201
|
Doxey BW, Kuwada SK, Burt RW. Inherited polyposis syndromes: molecular mechanisms, clinicopathology, and genetic testing. Clin Gastroenterol Hepatol 2005; 3:633-41. [PMID: 16206494 DOI: 10.1016/s1542-3565(05)00370-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The inherited polyposis syndromes are a group of conditions in which multiple gastrointestinal polyps occur in the lumen of the gastrointestinal tract, most exhibit an increased risk of colon cancer. Benign and malignant extraintestinal tumors might also be observed. Recent elucidation of the underlying gene mutations has contributed to our understanding of the cell biology and molecular mechanisms associated with gastrointestinal tumorigenesis. Developments have also allowed genetic testing to become an integral component in accurate diagnosis, categorization, and management of inherited polyposis syndromes. In this review, we will focus on familial adenomatous polyposis, mutY human homologue-associated polyposis, Peutz-Jeghers syndrome, juvenile polyposis, and Cowden syndrome. It is essential that both physician and patient understand the benefits and limitations of genetic testing before submission of samples to the laboratory. There are many issues accompanying molecular diagnosis of cancer syndromes, and genetic counseling is an essential prelude to genetic testing.
Collapse
Affiliation(s)
- Brett W Doxey
- Division of Gastroenterology, University of Utah Health Sciences Center, USA
| | | | | |
Collapse
|
202
|
Luo Y, Shoemaker AR, Liu X, Woods KW, Thomas SA, de Jong R, Han EK, Li T, Stoll VS, Powlas JA, Oleksijew A, Mitten MJ, Shi Y, Guan R, McGonigal TP, Klinghofer V, Johnson EF, Leverson JD, Bouska JJ, Mamo M, Smith RA, Gramling-Evans EE, Zinker BA, Mika AK, Nguyen PT, Oltersdorf T, Rosenberg SH, Li Q, Giranda VL. Potent and selective inhibitors of Akt kinases slow the progress of tumors in vivo. Mol Cancer Ther 2005; 4:977-86. [PMID: 15956255 DOI: 10.1158/1535-7163.mct-05-0005] [Citation(s) in RCA: 187] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The Akt kinases are central nodes in signal transduction pathways that are important for cellular transformation and tumor progression. We report the development of a series of potent and selective indazole-pyridine based Akt inhibitors. These compounds, exemplified by A-443654 (K(i) = 160 pmol/L versus Akt1), inhibit Akt-dependent signal transduction in cells and in vivo in a dose-responsive manner. In vivo, the Akt inhibitors slow the progression of tumors when used as monotherapy or in combination with paclitaxel or rapamycin. Tumor growth inhibition was observed during the dosing interval, and the tumors regrew when compound administration was ceased. The therapeutic window for these compounds is narrow. Efficacy is achieved at doses approximately 2-fold lower than the maximally tolerated doses. Consistent with data from knockout animals, the Akt inhibitors induce an increase in insulin secretion. They also induce a reactive increase in Akt phosphorylation. Other toxicities observed, including malaise and weight loss, are consistent with abnormalities in glucose metabolism. These data show that direct Akt inhibition may be useful in cancer therapy, but significant metabolic toxicities are likely dose limiting.
Collapse
Affiliation(s)
- Yan Luo
- Cancer Research, Abbott Laboratories, IL 60064-6117, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Zhang L, Yu Q, He J, Zha X. Study of the PTEN gene expression and FAK phosphorylation in human hepatocarcinoma tissues and cell lines. Mol Cell Biochem 2005; 262:25-33. [PMID: 15532706 DOI: 10.1023/b:mcbi.0000038212.78008.7f] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The tumor suppressor PTEN gene maps to chromosome 10q23.3 and encodes a dual specificity phosphatase. Mutations of this gene had been found in a variety of human tumors. In the present study, we analyzed the structure and expression of the PTEN gene in 34 hepatocellular carcinoma tissues and two hepatoma cell lines. We found neither homozygous nor hemizygous deletions in these samples. We, however, found point mutations in 4 of the 34 tissue samples. Five of ten hepatocellular carcinoma tissues showed reduced PTEN expression at mRNA level. HepG2 and SMMC-7721 hepatoma cells showed decreased PTEN expression at both mRNA and protein levels compared with immortalized L02 hepatic cells. PTEN mRNA in SMMC-7721 hepatoma cells could be reduced by TGF-betaI treatment. We also found that the phosphorylation levels of FAK in both of the hepatoma cell lines were higher than that in L02 hepatic cells. Transient expression of the PTEN gene in SMMC-7721 and HepG2 hepatoma cells resulted in decreased FAK phosphorylation. The level of FAK tyrosine phosphorylation appeared to be inversely correlated with the level of the PTEN protein. In summary, our results indicated that the function of the PTEN gene in hepatocarcinomas may be impaired mainly through point mutations and expression deficiency and that the defect of PTEN in tumor cells could alter the phosphorylation of FAK.
Collapse
Affiliation(s)
- Lineng Zhang
- Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, PR China
| | | | | | | |
Collapse
|
204
|
Cohen MM. Beckwith-Wiedemann syndrome: historical, clinicopathological, and etiopathogenetic perspectives. Pediatr Dev Pathol 2005; 8:287-304. [PMID: 16010495 DOI: 10.1007/s10024-005-1154-9] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2005] [Accepted: 02/07/2005] [Indexed: 01/18/2023]
Abstract
Macroglossia, prenatal or postnatal overgrowth, and abdominal wall defects (omphalocele, umbilical hernia, or diastasis recti) permit early recognition of Beckwith-Wiedemann syndrome. Complications include neonatal hypoglycemia and an increased risk for Wilms tumor, adrenal cortical carcinoma, hepatoblastoma, rhabdomyosarcoma, and neuroblastoma, among others. Perinatal mortality can result from complications of prematurity, pronounced macroglossia, and rarely cardiomyopathy. The molecular basis of Beckwith-Wiedemann syndrome is complex, involving deregulation of imprinted genes found in 2 domains within the 11p15 region: telomeric Domain 1 (IGF2 and H19) and centromeric Domain 2 (KCNQ1, KCNQ1OT1, and CDKN1C). Topics discussed in this article are organized as a series of perspectives: general, historical, epidemiologic, clinical, pathologic, genetic/molecular, diagnostic, and differential diagnostic.
Collapse
Affiliation(s)
- M Michael Cohen
- Department of Pediatrics, Dalhousie Univesity, Halifax, Nova Scotia B3H 3J5, Canada.
| |
Collapse
|
205
|
Hang Y, Zheng YC, Cao Y, Li QS, Sui YJ. Suppression of gastric cancer growth by adenovirus-mediated transfer of the PTEN gene. World J Gastroenterol 2005; 11:2224-9. [PMID: 15818730 PMCID: PMC4305803 DOI: 10.3748/wjg.v11.i15.2224] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the tumor-suppressive effect of the phosphatase and tensin homologue deleted from chromosome (PTEN) in human gastric cancer cells that were wild type for PTEN.
METHODS: Adenoviruses expressing PTEN or luciferase as a control were introduced into gastric cancer cells. The effect of exogenous PTEN gene on the growth and apoptosis of gastric cancer cells that are wtPTEN were examined in vitro and in vivo.
RESULTS: Adenovirus-mediated transfer of PTEN (Ad-PTEN) suppressed cell growth and induced apoptosis significantly in gastric cancer cells (MGC-803, SGC-7901) carrying wtPTEN in comparison with that in normal gastric epithelial cells (GES-1) carrying wtPTEN. This suppression was induced through downregulation of the Akt/PKB pathway, dephosphorylation of focal adhesion kinase and mitogen-activated protein kinase and cell-cycle arrest at the G2/M phase but not at the G1 phase. Furthermore, treatment of human gastric tumor xenografts (MGC-803, SGC-7901) with Ad-PTEN resulted in a significant (P<0.01) suppression of tumor growth.
CONCLUSION: These results indicate a significant tumor-suppressive effect of Ad-PTEN against human gastric cancer cells. Thus, Ad-PTEN may be used as a potential therapeutic strategy for treatment of gastric cancers.
Collapse
Affiliation(s)
- Ying Hang
- Central Laboratory, The Second Hospital of Jilin University, 18 Zi Qiang Street, Chuangchun 130041, Jilin Province, China.
| | | | | | | | | |
Collapse
|
206
|
Di Vizio D, Cito L, Boccia A, Chieffi P, Insabato L, Pettinato G, Motti ML, Schepis F, D'Amico W, Fabiani F, Tavernise B, Venuta S, Fusco A, Viglietto G. Loss of the tumor suppressor gene PTEN marks the transition from intratubular germ cell neoplasias (ITGCN) to invasive germ cell tumors. Oncogene 2005; 24:1882-94. [PMID: 15674339 DOI: 10.1038/sj.onc.1208368] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
PTEN/MMAC1/TEP1: (hereafter PTEN) is a tumor suppressor gene (located at 10q23) that is frequently mutated or deleted in sporadic human tumors. PTEN encodes a multifunctional phosphatase, which negatively regulates cell growth, migration and survival via the phosphatidylinositol 3'-kinase/AKT signalling pathway. Accordingly, Pten+/- mice develop various types of tumors including teratocarcinomas and teratomas. We have investigated PTEN expression in 60 bioptic specimens of germ cell tumors (32 seminomas, 22 embryonal carcinomas and six teratomas) and 22 intratubular germ cell neoplasias (ITGCN) adjacent to the tumors for PTEN protein and mRNA expression. In total, 10 testicular biopsies were used as controls. In the testis, PTEN was abundantly expressed in germ cells whereas it was virtually absent from 56% of seminomas as well as from 86% of embryonal carcinomas and virtually all teratomas. On the contrary, ITGCN intensely expressed PTEN, indicating that loss of PTEN expression is not an early event in testicular tumor development. The loss of PTEN expression occurs mainly at the RNA level as determined by in situ hybridization of cellular mRNA (17/22) but also it may involve some kind of post-transcriptional mechanisms in the remaining 25% of cases. Analysis of microsatellites D10S551, D10S541 and D10S1765 in GCTs (n=22) showed LOH at the PTEN locus at 10q23 in at least 36% of GCTs (three embryonal carcinoma, three seminoma, two teratoma); one seminoma and one embryonal (9%) carcinoma presented an inactivating mutation in the PTEN gene (2/22). Finally, we demonstrated that the phosphatidylinositol 3'-kinase/AKT pathway, which is regulated by the PTEN phosphatase, is crucial in regulating the proliferation of the NT2/D1 embryonal carcinoma cells, and that the cyclin-dependent kinase inhibitor p27(kip1) is a key downstream target of this pathway.
