351
|
Lukong KE, Richard S. Targeting the RNA-binding protein Sam68 as a treatment for cancer? Future Oncol 2008; 3:539-44. [PMID: 17927519 DOI: 10.2217/14796694.3.5.539] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The contradictory properties of RNA-binding proteins (RBPs) have mystified their roles in human diseases including cancer. Are certain RBPs oncogenes or tumor suppressors? In the case of the signal transduction activator of RNA metabolism (STAR) family of hnRNP K homology (KH)-domain-containing RBPs, the dominant view with loose experimental evidence is that these proteins are tumor suppressors. However, recent developments support a pro-oncogenic role for archetypical STAR protein Sam68. Sam68-null mice are not prone to cancer, but instead display pronounced defects in mammary gland ductal development, and haploinsufficiency of Sam68 impedes mammary tumor onset and tumor multiplicity in mouse models expressing the mammary-targeted polyoma middle T antigen oncogene. These advances have increased the interest in the role of Sam68 as a positive regulator of cancer progression and position Sam68 as a viable therapeutic target. Retrospective and perspective implications of Sam68 in cancer are discussed.
Collapse
Affiliation(s)
- Kiven E Lukong
- Lady Davis Institute, 3755 Côte Ste.-Catherine Road, Montréal, Québec H3T 1E2, Canada.
| | | |
Collapse
|
352
|
Fraser MM, Bayazitov IT, Zakharenko SS, Baker SJ. Phosphatase and tensin homolog, deleted on chromosome 10 deficiency in brain causes defects in synaptic structure, transmission and plasticity, and myelination abnormalities. Neuroscience 2008; 151:476-88. [PMID: 18082964 PMCID: PMC2278004 DOI: 10.1016/j.neuroscience.2007.10.048] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 10/10/2007] [Accepted: 10/17/2007] [Indexed: 01/08/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K) signaling pathway modulates growth, proliferation and cell survival in diverse tissue types and plays specialized roles in the nervous system including influences on neuronal polarity, dendritic branching and synaptic plasticity. The tumor-suppressor phosphatase with tensin homology (PTEN) is the central negative regulator of the PI3K pathway. Germline PTEN mutations result in cancer predisposition, macrocephaly and benign hamartomas in many tissues, including Lhermitte-Duclos disease, a cerebellar growth disorder. Neurological abnormalities including autism, seizures and ataxia have been observed in association with inherited PTEN mutation with variable penetrance. It remains unclear how loss of PTEN activity contributes to neurological dysfunction. To explore the effects of Pten deficiency on neuronal structure and function, we analyzed several ultra-structural features of Pten-deficient neurons in Pten conditional knockout mice. Using Golgi stain to visualize full neuronal morphology, we observed that increased size of nuclei and somata in Pten-deficient neurons was accompanied by enlarged caliber of neuronal projections and increased dendritic spine density. Electron microscopic evaluation revealed enlarged abnormal synaptic structures in the cerebral cortex and cerebellum. Severe myelination defects included thickening and unraveling of the myelin sheath surrounding hypertrophic axons in the corpus callosum. Defects in myelination of axons of normal caliber were observed in the cerebellum, suggesting intrinsic abnormalities in Pten-deficient oligodendrocytes. We did not observe these abnormalities in wild-type or conditional Pten heterozygous mice. Moreover, conditional deletion of Pten drastically weakened synaptic transmission and synaptic plasticity at excitatory synapses between CA3 and CA1 pyramidal neurons in the hippocampus. These data suggest that Pten is involved in mechanisms that control development of neuronal and synaptic structures and subsequently synaptic function.
Collapse
Affiliation(s)
- Melissa M. Fraser
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 332, N. Lauderdale, Memphis, TN 38105
| | - Ildar T. Bayazitov
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 332, N. Lauderdale, Memphis, TN 38105
| | - Stanislav S. Zakharenko
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 332, N. Lauderdale, Memphis, TN 38105
| | - Suzanne J. Baker
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 332, N. Lauderdale, Memphis, TN 38105
| |
Collapse
|
353
|
Janas ML, Hodson D, Stamataki Z, Hill S, Welch K, Gambardella L, Trotman LC, Pandolfi PP, Vigorito E, Turner M. The effect of deleting p110delta on the phenotype and function of PTEN-deficient B cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 180:739-46. [PMID: 18178811 DOI: 10.4049/jimmunol.180.2.739] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Control of the intracellular levels of phosphatidylinositol-(3, 4, 5)-trisphosphate by PI3K and phosphatase and tensin homolog (PTEN) is essential for B cell development and differentiation. Deletion of the PI3K catalytic subunit p110delta leads to a severe reduction in B1 and marginal zone (MZ) B cells, whereas deletion of PTEN results in their expansion. We have examined the relationship between these two molecules by generating mice with a B cell-specific deletion of PTEN (PTENB) and a concurrent germline deletion of p110delta. The expanded B1 cell population of PTENB mice was reduced to normal levels in PTENB/p110delta mutant mice, indicating a critical role for the p110delta isoform in the expansion of B1 cells. However, numbers of MZ B cells in the PTENB/p110delta mutants was intermediate between wild-type and PTENB-deficient mice, suggesting an additional role for other PI3K catalytic isoforms in MZ differentiation. Furthermore, the defective class switch recombination in PTENB B cells was only partially reversed in PTENB/p110delta double mutant B cells. These results demonstrate an epistatic relationship between p110delta and PTEN. In addition, they also suggest that additional PI3K catalytic subunits contribute to B cell development and function.
Collapse
Affiliation(s)
- Michelle L Janas
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Babraham, Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
354
|
PIP3 pathway in regulatory T cells and autoimmunity. Immunol Res 2008; 39:194-224. [PMID: 17917066 DOI: 10.1007/s12026-007-0075-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/07/2023]
Abstract
Regulatory T cells (Tregs) play an important role in preventing both autoimmune and inflammatory diseases. Many recent studies have focused on defining the signal transduction pathways essential for the development and the function of Tregs. Increasing evidence suggest that T-cell receptor (TCR), interleukin-2 (IL-2) receptor (IL-2R), and co-stimulatory receptor signaling are important in the early development, peripheral homeostasis, and function of Tregs. The phosphoinositide-3 kinase (PI3K)-regulated pathway (PIP3 pathway) is one of the major signaling pathways activated upon TCR, IL-2R, and CD28 stimulation, leading to T-cell activation, proliferation, and cell survival. Activation of the PIP3 pathway is also negatively regulated by two phosphatidylinositol phosphatases SHIP and PTEN. Several mouse models deficient for the molecules involved in PIP3 pathway suggest that impairment of PIP3 signaling leads to dysregulation of immune responses and, in some cases, autoimmunity. This review will summarize the current understanding of the importance of the PIP3 pathway in T-cell signaling and the possible roles this pathway performs in the development and the function of Tregs.
Collapse
|
355
|
Tokyol C, Aktepe F, Hüsniye Dilek F, Yilmazer M. Comparison of placental PTEN and beta1 integrin expression in early spontaneous abortion, early and late normal pregnancy. Ups J Med Sci 2008; 113:235-42. [PMID: 18509818 DOI: 10.3109/2000-1967-231] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND PTEN seems to play an important role in cell cycle, growth, migration, and death. Integrins are cell surface receptors that play a role in the regulation of cell proliferation, differentiation, implantation, and embryogenesis. PTEN inhibits beta1 integrin signaling. The objective of this study is to investigate the expression of PTEN and beta1 integrin in placental tissues of early spontaneous abortion and first and third trimesters of normal pregnancy. METHOD A total of 43 placental tissue samples were evaluated using immunohistochemistry for PTEN and beta1 integrin. Group 1 included placental tissues of volunteer termination of normal pregnancy during the first trimester (5-10 wk gestation). Group 2 included placental tissues of normal vaginal delivery at the third trimester of pregnancy (36-40 wk gestation). Group 3 included placental tissues of pregnancy termination because of spontaneous abortion during the first trimester (5-10 wk gestation). RESULTS PTEN expression of villous trophoblast was decreasing as the pregnancy advanced. PTEN staining of decidual cells was significantly stronger in tissue samples from early spontaneous abortion than in tissue samples from early and late normal pregnancy (p=0.003, p=0.001, respectively). There was no significant difference between beta1 integrin expression of villous trophoblast and decidual cells in three groups. CONCLUSION Our findings suggest that altered patterns of PTEN expression may be associated with abortion, but it seems that beta1 integrin does not contribute to this process as a signaling protein. Further evaluation is needed to highlight this subject.
Collapse
Affiliation(s)
- Cigdem Tokyol
- Department of Pathology, Afyon Kocatepe University School of Medicine, Afyonkarahisar, Turkey.
| | | | | | | |
Collapse
|
356
|
van Diepen MT, Eickholt BJ. Function of PTEN during the formation and maintenance of neuronal circuits in the brain. Dev Neurosci 2008; 30:59-64. [PMID: 18075255 DOI: 10.1159/000109852] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2006] [Accepted: 01/30/2007] [Indexed: 11/19/2022] Open
Abstract
PTEN (phosphatase and tensin homologue deleted on chromosome 10) is a tumor suppressor that can inhibit proliferation and migration and controls apoptosis in a number of cell types, mainly through inhibition of the phosphoinositide 3-kinase (PI3K) signaling pathway. Patients carrying inactivating mutations of PTEN show a prevalence to develop tumors that can coincide with neurological defects such as mental retardation, ataxia and seizures. A number of in vitro and in vivo studies were instrumental in uncovering a direct correlation between deregulated PI3K/PTEN signaling and changes in neuronal morphogenesis, which is likely to have profound bearings upon the pathogenesis of neurological symptoms. This review outlines recent work on the function of PTEN during vertebrate brain development and the current understanding of the signaling pathways downstream of PTEN that control neuronal connectivity in the brain.
Collapse
|
357
|
Hormone refractory prostate cancer: Lessons learned from the PTEN prostate cancer model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 617:87-95. [PMID: 18497033 DOI: 10.1007/978-0-387-69080-3_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
358
|
Strumane K, Song JY, Baas I, Collard JG. Increased Rac activity is required for the progression of T-lymphomas induced by Pten-deficiency. Leuk Res 2008; 32:113-20. [PMID: 17521720 DOI: 10.1016/j.leukres.2007.03.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Revised: 01/30/2007] [Accepted: 03/30/2007] [Indexed: 10/23/2022]
Abstract
Mutation of the tumor suppressor PTEN results in loss of its PI3-kinase counteracting function. PI3-kinase stimulates tumor formation by PKB/Akt-mediated cell proliferation and prevention of apoptosis. PI3-kinase may also activate Rho-GTPases and their regulatory GEFs to promote invasion. Here we have analyzed the function of the Rac-specific activator, Tiam1, in PI3-kinase-induced T-lymphomagenesis. Mice with a T cell-specific Pten deletion developed T-lymphomas with enhanced PKB/Akt phosphorylation. However, these T-lymphomas infiltrated more frequently into various organs in Tiam1-deficient mice compared to wild type mice. Surprisingly, Tiam1-deficient lymphomas showed increased Rac activity, suggesting that the lack of Tiam1 is compensated by alternative Rac-activating mechanisms that lead to increased progression of PI3-kinase-induced T-lymphomas.
