, Aurora kinase, Nedd4, and Trim24, that bind mutant p53 and may shift its conformation to a WT form. We propose this conformational shift as a novel mechanism of maintenance of genomic integrity, despite p53 mutation. Harnessing the ability of these protein interactors to transform the oncogenic mutant p53 to the tumor suppressor WT form can be the basis for future development of p53-targeted cancer therapy.


Subject(s)
Cell Transformation, Neoplastic/genetics , Embryonic Stem Cells/cytology , Li-Fraumeni Syndrome/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Adenocarcinoma , Animals , Breast Neoplasms , Cell Line , Cell Line, Tumor , Cell Proliferation , Embryonic Development/genetics , Embryonic Stem Cells/physiology , Homeodomain Proteins/genetics , Humans , Li-Fraumeni Syndrome/metabolism , Loss of Heterozygosity/physiology , Mice , Mice, Knockout , Nanog Homeobox Protein , Protein Conformation , Proteomics , Tumor Suppressor Protein p53/metabolism
15.
Sci Rep ; 4: 4223, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24573247

ABSTRACT

Mutations of p53 cause not only loss of wild-type function but also gain of novel oncogenic functions (GOF). Accumulating evidence suggest that p53 hotspot mutations may confer different types and magnitudes of GOF. Here we add support to the heterogeneity of mutant p53 GOF by showing their unequal association with early tumor onset and spectrum of tumor types. We stratified Li-Fraumeni syndrome (LFS) patients according to carried p53 mutations using data from the updated p53 germline mutation database. When compared to loss-of-function nonsense mutations, the R282 GOF mutation associated with significantly earlier onset, while the G245 mutation displayed later onset. The R175, Y220, R248, R282 and nonsense mutations showed preferential distribution in certain cancer types, which varied in the age of onset. Multivariate COX regression model adjusting for cancer types and patient sex suggested that nonsense and G245 mutations had lower risk than R248 for early onset, suggesting unequal strengths of mutant GOF effects. Our results suggest that Li-Fraumeni syndrome can be subdivided into subtypes linking to unequal GOF effects of p53 mutations. These findings have potential implications in the prevention, early detection and targeted treatment of LFS tumors.


Subject(s)
Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Age of Onset , Databases, Genetic , Female , Germ-Line Mutation , Humans , Li-Fraumeni Syndrome/epidemiology , Male , Proportional Hazards Models
16.
J Cutan Pathol ; 41(3): 303-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24299451

ABSTRACT

Patients with Li-Fraumeni syndrome (LFS) have a germ-line mutation of p53 (TP53) and are predisposed to develop a variety of malignancies at an early age. In this report, we describe an 18-year-old woman with LFS who developed an atypical fibroxanthoma (AFX) on her left arm. This tumor was based in the dermis, sparsely cellular and had ill-defined borders. It was composed predominantly of medium-sized spindled-shaped cells, but many large cells with pleomorphic nuclei were also present. Immunohistochemical stains showed that the tumor cells lacked expression of keratin, S-100 protein, desmin and CD34. Array-based comparative genomic hybridization (aCGH) revealed marked genomic instability with multiple whole chromosome losses, including chromosomes 8, 10, 13 and 22, as well as a partial loss of 17p. This represents one of a few reports of a cutaneous tumor in a patient with LFS and a rare example of an AFX occurring at a young age.


Subject(s)
Biomarkers, Tumor , Fibrosarcoma , Gene Expression Regulation, Neoplastic , Li-Fraumeni Syndrome , Tumor Suppressor Protein p53 , Adolescent , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Female , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/pathology , Mutation , Skin Neoplasms , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Cancer Discov ; 3(11): 1238-44, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23950206

ABSTRACT

UNLABELLED: Genomic characterization of recurrent breast and lung tumors developed over the course of 10 years in a 29-year-old patient with a germline TP53 mutation (Li-Fraumeni Syndrome) identified oncogenic alterations in the HER2 and EGFR genes across all tumors, including HER2 amplifications, an EGFR-exon 20 insertion, and the first-in-humans HER2V659E mutation showing a phenotypic convergent evolution toward HER2 and EGFR alterations. Following the identification of HER2-activating events in the most recent lung carcinoma and in circulating tumor cells, we treated the reminiscent metastatic lesions with a lapatinib-based therapy. A symptomatic and radiologic clinical response was achieved. HER2V659E sensitivity to lapatinib was confirmed in the laboratory. SIGNIFICANCE: The precise knowledge of the genomic alterations present in tumors is critical to selecting the optimal treatment for each patient. Here, we report the molecular characterization and clinical response to a lapatinib-based therapy for the tumors of a Li-Fraumeni patient showing prevalence of HER2 and EGFR genomic alterations.