Collapse
Affiliation(s)
- Dolores Di Vizio
- Dipartimento di Scienze Biomorfologiche e Funzionali, Facoltà di Medicina e Chirurgia, Università di Napoli Federico II, via S. Pansini, 5, 80131 Napoli, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
207
|
Inoki K, Ouyang H, Li Y, Guan KL. Signaling by target of rapamycin proteins in cell growth control. Microbiol Mol Biol Rev 2005; 69:79-100. [PMID: 15755954 PMCID: PMC1082789 DOI: 10.1128/mmbr.69.1.79-100.2005] [Citation(s) in RCA: 259] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Target of rapamycin (TOR) proteins are members of the phosphatidylinositol kinase-related kinase (PIKK) family and are highly conserved from yeast to mammals. TOR proteins integrate signals from growth factors, nutrients, stress, and cellular energy levels to control cell growth. The ribosomal S6 kinase 1 (S6K) and eukaryotic initiation factor 4E binding protein 1(4EBP1) are two cellular targets of TOR kinase activity and are known to mediate TOR function in translational control in mammalian cells. However, the precise molecular mechanism of TOR regulation is not completely understood. One of the recent breakthrough studies in TOR signaling resulted in the identification of the tuberous sclerosis complex gene products, TSC1 and TSC2, as negative regulators for TOR signaling. Furthermore, the discovery that the small GTPase Rheb is a direct downstream target of TSC1-TSC2 and a positive regulator of the TOR function has significantly advanced our understanding of the molecular mechanism of TOR activation. Here we review the current understanding of the regulation of TOR signaling and discuss its function as a signaling nexus to control cell growth during normal development and tumorigenesis.
Collapse
Affiliation(s)
- Ken Inoki
- Life Science Institute, University of Michigan Medical School, 5450 Medical Science I Bldg., Ann Arbor, MI 48109-0606, USA
| | | | | | | |
Collapse
|
208
|
Kim RH, Peters M, Jang Y, Shi W, Pintilie M, Fletcher GC, DeLuca C, Liepa J, Zhou L, Snow B, Binari RC, Manoukian AS, Bray MR, Liu FF, Tsao MS, Mak TW. DJ-1, a novel regulator of the tumor suppressor PTEN. Cancer Cell 2005; 7:263-73. [PMID: 15766664 DOI: 10.1016/j.ccr.2005.02.010] [Citation(s) in RCA: 446] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2004] [Revised: 01/27/2005] [Accepted: 02/22/2005] [Indexed: 11/27/2022]
Abstract
The phosphatidylinositol 3' kinase (PI3'K) pathway, which regulates cell survival, is antagonized by the PTEN tumor suppressor. The regulation of PTEN is unclear. A genetic screen of Drosophila gain-of-function mutants identified DJ-1 as a suppressor of PTEN function. In mammalian cells, DJ-1 underexpression results in decreased phosphorylation of PKB/Akt, while DJ-1 overexpression leads to hyperphosphorylation of PKB/Akt and increased cell survival. In primary breast cancer samples, DJ-1 expression correlates negatively with PTEN immunoreactivity and positively with PKB/Akt hyperphosphorylation. In 19/23 primary non-small cell lung carcinoma samples, DJ-1 expression was increased compared to paired nonneoplastic lung tissue, and correlated positively with relapse incidence. DJ-1 is thus a key negative regulator of PTEN that may be a useful prognostic marker for cancer.
Collapse
Affiliation(s)
- Raymond H Kim
- Advanced Medical Discovery Institute, The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, Toronto, Canada M5G 2C1
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
209
|
DeGraffenried LA, Fulcher L, Friedrichs WE, Grünwald V, Ray RB, Hidalgo M. Reduced PTEN expression in breast cancer cells confers susceptibility to inhibitors of the PI3 kinase/Akt pathway. Ann Oncol 2005; 15:1510-6. [PMID: 15367412 DOI: 10.1093/annonc/mdh388] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The PTEN protein is a lipid phosphatase with putative tumor suppressing abilities, including inhibition of the PI3K/Akt signaling pathway. Inactivating mutations or deletions of the PTEN gene, which result in hyper-activation of the PI3K/Akt signaling pathway, are increasingly being reported in human malignancies, including breast cancer, and have been related to features of poor prognosis and resistance to chemotherapy and hormone therapy. Prior studies in different tumor models have shown that, under conditions of PTEN deficiency, the PI3K/Akt signaling pathway becomes a fundamental proliferative and survival pathway, and that pharmacological inhibition of this pathway results in tumor growth inhibition. This study aimed to explore further this hypothesis in breast cancer cells. To this end, we have determined the growth response to inhibition of the PI3K/Akt signaling pathway in a series of breast cancer cell lines with different PTEN levels. The PTEN-negative cell line displayed greater sensitivity to the growth inhibitory effects of the PI3K inhibitor, LY294002 and rapamycin, an inhibitor of the PI3K/Akt downstream mediator mTOR, compared with the PTEN-positive cell lines. To determine whether or not these differences in response are specifically due to effects of PTEN, we developed a series of cell lines with reduced PTEN protein expression compared with the parental cell line. These reduced PTEN cells demonstrated an increased sensitivity to the anti-proliferative effects induced by LY294002 and rapamycin compared with the parental cells, which corresponded to alterations in cell cycle response. These findings indicate that inhibitors of mTOR, some of which are already in clinical development (CCI-779, an ester of rapamycin), have the potential to be effective in the treatment of breast cancer patients with PTEN-negative tumors and should be evaluated in this setting.
Collapse
Affiliation(s)
- L A DeGraffenried
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | | | | | | | | |
Collapse
|
210
|
Abstract
Hereditary cancer syndromes are a group of disorders characterized by a genetic susceptibility to the development of malignant tumors. Multiple cancers in the family or an abnormally early onset for the given cancer may suggest an underlying inherited predisposition. Awareness of their associated dermatologic manifestations can facilitate early detection of risk for neoplasms. This article provides an update on the clinical features, diagnostic criteria, and the use of genetic analysis in the detection of causative mutations of those hereditary cancer syndromes with cutaneous manifestations.
Collapse
|
211
|
Abstract
Dysmorphology is the study of abnormal patterns of human development. A recurrent and recognizable combination of physical and behavioural abnormalities makes up a syndrome. Accurate recognition and diagnosis of syndromes is important because it influences medical management of patients, provides information about prognosis, and allows for genetic counselling including accurate estimation of genetic risk within families and, where possible, prenatal diagnosis. This review examines the diagnostic process in dysmorphology and indicates how skin signs may provide important clues to the clinician.
Collapse
Affiliation(s)
- S F Smithson
- Department of Clinical Genetics, St Michael's Hospital, Southwell Street, Bristol BS2 8EG, UK.
| | | |
Collapse
|
212
|
Cully M, Elia A, Ong SH, Stambolic V, Pawson T, Tsao MS, Mak TW. grb2 heterozygosity rescues embryonic lethality but not tumorigenesis in pten+/- mice. Proc Natl Acad Sci U S A 2004; 101:15358-63. [PMID: 15492213 PMCID: PMC524460 DOI: 10.1073/pnas.0406613101] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PTEN is a tumor suppressor gene implicated in both sporadic cancers and inherited tumor-prone syndromes. Here we show that pten+/- mice display a partially penetrant embryonic lethality. This lethality is associated with defects in both neural and placental development. Notably, this lethality is completely rescued by grb2 haploinsufficiency. In contrast, grb2 heterozygosity did not alter tumorigenesis in either pten+/- or T cell-specific pten-/- mice. grb2-/hypomorph murine embryonic fibroblasts (MEFs) show decreased activation of both PKB and Erk upon stimulation with epidermal growth factor, whereas grb2-/hypomorph; pten+/- MEFs activate PKB but not Erk normally. Similarly, grb2-/hypomorph fibroblasts die in low serum, and this phenotype is rescued by pten haploinsufficiency. Activation of both PKB and Erk as well as survival in low serum-containing media are all rescued by reexpression of Grb2 containing mutations within the N-terminal Src homology 3 (SH3) domain, but not by C-terminal SH3 domain mutants. The N-terminal SH3 domain mutants fail to bind to Sos, whereas the C-terminal SH3 domain mutants fail to bind to Gab1, suggesting that Erk and PKB activation in fibroblasts in response to epidermal growth factor depends on Gab1 or other C-terminal SH3 domain-interacting proteins, but not on Sos. Thus, PTEN/phosphatidylinositol 3' kinase signaling requires Grb2 during both embryonic development and fibroblast survival, but Grb2 heterozygosity does not effect tumorigenesis in pten-deficient mice. In fibroblasts, survival signals emanating from the epidermal growth factor receptor appear to be PKB-dependent, and this activation depends on the C-terminal SH3 domain of Grb2, likely through the interaction of Grb2 with Gab1.
Collapse
Affiliation(s)
- Megan Cully
- Department of Medical Biophysics, University of Toronto, Ontario Cancer Institute, Princess Margaret Hospital, 610 University Avenue, Room 7-411, Toronto, ON, Canada M5G 2M9
| | | | | | | | | | | | | |
Collapse
|
213
|
Abstract
Development of any cancer reflects a progressive accumulation of alterations in various genes. Oncogenes, tumour suppressor genes, DNA repair genes and metastasis suppressor genes have been investigated in prostate cancer. Here, we review current understanding of the molecular biology of prostate cancer. Detailed understanding of the molecular basis of prostate cancer will provide insights into the aetiology and prognosis of the disease, and suggest avenues for therapeutic intervention in the future.
Collapse
Affiliation(s)
- M K Karayi
- Molecular Medicine Unit, University of Leeds, St James's University Hospital, Leeds, UK.
| | | |
Collapse
|
214
|
García JM, Silva J, Peña C, Garcia V, Rodríguez R, Cruz MA, Cantos B, Provencio M, España P, Bonilla F. Promoter methylation of the PTEN gene is a common molecular change in breast cancer. Genes Chromosomes Cancer 2004; 41:117-24. [PMID: 15287024 DOI: 10.1002/gcc.20062] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
About 25-50% of women with Cowden disease, a syndrome associated with germ-line mutations of the PTEN gene (at 10q23), develop breast cancer (BC), but PTEN mutations have been found in only 5% of sporadic BCs. However, 29-48% of BCs display loss of heterozygosity in 10q23, and about 40% of BCs show a decrease or absence of PTEN protein levels at the time of diagnosis. Promoter hypermethylation has been identified as an alternative mechanism of tumor-suppressor gene inactivation, but its importance in PTEN silencing in sporadic BC is unknown. We investigated PTEN promoter hypermethylation in 90 sporadic BCs and its correlations with 11 molecular and pathologic parameters, including mRNA levels of PTEN. The study, a methylation-specific PCR assay, was carried out with methylated specific primers designed in a region with scarce homology with the psiPTEN pseudogene. Expression was analyzed by real-time PCR. We found that the PTEN promoter was hypermethylated in 43 BCs (48%). PTEN hypermethylation was associated with ERBB2 overexpression, larger size, and higher histologic grade (P=0.012, 0.03, and 0.009, respectively). We concluded that PTEN promoter hypermethylation is a common event in sporadic BC, correlating with other well-established prognostic factors of this malignancy. Additionally, PTEN mRNA expression was lower in tumors with aberrant methylation.