Collapse
Affiliation(s)
- Kristin Strumane
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
359
|
Hagenbeek TJ, Spits H. T-cell lymphomas in T-cell-specific Pten-deficient mice originate in the thymus. Leukemia 2007; 22:608-19. [PMID: 18046443 DOI: 10.1038/sj.leu.2405056] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (Pten) is a tumor suppressor protein whose loss of lipid phosphatase activity is associated with lymphomagenesis. We made use of the Cre-loxP system to delete Pten expression in Lck- or CD4-expressing T-lineage cells. Mice initially showed modest thymic hyperplasia and subsequently developed expanding and infiltrating T-cell lymphomas, leading to a premature death within 5 to 23 weeks. Frequently, all thymocyte and peripheral T-cell populations displayed phenotypes characteristic for immature developing thymocyte precursors and shared elevated levels of clonally rearranged T-cell receptor (TCR) beta chains. In concert, CD2, CD5, CD3epsilon and CD44, proteins associated with increased expression and signaling capacity of both the immature pre-TCR and the mature alphabetaTCR, were more abundantly expressed, reflecting a constitutive state of activation. Although most T-cell lymphomas had acquired the capability to infiltrate the periphery, not all populations left the thymus and expanded clonally exclusively in the thymus. In line with this, only transplantation of thymocytes with infiltrating capacity gave rise to T-cell lymphoma in immunodeficient recipients. These results indicate that T-cell-specific Pten deletion during various stages of thymocyte development gives rise to clonally expanding T-cell lymphomas that frequently infiltrate the periphery, but originate in the thymus.
Collapse
Affiliation(s)
- T J Hagenbeek
- Department of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
360
|
Lu TL, Chang JL, Liang CC, You LR, Chen CM. Tumor spectrum, tumor latency and tumor incidence of the Pten-deficient mice. PLoS One 2007; 2:e1237. [PMID: 18043744 PMCID: PMC2077932 DOI: 10.1371/journal.pone.0001237] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Accepted: 11/01/2007] [Indexed: 11/18/2022] Open
Abstract
Background Pten functionally acts as a tumor suppressor gene. Lately, tissue-specific ablation of Pten gene in mice has elucidated the role of Pten in different tumor progression models. However, a temporally controlled Pten loss in all adult tissues to examine susceptibility of various tissues to Pten-deficient tumorigenesis has not been addressed yet. Our goal was to explore the genesis of Pten-deficient malignancies in multiple tissue lineages of the adult mouse. Methods and Findings We utilized an inducible Cre/loxP system to delete Pten exon 5 in the systemic organs of ROSA26 (R26)-CreERT;Ptenfx/fx mice. On reaching 45 weeks 4OHT-induced Pten loss, we found that the R26-CreERT;Ptenfx/fx mice developed a variety of malignancies. Overall tumor mean latency was 17 weeks in the Pten-deficient mice. Interestingly, mutant females developed malignancies more quickly at 10∼11 weeks compared with a tumor latency of 21 weeks for mutant males. Lymphoma incidence (76.9% in females; 40.0% in males) was higher than the other malignancies found in the mutant mice. Mutant males developed prostate (20.0%), intestinal cancer (35.0%) and squamous cell carcinoma (10.0%), whereas the mutant females developed squamous cell carcinoma (15.4%) and endometrial cancer (46.1%) in addition to lymphomas. Furthermore, we tested the pharmacological inhibition of the PTEN downstream effectors using LY294002 on Pten-deficient prostate hyperplasia. Our data revealed that, indeed, the prostate hyperplasia resulting from the induced Pten loss was significantly suppressed by LY294002 (p = 0.007). Conclusions Through monitoring a variety of Pten-deficient tumor formation, our results revealed that the lymphoid lineages and the epithelium of the prostate, endometrium, intestine and epidermis are highly susceptible to tumorigenesis after the Pten gene is excised. Therefore, this R26-CreERT; Ptenfx/fx mouse model may provide an entry point for understanding the role of Pten in the tumorigenesis of different organs and extend the search for potential therapeutic approaches to prevent Pten-deficient malignancies.
Collapse
Affiliation(s)
- Tsai-Ling Lu
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Junn-Liang Chang
- Department of Pathology, Taoyuan Armed Forces General Hospital, Lungtan, Taoyuan County, Taiwan
| | - Chih-Chia Liang
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Li-Ru You
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Chun-Ming Chen
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
361
|
Yanagi S, Kishimoto H, Kawahara K, Sasaki T, Sasaki M, Nishio M, Yajima N, Hamada K, Horie Y, Kubo H, Whitsett JA, Mak TW, Nakano T, Nakazato M, Suzuki A. Pten controls lung morphogenesis, bronchioalveolar stem cells, and onset of lung adenocarcinomas in mice. J Clin Invest 2007; 117:2929-40. [PMID: 17909629 PMCID: PMC1994617 DOI: 10.1172/jci31854] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2007] [Accepted: 07/12/2007] [Indexed: 12/22/2022] Open
Abstract
PTEN is a tumor suppressor gene mutated in many human cancers. We generated a bronchioalveolar epithelium-specific null mutation of Pten in mice [SP-C-rtTA/(tetO)(7)-Cre/Pten(flox/flox) (SOPten(flox/flox)) mice] that was under the control of doxycycline. Ninety percent of SOPten(flox/flox) mice that received doxycycline in utero [SOPten(flox/flox)(E10-16) mice] died of hypoxia soon after birth. Surviving SOPten(flox/flox)(E10-16) mice and mice that received doxycycline postnatally [SOPten(flox/flox)(P21-27) mice] developed spontaneous lung adenocarcinomas. Urethane treatment accelerated number and size of lung tumors developing in SOPten(flox/flox) mice of both ages. Histological and biochemical examinations of the lungs of SOPten(flox/flox)(E10-16) mice revealed hyperplasia of bronchioalveolar epithelial cells and myofibroblast precursors, enlarged alveolar epithelial cells, and impaired production of surfactant proteins. Numbers of bronchioalveolar stem cells (BASCs), putative initiators of lung adenocarcinomas, were increased. Lungs of SOPten(flox/flox)(E10-16) mice showed increased expression of Spry2, which inhibits the maturation of alveolar epithelial cells. Levels of Akt, c-Myc, Bcl-2, and Shh were also elevated in SOPten(flox/flox)(E10-16) and SOPten(flox/flox)(P21-27) lungs. Furthermore, K-ras was frequently mutated in adenocarcinomas observed in SOPten(flox/flox)(P21-27) lungs. These results indicate that Pten is essential for both normal lung morphogenesis and the prevention of lung carcinogenesis, possibly because this tumor suppressor is required for BASC homeostasis.
Collapse
Affiliation(s)
- Shigehisa Yanagi
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Hiroyuki Kishimoto
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Kohichi Kawahara
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Takehiko Sasaki
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Masato Sasaki
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Miki Nishio
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Nobuyuki Yajima
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Koichi Hamada
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Yasuo Horie
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Hiroshi Kubo
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Jeffrey A. Whitsett
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Tak Wah Mak
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Toru Nakano
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Masamitsu Nakazato
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Akira Suzuki
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
Division of Neurology, Respirology, Endocrinology and Metabolism, Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
Division of Embryonic and Genetic Engineering, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
Department of Microbiology and
Department of Gastroenterology, Akita University School of Medicine, Akita, Japan.
Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
The Campbell Family Institute for Breast Cancer Research and Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
Department of Pathology, Medical School, and Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
362
|
Tsuruta H, Kishimoto H, Sasaki T, Horie Y, Natsui M, Shibata Y, Hamada K, Yajima N, Kawahara K, Sasaki M, Tsuchiya N, Enomoto K, Mak TW, Nakano T, Habuchi T, Suzuki A. Hyperplasia and carcinomas in Pten-deficient mice and reduced PTEN protein in human bladder cancer patients. Cancer Res 2007; 66:8389-96. [PMID: 16951148 DOI: 10.1158/0008-5472.can-05-4627] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PTEN is a tumor suppressor gene mutated in many human cancers. We used the Cre-loxP system to generate an urothelium-specific null mutation of Pten in mice [FabpCrePten(flox/flox) (FPten(flox/flox)) mice]. Histologic examination revealed that all FPten(flox/flox) mice exhibited urothelial hyperplasia in which component cells showed enlarged nuclei and increased cell size. With time, 10% of FPten(flox/flox) mice spontaneously developed pedicellate papillary transitional cell carcinomas (TCC). This type of tumor also arose in FPten(flox/flox) mice treated with the chemical carcinogen N-butyl-N-(4-hydroxybutyl) nitrosamine. FPten(flox/flox) urothelial cells were hyperproliferative and showed increased activation of the survival signaling molecules Akt and extracellular signal-regulated kinase. In humans, 53% of primary bladder cancer patients exhibited decreased or absent expression of PTEN protein in either the cytoplasm or nucleus of tumor cells. In early bladder cancers, PTEN expression was repressed in 42% of superficial papillary TCC but in only 8% of cases of carcinoma in situ (CIS). In advanced bladder cancers, PTEN protein was significantly reduced (particularly in the nucleus) in 94% of cases, and this decrease in PTEN correlated with disease stage and grade. Thus, PTEN deficiency may contribute to bladder cancer both by initiating superficial papillary TCC and by promoting the progression of CIS to advanced invasive and metastatic forms.
Collapse
Affiliation(s)
- Hiroshi Tsuruta
- Department of Urology, Akita University School of Medicine, Akita, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
363
|
Zbuk KM, Patocs A, Shealy A, Sylvester H, Miesfeldt S, Eng C. Germline mutations in PTEN and SDHC in a woman with epithelial thyroid cancer and carotid paraganglioma. ACTA ACUST UNITED AC 2007; 4:608-12. [PMID: 17898811 DOI: 10.1038/ncponc0935] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Accepted: 03/12/2007] [Indexed: 01/29/2023]
Abstract
BACKGROUND A 43-year-old woman presented to a cancer genetics clinic for a genetic risk assessment because of her personal history of multiple neoplasias. At 37 years of age, she was diagnosed with multifocal papillary thyroid cancer, and within a year was further diagnosed with a paraganglioma of the left common carotid artery. Two years later, she was diagnosed with a paraganglioma of the right carotid body. All three tumors were treated with surgical resection. There was no family history of malignancy. Past medical history includes uterine leiomyoma and fibrocystic breast disease. Physical examination revealed macrocephaly and papillomatous papules. INVESTIGATIONS CT scan of the neck and thorax, 24-hour urine collection for measurement of metanephrines and catecholamines, MRI of the neck, thorax, and abdomen, metaiodobenzylguanidine scan, germline mutation analysis of PTEN, SDHB, SDHC and SDHD. DIAGNOSIS Cowden syndrome due to a germline mutation of PTEN, and pheochromocytoma-paraganglioma syndrome due to a germline mutation of SDHC. MANAGEMENT Clinical surveillance for breast, endometrial, thyroid, and renal cell carcinoma risks associated with Cowden syndrome according to the National Comprehensive Cancer Network guidelines, annual MRI of the neck, thorax, abdomen and pelvis, annual metabolic screening, and where available, annual 18-fluorodopamine PET scanning, predictive genetic testing of both PTEN and SDHC for the patient's daughter and parents.