Subject(s)
Li-Fraumeni Syndrome/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Receptor, ErbB-2/genetics , Adult , Exome , Female , Humans , Lapatinib , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/pathology , Mutation , Neoplasm Metastasis , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured
18.
N Engl J Med ; 368(11): 1027-32, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23484829

ABSTRACT

There is growing evidence that alterations in metabolism may contribute to tumorigenesis. Here, we report on members of families with the Li-Fraumeni syndrome who carry germline mutations in TP53, the gene encoding the tumor-suppressor protein p53. As compared with family members who are not carriers and with healthy volunteers, family members with these mutations have increased oxidative phosphorylation of skeletal muscle. Basic experimental studies of tissue samples from patients with the Li-Fraumeni syndrome and a mouse model of the syndrome support this in vivo finding of increased mitochondrial function. These results suggest that p53 regulates bioenergetic homeostasis in humans. (Funded by the National Heart, Lung, and Blood Institute and the National Institutes of Health; ClinicalTrials.gov number, NCT00406445.).


Subject(s)
Energy Metabolism/genetics , Exercise/physiology , Genes, p53 , Li-Fraumeni Syndrome/metabolism , Mitochondria, Muscle/metabolism , Phosphocreatine/metabolism , Animals , Case-Control Studies , Disease Models, Animal , Female , Germ-Line Mutation , Heterozygote , Humans , Li-Fraumeni Syndrome/genetics , Male , Mice , Muscle, Skeletal/metabolism , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Pilot Projects , Weight Lifting/physiology
19.
Mol Carcinog ; 52(10): 770-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-22495821

ABSTRACT

Most p53 mutations identified in Li-Fraumeni syndrome (LFS) are missense mutations; splicing mutations have rarely been reported. A novel splicing p53 mutation was identified in a patient with Li-Fraumeni-like syndrome (LFL). Usually, p53 missense mutants identified in LFS and cancer cells function as dominant negative mutations interfering with wild-type p53 function. However, the mechanism by which p53 haploinsufficiency causes carcinogenesis is not well characterized. In this study, we describe a novel splicing mutation that results in the loss-of-function of p53. These findings suggest a linkage between the loss-of-function type p53 mutation and a LFL phenotype.


Subject(s)
Breast Neoplasms/genetics , Germ-Line Mutation/genetics , Li-Fraumeni Syndrome/genetics , Osteosarcoma/genetics , RNA Splicing/genetics , Tumor Suppressor Protein p53/genetics , Adolescent , Animals , Blotting, Western , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation , Cell Proliferation , Cells, Cultured , DNA Primers , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Predisposition to Disease , Humans , Immunoenzyme Techniques , Immunoprecipitation , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/pathology , Luciferases/metabolism , Mice , Mice, Knockout , Osteosarcoma/metabolism , Osteosarcoma/pathology , Peptide Fragments , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
20.
Cancer Biol Ther ; 14(1): 29-38, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23114650

ABSTRACT

Li-Fraumeni syndrome (LFS) is primarily characterized by development of tumors exhibiting germ-line mutations in the p53 gene. Cell lines developed from patients of a LFS family have decreased p53 activity as evidenced by the absence of apoptosis upon etoposide treatment. To test our hypothesis that changes in gene expression beyond p53 per se are contributing to the development of tumors, we compared gene expression in non-cancerous skin fibroblasts of LFS-affected (p53 heterozygous) vs. non-affected (p53 wild-type homozygous) family members. Expression analysis showed that several genes were differentially regulated in the p53 homozygous and heterozygous cell lines. We were particularly intrigued by the decreased expression (~88%) of a putative tumor-suppressor protein, caveolin-1 (Cav-1), in the p53-mutant cells. Decreased expression of Cav-1 was also seen in both p53-knockout and p21-knockout HTC116 cells suggesting that p53 controls Cav-1 expression through p21 and leading to the speculation that p53, Cav-1 and p21 may be part of a positive auto-regulatory feedback loop. The direct relationship between p53 and Cav-1 was also tested with HeLa cells (containing inactive p53), which expressed a significantly lower Cav-1 protein. A panel of nonfunctional and p53-deficient colon and epithelial breast cancer cell lines showed undetectable expression of Cav-1 supporting the role of p53 in the control of Cav-1. However, in two aggressively metastasizing breast cancer cell lines, Cav-1 was strongly expressed suggesting a possible role in tumor metastasis. Thus, there is a divergent control of Cav-1 expression as evidenced in non-cancerous Li-Fraumeni syndrome and some aggressive human cancer cell lines.


Subject(s)
Caveolin 1/genetics , Gene Expression Regulation, Neoplastic , Li-Fraumeni Syndrome/metabolism , Caveolin 1/metabolism , Cell Cycle , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation , Feedback, Physiological , Female , Gene Expression , Humans , Li-Fraumeni Syndrome/genetics , Male , Oligonucleotide Array Sequence Analysis , Transcriptome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...