Collapse
Affiliation(s)
- Jose M García
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
215
|
Abstract
The past two decades have brought many important advances in our understanding of the hereditary susceptibility to cancer. Approximately 5-10% of all cancers are inherited, the majority in an autosomal dominant manner with incomplete penetrance. While this is a small fraction of the overall cancer burden worldwide, the molecular genetic discoveries that have resulted from the study of families with heritable cancer have not only changed the way these families are counselled and managed, but have shed light on molecular regulatory pathways important in sporadic tumour development as well. In this review, we consider 10 of the more highly penetrant cancer syndromes, with emphasis on those predisposing to breast, colon, and/or endocrine neoplasia. We discuss the prevalence, penetrance, and tumour spectrum associated with these syndromes, as well as their underlying genetic defects.
Collapse
Affiliation(s)
- Rebecca Nagy
- Clinical Cancer Genetics Program, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, The Ohio State University, Columbus 43221, USA.
| | | | | |
Collapse
|
216
|
Tsao H, Mihm MC, Sheehan C. PTEN expression in normal skin, acquired melanocytic nevi, and cutaneous melanoma. J Am Acad Dermatol 2004; 49:865-72. [PMID: 14576666 DOI: 10.1016/s0190-9622(03)02473-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Although various studies have shown mutations of the tumor suppressor gene, PTEN/MMAC1, in primary, metastatic, and cultured cutaneous melanoma specimens, little is known about the pattern of PTEN protein expression in early melanocytic tumor progression. OBJECTIVE To further investigate the role of PTEN in melanocytic tumor development. METHODS We assessed the level and distribution of PTEN in normal skin, 39 acquired melanocytic nevi, and 30 primary cutaneous melanomas, including lentigo malignas, by immunostaining. RESULTS We found high levels of PTEN expression in cutaneous muscles, nerves, and muscular arteries, and moderate-to-high amounts of PTEN in the epidermis, follicular epithelium, and sebaceous and eccrine glands. PTEN staining in cutaneous lymphatics, dermal and periadnexal adventitial fibroblasts, and chondrocytes were variably absent. Junctional melanocytes and chondrocytes frequently exhibited preferential nuclear staining. We found uniformly strong PTEN expression in the cytoplasm of almost all benign and dysplastic nevi. However, there was some evidence of nuclear PTEN loss even in the benign melanocytic proliferations. In addition, out of 30 primary cutaneous melanomas and lentigo malignas, we detected diffuse expression of PTEN in 11 (37%) tumors, widespread loss of PTEN in 11 (37%) tumors and mixed PTEN expression in 8 (27%) lesions. In the primary cutaneous melanomas, PTEN was largely localized to the cytoplasm. CONCLUSIONS The presence of PTEN in benign melanocytic tumors and the absence of PTEN in a significant proportion of primary cutaneous melanomas support a role for PTEN loss in the pathogenesis of melanoma.
Collapse
Affiliation(s)
- Hensin Tsao
- Department of Dermatology, Massachusetts General Hospital, Boston 02114, USA.
| | | | | |
Collapse
|
217
|
Komazawa N, Suzuki A, Sano S, Horie K, Matsuura N, Mak TW, Nakano T, Takeda J, Kondoh G. Tumorigenesis facilitated by Pten deficiency in the skin: evidence of p53-Pten complex formation on the initiation phase. Cancer Sci 2004; 95:639-43. [PMID: 15298725 PMCID: PMC11159272 DOI: 10.1111/j.1349-7006.2004.tb03322.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2004] [Revised: 06/09/2004] [Accepted: 06/19/2004] [Indexed: 11/30/2022] Open
Abstract
Pten, a tumor suppressor gene, is mutated in various human cancers and in hereditary cancer syndromes, such as Cowden disease. We have previously developed a knockout mouse in which Pten is specifically disrupted in the skin, resulting in hyperproliferation and spontaneous tumorigenesis of the skin keratinocytes. In this study, we further clarified the effects of Pten deficiency in tumorigenesis, by using a two-step model in intact skin of Pten knockout mouse. Although the conventional protocol requires serial exposures to DMBA and TPA, mice deficient for Pten developed skin papilloma within 6 weeks after a single exposure to DMBA, indicating that loss of Pten has a tumor-promoting effect. Serial exposure to DMBA-TPA ointments produced 10-fold more papillomas in the skin of knockout mice than in the wild-type counterpart, suggesting an increased rate of initiation. Therefore, we precisely examined the effect of DMBA. This treatment was highly apoptotic in wild-type mice, whereas the number of apoptotic cells was diminished in Pten-deficient skin. Moreover, primary keratinocytes isolated from Pten-deficient mice were also resistant to the apoptotic effect of DMBA. The status of p53, Pten proteins and downstream targets of p53, such as p21, 14-3-3, and Reprimo, were also examined, and we found that accumulation of p53 protein and up-regulation of p53 targets were delayed in Pten-knockout skin. These observations suggest that Pten is involved in rapid recruitment of p53 in the tumor initiation phase.
Collapse
Affiliation(s)
- Nobuyasu Komazawa
- Department of Social and Environmental Medicine, Osaka University, Suita, 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Abstract
Vascular endothelial growth factor (VEGF) is an endothelial cell-specific mitogen in vitro and an angiogenic inducer in a variety of in vivo models. Hypoxia has been shown to be a major inducer of VEGF gene transcription. The tyrosine kinases Flt-1 (VEGFR-1) and Flk-1/KDR (VEGFR-2) are high-affinity VEGF receptors. The role of VEGF in developmental angiogenesis is emphasized by the finding that loss of a single VEGF allele results in defective vascularization and early embryonic lethality. VEGF is critical also for reproductive and bone angiogenesis. Substantial evidence also implicates VEGF as a mediator of pathological angiogenesis. In situ hybridization studies demonstrate expression of VEGF mRNA in the majority of human tumors. Anti-VEGF monoclonal antibodies and other VEGF inhibitors block the growth of several tumor cell lines in nude mice. Clinical trials with various VEGF inhibitors in a variety of malignancies are ongoing. Very recently, an anti-VEGF monoclonal antibody (bevacizumab; Avastin) has been approved by the Food and Drug Administration as a first-line treatment for metastatic colorectal cancer in combination with chemotherapy. Furthermore, VEGF is implicated in intraocular neovascularization associated with diabetic retinopathy and age-related macular degeneration.
Collapse
Affiliation(s)
- Napoleone Ferrara
- Department of Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
219
|
Mao JH, To MD, Perez-Losada J, Wu D, Del Rosario R, Balmain A. Mutually exclusive mutations of the Pten and ras pathways in skin tumor progression. Genes Dev 2004; 18:1800-1805. [PMID: 15289454 PMCID: PMC517400 DOI: 10.1101/gad.1213804] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2004] [Accepted: 06/02/2004] [Indexed: 12/31/2022]
Abstract
Pten heterozygous (Pten+/-) mice develop increased papilloma numbers and show decreased carcinoma latency time in comparison with controls after skin treatment with dimethyl benzanthracene (DMBA) and tetradecanoyl-phorbol acetate (TPA). H-ras mutation is normally a hallmark of DMBA-TPA-induced skin tumors, but 70% of carcinomas from Pten+/- mice do not exhibit this mutation, and in all cases have lost the wild-type Pten allele. Tumors that retain the Pten wild-type allele also have H-ras mutations, indicating that activation of H-ras and complete loss of Pten are mutually exclusive events in skin carcinomas. Mitogen-activated protein kinase (MAPK) is consistently activated in the tumors with H-ras mutations, but is strongly down-regulated in Pten-/- tumors, suggesting that this pathway is dispensable for skin carcinoma formation. These data have important implications in designing individual therapeutic strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Jian-Hua Mao
- University of California at San Francisco Comprehensive Cancer Center, San Francisco, California 94143, USA
| | | | | | | | | | | |
Collapse
|
220
|
Abstract
Genetic alterations targeting the PTEN tumor suppressor gene are among the most frequently noted somatic mutations in human cancers. Such lesions have been noted in cancers of the prostate and endometrium and in glioblastoma multiforme, among many others. Moreover, germline mutation of PTEN leads to the development of the related hereditary cancer predisposition syndromes, Cowden disease, and Bannayan-Zonana syndrome, wherein breast and thyroid cancer incidence is elevated. The protein product, PTEN, is a lipid phosphatase, the enzymatic activity of which primarily serves to remove phosphate groups from key intracellular phosphoinositide signaling molecules. This activity normally serves to restrict growth and survival signals by limiting activity of the phosphoinositide-3 kinase (PI3K) pathway. Multiple lines of evidence support the notion that this function is critical to the ability of PTEN to maintain cell homeostasis. Indeed, the absence of functional PTEN in cancer cells leads to constitutive activation of downstream components of the PI3K pathway including the Akt and mTOR kinases. In model organisms, inactivation of these kinases can reverse the effects of PTEN loss. These data raise the possibility that drugs targeting these kinases, or PI3K itself, might have significant therapeutic activity in PTEN-null cancers. Akt kinase inhibitors are still in development; however, as a first test of this hypothesis, phase I and phase II trials of inhibitors of mTOR, namely, rapamycin and rapamycin analogs are underway.