Collapse
Affiliation(s)
- Kevin M Zbuk
- Genomic Medicine Institute at the Cleveland Clinic, Cleveland, OH, USA
| | | | | | | | | | | |
Collapse
|
364
|
Fayard E, Gill J, Paolino M, Hynx D, Holländer GA, Hemmings BA. Deletion of PKBalpha/Akt1 affects thymic development. PLoS One 2007; 2:e992. [PMID: 17912369 PMCID: PMC1991598 DOI: 10.1371/journal.pone.0000992] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Accepted: 09/04/2007] [Indexed: 12/31/2022] Open
Abstract
Background The thymus constitutes the primary lymphoid organ for the majority of T cells. The phosphatidyl-inositol 3 kinase (PI3K) signaling pathway is involved in lymphoid development. Defects in single components of this pathway prevent thymocytes from progressing beyond early T cell developmental stages. Protein kinase B (PKB) is the main effector of the PI3K pathway. Methodology/Principal Findings To determine whether PKB mediates PI3K signaling in the thymus, we characterized PKB knockout thymi. Our results reveal a significant thymic hypocellularity in PKBα−/− neonates and an accumulation of early thymocyte subsets in PKBα−/− adult mice. Using thymic grafting and fetal liver cell transfer experiments, the latter finding was specifically attributed to the lack of PKBα within the lymphoid component of the thymus. Microarray analyses show that the absence of PKBα in early thymocyte subsets modifies the expression of genes known to be involved in pre-TCR signaling, in T cell activation, and in the transduction of interferon-mediated signals. Conclusions/Significance This report highlights the specific requirements of PKBα for thymic development and opens up new prospects as to the mechanism downstream of PKBα in early thymocytes.
Collapse
Affiliation(s)
- Elisabeth Fayard
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Jason Gill
- Pediatric Immunology, Center for Biomedicine, Department of Clinical-Biological Sciences, The University of Basel, The University Children's Hospital, Basel, Switzerland
| | - Magdalena Paolino
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Debby Hynx
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Georg A. Holländer
- Pediatric Immunology, Center for Biomedicine, Department of Clinical-Biological Sciences, The University of Basel, The University Children's Hospital, Basel, Switzerland
| | - Brian A. Hemmings
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
365
|
Palomero T, Sulis ML, Cortina M, Real PJ, Barnes K, Ciofani M, Caparros E, Buteau J, Brown K, Perkins SL, Bhagat G, Mishra A, Basso G, Castillo M, Nagase S, Cordon-Cardo C, Parsons R, Zúñiga-Pflücker JC, Dominguez M, Ferrando AA. Mutational loss of PTEN induces resistance to NOTCH1 inhibition in T-cell leukemia. Nat Med 2007; 13:1203-10. [PMID: 17873882 PMCID: PMC2600418 DOI: 10.1038/nm1636] [Citation(s) in RCA: 722] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2007] [Accepted: 07/20/2007] [Indexed: 01/19/2023]
Abstract
Gain-of-function mutations in NOTCH1 are common in T-cell lymphoblastic leukemias and lymphomas (T-ALL), making this receptor a promising target for drugs such as gamma-secretase inhibitors, which block a proteolytic cleavage required for NOTCH1 activation. However, the enthusiasm for these therapies has been tempered by tumor resistance and the paucity of information on the oncogenic programs regulated by oncogenic NOTCH1. Here we show that NOTCH1 regulates the expression of PTEN (encoding phosphatase and tensin homolog) and the activity of the phosphoinositol-3 kinase (PI3K)-AKT signaling pathway in normal and leukemic T cells. Notch signaling and the PI3K-AKT pathway synergize in vivo in a Drosophila melanogaster model of Notch-induced tumorigenesis, and mutational loss of PTEN is associated with human T-ALL resistance to pharmacological inhibition of NOTCH1. Overall, these findings identify transcriptional control of PTEN and regulation of the PI3K-AKT pathway as key elements of the leukemogenic program activated by NOTCH1 and provide the basis for the design of new therapeutic strategies for T-ALL.
Collapse
Affiliation(s)
- Teresa Palomero
- Institute for Cancer Genetics-Columbia University, New York, NY, 10032, USA
- Department of Pathology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Maria Luisa Sulis
- Institute for Cancer Genetics-Columbia University, New York, NY, 10032, USA
- Department of Pediatrics, Columbia University Medical Center, New York, NY, 10032, USA
| | - Maria Cortina
- Instituto de Neurociencias de Alicante, Alicante, 03550, Spain
| | - Pedro J. Real
- Institute for Cancer Genetics-Columbia University, New York, NY, 10032, USA
| | - Kelly Barnes
- Institute for Cancer Genetics-Columbia University, New York, NY, 10032, USA
| | - Maria Ciofani
- Department of Immunology, University of Toronto, Sunnybrook Research Institute, Toronto, M4N 3M5, Canada
| | - Esther Caparros
- Instituto de Neurociencias de Alicante, Alicante, 03550, Spain
| | - Jean Buteau
- Departments of Medicine and Endocrinology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Kristy Brown
- Department of Pathology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Sherrie L. Perkins
- Pathology Department, University of Utah Health Sciences Center, Salt Lake City, UT, 84132, USA
| | - Govind Bhagat
- Department of Pathology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Archana Mishra
- Pathology Department, University of Utah Health Sciences Center, Salt Lake City, UT, 84132, USA
| | - Giuseppe Basso
- Hemato-Oncology Laboratory, Department of Pediatrics, University of Padua, Padua, 35128, Italy
| | - Mireia Castillo
- Department of Pathology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Satoru Nagase
- Department of Obstetrics and Gynecology Tohoku University School of Medicine Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Carlos Cordon-Cardo
- Department of Pathology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Ramon Parsons
- Institute for Cancer Genetics-Columbia University, New York, NY, 10032, USA
| | | | - Maria Dominguez
- Instituto de Neurociencias de Alicante, Alicante, 03550, Spain
| | - Adolfo A. Ferrando
- Institute for Cancer Genetics-Columbia University, New York, NY, 10032, USA
- Department of Pathology, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Pediatrics, Columbia University Medical Center, New York, NY, 10032, USA
| |
Collapse
|
366
|
Jiang BH, Liu LZ. PI3K/PTEN signaling in tumorigenesis and angiogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2007; 1784:150-8. [PMID: 17964232 DOI: 10.1016/j.bbapap.2007.09.008] [Citation(s) in RCA: 255] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Revised: 08/22/2007] [Accepted: 09/21/2007] [Indexed: 11/27/2022]
Abstract
The phosphatidyl inositol 3-kinase (PI3K) can be activated by a variety of extracellular signals and involved in a number of cellular processes including cell proliferation, survival, protein synthesis, and tumor growth. Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) is an antagonist of PI3K. The alterations of PI3K pathway such as activation of oncogenes, gene amplification, and inactivation of tumor suppressors, commonly occur in many human cancers. Angiogenesis is required for tumor growth and metastasis when the tumor reaches more than 1 mm in diameter. Recent studies have shown that PI3K and Akt play an important role in regulating tumor growth and angiogenesis through VEGF and HIF-1 expression. PI3K regulates the expression of these two proteins through HDM2 and p70S6K1 in human cancer cells. The frequent dysregulation of the PI3K/PTEN pathway in human cancer demonstrates that this pathway is an appropriate target for cancer therapeutics. In this review, we describe the recent advances in understanding the PI3K/PTEN pathway, the role and mechanism of PI3K in regulating tumor growth and angiogenesis, and the potential therapeutic opportunities for targeting this pathway for cancer treatment.
Collapse
Affiliation(s)
- Bing-Hua Jiang
- Cancer Center, Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu, China.
| | | |
Collapse
|
367
|
Suzuki A, Hamada K, Sasaki T, Mak TW, Nakano T. Role of PTEN/PI3K pathway in endothelial cells. Biochem Soc Trans 2007; 35:172-6. [PMID: 17371230 DOI: 10.1042/bst0350172] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PTEN (phosphatase and tensin homologue deleted on chromosome 10) is an important tumour-suppressor gene that encodes a 3-phosphatase. The major substrate of PTEN is PIP(3) (phosphatidylinositol 3,4,5-trisphosphate) generated by the action of PI3Ks (phosphoinositide 3-kinases). Hereditary mutation of PTEN causes tumour-susceptibility diseases such as Cowden disease. We used the Cre-loxP system to generate an endothelial cell-specific mutation of PTEN in mice. Heterozygous mutation of PTEN in endothelial cells enhances postnatal neovascularization, including tumour angiogenesis necessary for tumour growth. This observation suggests that Cowden disease patients are not only at risk for additional tumorigenic mutations due to complete loss of PTEN function, but may also experience accelerated growth of incipient tumours due to enhanced angiogenesis. Homozygous mutation of Pten in murine endothelial cells impairs cardiovascular morphogenesis and is embryonic lethal due to endothelial cell hyperproliferation and impaired vascular remodelling. Additional homozygous mutation of p85alpha, the regulatory subunit of class IA PI3Ks, or p110gamma, the catalytic subunit of the sole class IB PI3K, led to a partial rescue of all phenotypes in our PTEN-deficient mice. Thus inhibition of the PI3K pathway, including the targeting of PI3Kgamma, may be an attractive therapeutic strategy for the treatment of various malignancies.
Collapse
Affiliation(s)
- A Suzuki
- Department of Molecular Biology, Akita University School of Medicine, Akita, Japan.
| | | | | | | | | |
Collapse
|
368
|
Zbuk KM, Eng C. Hamartomatous polyposis syndromes. ACTA ACUST UNITED AC 2007; 4:492-502. [PMID: 17768394 DOI: 10.1038/ncpgasthep0902] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Accepted: 06/14/2007] [Indexed: 12/13/2022]
Abstract
The hamartomatous polyposis syndromes are a heterogeneous group of disorders that share an autosomal-dominant pattern of inheritance and are characterized by hamartomatous polyps of the gastrointestinal tract. These syndromes include juvenile polyposis syndrome, Peutz-Jeghers syndrome and the PTEN hamartoma tumor syndrome. The frequency and location of the polyps vary considerably among syndromes, as does the affected patient's predisposition to the development of gastrointestinal and other malignancies. Although the syndromes are uncommon, it is important for the clinician to recognize these disorders because they are associated with considerable morbidity and mortality, not only from malignancy but also from nonmalignant manifestations such as bleeding, intussusception, and bowel obstruction. Each hamartomatous polyposis syndrome has its own distinctive organ-specific manifestations and each requires a different surveillance strategy, which makes accurate diagnosis crucial for appropriate patient management. The availability of clinical genetic testing for these disorders means that appropriate recognition allows for timely referral for cancer genetic counseling, and often allows for predicative testing in at-risk family members. Promisingly, an understanding of the molecular pathogenesis of these disorders offers insights into the mechanisms underlying the development of sporadic malignancy, and enables rational selection of targeted therapies that warrant further investigation.
Collapse
Affiliation(s)
- Kevin M Zbuk
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | |
Collapse
|
369
|
Faucherre A, Taylor GS, Overvoorde J, Dixon JE, Hertog JD. Zebrafish pten genes have overlapping and non-redundant functions in tumorigenesis and embryonic development. Oncogene 2007; 27:1079-86. [PMID: 17704803 DOI: 10.1038/sj.onc.1210730] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In human cancer, PTEN (Phosphatase and TENsin homolog on chromosome 10, also referred to as MMAC1 and TEP1) is a frequently mutated tumor suppressor gene. We have used the zebrafish as a model to investigate the role of Pten in embryonic development and tumorigenesis. The zebrafish genome encodes two pten genes, ptena and ptenb. Here, we report that both Pten gene products from zebrafish are functional. Target-selected inactivation of ptena and ptenb revealed that Ptena and Ptenb have redundant functions in embryonic development, in that ptena-/- and ptenb-/- mutants did not show embryonic phenotypes. Homozygous single mutants survived as adults and they were viable and fertile. Double homozygous ptena-/-ptenb-/- mutants died at 5 days post fertilization with pleiotropic defects. These defects were rescued by treatment with the phosphatidylinositol-3-kinase inhibitor, LY294002. Double homozygous embryos showed enhanced cellular proliferation. In addition, cell survival was dramatically enhanced in embryos that lack functional Pten upon gamma-irradiation. Surprisingly, adult ptenb-/- zebrafish developed ocular tumors later in life, despite the expression of ptena in adult eyes. We conclude that whereas Ptena and Ptenb have redundant functions in embryonic development, they apparently do not have completely overlapping functions later in life. These pten mutant zebrafish represent a unique model to screen for genetic and/or chemical suppressors of Pten loss-of-function.
Collapse
Affiliation(s)
- A Faucherre
- Hubrecht Institute, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
370
|
Lopiccolo J, Ballas MS, Dennis PA. PTEN hamartomatous tumor syndromes (PHTS): rare syndromes with great relevance to common cancers and targeted drug development. Crit Rev Oncol Hematol 2007; 63:203-14. [PMID: 17643312 DOI: 10.1016/j.critrevonc.2007.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2007] [Accepted: 06/07/2007] [Indexed: 12/22/2022] Open
Abstract
Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) is a tumor suppressor gene located on chromosome 10q22-23 that negatively regulates the pro-survival PI3K/Akt/mTOR pathway by functioning as a lipid phosphatase. Signaling through this pathway promotes cellular transformation and survival as well as resistance to chemotherapy and radiation. Loss of PTEN function is commonly observed in human cancers through somatic mutation, hypermethylation, and/or enhanced degradation. PTEN hamartomatous tumor syndromes (PHTS) are a collection of rare clinical syndromes marked by germline PTEN loss. Compared to the general population, PHTS patients have an increased risk of developing certain cancers and can develop benign tumors in virtually any organ. These patients provide a unique opportunity to examine the role of PTEN in human tumorigenesis, as well as study genotype-phenotype relationships. Because these patients are at higher risk of developing malignancies and have no established medical therapies, early screening, surveillance, and preventive care are important issues. Inhibitors of the PI3K/Akt/mTOR pathway that are being developed as cancer therapeutics could provide new therapeutic options for these rare patients, and could be credentialed as pathway inhibitors prior to testing in the general oncology population.
Collapse
Affiliation(s)
- Jaclyn Lopiccolo
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20889, United States
| | | | | |
Collapse
|
371
|
He L, Fan C, Gillis A, Feng X, Sanatani M, Hotte S, Kapoor A, Tang D. Co-existence of high levels of the PTEN protein with enhanced Akt activation in renal cell carcinoma. Biochim Biophys Acta Mol Basis Dis 2007; 1772:1134-42. [PMID: 17681738 DOI: 10.1016/j.bbadis.2007.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Revised: 06/29/2007] [Accepted: 07/02/2007] [Indexed: 10/23/2022]
Abstract
Recruiting Akt to the membrane-bound phosphatidylinositol (3,4,5) trisphosphate (PIP3) is required for Akt activation. While PI3 kinase (PI3K) produces PIP3, PTEN dephosphorylates the 3-position phosphate from PIP3, thereby directly inhibiting Akt activation. PTEN is the dominant PIP3 phosphatase, as knockdown of PTEN results in increases in Akt activation in mice. The PTEN tumor suppressor gene is frequently mutated in a variety of human cancers, consistent with an inverse correlation between levels of the PTEN protein and Akt activation. We have examined PTEN expression and Akt activation in 35 primary clear cell renal cell carcinomas RCCs (ccRCCs) and 9 papillary RCCs (pRCCs) and their respective non-tumor kidney tissues. The PTEN protein was reduced in 16 ccRCCs (16/35=45.7%) and 8 pRCCs (8/9=88.9%). In these RCCs, 25.0% (4/16) of ccRCCs and 25.0% (2/8) of pRCCs expressed elevated Akt activation. 19 ccRCCc (19/35=54.3%) expressed comparable or higher levels of PTEN. Of these ccRCCs, 31.6% (6/19) showed increases in Akt activation. As PTEN dominantly inhibits Akt activation, the coexistence of high levels of the PTEN protein with enhanced Akt activation suggests the existence of novel mechanisms which attenuate PTEN function in ccRCC. These mechanisms may reduce PTEN function or increase PIP3 production.
Collapse
Affiliation(s)
- Lizhi He
- Division of Nephrology, Department of Medicine, McMaster University, and Father Sean O'Sullivan Research Centre, St. Joseph's Hospital, 50 Charlton Ave East, Hamilton, ON, Canada L8N 4A6
| | | | | | | | | | | | | | | |
Collapse
|
372
|
Dunlap SM, Celestino J, Wang H, Jiang R, Holland EC, Fuller GN, Zhang W. Insulin-like growth factor binding protein 2 promotes glioma development and progression. Proc Natl Acad Sci U S A 2007; 104:11736-41. [PMID: 17606927 PMCID: PMC1913900 DOI: 10.1073/pnas.0703145104] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Overexpression of insulin-like growth factor binding protein 2 (IGFBP2) is associated with progression in many types of human cancer. In this study we used a glial-specific transgenic mouse model to examine the active role of IGFBP2 in tumorigenesis and progression. Our studies show that IGFBP2 coexpression results in progression to a higher-grade glioma in platelet-derived growth factor beta (PDGFB)-driven tumors. These anaplastic oligodendrogliomas are characterized by increased cellularity, vascular proliferation, small regions of necrosis, increased mitotic activity, and increased activation of the Akt pathway. Combined expression of IGFBP2 or Akt with K-Ras was required to form astrocytomas, indicating that activation of two separate pathways is necessary for gliomagenesis. In ex vivo experiments, blockade of Akt by an inhibitor led to decreased viability of cells coexpressing IGFBP2 versus PDGFB expression alone. Thus, this study provides definitive evidence that IGFBP2 plays a key role in activation of the Akt pathway and collaborates with K-Ras or PDGFB in the development and progression of two major types of glioma.
Collapse
Affiliation(s)
- Sarah M. Dunlap
- *Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
| | - Joseph Celestino
- *Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
| | - Hua Wang
- *Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
| | - Rongcai Jiang
- *Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
| | - Eric C. Holland
- Department of Neurosurgery, Memorial Sloan–Kettering Cancer Center, New York, NY 10021
| | - Gregory N. Fuller
- *Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
- To whom correspondence may be addressed at:
Department of Pathology, University of Texas M. D. Anderson Cancer Center, Unit 85, 1515 Holcombe Boulevard, Houston, TX 77030. E-mail: or
| | - Wei Zhang
- *Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
- To whom correspondence may be addressed at:
Department of Pathology, University of Texas M. D. Anderson Cancer Center, Unit 85, 1515 Holcombe Boulevard, Houston, TX 77030. E-mail: or
| |
Collapse
|
373
|
Guertin DA, Sabatini DM. Defining the role of mTOR in cancer. Cancer Cell 2007; 12:9-22. [PMID: 17613433 DOI: 10.1016/j.ccr.2007.05.008] [Citation(s) in RCA: 2292] [Impact Index Per Article: 127.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2007] [Revised: 04/10/2007] [Accepted: 05/18/2007] [Indexed: 11/21/2022]
Abstract
The mammalian target of rapamycin (mTOR) has emerged as a critical effector in cell-signaling pathways commonly deregulated in human cancers. This has led to the prediction that mTOR inhibitors may be useful in oncology, and derivatives of one such molecule, rapamycin (from which mTOR derives its name), are currently in clinical development. In this review, we discuss recent progress in understanding mTOR signaling, paying particular attention to its relevance in cancer. We further discuss the use of rapamycin in oncology and conclude with a discussion on the future of mTOR-targeted therapy.
Collapse
Affiliation(s)
- David A Guertin
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, Cambridge, MA 02141, USA
| | | |
Collapse
|
374
|
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN)/phosphatidylinositol 3-kinase (PI3K)/AKT constitute an important pathway regulating the signaling of multiple biological processes such as apoptosis, metabolism, cell proliferation and cell growth. PTEN is a dual protein/lipid phosphatase and its main substrate phosphatidyl-inositol 3,4,5 triphosphate (PIP3) is the product of PI3K. Increase in PIP3 recruits AKT to the membrane where is activated by other kinases also dependent on PIP3. Many components of this pathway have been described as causal forces in cancer. PTEN activity is lost by mutations, deletions or promoter methylation silencing at high frequency in many primary and metastatic human cancers. Germ line mutations of PTEN are found in several familial cancer predisposition syndromes. Recently, many activating mutations in the PI3KCA gene (coding for the p110alpha catalytic subunit of PI3K) have been described in human tumors. Activation of PI3K and AKT are reported to occur in breast, ovarian, pancreatic, esophageal and other cancers. Genetically modified mice confirm these PTEN activities. Tissue-specific deletions of PTEN usually provoke cancer. Moreover, an absence of PTEN cooperates with an absence of p53 to promote cancer. However, we have observed very different results with the expression of activated versions of AKT in several tissues. Activated AKT transgenic lines do not develop tumors in breast or prostate tissues and do not cooperate with an absence of p53. This data suggest that an AKT-independent mechanism contributes to PTEN tumorigenesis. Crosses with transgenic mice expressing possible PTEN targets indicate that neither cyclin D1 nor p53 are these AKT-independent targets. However, AKT is more than a passive bridge toward PTEN tumorigenesis, since its expression not only allows but also enforces and accelerates the tumorigenic process in combination with other oncogenes.