Collapse
Affiliation(s)
- Isabelle Sansal
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | | |
Collapse
|
221
|
Minaguchi T, Yoshikawa H, Nakagawa S, Yasugi T, Yano T, Iwase H, Mizutani K, Shiromizu K, Ohmi K, Watanabe Y, Noda K, Nishiu M, Nakamura Y, Taketani Y. Association of PTEN mutation with HPV-negative adenocarcinoma of the uterine cervix. Cancer Lett 2004; 210:57-62. [PMID: 15172121 DOI: 10.1016/j.canlet.2004.03.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2003] [Revised: 03/03/2004] [Accepted: 03/17/2004] [Indexed: 10/26/2022]
Abstract
Serous, mucinous, endometrioid, and clear cell adenocarcinomas arise from reproductive organs of mullerian origin. Although the mutation of PTEN, a tumor suppressor, is known to be involved in tumorigenesis of endometrioid adenocarcinomas of the endometrium and ovary, the role of PTEN alteration in endometrioid adenocarcinoma of the cervix remains to be investigated. To elucidate the molecular pathogenesis of cervical adenocarcinoma and adenosquamous carcinoma, and in particular to examine the potential role of PTEN mutation in endometrioid-type cancer of the cervix, we analyzed 32 cervical adeno- or adenosquamous carcinomas (8 endometrioid adenocarcinomas, 14 mucinous adenocarcinomas and 10 adenosquamous carcinomas) for PTEN mutations and HPV infections. PTEN mutation was detected in 2 of 8 (25.0%) endometrioid cases, 2 of 14 (14.3%) mucinous cases, and none of 10 (0%) adenosquamous cases. HPV DNA was detected in 11 out of 18 (61.1%) PTEN wild-type adenocarcinomas and 8 out of 10 (80.0%) adenosquamous carcinomas. Among 11 HPV-negative adenocarcinomas, 40.0% (2/5) endometrioid cases and 33.3% (2/6) mucinous cases were shown to be PTEN mutated, while no cases (0/21) were PTEN-mutant in the remainder (i.e. adenosquamous carcinomas and HPV-positive adenocarcinomas). The current observations suggest that PTEN mutation is frequently detected in HPV-negative adenocarcinomas of the cervix and the most prevalent occurrence of PTEN mutation in endometrioid subtype is keeping with endometrial and ovarian carcinomas.
Collapse
Affiliation(s)
- Takeo Minaguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Cohen MM. Molecular dimensions of gastrointestinal tumors: some thoughts for digestion. Am J Med Genet A 2004; 122A:303-14. [PMID: 14518068 DOI: 10.1002/ajmg.a.20473] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Topics discussed here include PTEN mutations and colonic polyps; WNT signaling, APC, beta-catenin, and gastrointestinal neoplasms; mismatch-repair genes (MLH1, MSH2, PMS1, MSH6) and hereditary nonpolyposis colorectal cancer; MYH mutations and autosomal recessive colorectal tumors; STK11 mutations and Peutz-Jeghers syndrome; TGFbeta and gastrointestinal cancer; BMPR1A mutations and juvenile polyposis; FGF/FGFR alterations in gastrointestinal neoplasms; PTCH mutations and gastrointestinal neoplasms; RUNX3 expression and gastric cancer; role of mucins in gastric carcinogenesis; KIT, PDGFRalpha, and gastrointestinal stromal tumors; intestinal neurofibromatosis; and gastrointestinal tumors in other disorders.
Collapse
Affiliation(s)
- M Michael Cohen
- Department of Oral & Maxillofacial Sciences, Pediatrics, Community Health & Epidemiology, Health Services Administration, and Sociology & Social Anthropology, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
223
|
Tanaka M, Grossman HB. Tumor suppressor genes of bladder cancer and potential for gene therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2004; 539:185-91. [PMID: 15088905 DOI: 10.1007/978-1-4419-8889-8_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Affiliation(s)
- Motoyoshi Tanaka
- Department of Urology, M. D. Anderson Cancer Center, The University of Texas, Houston 77030, USA
| | | |
Collapse
|
224
|
Karoui M, Tresallet C, Julie C, Zimmermann U, Staroz F, Brams A, Muti C, Boulard C, Robreau AM, Puy H, Malafosse R, Penna C, Pruvot FR, Thiery JP, Boileau C, Rougier P, Nordlinger B, Radvanyi F, Franc B, Hofmann-Radvanyi H. Loss of heterozygosity on 10q and mutational status of PTEN and BMPR1A in colorectal primary tumours and metastases. Br J Cancer 2004; 90:1230-4. [PMID: 15026806 PMCID: PMC2409663 DOI: 10.1038/sj.bjc.6601687] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We investigated the possible role of chromosome 10q losses in colorectal cancer metastasis by carrying out an allelic imbalance study on a series of microsatellite instability-negative (MSI−) primary tumours (n=32) and metastases (n=36) from 49 patients. Our results demonstrate that 10q allelic losses are associated with a significant proportion (25%) of MSI− colorectal tumours, but are not involved in the metastatic process. PTEN and BMPR1A, two genes located in the common deleted region, were screened for mutations in samples with loss of heterozygosity. The absence or low frequency of mutations indicates that the inactivation of these genes by deletion of one allele and mutation of the other one plays only a minor role in MSI− tumours.
Collapse
Affiliation(s)
- M Karoui
- Fédération des Spécialités Digestives, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 92104 Boulogne Cedex, France
- UMR 144, CNRS-Institut Curie, 75248 Paris Cedex 05, France
- Service de Chirurgie Digestive et de Transplantation, Hôpital Claude Huriez, CHRU de Lille, 59037 Lille Cedex, France
| | - C Tresallet
- Fédération des Spécialités Digestives, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 92104 Boulogne Cedex, France
- Service de Chirurgie Digestive, Hôpital Pitié-Salpêtrière, AP-HP, 75651 Paris Cedex 13, France
| | - C Julie
- Service d'Anatomie et de Cytologie Pathologiques, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 92104 Boulogne Cedex, France
| | - U Zimmermann
- Service d'Anatomie et de Cytologie Pathologiques, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 92104 Boulogne Cedex, France
| | - F Staroz
- Service d'Anatomie et de Cytologie Pathologiques, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 92104 Boulogne Cedex, France
| | - A Brams
- Fédération des Spécialités Digestives, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 92104 Boulogne Cedex, France
- UMR 144, CNRS-Institut Curie, 75248 Paris Cedex 05, France
| | - C Muti
- Service de Génétique, Hôpital Ambroise Paré, AP-HP, 92104 Boulogne Cedex, France
| | - C Boulard
- UMR 144, CNRS-Institut Curie, 75248 Paris Cedex 05, France
| | - A-M Robreau
- Laboratoire de Biochimie, Hôpital Louis Mourier, AP-HP, 92701 Colombes Cedex, France
| | - H Puy
- Laboratoire de Biochimie, Hôpital Louis Mourier, AP-HP, 92701 Colombes Cedex, France
- Laboratoire de Biochimie et de Génétique Moléculaire, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 9 Avenue Charles de Gaulle, 92104 Boulogne Cedex, France
| | - R Malafosse
- Fédération des Spécialités Digestives, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 92104 Boulogne Cedex, France
| | - C Penna
- Fédération des Spécialités Digestives, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 92104 Boulogne Cedex, France
| | - F-R Pruvot
- Service de Chirurgie Digestive et de Transplantation, Hôpital Claude Huriez, CHRU de Lille, 59037 Lille Cedex, France
| | - J P Thiery
- UMR 144, CNRS-Institut Curie, 75248 Paris Cedex 05, France
| | - C Boileau
- Laboratoire de Biochimie et de Génétique Moléculaire, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 9 Avenue Charles de Gaulle, 92104 Boulogne Cedex, France
| | - P Rougier
- Fédération des Spécialités Digestives, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 92104 Boulogne Cedex, France
| | - B Nordlinger
- Fédération des Spécialités Digestives, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 92104 Boulogne Cedex, France
| | - F Radvanyi
- UMR 144, CNRS-Institut Curie, 75248 Paris Cedex 05, France
| | - B Franc
- Service d'Anatomie et de Cytologie Pathologiques, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 92104 Boulogne Cedex, France
| | - H Hofmann-Radvanyi
- UMR 144, CNRS-Institut Curie, 75248 Paris Cedex 05, France
- Laboratoire de Biochimie et de Génétique Moléculaire, Hôpital Ambroise Paré, AP-HP, Université Versailles-Saint Quentin en Yvelines, 9 Avenue Charles de Gaulle, 92104 Boulogne Cedex, France
- UMR 144, CNRS-Institut Curie, 75248 Paris Cedex 05, France. E-mail:
| |
Collapse
|
225
|
Vasudevan KM, Gurumurthy S, Rangnekar VM. Suppression of PTEN expression by NF-kappa B prevents apoptosis. Mol Cell Biol 2004; 24:1007-21. [PMID: 14729949 PMCID: PMC321419 DOI: 10.1128/mcb.24.3.1007-1021.2004] [Citation(s) in RCA: 203] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
NF-kappa B is a heterodimeric transcription activator consisting of the DNA binding subunit p50 and the transactivation subunit p65/RelA. NF-kappa B prevents cell death caused by tumor necrosis factor (TNF) and other genotoxic insults by directly inducing antiapoptotic target genes. We report here that the tumor suppressor PTEN, which functions as a negative regulator of phosphatidylinositol (PI)-3 kinase/Akt-mediated cell survival pathway, is down regulated by p65 but not by p50. Moreover, a subset of human lung or thyroid cancer cells expressing high levels of endogenous p65 showed decreased expression of PTEN that could be rescued by specific inhibition of the NF-kappa B pathway with I kappa B overexpression as well as with small interfering RNA directed against p65. Importantly, TNF, a potent inducer of NF-kappa B activity, suppressed PTEN gene expression in IKK beta(+/+) cells but not in IKK beta(-/-) cells, which are deficient in the NF-kappa B activation pathway. These findings indicated that NF-kappa B activation was necessary and sufficient for inhibition of PTEN expression. The promoter, RNA, and protein levels of PTEN are down-regulated by NF-kappa B. The mechanism underlying suppression of PTEN expression by NF-kappa B was independent of p65 DNA binding or transcription function and involved sequestration of limiting pools of transcriptional coactivators CBP/p300 by p65. Restoration of PTEN expression inhibited NF-kappa B transcriptional activity and augmented TNF-induced apoptosis, indicating a negative regulatory loop involving PTEN and NF-kappa B. PTEN is, thus, a novel target whose suppression is critical for antiapoptosis by NF-kappa B.