Collapse
Affiliation(s)
- Carmen Blanco-Aparicio
- Experimental Therapeutics Programme, Spanish National Cancer Centre (CNIO), C/Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | | | | | | |
Collapse
|
375
|
Blero D, Payrastre B, Schurmans S, Erneux C. Phosphoinositide phosphatases in a network of signalling reactions. Pflugers Arch 2007; 455:31-44. [PMID: 17605038 DOI: 10.1007/s00424-007-0304-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Revised: 05/18/2007] [Accepted: 05/29/2007] [Indexed: 12/18/2022]
Abstract
Phosphoinositide phosphatases dephosphorylate the three positions (D-3, 4 and 5) of the inositol ring of the poly-phosphoinositides. They belong to different families of enzymes. The PtdIns(3,4)P(2) 4-phosphatase family, the tumour suppressor phosphatase and tensin homolog deleted on chromosome 10 (PTEN), SAC1 domain phosphatases and myotubularins belong to the tyrosine protein phosphatases superfamily. They share the presence of a conserved cysteine residue in the consensus CX(5)RT/S. Another family consists of the inositol polyphosphate 5-phosphatase isoenzymes. The importance of these phosphoinositide phosphatases in cell regulation is illustrated by multiple examples of their implications in human diseases such as Lowe syndrome, X-linked myotubular myopathy, cancer, diabetes or bacterial infection.
Collapse
Affiliation(s)
- Daniel Blero
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, Campus Erasme, Bldg C, 808 Route de Lennik, 1070, Brussels, Belgium
| | | | | | | |
Collapse
|
376
|
Maser RS, Choudhury B, Campbell PJ, Feng B, Wong KK, Protopopov A, O'Neil J, Gutierrez A, Ivanova E, Perna I, Lin E, Mani V, Jiang S, McNamara K, Zaghlul S, Edkins S, Stevens C, Brennan C, Martin ES, Wiedemeyer R, Kabbarah O, Nogueira C, Histen G, Aster J, Mansour M, Duke V, Foroni L, Fielding AK, Goldstone AH, Rowe JM, Wang YA, Look AT, Stratton MR, Chin L, Futreal PA, DePinho RA. Chromosomally unstable mouse tumours have genomic alterations similar to diverse human cancers. Nature 2007; 447:966-71. [PMID: 17515920 PMCID: PMC2714968 DOI: 10.1038/nature05886] [Citation(s) in RCA: 312] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2007] [Accepted: 04/30/2007] [Indexed: 01/07/2023]
Abstract
Highly rearranged and mutated cancer genomes present major challenges in the identification of pathogenetic events driving the neoplastic transformation process. Here we engineered lymphoma-prone mice with chromosomal instability to assess the usefulness of mouse models in cancer gene discovery and the extent of cross-species overlap in cancer-associated copy number aberrations. Along with targeted re-sequencing, our comparative oncogenomic studies identified FBXW7 and PTEN to be commonly deleted both in murine lymphomas and in human T-cell acute lymphoblastic leukaemia/lymphoma (T-ALL). The murine cancers acquire widespread recurrent amplifications and deletions targeting loci syntenic to those not only in human T-ALL but also in diverse human haematopoietic, mesenchymal and epithelial tumours. These results indicate that murine and human tumours experience common biological processes driven by orthologous genetic events in their malignant evolution. The highly concordant nature of genomic events encourages the use of genomically unstable murine cancer models in the discovery of biological driver events in the human oncogenome.
Collapse
Affiliation(s)
- Richard S Maser
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
377
|
Guo J, Zhu T, Xiao ZXJ, Chen CY. Modulation of intracellular signaling pathways to induce apoptosis in prostate cancer cells. J Biol Chem 2007; 282:24364-72. [PMID: 17573344 DOI: 10.1074/jbc.m702938200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
An understanding of the molecular pathways defining the susceptibility of prostate cancer, especially refractory prostate cancer, to apoptosis is the key for developing a cure for this disease. We previously demonstrated that up-regulating Ras signaling, together with suppression of protein kinase C (PKC), induces apoptosis. Dysregulation of various intracellular signaling pathways, including those governed by Ras, is the important element in the development of prostate cancer. In this study, we tested whether it is possible to modulate the activities of these pathways and induce an apoptotic crash among them in prostate cancer cells. Our data showed that DU145 cells express a high amount of JNK1 that is phosphorylated after endogenous PKC is suppressed, which initiates caspase 8 cleavage and cytochrome c release, leading to apoptosis. PC3 and LNCaP cells contain an activated Akt. The inhibition of PKC further augments Akt activity, which in turn induces ROS production and the accumulation of unfolded proteins in the endoplasmic reticulum, resulting in cell death. However, the concurrent activation of JNK1 and Akt, under the condition of PKC abrogation, dramatically augment the magnitude of apoptosis in the cells. Thus, our study suggests that Akt, JNK1, and PKC act in concert to signal the intracellular apoptotic machinery for a full execution of apoptosis in prostate cancer cells.
Collapse
Affiliation(s)
- Jinjin Guo
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
378
|
Salmena L, Pandolfi PP. Changing venues for tumour suppression: balancing destruction and localization by monoubiquitylation. Nat Rev Cancer 2007; 7:409-13. [PMID: 17508027 DOI: 10.1038/nrc2145] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent studies have shown that three major tumour-suppressor proteins undergo monoubiquitylation-mediated nuclear-cytoplasmic shuttling. Importantly, this mechanism has consequences for cancer and implies that proper localization is central to the function of tumour suppressors. This Progress article highlights recent efforts demonstrating that monoubiquitylation coupled to nuclear-cytoplasmic shuttling might be a novel regulatory mechanism that directly influences the function of tumour suppressors.
Collapse
Affiliation(s)
- Leonardo Salmena
- Cancer Biology and Genetics Program and Department of Pathology, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | |
Collapse
|
379
|
Abstract
The regulatory circuits that control the activities of the two distinct target of rapamycin (TOR) complexes, TORC1 and TORC2, and of Akt have been a focus of intense research in recent years. It has become increasingly evident that these regulatory circuits control some of the most fundamental aspects of metabolism, cell growth, proliferation, survival, and differentiation at both the cellular and organismal levels. As such, they also play a pivotal role in the genesis of diseases including cancer, diabetes, aging, and degenerative diseases. This review highlights recent developments aimed at deciphering the interplay between Akt and mTORCs as well as their role in embryonic development and in cancer.
Collapse
Affiliation(s)
- Prashanth T Bhaskar
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago College of Medicine, 900 South Ashland Avenue, Chicago, IL 60607, USA
| | | |
Collapse
|
380
|
Sasaki T, Sasaki J, Watanabe K, Suzuki A. Non-invasive visualization of the lipid product of class I PI3K in transgenic mouse models. Biochem Soc Trans 2007; 35:215-8. [PMID: 17371241 DOI: 10.1042/bst0350215] [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] [Indexed: 12/17/2022]
Abstract
PI3Ks (phosphoinositide 3-kinases) regulate many critical cellular responses by producing PI(3,4,5)P3 (phosphatidylinositol 3,4,5-trisphosphate). To facilitate the spatio-temporal characterization of PI(3,4,5)P3 in living primary cells, we generated a novel strain of transgenic mice [AktPH (Akt pleckstrin homology domain)–GFP (green fluorescent protein) Tg (transgenic) mice] that express a fluorescent bioprobe for PI(3,4,5)P3/PI(3,4)P2 (phosphatidylinositol 3,4-bisphosphate). By crossing AktPH–GFP Tg mice with strains of gene-targeted ‘knockout’ mice lacking a particular phosphoinositide-metabolizing enzyme, we have been able to evaluate the contribution of each enzyme to PI(3,4,5)P3 localization in migrating neutrophils. Our results indicate that PI3Kγ and the PI(3,4,5)P3 phosphatase SHIP1 [SH2 (Src homology 2)-containing inositol phosphatase-1] are the key regulators of PI(3,4,5)P3 dynamics during fMet-Leu-Phe (N-formylmethionyl-leucylphenylalanine; ‘chemotactic peptide’)-stimulated neutrophil migration. Our study has also validated the fluorescent transgenic strategy for studying PI(3,4,5)P3 metabolism in physiological and pathological situations.
Collapse
Affiliation(s)
- T Sasaki
- Department of Pathology and Immunology, Akita University School of Medicine, 1-1-1 Hondo, Akita, Japan.
| | | | | | | |
Collapse
|
381
|
Trotman LC, Wang X, Alimonti A, Chen Z, Teruya-Feldstein J, Yang H, Pavletich NP, Carver BS, Cordon-Cardo C, Erdjument-Bromage H, Tempst P, Chi SG, Kim HJ, Misteli T, Jiang X, Pandolfi PP. Ubiquitination regulates PTEN nuclear import and tumor suppression. Cell 2007; 128:141-56. [PMID: 17218261 PMCID: PMC1855245 DOI: 10.1016/j.cell.2006.11.040] [Citation(s) in RCA: 608] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Revised: 07/02/2006] [Accepted: 11/01/2006] [Indexed: 12/13/2022]
Abstract
The PTEN tumor suppressor is frequently affected in cancer cells, and inherited PTEN mutation causes cancer-susceptibility conditions such as Cowden syndrome. PTEN acts as a plasma-membrane lipid-phosphatase antagonizing the phosphoinositide 3-kinase/AKT cell survival pathway. However, PTEN is also found in cell nuclei, but mechanism, function, and relevance of nuclear localization remain unclear. We show that nuclear PTEN is essential for tumor suppression and that PTEN nuclear import is mediated by its monoubiquitination. A lysine mutant of PTEN, K289E associated with Cowden syndrome, retains catalytic activity but fails to accumulate in nuclei of patient tissue due to an import defect. We identify this and another lysine residue as major monoubiquitination sites essential for PTEN import. While nuclear PTEN is stable, polyubiquitination leads to its degradation in the cytoplasm. Thus, we identify cancer-associated mutations of PTEN that target its posttranslational modification and demonstrate how a discrete molecular mechanism dictates tumor progression by differentiating between degradation and protection of PTEN.