Collapse
Affiliation(s)
- Krishna Murthi Vasudevan
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | |
Collapse
|
226
|
Pfeil K, Eder IE, Putz T, Ramoner R, Culig Z, Ueberall F, Bartsch G, Klocker H. Long-term androgen-ablation causes increased resistance to PI3K/Akt pathway inhibition in prostate cancer cells. Prostate 2004; 58:259-68. [PMID: 14743465 DOI: 10.1002/pros.10332] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND In advanced stages of prostate cancer, the phosphatidylinositol-3' kinase (PI3K)/Akt signaling cascade, one of the major survival pathways in the cell, is frequently constitutively activated due to mutation or loss of the tumor suppressor protein phosphatase and tensin homolog deleted on chromosome 10 (PTEN). Using cell culture models representing different tumor stages, we explored the effect of inhibition of this survival pathway on the induction of apoptosis. METHODS Inhibition of the survival kinase Akt and induction of apoptosis was analyzed in androgen-insensitive DU145 and PC-3 cells, in androgen-responsive LNCaP, and in androgen-independent long-term androgen-ablated LNCaP-abl cells representing therapy-resistant prostate cancer cells. Activated Akt was determined by immunoblotting using a phospho-Akt specific antibody. Induction of apoptosis was analyzed employing annexing V and propidium iodide staining and flow cytometry and measurement of cleavage of the caspases substrate poly-ADP-ribose polymerase (PARP). RESULTS IGF-1, EGF, and heregulin but not PDGF or activators of protein kinase A induced phosphorylation of Akt in DU145 cells and activation was completely blocked by the PI3K inhibitor LY294002. In the hormone-responsive prostate cancer cell line LNCaP that has a constitutively switched-on Akt kinase, LY294002 caused a dose- and time-dependent Akt inhibition, which was absent in long-term androgen-ablated LNCaP sublines. In agreement with the resistance to inhibition of the PI3K/Akt pathway, long-term androgen-ablated LNCaP sublines remained relatively resistant to induction of cell death by LY294002 or the cytotoxic drug etoposide. Inhibition of the PI3K/Akt pathway restored the sensitivity of long-term androgen-ablated cells to induction of apoptosis by a cytotoxic drug almost completely. CONCLUSION These results suggest that long-term androgen ablation therapy for prostate cancer reinforces the PI3K/Akt pathway and impedes its inhibition thus contributing to increased resistance of tumor cells to induction of apoptosis. With regard to treatment of therapy-refractory prostate cancer, these findings suggest effectiveness of a combination of cytotoxic treatment and inhibition of the PI3K-Akt survival pathway in tumor cells after failure of androgen-ablation therapy.
Collapse
Affiliation(s)
- Karina Pfeil
- Department of Urology, University of Innsbruck, Austria
| | | | | | | | | | | | | | | |
Collapse
|
227
|
Kim S, Domon-Dell C, Kang J, Chung DH, Freund JN, Evers BM. Down-regulation of the tumor suppressor PTEN by the tumor necrosis factor-alpha/nuclear factor-kappaB (NF-kappaB)-inducing kinase/NF-kappaB pathway is linked to a default IkappaB-alpha autoregulatory loop. J Biol Chem 2004; 279:4285-91. [PMID: 14623898 DOI: 10.1074/jbc.m308383200] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The PTEN (phosphatase and tensin homolog deleted on chromosome ten) tumor suppressor gene affects multiple cellular processes including cell growth, proliferation, and cell migration by antagonizing phosphatidylinositol 3-kinase (PI3K). However, mechanisms by which PTEN expression is regulated have not been studied extensively. Similar to PTEN, tumor necrosis factor-alpha (TNF-alpha) affects a wide spectrum of diseases including inflammatory processes and cancer by acting as a mediator of apoptosis, inflammation, and immunity. In this study, we show that treatment of cancer cell lines with TNF-alpha decreases PTEN expression. In addition, overexpression of TNF-alpha downstream signaling targets, nuclear factor-kappaB (NF-kappaB)-inducing kinase (NIK) and p65 nuclear factor NF-kappaB, lowers PTEN expression, suggesting that TNF-alpha-induced down-regulation of PTEN is mediated through a TNF-alpha/NIK/NF-kappaB pathway. Down-regulation of PTEN by NIK/NF-kappaB results in activation of the PI3K/Akt pathway and augmentation of TNF-alpha-induced PI3K/Akt stimulation. Importantly, we demonstrate that this effect is associated with a lack of an inhibitor of kappaB (IkappaB)-alpha autoregulatory loop. Moreover, these findings suggest the interaction between PI3K/Akt and NF-kappaB via transcriptional regulation of PTEN and offer one possible explanation for increased tumorigenesis in systems in which NF-kappaB is chronically activated. In such a tumor system, these findings suggest a positive feedback loop whereby Akt activation of NF-kappaB further stimulates Akt via down-regulation of the PI3K inhibitor PTEN.
Collapse
Affiliation(s)
- Sunghoon Kim
- Department of Surgery, The University of Texas Medical Branch, Galveston, Texas 77555-0536, USA
| | | | | | | | | | | |
Collapse
|
228
|
Nassif NT, Lobo GP, Wu X, Henderson CJA, Morrison CD, Eng C, Jalaludin B, Segelov E. PTEN mutations are common in sporadic microsatellite stable colorectal cancer. Oncogene 2004; 23:617-28. [PMID: 14724591 DOI: 10.1038/sj.onc.1207059] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The tumour suppressor gene PTEN, located at chromosome sub-band 10q23.3, encodes a dual-specificity phosphatase that negatively regulates the phosphatidylinositol 3'-kinase (PI3 K)/Akt-dependent cellular survival pathway. PTEN is frequently inactivated in many tumour types including glioblastoma, prostate and endometrial cancers. While initial studies reported that PTEN gene mutations were rare in colorectal cancer, more recent reports have shown an approximate 18% incidence of somatic PTEN mutations in colorectal tumours exhibiting microsatellite instability (MSI+). To verify the role of this gene in colorectal tumorigenesis, we analysed paired normal and tumour DNA from 41 unselected primary sporadic colorectal cancers for PTEN inactivation by mutation and/or allelic loss. We now report PTEN gene mutations in 19.5% (8/41) of tumours and allele loss, including all or part of the PTEN gene, in a further 17% (7/41) of the cases. Both PTEN alleles were affected in over half (9/15) of these cases showing PTEN genetic abnormalities. Using immunohistochemistry, we have further shown that all tumours harbouring PTEN alterations have either reduced or absent PTEN expression and this correlated strongly with later clinical stage of tumour at presentation (P=0.02). In contrast to previous reports, all but one of the tumours with PTEN gene mutations were microsatellite stable (MSI-), suggesting that PTEN is involved in a distinct pathway of colorectal tumorigenesis that is separate from the pathway of mismatch repair deficiency. This work therefore establishes the importance of PTEN in primary sporadic colorectal cancer.
Collapse
Affiliation(s)
- Najah T Nassif
- Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, NSW 2170, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
229
|
Mazereeuw-Hautier J, Assouère MN, Moreau-Cabarrot A, Longy M, Bonafé JL. Cowden's syndrome: possible association with testicular seminoma. Br J Dermatol 2004; 150:378-9. [PMID: 14996122 DOI: 10.1111/j.1365-2133.2003.05770.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
230
|
Hendriks YMC, Verhallen JTCM, van der Smagt JJ, Kant SG, Hilhorst Y, Hoefsloot L, Hansson KBM, van der Straaten PJC, Boutkan H, Breuning MH, Vasen HFA, Bröcker-Vriends AHJT. Bannayan-Riley-Ruvalcaba syndrome: further delineation of the phenotype and management of PTEN mutation-positive cases. Fam Cancer 2004; 2:79-85. [PMID: 14574156 DOI: 10.1023/a:1025713815924] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Bannayan-Riley-Ruvalcaba syndrome (BRRS) is characterised by macrocephaly, intestinal hamartomatous polyps, lipomas, pigmented maculae of the glans penis, developmental delay and mental retardation. The syndrome follows an autosomal dominant pattern of inheritance. In 1997 reports on two BRRS patients with a deletion at 10q23.2-q24.1 were published. In the same year, the first two families with BRRS and a mutation of the PTEN gene were reported. Mutations in the PTEN gene have also been demonstrated in patients with Cowden syndrome (CS), which shows partial clinical overlap with BRRS, and in families with cases both of BRRS and CS. PTEN mutation positive BRRS and CS are likely to be different phenotypic presentations of the same syndrome. If BRRS and CS are one single condition, the question arises whether patients with BRRS should be screened for malignant tumours, since patients with Cowden syndrome have an increased risk of breast, endometrial, thyroid and renal cancer. We present two isolated cases and one family and confirm that BRRS and CS are allelic. Furthermore, we review the PTEN mutation positive BRRS cases, to further delineate the phenotype and to compare the cases with a genomic deletion with the cases with a point mutation. We recommend offering BRRS cases with a mutation in PTEN the same surveillance protocol for (malignant) tumours as is currently recommended for CS. In addition, we propose a yearly haemoglobin test from early infancy for the early detection of intestinal hamartomas, which are likely to give severe complications, especially in BRRS cases.
Collapse
Affiliation(s)
- Y M C Hendriks
- Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Ouellet M, Barbeau B, Tremblay MJ. Protein tyrosyl phosphatases in T cell activation: implication for human immunodeficiency virus transcriptional activity. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 2004; 73:69-105. [PMID: 12882515 DOI: 10.1016/s0079-6603(03)01003-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The protein tyrosine phosphatases (PTPs) superfamily is a large group of enzymes showing a wide diversity of structure and biological functions. Their implication in the regulation of signal transduction processes is critical for homeostasis and efficient cellular activation. Disturbance of the delicate balance between protein tyrosine kinase and protein tyrosine phosphatase activities is at the heart of a large number of diseases. Control of cellular activation is especially important for human immunodeficiency virus type 1 (HIV-1) since this retrovirus requires activated T cells in order to replicate efficiently. Identification of PTPs implicated in signaling pathways leading to upregulation of HIV-1 gene transcription therefore contributes to the general understanding of cellular factors needed for strong HIV-1 replication and progression to AIDS. The use of bisperoxovanadium compounds as potent, specific, and highly purified PTP inhibitors releases HIV-1 from PTP control and strongly increases HIV-1 gene expression. These inhibitors can thus be used to study signal transduction mechanisms regulated by PTP activity that are important for HIV-1 replication and provide new and interesting therapeutic avenues for the efficient control of this debilitating retroviral infection.
Collapse
Affiliation(s)
- Michel Ouellet
- Centre de Recherche en Infectiologie, Hôpital CHUL, Centre Hospitalier Universitaire de Québec, Canada, G1V 4G2
| | | | | |
Collapse
|
232
|
Sawada T, Okada T, Miwa K, Satoh H, Asano A, Mabuchi H. Two novel mutations ofPTEN gene in Japanese patients with Cowden syndrome. ACTA ACUST UNITED AC 2004; 128A:12-4. [PMID: 15211648 DOI: 10.1002/ajmg.a.20309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Cowden syndrome (CS), also known as multiple hamartoma syndrome, is an autosomal dominant cancer syndrome associated with high risk of breast and thyroid cancer. In three unrelated Japanese CS patients, three PTEN germline mutations were identified, including two novel ones: 589A --> T, resulting in Lys197Stop, and 219-222delAAGA. We also detected a previously reported mutation: 697C --> T, resulting in Arg233Stop. Reports from Western countries have indicated that approximately two-thirds of mutations are found in exons 5, 7, and 8, which is almost the same frequency as found in Japanese CS. No genotype-phenotype correlations have been found in CS patients from 21 Japanese families.
Collapse
Affiliation(s)
- Takeshi Sawada
- Second Department of Internal Medicine, Kanazawa University School of Medicine, 13-1 Takaramachi, Kanazawa 920-8640, Japan
| | | | | | | | | | | |
Collapse
|
233
|
Mcheik JN, Cordier MP, Longy M, Levard G. [Macrocephaly and multiple hamartoma: a very variable entity]. Arch Pediatr 2004; 11:33-6. [PMID: 14700758 DOI: 10.1016/j.arcped.2003.09.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
UNLABELLED The Bannayan-Zonana is a dominant autosomal polymalformation syndrome. CASE REPORT We report a case of Bannayan-Zonana syndrome in a 3-year-old girl, who presented with macrocephaly and a cervical lipoma. The patient's mother had neurofibromatosis I. No mutation in the PTEN gene was found. The slowly progressive lipoma increased with age and the surgical reduction was necessary. COMMENTARIES Macrocephaly is constant in different syndromes characterized with multiple hamartomas. The multiple hamartomas syndrome represents an entity with a high variability of expression.
Collapse
Affiliation(s)
- J N Mcheik
- Service de chirurgie pédiatrique, centre hospitalo-universitaire de Poitiers, 350, avenue Jacques-Coeur, BP 577, 86021 Poitiers, France.
| | | | | | | |
Collapse
|
234
|
Müller H, Plasilova M, Russell AM, Heinimann K. Genetic predisposition as a basis for chemoprevention, surgical and other interventions in colorectal cancer. Recent Results Cancer Res 2003; 163:235-47; discussion 264-6. [PMID: 12903858 DOI: 10.1007/978-3-642-55647-0_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Strategies of cancer prevention are generally developed with the population at large in mind. However, special attention is warranted for those persons with rare genetic traits associated with a greatly elevated risk of developing colorectal cancer (CRC) and some other malignancies: Orphan diseases demand Orphan preventive measures! Recent advances in modern genetics have enhanced our understanding of several genes and the specific germ-line mutations responsible for colorectal carcinogenesis. A number of features provide evidence for a genetic predisposition to CRC. These include typical clinical and histological features of a particular syndrome, a familial aggregation of CRC and associated malignancies, young age at onset of CRC, occurrence of multiple neoplasias and/or unusual localisation of the tumour (e.g., right side of the colon). In hereditary colorectal cancer, genetic testing can easily be demonstrated as cost-effective.
Collapse
Affiliation(s)
- Hansjakob Müller
- Research Group Human Genetics, Division of Medical Genetics UKBB, Department of Clinical-Biological Sciences, University of Basel, Vesalgasse 1, 4051 Basel, Switzerland
| | | | | | | |
Collapse
|
235
|
Narayan S, Roy D. Role of APC and DNA mismatch repair genes in the development of colorectal cancers. Mol Cancer 2003; 2:41. [PMID: 14672538 PMCID: PMC317355 DOI: 10.1186/1476-4598-2-41] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2003] [Accepted: 12/12/2003] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is the third most common cause of cancer-related death in both men and women in the western hemisphere. According to the American Cancer Society, an estimated 105,500 new cases of colon cancer with 57,100 deaths will occur in the U.S. in 2003, accounting for about 10% of cancer deaths. Among the colon cancer patients, hereditary risk contributes approximately 20%. The main inherited colorectal cancers are the familial adenomatous polyposis (FAP) and the hereditary nonpolyposis colorectal cancers (HNPCC). The FAP and HNPCC are caused due to mutations in the adenomatous polyposis coli (APC) and DNA mismatch repair (MMR) genes. The focus of this review is to summarize the functions of APC and MMR gene products in the development of colorectal cancers.
Collapse
Affiliation(s)
- Satya Narayan
- Department of Anatomy and Cell Biology and UF Shands Cancer Center, College of Medicine, Academic Research Building, Room R4-216, 1600 SW Archer Road, University of Florida, Gainesville, FL 32610, USA
| | - Deodutta Roy
- Environmental Health Sciences, University of Alabama at Birmingham, 317 Ryals Building, 1665 University Boulevard, Birmingham, AL 35294-0022, USA
| |
Collapse
|
236
|
Tapparel C, Reymond A, Girardet C, Guillou L, Lyle R, Lamon C, Hutter P, Antonarakis SE. The TPTE gene family: cellular expression, subcellular localization and alternative splicing. Gene 2003; 323:189-99. [PMID: 14659893 DOI: 10.1016/j.gene.2003.09.038] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The human TPTE (Transmembrane Phosphatase with TEnsin homology) gene family encodes a PTEN-related tyrosine phosphatase with four potential transmembrane domains. Chromosomal mapping revealed multiple copies of the TPTE gene on chromosomes 13, 15, 21, 22 and Y. Human chromosomes 13 and 21 copies encode two functional proteins, TPIP (TPTE and PTEN homologous Inositol lipid Phosphatase) and TPTE, respectively, whereas only one copy of the gene exists in the mouse genome. In the present study, we show that TPTE and TPIP proteins are expressed in secondary spermatocytes and/or prespermatids. In addition, we report the existence of several novel alternatively spliced isoforms of these two proteins with variable number of transmembrane domains. The latter has no influence on the subcellular localization of these different peptides as shown by co-immunofluorescence experiments. Finally, we identify another expressed TPTE copy, mapping to human chromosome 22, whose transcription appears to be under the control of the LTR of human endogenous retrovirus RTVL-H3.
Collapse
Affiliation(s)
- Caroline Tapparel
- Division of Medical Genetics, Centre Medical Universitaire, University of Geneva Medical School, 1, Rue Michel Servet CH-1211, Geneva, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
237
|
Zhou XP, Marsh DJ, Morrison CD, Chaudhury AR, Maxwell M, Reifenberger G, Eng C. Germline inactivation of PTEN and dysregulation of the phosphoinositol-3-kinase/Akt pathway cause human Lhermitte-Duclos disease in adults. Am J Hum Genet 2003; 73:1191-8. [PMID: 14566704 PMCID: PMC1180498 DOI: 10.1086/379382] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2003] [Accepted: 08/29/2003] [Indexed: 01/17/2023] Open
Abstract
Lhermitte-Duclos disease (LDD), or dysplastic gangliocytoma of the cerebellum, is an unusual hamartomatous overgrowth disorder. LDD can be familial or, more commonly, sporadic. It has been only recently recognized that LDD may be associated with Cowden syndrome (CS). Over 80% of patients with CS carry germline mutations in PTEN. It remains unclear whether all cases of LDD, even without features of CS, are caused by germline PTEN mutation and whether somatic PTEN mutation occurs in sporadic LDD. We obtained paraffin-embedded LDD lesions from 18 unselected, unrelated patients and performed mutational analysis of PTEN. Overall, 15 (83%) of 18 samples were found to carry a PTEN mutation. All individuals with mutations were adult-onset patients, but the three without mutations were diagnosed at the ages of 1, 3, and 11 years. Germline DNA was available from six adult-onset cases, and all had germline PTEN mutations. Of these six, two had CS features, one did not have CS features, and three were of unknown CS status. Immunohistochemistry revealed that 75% of the LDD samples had complete or partial loss of PTEN expression accompanied by elevated phosphorylated Akt, specifically in the dysplastic gangliocytoma cells. These data suggest that the loss of PTEN function is sufficient to cause LDD. The high frequency and spectrum of germline PTEN mutations in patients ascertaining by LDD alone confirm that LDD is an important defining feature of CS. Individuals with LDD, even without apparent CS features, should be counseled as in CS.