Collapse
Affiliation(s)
- Lloyd C Trotman
- Cancer Biology and Genetics Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
382
|
Shen WH, Balajee AS, Wang J, Wu H, Eng C, Pandolfi PP, Yin Y. Essential role for nuclear PTEN in maintaining chromosomal integrity. Cell 2007; 128:157-70. [PMID: 17218262 DOI: 10.1016/j.cell.2006.11.042] [Citation(s) in RCA: 748] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 08/10/2006] [Accepted: 11/22/2006] [Indexed: 10/23/2022]
Abstract
A broad spectrum of mutations in PTEN, encoding a lipid phosphatase that inactivates the P13-K/AKT pathway, is found associated with primary tumors. Some of these mutations occur outside the phosphatase domain, suggesting that additional activities of PTEN function in tumor suppression. We report a nuclear function for PTEN in controlling chromosomal integrity. Disruption of Pten leads to extensive centromere breakage and chromosomal translocations. PTEN was found localized at centromeres and physically associated with CENP-C, an integral component of the kinetochore. C-terminal PTEN mutants disrupt the association of PTEN with centromeres and cause centromeric instability. Furthermore, Pten null cells exhibit spontaneous DNA double-strand breaks (DSBs). We show that PTEN acts on chromatin and regulates expression of Rad51, which reduces the incidence of spontaneous DSBs. Our results demonstrate that PTEN plays a fundamental role in the maintenance of chromosomal stability through the physical interaction with centromeres and control of DNA repair. We propose that PTEN acts as a guardian of genome integrity.
Collapse
Affiliation(s)
- Wen Hong Shen
- Department of Radiation Oncology, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | | | | | | | | | | | | |
Collapse
|
383
|
Liu X, Bruxvoort KJ, Zylstra CR, Liu J, Cichowski R, Faugere MC, Bouxsein ML, Wan C, Williams BO, Clemens TL. Lifelong accumulation of bone in mice lacking Pten in osteoblasts. Proc Natl Acad Sci U S A 2007; 104:2259-64. [PMID: 17287359 PMCID: PMC1892939 DOI: 10.1073/pnas.0604153104] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Bone formation is carried out by the osteoblast, a mesenchymal cell whose lifespan and activity are regulated by growth factor signaling networks. Growth factors activate phosphatidylinositol 3-kinase (PI3K), which enhances cell survival and antagonizes apoptosis through activation of Akt/PKB. This process is negatively regulated by the Pten phosphatase, which inhibits the activity of PI3K. In this study, we investigated the effects of Akt activation in bone in vivo by conditionally disrupting the Pten gene in osteoblasts by using Cre-mediated recombination. Mice deficient in Pten in osteoblasts were of normal size but demonstrated a dramatic and progressively increasing bone mineral density throughout life. In vitro osteoblasts lacking Pten differentiated more rapidly than controls and exhibited greatly reduced apoptosis in association with markedly increased levels of phosphorylated Akt and activation of signaling pathways downstream of activated Akt. These findings support a critical role for this tumor-suppressor gene in regulating osteoblast lifespan and likely explain the skeletal abnormalities in patients carrying germ-line mutations of PTEN.
Collapse
Affiliation(s)
- Ximeng Liu
- *Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Katia J. Bruxvoort
- Laboratory of Cell Signaling and Carcinogenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Cassandra R. Zylstra
- Laboratory of Cell Signaling and Carcinogenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Jiarong Liu
- *Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Rachel Cichowski
- Laboratory of Cell Signaling and Carcinogenesis, Van Andel Research Institute, Grand Rapids, MI 49503
| | | | - Mary L. Bouxsein
- Orthopedic Biomechanics Laboratory, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Chao Wan
- *Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Bart O. Williams
- Laboratory of Cell Signaling and Carcinogenesis, Van Andel Research Institute, Grand Rapids, MI 49503
- To whom correspondence may be addressed. E-mail:
or
| | - Thomas L. Clemens
- *Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- To whom correspondence may be addressed. E-mail:
or
| |
Collapse
|
384
|
Shiroki F, Matsuda S, Doi T, Fujiwara M, Mochizuki Y, Kadowaki T, Suzuki H, Koyasu S. The p85α Regulatory Subunit of Class IA Phosphoinositide 3-Kinase Regulates β-Selection in Thymocyte Development. THE JOURNAL OF IMMUNOLOGY 2007; 178:1349-56. [PMID: 17237381 DOI: 10.4049/jimmunol.178.3.1349] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We examined the role of class IA PI3K in pre-TCR controlled beta-selection and TCR-controlled positive/negative selection in thymic development. Using mice deficient for p85alpha, a major regulatory subunit of the class IA PI3K family, the role of class IA PI3K in beta-selection was examined by injection of anti-CD3epsilon mAb into p85alpha(-/-)Rag-2(-/-) mice, which mimics pre-TCR signals. Transition of CD4(-)CD8(-) double-negative (DN) to CD4(+)CD8(+) double-positive (DP) thymocytes triggered by anti-CD3epsilon mAb was significantly impaired in p85alpha(-/-)Rag-2(-/-) compared with p85alpha(+/-)Rag-2(-/-) mice. Furthermore, DP cell numbers were lower in p85alpha(-/-)DO11.10/Rag-2(-/-) TCR-transgenic mice than in DO11.10/Rag-2(-/-) mice. In addition, inhibition by IC87114 of the major class IA PI3K catalytic subunit expressed in lymphocytes, p110delta, blocked transition of DN to DP cells in embryonic day 14.5 fetal thymic organ culture without affecting cell viability. In the absence of phosphatase and tensin homolog deleted on chromosome 10, where class IA PI3K signals would be amplified, the DN to DP transition was accelerated. In contrast, neither positive nor negative selection in Rag-2(-/-)TCR-transgenic mice was perturbed by the lack of p85alpha. These findings establish an important function of class IA PI3K in the pre-TCR-controlled developmental transition of DN to DP thymocytes.
Collapse
Affiliation(s)
- Fumiko Shiroki
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
385
|
Nishio M, Watanabe KI, Sasaki J, Taya C, Takasuga S, Iizuka R, Balla T, Yamazaki M, Watanabe H, Itoh R, Kuroda S, Horie Y, Förster I, Mak TW, Yonekawa H, Penninger JM, Kanaho Y, Suzuki A, Sasaki T. Control of cell polarity and motility by the PtdIns(3,4,5)P3 phosphatase SHIP1. Nat Cell Biol 2006; 9:36-44. [PMID: 17173042 DOI: 10.1038/ncb1515] [Citation(s) in RCA: 245] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Accepted: 10/23/2006] [Indexed: 01/01/2023]
Abstract
Proper neutrophil migration into inflammatory sites ensures host defense without tissue damage. Phosphoinositide 3-kinase (PI(3)K) and its lipid product phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P(3)) regulate cell migration, but the role of PtdIns(3,4,5)P(3)-degrading enzymes in this process is poorly understood. Here, we show that Src homology 2 (SH2) domain-containing inositol-5-phosphatase 1 (SHIP1), a PtdIns(3,4,5)P(3) phosphatase, is a key regulator of neutrophil migration. Genetic inactivation of SHIP1 led to severe defects in neutrophil polarization and motility. In contrast, loss of the PtdIns(3,4,5)P(3) phosphatase PTEN had no impact on neutrophil chemotaxis. To study PtdIns(3,4,5)P(3) metabolism in living primary cells, we generated a novel transgenic mouse (AktPH-GFP Tg) expressing a bioprobe for PtdIns(3,4,5)P(3.) Time-lapse footage showed rapid, localized binding of AktPH-GFP to the leading edge membrane of chemotaxing ship1(+/+)AktPH-GFP Tg neutrophils, but only diffuse localization in ship1(-/-)AktPH-GFP Tg neutrophils. By directing where PtdIns(3,4,5)P(3) accumulates, SHIP1 governs the formation of the leading edge and polarization required for chemotaxis.
Collapse
Affiliation(s)
- Miki Nishio
- Department of Pathology and Immunology, Akita University School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
386
|
Kishimoto H, Ohteki T, Yajima N, Kawahara K, Natsui M, Kawarasaki S, Hamada K, Horie Y, Kubo Y, Arase S, Taniguchi M, Vanhaesebroeck B, Mak TW, Nakano T, Koyasu S, Sasaki T, Suzuki A. The Pten/PI3K pathway governs the homeostasis of Valpha14iNKT cells. Blood 2006; 109:3316-24. [PMID: 17170126 DOI: 10.1182/blood-2006-07-038059] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The tumor suppressor PTEN is mutated in many human cancers. We previously used the Cre-loxP system to generate mice (LckCrePten mice) with a Pten mutation in T-lineage cells. Here we describe the phenotype of Pten-deficient Valpha14iNKT cells. A failure in the development of Valpha14iNKT cells occurs in the LckCrePten thymus between stage 2 (CD44(high)NK1.1(-)) and stage 3 (CD44(high)NK1.1(+)), resulting in decreased numbers of peripheral Valpha14iNKT cells. In vitro, Pten-deficient Valpha14iNKT cells show reduced proliferation and cytokine secretion in response to alphaGalCer stimulation but enhanced inhibitory Ly49 receptor expression. Following interaction with dendritic cells (DCs) loaded with alphaGalCer, Pten-deficient Valpha14iNKT cells demonstrate activation of PI3K. Indeed, the effects of the Pten mutation require intact function of the PI3K subunits p110gamma and p110delta. In vivo, LckCrePten mice display reduced serum IFNgamma after alphaGalCer administration. Importantly, Valpha14iNKT cell-mediated protection against the metastasis of melanoma cells to the lung was impaired in the absence of Pten. Thus, the Pten/PI3K pathway is indispensable for the homeostasis and antitumor surveillance function of Valpha14iNKT cells.
Collapse
MESH Headings
- Animals
- Antigens, Ly/immunology
- Antigens, Surface/immunology
- Cell Line, Tumor
- Class I Phosphatidylinositol 3-Kinases
- Dendritic Cells/immunology
- Homeostasis/genetics
- Homeostasis/immunology
- Hyaluronan Receptors/immunology
- Immunologic Surveillance/genetics
- Immunologic Surveillance/immunology
- Killer Cells, Natural/immunology
- Lectins, C-Type/immunology
- Mice
- Mice, Transgenic
- Mutation
- NK Cell Lectin-Like Receptor Subfamily B
- PTEN Phosphohydrolase/genetics
- PTEN Phosphohydrolase/immunology
- Phosphatidylinositol 3-Kinases/immunology
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, NK Cell Lectin-Like
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Hiroyuki Kishimoto
- Department of Molecular Biology, Akita University School of Medicine, Hondo 1-1-1, Akita 0108-543, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
387
|
Puc J, Placha G, Wocial B, Podsypanina K, Parsons R, Gaciong Z. Analysis of PTEN mutation in non-familial pheochromocytoma. Ann N Y Acad Sci 2006; 1073:317-31. [PMID: 17102102 DOI: 10.1196/annals.1353.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PTEN, a tumor suppressor gene, is frequently mutated in a variety of human tumors. In mice, monoallelic inactivation of this gene predisposes animals to neoplasia of multiple organs. Interestingly, Pten heterozygous mice develop bilateral hyperplasia of the adrenal medulla. In this report we demonstrate that these neoplasms are hormonally active pheochromocytomas that secrete increased amounts of bioactive catecholamines: norepinephrine and epinephrine. To test a possibility that PTEN might be one of the genes responsible for human sporadic pheochromocytoma, we performed mutation analysis of DNA obtained from tumors of 29 patients. However, direct sequencing of all nine exons of the PTEN gene, including the splice junctions, revealed no mutations. Examination of protein expression by immunohistochemistry using 8 normal adrenals and 11 sporadic pheochromocytomas showed no decrease in the PTEN protein expression in the tumor tissue, but upregulation of insulin-like growth factor II, a peptide implicated in growth of adrenal tissue, was observed in four cases (36%).