Collapse
Affiliation(s)
- Xiao-Ping Zhou
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology, and Medical Genetics, and the Division of Human Genetics, Department of Internal Medicine, and Department of Pathology, The Ohio State University, Columbus; Translational Research Laboratory, Department of Adult Oncology, Charles A. Dana Human Cancer Genetics Unit, Dana-Farber Cancer Institute, Boston; Neurosurgical Service, Massachusetts General Hospital, Department of Surgery, Harvard Medical School, Boston; Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, and the Department of Molecular Medicine, University of Sydney, Sydney, Australia; Institut für Neuropathologie, Heinrich Heine Universität Düsseldorf, Düsseldorf; and Cancer Research UK, Human Cancer Genetics Research Group, University of Cambridge, Cambridge
| | - Deborah J. Marsh
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology, and Medical Genetics, and the Division of Human Genetics, Department of Internal Medicine, and Department of Pathology, The Ohio State University, Columbus; Translational Research Laboratory, Department of Adult Oncology, Charles A. Dana Human Cancer Genetics Unit, Dana-Farber Cancer Institute, Boston; Neurosurgical Service, Massachusetts General Hospital, Department of Surgery, Harvard Medical School, Boston; Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, and the Department of Molecular Medicine, University of Sydney, Sydney, Australia; Institut für Neuropathologie, Heinrich Heine Universität Düsseldorf, Düsseldorf; and Cancer Research UK, Human Cancer Genetics Research Group, University of Cambridge, Cambridge
| | - Carl D. Morrison
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology, and Medical Genetics, and the Division of Human Genetics, Department of Internal Medicine, and Department of Pathology, The Ohio State University, Columbus; Translational Research Laboratory, Department of Adult Oncology, Charles A. Dana Human Cancer Genetics Unit, Dana-Farber Cancer Institute, Boston; Neurosurgical Service, Massachusetts General Hospital, Department of Surgery, Harvard Medical School, Boston; Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, and the Department of Molecular Medicine, University of Sydney, Sydney, Australia; Institut für Neuropathologie, Heinrich Heine Universität Düsseldorf, Düsseldorf; and Cancer Research UK, Human Cancer Genetics Research Group, University of Cambridge, Cambridge
| | - Abhik R. Chaudhury
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology, and Medical Genetics, and the Division of Human Genetics, Department of Internal Medicine, and Department of Pathology, The Ohio State University, Columbus; Translational Research Laboratory, Department of Adult Oncology, Charles A. Dana Human Cancer Genetics Unit, Dana-Farber Cancer Institute, Boston; Neurosurgical Service, Massachusetts General Hospital, Department of Surgery, Harvard Medical School, Boston; Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, and the Department of Molecular Medicine, University of Sydney, Sydney, Australia; Institut für Neuropathologie, Heinrich Heine Universität Düsseldorf, Düsseldorf; and Cancer Research UK, Human Cancer Genetics Research Group, University of Cambridge, Cambridge
| | - Marius Maxwell
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology, and Medical Genetics, and the Division of Human Genetics, Department of Internal Medicine, and Department of Pathology, The Ohio State University, Columbus; Translational Research Laboratory, Department of Adult Oncology, Charles A. Dana Human Cancer Genetics Unit, Dana-Farber Cancer Institute, Boston; Neurosurgical Service, Massachusetts General Hospital, Department of Surgery, Harvard Medical School, Boston; Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, and the Department of Molecular Medicine, University of Sydney, Sydney, Australia; Institut für Neuropathologie, Heinrich Heine Universität Düsseldorf, Düsseldorf; and Cancer Research UK, Human Cancer Genetics Research Group, University of Cambridge, Cambridge
| | - Guido Reifenberger
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology, and Medical Genetics, and the Division of Human Genetics, Department of Internal Medicine, and Department of Pathology, The Ohio State University, Columbus; Translational Research Laboratory, Department of Adult Oncology, Charles A. Dana Human Cancer Genetics Unit, Dana-Farber Cancer Institute, Boston; Neurosurgical Service, Massachusetts General Hospital, Department of Surgery, Harvard Medical School, Boston; Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, and the Department of Molecular Medicine, University of Sydney, Sydney, Australia; Institut für Neuropathologie, Heinrich Heine Universität Düsseldorf, Düsseldorf; and Cancer Research UK, Human Cancer Genetics Research Group, University of Cambridge, Cambridge
| | - Charis Eng
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology, and Medical Genetics, and the Division of Human Genetics, Department of Internal Medicine, and Department of Pathology, The Ohio State University, Columbus; Translational Research Laboratory, Department of Adult Oncology, Charles A. Dana Human Cancer Genetics Unit, Dana-Farber Cancer Institute, Boston; Neurosurgical Service, Massachusetts General Hospital, Department of Surgery, Harvard Medical School, Boston; Cancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, and the Department of Molecular Medicine, University of Sydney, Sydney, Australia; Institut für Neuropathologie, Heinrich Heine Universität Düsseldorf, Düsseldorf; and Cancer Research UK, Human Cancer Genetics Research Group, University of Cambridge, Cambridge
| |
Collapse
|
238
|
Waite KA, Eng C. From developmental disorder to heritable cancer: it's all in the BMP/TGF-beta family. Nat Rev Genet 2003; 4:763-73. [PMID: 14526373 DOI: 10.1038/nrg1178] [Citation(s) in RCA: 217] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Transforming growth factor-beta (TGF-beta) regulates many cellular processes through complex signal-transduction pathways that have crucial roles in normal development. Disruption of these pathways can lead to a range of diseases, including cancer. Mutations in the genes that encode members of the TGF-beta pathway are involved in vascular diseases as well as gastrointestinal neoplasia. More recently, they have been implicated in Cowden syndrome, which is normally associated with mutations in the phosphatase and tensin homologue gene PTEN. Molecular studies of TGF-beta signalling are now showing why mutations in genes that encode components of this pathway result in inherited cancer and developmental diseases.
Collapse
Affiliation(s)
- Kristin A Waite
- Human Cancer Genetics and Clinical Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, Ohio 43210, USA
| | | |
Collapse
|
239
|
Saito Y, Swanson X, Mhashilkar AM, Oida Y, Schrock R, Branch CD, Chada S, Zumstein L, Ramesh R. Adenovirus-mediated transfer of the PTEN gene inhibits human colorectal cancer growth in vitro and in vivo. Gene Ther 2003; 10:1961-9. [PMID: 14528320 DOI: 10.1038/sj.gt.3302100] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The tumor-suppressor gene PTEN encodes a multifunctional phosphatase that is mutated in a variety of human cancers. PTEN inhibits the phosphatidylinositol 3-kinase pathway and downstream functions, including activation of Akt/protein kinase B (PKB), cell survival, and cell proliferation in tumor cells carrying mutant- or deletion-type PTEN. In such tumor cells, enforced expression of PTEN decreases cell proliferation through cell-cycle arrest at G1 phase accompanied, in some cases, by induction of apoptosis. More recently, the tumor-suppressive effect of PTEN has been reported in ovarian and thyroid tumors that are wild type for PTEN. In the present study, we examined the tumor-suppressive effect of PTEN in human colorectal cancer cells that are wild type for PTEN. Adenoviral-mediated transfer of PTEN (Ad-PTEN) suppressed cell growth and induced apoptosis significantly in colorectal cancer cells (DLD-1, HT29, and SW480) carrying wtPTEN than in normal colon fibroblast cells (CCD-18Co) carrying wtPTEN. This suppression was induced through downregulation of the Akt/PKB pathway, dephosphorylation of focal adhesion kinase (FAK) and mitogen-activated protein kinase (MAPK) and cell-cycle arrest at the G2/M phase, but not the G1 phase. Furthermore, treatment of human colorectal tumor xenografts (HT-29, and SW480) with Ad-PTEN resulted in significant (P=0.01) suppression of tumor growth. These results indicate that Ad-PTEN exerts its tumor-suppressive effect on colorectal cancer cells through inhibition of cell-cycle progression and induction of cell death. Thus Ad-PTEN may be a potential therapeutic for treatment of colorectal cancers.
Collapse
Affiliation(s)
- Y Saito
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Abstract
The inherited hamartoma polyposis syndromes encompass several distinct clinical syndromes with different genetic bases, Cowden syndrome (CS), Bannayan-Riley-Ruvalcaba syndrome (BRRS), juvenile polyposis syndrome (JPS), and Peutz-Jeghers syndrome (PJS). Germline mutations in PTEN, encoding a tumor suppressor phosphatase on 10q23.3, is associated with 80% of CS and 60% of BRRS. JPS is caused by mutations in MADH4 and BMPR1A, encoding two members of the TGFB superfamily. Germline mutations in LKB1 (STK11) are associated with a subset of PJS. The number, distribution, and histologic type of polyps differ amongst these syndromes as do component cancer risks. While rare, usually asymptomatic, hamartomatous polyps are felt to be component to CS. Hamartomatous polyposis is usually prominent and symptomatic in BRRS. Polyposis, which can be quite symptomatic, is a cardinal component feature of PJS and JPS. Interestingly, glycogenic acanthosis of the esophagus is highly predictive of CS and the presence of PTEN mutation. PTEN mutation positive CS have been shown to be at increased risk of breast, thyroid, and endometrial cancer. PTEN mutation positive BRRS are at increased risk of at least breast cancer, possibly that of the thyroid as well. In contrast, JPS and PJS have increased risk of gastrointestinal cancers in particular. Thus, molecular-based diagnoses to differentiate each of these syndromes are important for medical management.
Collapse
Affiliation(s)
- Charis Eng
- Clinical Cancer Genetics Program and Human Cancer Genetics Program, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, USA.
| |
Collapse
|
241
|
Lowichik A, Jackson WD, Coffin CM. Gastrointestinal polyposis in childhood: clinicopathologic and genetic features. Pediatr Dev Pathol 2003; 6:371-91. [PMID: 14708731 DOI: 10.1007/s10024-002-0701-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Gastrointestinal polyps and certain extraintestinal lesions in children may herald a hereditary polyposis syndrome, with an increased risk of neoplasia and other health problems for both children and their relatives. The availability of molecular/genetic screening tests has increased early diagnosis of younger members of known polyposis families. This article reviews the gross and microscopic features of polyposis syndromes of childhood and summarizes the molecular/genetic advances in this field. Clinical management is also briefly discussed.
Collapse
Affiliation(s)
- Amy Lowichik
- Department of Pathology, University of Utah Health Sciences Center, 30 N. 1900 E, Salt Lake City, UT 84132-2501, USA.
| | | | | |
Collapse
|
242
|
Abstract
PTEN, on 10q23.3, encodes a major lipid phosphatase which signals down the phosphoinositol-3-kinase/Akt pathway and effects G1 cell cycle arrest and apoptosis. Germline PTEN mutations have been found to occur in 80% of classic Cowden syndrome (CS), 60% of Bannayan-Riley-Ruvalcaba syndrome (BRRS), up to 20% of Proteus syndrome (PS), and approximately 50% of a Proteus-like syndrome (PSL). CS is a heritable multiple hamartoma syndrome with a high risk of breast, thyroid, and endometrial carcinomas. BRRS is a congenital autosomal dominant disorder characterized by megencephaly, developmental delay, lipomatosis, and speckled penis. PS and PSL had never been associated with risk of malignancy. Finding germline PTEN mutations in patients with BRRS, PS, and PSL suggests equivalent risks of developing malignancy as in CS with implications for medical management. The mutational spectra of CS and BRRS overlap, with many of the mutations occurring in exons 5, 7, and 8. Genotype-phenotype association analyses have revealed that the presence of germline PTEN mutations is associated with breast tumor development, and that mutations occurring within and 5' of the phosphatase motif were associated with multi-organ involvement. Pooled analysis of PTEN mutation series of CS and BRRS occurring in the last five years reveals that 65% of CS-associated mutations occur in the first five exons encoding the phosphatase domain and the promoter region, while 60% of BRRS-associated mutations occur in the 3' four exons encoding mainly the C2 domain. Somatic PTEN mutations occur with a wide distribution of frequencies in sporadic primary tumors, with the highest frequencies in endometrial carcinomas and glioblastoma multiform. Several mechanisms of PTEN inactivation occur in primary malignancies derived from different tissues, but a favored mechanism appears to occur in a tissue-specific manner. Inappropriate subcellular compartmentalization and increased/decreased proteosome degradation may be two novel mechanisms of PTEN inactivation. Further functional work could reveal more effective means of molecular-directed therapy and prevention.
Collapse
Affiliation(s)
- Charis Eng
- Clinical Cancer Genetics Program and Human Cancer Genetics Program, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, USA.
| |
Collapse
|
243
|
Zhou XP, Waite KA, Pilarski R, Hampel H, Fernandez MJ, Bos C, Dasouki M, Feldman GL, Greenberg LA, Ivanovich J, Matloff E, Patterson A, Pierpont ME, Russo D, Nassif NT, Eng C. Germline PTEN promoter mutations and deletions in Cowden/Bannayan-Riley-Ruvalcaba syndrome result in aberrant PTEN protein and dysregulation of the phosphoinositol-3-kinase/Akt pathway. Am J Hum Genet 2003; 73:404-11. [PMID: 12844284 PMCID: PMC1180378 DOI: 10.1086/377109] [Citation(s) in RCA: 195] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2003] [Accepted: 05/22/2003] [Indexed: 01/18/2023] Open
Abstract
Germline intragenic mutations in PTEN are associated with 80% of patients with Cowden syndrome (CS) and 60% of patients with Bannayan-Riley-Ruvalcaba syndrome (BRRS). The underlying genetic causes remain to be determined in a considerable proportion of classic CS and BRRS without a polymerase chain reaction (PCR)-detectable PTEN mutation. We hypothesized that gross gene deletions and mutations in the PTEN promoter might alternatively account for a subset of apparently mutation-negative patients with CS and BRRS. Using real time and multiplex PCR techniques, we identified three germline hemizygous PTEN deletions in 122 apparently mutation-negative patients with classic CS (N=95) or BRRS (N=27). Fine mapping suggested that one deletion encompassed the whole gene and the other two included exon 1 and encompassed exons 1-5 of PTEN, respectively. Two patients with the deletion were diagnosed with BRRS, and one patient with the deletion was diagnosed with BRRS/CS overlap (features of both). Thus 3 (11%) of 27 patients with BRRS or BRRS/CS-overlap had PTEN deletions. Analysis of the PTEN promoter revealed nine cases (7.4%) harboring heterozygous germline mutations. All nine had classic CS, representing almost 10% of all subjects with CS. Eight had breast cancers and/or benign breast tumors but, otherwise, oligo-organ involvement. PTEN protein analysis, from one deletion-positive and five PTEN-promoter-mutation-positive samples, revealed a 50% reduction in protein and multiple bands of immunoreactive protein, respectively. In contrast, control samples showed only the expected band. Further, an elevated level of phosphorylated Akt was detected in the five promoter-mutation-positive samples, compared with controls, indicating an absence of or marked reduction in functional PTEN. These data suggest that patients with BRRS and CS without PCR-detected intragenic PTEN mutations be offered clinical deletion analysis and promoter-mutation analysis, respectively.
Collapse
Affiliation(s)
- Xiao-Ping Zhou
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Kristin A. Waite
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Robert Pilarski
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Heather Hampel
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Magali J. Fernandez
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Cindy Bos
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Majed Dasouki
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Gerald L. Feldman
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Lois A. Greenberg
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Jennifer Ivanovich
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Ellen Matloff
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Annette Patterson
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Mary Ella Pierpont
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Donna Russo
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Najah T. Nassif
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Charis Eng
- Clinical Cancer Genetics and Human Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Genetics, Department of Internal Medicine, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus; Ferguson Inherited Colon Cancer Registry, Grand Rapids, MI; Division of Genetics, Children’s Mercy Hospital, Kansas City, MO; Molecular Medicine and Genetics and Karmanos Cancer Center Institute, Detroit Medical Center/Wayne State University School of Medicine, Detroit; Cancer Genetics Program, Bennett Cancer Center, Stamford Hospital, Stamford, CT; Pediatric Clinic, Washington University School of Medicine, St. Louis; Department of Genetics, Yale Cancer Center, New Haven, CT; University of Texas Southwestern Medical Center, Dallas; Children’s Hospital, St. Paul; University of Minnesota, Minneapolis; Cancer Genetics Program, NYP Columbia Presbyterian Hospital, New York; Cancer Research Laboratories, South West Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, Australia; and Cancer Research UK Human Cancer Genetics Research Group, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
244
|
Abstract
Therapeutic resistance and proclivity for metastasis are hallmarks of malignant melanoma. Genetic, epidemiological and genomic investigations are uncovering the spectrum of stereotypical mutations that are associated with melanoma and how these mutations relate to risk factors such as ultraviolet exposure. The ability to validate the pathogenetic relevance of these mutations in the mouse, coupled with advances in rational drug design, has generated optimism for the development of effective prevention programmes, diagnostic measures and targeted therapeutics in the near future.
Collapse
Affiliation(s)
- Lynda Chin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
245
|
Marchese C, Montera M, Torrini M, Goldoni F, Mareni C, Forni M, Locatelli L. Granular cell tumor in a PHTS patient with a novel germline PTEN mutation. Am J Med Genet A 2003; 120A:286-8. [PMID: 12833416 DOI: 10.1002/ajmg.a.20179] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
246
|
Goldsmith MI, Fisher S, Waterman R, Johnson SL. Saltatory control of isometric growth in the zebrafish caudal fin is disrupted in long fin and rapunzel mutants. Dev Biol 2003; 259:303-17. [PMID: 12871703 DOI: 10.1016/s0012-1606(03)00186-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Zebrafish fins grow by sequentially adding new segments of bone to the distal end of each fin ray. In wild type zebrafish, segment addition is regulated such that an isometric relationship is maintained between fin length and body length over the lifespan of the growing fish. Using a novel, surrogate marker for fin growth in conjunction with cell proliferation assays, we demonstrate here that segment addition is not continuous, but rather proceeds by saltation. Saltation is a fundamental growth mechanism shared by disparate vertebrates, including humans. We further demonstrate that segment addition proceeds in conjunction with cyclic bursts of cell proliferation in the distal fin ray mesenchyme. In contrast, cells in the distal fin epidermis proliferate at a constant rate throughout the fin ray growth cycle. Finally, we show that two separate fin overgrowth mutants, long fin and rapunzel, bypass the stasis phase of the fin ray growth cycle to develop asymmetrical and symmetrical fin overgrowth, respectively.
Collapse
Affiliation(s)
- Matthew I Goldsmith
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | | | |
Collapse
|
247
|
Ricci R, Komminoth P, Bannwart F, Torhorst J, Wight E, Heitz PU, Caduff RF. PTEN as a molecular marker to distinguish metastatic from primary synchronous endometrioid carcinomas of the ovary and uterus. DIAGNOSTIC MOLECULAR PATHOLOGY : THE AMERICAN JOURNAL OF SURGICAL PATHOLOGY, PART B 2003; 12:71-8. [PMID: 12766611 DOI: 10.1097/00019606-200306000-00002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The distinction between two primary carcinomas on the one hand and a metastatic disease on the other hand in patients suffering from synchronous endometrioid carcinomas of the uterus and ovary is difficult. Exclusive histopathologic analysis appears to be insufficient and sometimes misleading. The tumor suppressor PTEN was found to be important in early neoplastic transformation in endometrioid carcinomas of the uterus. In this study, we screened synchronous endometrioid carcinomas of the uterus and ovary of 10 patients for loss of heterozygosity using seven different microsatellite markers at 10q23.3 and for mutations in the entire coding region of PTEN. Point mutations or microdeletions/insertions were found in six patients. Allelic loss at 10q23.3 was detected in eight patients. Based on conventional histology, a metastatic disease was diagnosed in seven patients and a concomitant uterine and ovarian carcinoma in three patients. After molecular analysis, the histopathologic diagnosis of three patients had to be revised. Histopathology represents the standard method to process tumor specimens from these patients. Nevertheless, mutation screen for PTEN and LOH analysis at 10q23.3 provide helpful genetic tools to establish a correct final diagnosis, which is important in view of prognosis and therapeutic implications.
Collapse
Affiliation(s)
- Romeo Ricci
- Department of Pathology, University Hospital of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
248
|
McGarrity TJ, Wagner Baker MJ, Ruggiero FM, Thiboutot DM, Hampel H, Zhou XP, Eng C. GI polyposis and glycogenic acanthosis of the esophagus associated with PTEN mutation positive Cowden syndrome in the absence of cutaneous manifestations. Am J Gastroenterol 2003; 98:1429-34. [PMID: 12818292 DOI: 10.1111/j.1572-0241.2003.07496.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A 62-yr-old man was referred for management of GI polyposis. Large bowel polyps were initially diagnosed >25 yr ago, and the patient had undergone multiple colonoscopies and polypectomies. Personal and family history were notable for thyroid goiter and hypothyroidism. Physical examination was notable for lingular papillomatosis. No cutaneous lesions were seen. Upper endoscopy revealed esophageal glycogen acanthosis. There were multiple polyps throughout the stomach and the small and large intestines. Histology of these polyps showed multiple cell types including juvenile polyps, inflammatory polyps with fibromuscular proliferation and lamina propria ganglion cells, and focal adenomatous change. A clinical diagnosis of Cowden syndrome was made. Mutation analysis revealed a variant in exon 8 of the PTEN gene. Direct sequencing revealed a germline heterozygous C.892-895InsA, which is predicted to result in a truncated PTEN protein. Cowden syndrome is an underdiagnosed, underrecognized, autosomal dominant, inherited syndrome. For the gastroenterologist, esophageal acanthosis and multiple hamartomatous polyps should suggest the diagnosis. Sensitive molecular diagnostic tests looking for mutations in the appropriate genes are clinically available. Together with genetic counseling, molecular diagnostic testing will allow more accurate risk assessment and surveillance for cancer for both the patient and family members.
Collapse
Affiliation(s)
- Thomas J McGarrity
- Department of Medicine, Penn State Cancer Institute, The Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
249
|
Hughes JL, Reidy JF. Imaging and treatment of uterine fibroids including the role of uterine artery embolisation. IMAGING 2003. [DOI: 10.1259/img.15.2.150079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
250
|
Gary DS, Mattson MP. PTEN regulates Akt kinase activity in hippocampal neurons and increases their sensitivity to glutamate and apoptosis. Neuromolecular Med 2003; 2:261-9. [PMID: 12622404 DOI: 10.1385/nmm:2:3:261] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The tumor suppressor phosphatase PTEN can promote apoptosis of mitotic cells by inhibiting activation of the cell survival kinase Akt. PTEN is essential for normal embryonic development, PTEN expression is associated with neuronal differentiation, and deletion of PTEN in the mouse brain results in seizures, ataxia, and other abnormalities. However, the possible roles of PTEN in regulating neuronal survival are not known. We provide evidence that PTEN sensitizes hippocampal neurons to excitotoxic death in culture and in vivo. Overexpression of wild-type PTEN decreased, while a dominant-negative PTEN increased, levels of activated Akt in cultured hippocampal neurons. Wild-type PTEN promoted, while dominant-negative PTEN prevented, apoptotic death of neurons exposed to the excitatory amino acid neurotransmitter glutamate. Hippocampal neurons of mice with reduced PTEN levels were more resistant to seizure-induced death compared to wild-type littermates. These findings demonstrate a cell death function of PTEN in hippocampal neurons and identify PTEN as a potential therapeutic target for neurodegenerative disorders that involve excitotoxicity and apoptosis. The ability of PTEN to modify neuronal sensitivity to glutamate also suggests possible roles for PTEN in regulating developmental and synaptic plasticity.
Collapse
Affiliation(s)
- Devin S Gary
- Laboratory of Neurosciences, National Institute on Aging Gerontology Research Center, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | |
Collapse
|