Collapse
Affiliation(s)
- Janusz Puc
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA.
| | | | | | | | | | | |
Collapse
|
388
|
Skeen JE, Bhaskar PT, Chen CC, Chen WS, Peng XD, Nogueira V, Hahn-Windgassen A, Kiyokawa H, Hay N. Akt deficiency impairs normal cell proliferation and suppresses oncogenesis in a p53-independent and mTORC1-dependent manner. Cancer Cell 2006; 10:269-80. [PMID: 17045205 DOI: 10.1016/j.ccr.2006.08.022] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2006] [Revised: 05/30/2006] [Accepted: 08/30/2006] [Indexed: 12/21/2022]
Abstract
Akt contributes to tumorigenesis by inhibiting apoptosis. Here we establish that Akt is required for normal cell proliferation and susceptibility to oncogenesis independently of its antiapoptotic activity. Partial ablation of Akt activity by deleting Akt1 inhibits cell proliferation and oncogenesis. These effects are compounded by deleting both Akt1 and Akt2. In vivo, Akt1 null mice are resistant to MMTV-v-H-Ras-induced tumors and to skin carcinogenesis. Thus, partial ablation of Akt activity is sufficient to suppress tumorigenesis in vitro and in vivo. The effect of Akt deficiency on cell proliferation and oncogenesis is p53 independent but mTORC1 dependent. Surprisingly, upon mTORC1 hyperactivation, the reduction in Akt activity does not impair cell proliferation and susceptibility to oncogenic transformation; thus, Akt may mediate these processes exclusively via mTORC1.
Collapse
Affiliation(s)
- Jennifer E Skeen
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois 60607, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
389
|
Pardal R, Molofsky AV, He S, Morrison SJ. Stem cell self-renewal and cancer cell proliferation are regulated by common networks that balance the activation of proto-oncogenes and tumor suppressors. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2006; 70:177-85. [PMID: 16869752 DOI: 10.1101/sqb.2005.70.057] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Networks of proto-oncogenes and tumor suppressors that control cancer cell proliferation also regulate stem cell self-renewal and possibly stem cell aging. Proto-oncogenes promote regenerative capacity by promoting stem cell function but must be balanced with tumor suppressor activity to avoid neoplastic proliferation. Conversely, tumor suppressors inhibit regenerative capacity by promoting cell death or senescence in stem cells. For example, the polycomb family proto-oncogene, Bmi-1, is consistently required for the self-renewal of diverse adult stem cells, as well as for the proliferation of cancer cells in the same tissues. Bmi-1 promotes stem cell self-renewal partly by repressing the expression of Ink4a and Arf, tumor suppressor genes that are commonly deleted in cancer. Despite ongoing Bmi-1 expression, Ink4a expression increases with age, potentially reducing stem cell frequency and function. Increased tumor suppressor activity during aging therefore may partly account for age-related declines in stem cell function. Thus, networks of proto-oncogenes and tumor suppressors have evolved to coordinately regulate stem cell function throughout life. Imbalances within such networks cause cancer or premature declines in stem cell activity that resemble accelerated aging.
Collapse
Affiliation(s)
- R Pardal
- Howard Hughes Medical Institute and Department of Internal Medicine, University of Michigan, Ann Arbor, 48109-0934, USA
| | | | | | | |
Collapse
|
390
|
Freeman D, Lesche R, Kertesz N, Wang S, Li G, Gao J, Groszer M, Martinez-Diaz H, Rozengurt N, Thomas G, Liu X, Wu H. Genetic background controls tumor development in PTEN-deficient mice. Cancer Res 2006; 66:6492-6. [PMID: 16818619 DOI: 10.1158/0008-5472.can-05-4143] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PTEN is one of the most frequently mutated tumor suppressor genes in human cancers. Germ line mutations of PTEN have been detected in three rare autosomal-dominant disorders. However, identical mutations in the PTEN gene may lead to different symptoms that have traditionally been described as different disorders, such as Cowden disease, Lhermitte-Duclos disease, and Bannayan-Zonana syndromes. This lack of genotype-phenotype correlation prompted us to directly test the possible effects of genetic background or modifier genes on PTEN-controlled tumorigenesis using genetically engineered mouse models. In this study, we generated two animal models in which either exon 5 (Pten(Delta5)) or promoter to exon 3 (Pten(-)) of the murine Pten gene were deleted and compared phenotypes associated with individual mutations on two genetic backgrounds. We found that the onset and spectrum of tumor formation depend significantly on the genetic background but less on the type of mutation generated. Our results suggest that PTEN plays a critical role in cancer development, and genetic background may influence the onset, the spectrum, and the progression of tumorigenesis caused by Pten mutation.
Collapse
Affiliation(s)
- Dan Freeman
- Department of Molecular and Medical Pharmacology, University of California at Los Angeles School of Medicine, 650 C.E. Young Drive South, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
391
|
Chow LML, Baker SJ. PTEN function in normal and neoplastic growth. Cancer Lett 2006; 241:184-96. [PMID: 16412571 DOI: 10.1016/j.canlet.2005.11.042] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Revised: 11/21/2005] [Accepted: 11/22/2005] [Indexed: 01/19/2023]
Abstract
The PTEN tumor suppressor is a central negative regulator of the PI3K/AKT signaling cascade that influences multiple cellular functions including cell growth, survival, proliferation and migration in a context-dependent manner. Dysregulation of this signaling pathway contributes to many cancers in man. PTEN is the most commonly altered component of the PI3K pathway in human malignancies. Mutations occur in both heritable and sporadic settings, with high frequency in sporadic glioblastoma, prostate and endometrial cancer. Data from human tumors and animal models support the concept that the effects of PTEN inactivation are tissue-specific. Elucidation of the mechanisms regulating activation of unique downstream effectors that mediate distinct outcomes of PTEN loss will augment our understanding of tumorigenesis and ultimately lead to novel therapeutic options.
Collapse
Affiliation(s)
- Lionel M L Chow
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, 332 North Lauderdale Street, Memphis, TN 38105, USA
| | | |
Collapse
|
392
|
Zhang X, Zhang YW, Liu S, Bulloj A, Tong GG, Zhang Z, Liao FF, Xu H. Tumor suppressor PTEN affects tau phosphorylation: deficiency in the phosphatase activity of PTEN increases aggregation of an FTDP-17 mutant Tau. Mol Neurodegener 2006; 1:7. [PMID: 16930454 PMCID: PMC1559623 DOI: 10.1186/1750-1326-1-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2006] [Accepted: 07/31/2006] [Indexed: 01/24/2023] Open
Abstract
Background Aberrant hyperphosphorylation of tau protein has been implicated in a variety of neurodegenerative disorders. Although a number of protein kinases have been shown to phosphorylate tau in vitro and in vivo, the molecular mechanisms by which tau phosphorylation is regulated pathophysiologically are largely unknown. Recently, a growing body of evidence suggests a link between tau phosphorylation and PI3K signaling. In this study, phosphorylation, aggregation and binding to the microtubule of a mutant frontal temporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) tau in the presence of tumor suppressor PTEN, a major regulatory component in PI3K signaling, were investigated. Results Phosphorylation of the human mutant FTDP-17 tau, T40RW, was evaluated using different phospho-tau specific antibodies in the presence of human wild-type or phosphatase activity null mutant PTEN. Among the evaluated phosphorylation sites, the levels of Ser214 and Thr212 phospho-tau proteins were significantly decreased in the presence of wild-type PTEN, and significantly increased when the phosphatase activity null mutant PTEN was ectopically expressed. Fractionation of the mutant tau transfected cells revealed a significantly increased level of soluble tau in cytosol when wild-type PTEN was expressed, and an elevated level of SDS-soluble tau aggregates in the presence of the mutant PTEN. In addition, the filter/trap assays detected more SDS-insoluble mutant tau aggregates in the cells overexpressing the mutant PTEN compared to those in the cells overexpressing wild-type PTEN and control DNA. This notion was confirmed by the immunocytochemical experiment which demonstrated that the overexpression of the phosphatase activity null mutant PTEN caused the mutant tau to form aggregates in the COS-7 cells. Conclusion Tumor suppressor PTEN can alleviate the phosporylation of the mutant FTDP-17 tau at specific sites, and the phosphatase activity null PTEN increases the mutant tau phosphorylation at these sites. The changes of the tau phosphorylation status by ectopic expression of PTEN correlate to the alteration of the mutant tau's cellular distribution. In addition, the overexpression of the mutant PTEN can increase the level of the mutant tau aggregates and lead to the formation of visible aggregates in the cells.
Collapse
Affiliation(s)
- Xue Zhang
- Center for Neuroscience and Aging, Burnham Institute for Medical Research, 10901 N, Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | | | |
Collapse
|
393
|
Chen ML, Xu PZ, Peng XD, Chen WS, Guzman G, Yang X, Di Cristofano A, Pandolfi PP, Hay N. The deficiency of Akt1 is sufficient to suppress tumor development in Pten+/- mice. Genes Dev 2006; 20:1569-74. [PMID: 16778075 PMCID: PMC1482477 DOI: 10.1101/gad.1395006] [Citation(s) in RCA: 211] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The tumor suppressor PTEN is frequently inactivated in human cancers. A major downstream effector of PTEN is Akt, which is hyperactivated via PTEN inactivation. It is not known, however, whether diminished Akt activity is sufficient to inhibit tumorigenesis initiated by Pten deficiency. Here we showed that the deficiency of Akt1 is sufficient to dramatically inhibit tumor development in Pten+/- mice. Akt1 deficiency had a profound effect on endometrium and prostate neoplasia, two types of human cancer, in which PTEN is frequently mutated, and also affected thyroid and adrenal medulla tumors and intestinal polyps. Even haplodeficiency of Akt1 was sufficient to markedly attenuate the development of high-grade prostate intraepithelial neoplasia (PIN) and endometrial carcinoma. These results have significant implications for cancer therapy.
Collapse
Affiliation(s)
- Mei-Ling Chen
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
394
|
Lee JH, Shin SY, Kim S, Choo J, Lee YH. Suppression of PTEN expression during aggregation with retinoic acid in P19 mouse embryonal carcinoma cells. Biochem Biophys Res Commun 2006; 347:715-22. [PMID: 16842746 DOI: 10.1016/j.bbrc.2006.06.161] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Accepted: 06/25/2006] [Indexed: 12/19/2022]
Abstract
Apoptosis is thought to be involved in the maintenance of cellular homeostasis, as well as various pathological processes. However, little information is available about the regulation of apoptosis during the aggregation stage of P19 embryonal carcinoma (EC) cells. Here we report that aggregation-induced apoptosis is markedly attenuated by treatment with retinoic acid (RA). PTEN (phosphatase and tensin homolog deleted on chromosome 10) expression was down-regulated during the aggregation phase of P19 EC cells in the presence, but not in the absence, of RA. Suppression of PTEN expression during the aggregation was accompanied by increased phosphorylation of serine/threonine kinase Akt and glycogen synthase kinase-3beta (GSK-3beta). Our results suggest that RA attenuates the induction of apoptosis during the aggregation phase of P19 EC cells, probably by suppressing PTEN expression.
Collapse
Affiliation(s)
- Joon Ho Lee
- Division of Molecular and Life Sciences, College of Science and Technology, Hanyang University, Ansan, Gyeonggi-do, South Korea
| | | | | | | | | |
Collapse
|
395
|
Mahimainathan L, Das F, Venkatesan B, Choudhury GG. Mesangial cell hypertrophy by high glucose is mediated by downregulation of the tumor suppressor PTEN. Diabetes 2006; 55:2115-25. [PMID: 16804083 DOI: 10.2337/db05-1326] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Diabetic nephropathy is characterized early in its course by glomerular hypertrophy and, importantly, mesangial hypertrophy, which correlate with eventual glomerulosclerosis. The mechanism of hypertrophy, however, is not known. Gene disruption of the tumor suppressor PTEN, a negative regulator of the phosphatidylinositol 3-kinase/Akt pathway, in fruit flies and mice demonstrated its role in size control in a cell-specific manner. Here, we investigated the mechanism of mesangial hypertrophy in response to high extracellular glucose. We link early renal hypertrophy with significant reduction in PTEN expression in the streptozotocin-induced diabetic kidney cortex and glomeruli, concomitant with activation of Akt. Similarly, exposure of mesangial cells to high concentrations of glucose also decreased PTEN expression and its phosphatase activity, resulting in increased Akt activity. Expression of PTEN inhibited high-glucose-induced mesangial cell hypertrophy, and expression of dominant-negative PTEN was sufficient to induce hypertrophy. In diabetic nephropathy, the hypertrophic effect of hyperglycemia is thought to be mediated by transforming growth factor-beta (TGF-beta). TGF-beta significantly reduced PTEN expression in mesangial cells, with a reduction in its phosphatase activity and an increase in Akt activation. PTEN and dominant-negative Akt attenuated TGF-beta-induced hypertrophy of mesangial cells. Finally, we show that inhibition of TGF-beta signal transduction blocks the effect of high glucose on PTEN downregulation. These data identify a novel mechanism placing PTEN as a key regulator of diabetic mesangial hypertrophy involving TGF-beta signaling.
Collapse
Affiliation(s)
- Lenin Mahimainathan
- Department of Medicine, Mail Code 7882, 7703 Floyd Curl Dr., San Antonio, Texas 78229-3900, USA
| | | | | | | |
Collapse
|
396
|
Abstract
Human genetics offers new possibilities for understanding physiological regulatory mechanisms and disorders of the immune system. Genetic abnormalities of lymphocyte cell death programs have provided insights into mechanisms of receptor biology and principles of immune homeostasis and tolerance. Thus far, there are two major diseases of programmed cell death associated with inherited human mutations: the autoimmune lymphoproliferative syndrome and the caspase-eight deficiency state. We describe the details of their molecular pathogenesis and discuss how these diseases illustrate important concepts in immune regulation and genetics.
Collapse
Affiliation(s)
- Nicolas Bidère
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
397
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to examine the contribution of the PI3K signaling pathway to the development of human tumors and to propose further studies to elucidate how to develop therapeutics for patients with mutations in this pathway. RECENT FINDINGS More than 30% of various solid tumor types were recently found to contain mutations in PIK3CA, the catalytic subunit of PI3K. Further analysis of key genes in this pathway identified an additional eight genes altered in tumors. These were generally found to be mutated in a mutually exclusive manner, thus increasing the mutation frequency of the pathway to 40% in colorectal cancers and emphasizing the importance of the PI3K pathway in tumorigenesis. Functional analyses of PIK3CA mutations revealed that they increase its enzymatic activity, stimulate AKT signaling, allow growth factor-independent growth as well as increasing cell invasion and metastasis. SUMMARY The PI3K signaling pathway is dysregulated by a variety of mechanisms in a large fraction of human tumors. Both mutational and functional analyses have shown that PIK3CA is an oncogene that plays an important role in tumor progression. Mutant members of the PI3K pathway, including PIK3CA, are good targets for therapeutic intervention because most of them are kinases, making them attractive for drug development. Gaining further insights into PIK3CA oncogenic mechanisms may produce new biomarkers and help the development of targeted therapeutics.
Collapse
Affiliation(s)
- Yardena Samuels
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University Medical Institutions, Baltimore, Maryland 21231, USA.
| | | |
Collapse
|
398
|
Chalhoub N, Kozma SC, Baker SJ. S6k1 is not required for Pten-deficient neuronal hypertrophy. Brain Res 2006; 1100:32-41. [PMID: 16777079 DOI: 10.1016/j.brainres.2006.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 05/02/2006] [Accepted: 05/04/2006] [Indexed: 10/24/2022]
Abstract
The tumor suppressor PTEN (phosphatase and tensin homolog) plays a critical role in the development and maintenance of the mammalian nervous system. Effects of inherited mutation of PTEN are highly variable and include macrocephaly, Lhermitte-Duclos disease (LDD) caused by a hamartomatous enlargement of the cerebellum, ataxia, seizures and autism, in addition to cancer predisposition. In the mouse, selective inactivation of Pten in post-mitotic granule neurons of the cerebellum and dentate gyrus showed that Pten was required for proper regulation of neuronal nuclear and soma size. Hypertrophy of Pten-deficient neurons required the activity of the serine-threonine kinase mTor. mTor is a master regulator of cell and organ growth which can trigger a cascade of downstream signaling pathways involving, in part, components of the translational machinery, including S6k1 and its substrate the ribosomal protein S6. Deletion of S6k1 in mice results in decreased size. Therefore, to determine the relative contribution of S6k1 to Pten-deficient neuronal hypertrophy in vivo, we crossed Pten brain-conditional knockouts with S6k1 null mice. Double mutant mice show no reversion or improvement in their Pten-related size and neurological defects including enlarged cerebella and dentate gyri with increased size of neuronal nuclei and somata, ataxia, and premature death. The hypertrophic Pten/S6k1-deficient neurons contained high levels of phosphorylated S6, similar to Pten-deficient neurons, suggesting that the mTor/S6k/S6 branch of the pathway was still active. Thus, we conclude that S6k1 is not required to cause hypertrophy of Pten-deficient neurons. This study reveals a cell type-dependent role for S6k1 in PI3K-dependent hypertrophy.
Collapse
Affiliation(s)
- Nader Chalhoub
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 332 North Lauderdale, Memphis, TN 38105, USA
| | | | | |
Collapse
|
399
|
Carrière A, Liu X, Hekimi S. The age of heterozygosity. AGE (DORDRECHT, NETHERLANDS) 2006; 28:201-208. [PMID: 19943141 PMCID: PMC2464728 DOI: 10.1007/s11357-006-9006-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Accepted: 12/15/2005] [Indexed: 05/28/2023]
Abstract
Two mutant mouse models of longevity in which the loss of only one copy of the gene leads to a significantly increased lifespan have recently been described: Igf1r (+/-) and mclk1 (+/-). Igf1r encodes a transmembrane receptor kinase for the insulin-like growth factor-1, and mclk1 encodes a hydroxylase that is necessary for the biosynthesis of ubiquinone. Interestingly, the motivation for testing the longevity of both of these mutants came from observations in the nematode Caenorhabditis elegans. IGF-1R protein is homologous to DAF-2 and mCLK1 is the mouse orthologue of the C. elegans enzyme CLK-1. In worms, the homozygous inactivation of both of these longevity genes is viable and no dominant mutations are known. In addition to aging slowly, old mclk1 (+/-) mice were found to undergo loss-of-heterozygosity at the mclk1 locus, which results in clones of mclk1 (-/-) cells in the liver, presumably because mclk1 (-/-) cells can outcompete mclk1 (+/-) cells under certain conditions. We will discuss how these observations suggest novel directions of research, but also call for some caution in the interpretation of past and future results.
Collapse
Affiliation(s)
- Audrey Carrière
- Department of Biology, McGill University, 1205 avenue Docteur Penfield, H3A 1B1 Montreal, Quebec Canada
| | - Xingxing Liu
- Department of Biology, McGill University, 1205 avenue Docteur Penfield, H3A 1B1 Montreal, Quebec Canada
| | - Siegfried Hekimi
- Department of Biology, McGill University, 1205 avenue Docteur Penfield, H3A 1B1 Montreal, Quebec Canada
| |
Collapse
|
400
|
Trotman LC, Alimonti A, Scaglioni PP, Koutcher JA, Cordon-Cardo C, Pandolfi PP. Identification of a tumour suppressor network opposing nuclear Akt function. Nature 2006; 441:523-7. [PMID: 16680151 PMCID: PMC1976603 DOI: 10.1038/nature04809] [Citation(s) in RCA: 312] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Accepted: 04/13/2006] [Indexed: 01/24/2023]
Abstract
The proto-oncogene AKT (also known as PKB) is activated in many human cancers, mostly owing to loss of the PTEN tumour suppressor. In such tumours, AKT becomes enriched at cell membranes where it is activated by phosphorylation. Yet many targets inhibited by phosphorylated AKT (for example, the FOXO transcription factors) are nuclear; it has remained unclear how relevant nuclear phosphorylated AKT (pAKT) function is for tumorigenesis. Here we show that the PMLtumour suppressor prevents cancer by inactivating pAKT inside the nucleus. We find in a mouse model that Pml loss markedly accelerates tumour onset, incidence and progression in Pten-heterozygous mutants, and leads to female sterility with features that recapitulate the phenotype of Foxo3a knockout mice. We show that Pml deficiency on its own leads to tumorigenesis in the prostate, a tissue that is exquisitely sensitive to pAkt levels, and demonstrate that Pml specifically recruits the Akt phosphatase PP2a as well as pAkt into Pml nuclear bodies. Notably, we find that Pml-null cells are impaired in PP2a phosphatase activity towards Akt, and thus accumulate nuclear pAkt. As a consequence, the progressive reduction in Pml dose leads to inactivation of Foxo3a-mediated transcription of proapoptotic Bim and the cell cycle inhibitor p27(kip1). Our results demonstrate that Pml orchestrates a nuclear tumour suppressor network for inactivation of nuclear pAkt, and thus highlight the importance of AKT compartmentalization in human cancer pathogenesis and treatment.
Collapse
Affiliation(s)
- Lloyd C Trotman
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10021, USA
| | | | | | | | | | | |
Collapse
|