Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lloyd C. Trotman is active.

Publication


Featured researches published by Lloyd C. Trotman.


Nature | 2005

Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis

Zhenbang Chen; Lloyd C. Trotman; David R. Shaffer; Hui Kuan Lin; Zohar A. Dotan; Masaru Niki; Jason A. Koutcher; Howard I. Scher; Thomas Ludwig; William L. Gerald; Carlos Cordon-Cardo; Pier Paolo Pandolfi

Cellular senescence has been theorized to oppose neoplastic transformation triggered by activation of oncogenic pathways in vitro, but the relevance of senescence in vivo has not been established. The PTEN and p53 tumour suppressors are among the most commonly inactivated or mutated genes in human cancer including prostate cancer. Although they are functionally distinct, reciprocal cooperation has been proposed, as PTEN is thought to regulate p53 stability, and p53 to enhance PTEN transcription. Here we show that conditional inactivation of Trp53 in the mouse prostate fails to produce a tumour phenotype, whereas complete Pten inactivation in the prostate triggers non-lethal invasive prostate cancer after long latency. Strikingly, combined inactivation of Pten and Trp53 elicits invasive prostate cancer as early as 2 weeks after puberty and is invariably lethal by 7 months of age. Importantly, acute Pten inactivation induces growth arrest through the p53-dependent cellular senescence pathway both in vitro and in vivo, which can be fully rescued by combined loss of Trp53. Furthermore, we detected evidence of cellular senescence in specimens from early-stage human prostate cancer. Our results demonstrate the relevance of cellular senescence in restricting tumorigenesis in vivo and support a model for cooperative tumour suppression in which p53 is an essential failsafe protein of Pten-deficient tumours.


PLOS Biology | 2003

Pten Dose Dictates Cancer Progression in the Prostate

Lloyd C. Trotman; Masaru Niki; Zohar A. Dotan; Jason A. Koutcher; Antonio Di Cristofano; Andrew Xiao; Alan S Khoo; Pradip Roy-Burman; Norman M. Greenberg; Terry Van Dyke; Carlos Cordon-Cardo; Pier Paolo Pandolfi

Complete inactivation of the PTEN tumor suppressor gene is extremely common in advanced cancer, including prostate cancer (CaP). However, one PTEN allele is already lost in the vast majority of CaPs at presentation. To determine the consequence of PTEN dose variations on cancer progression, we have generated by homologous recombination a hypomorphic Pten mouse mutant series with decreasing Pten activity: Ptenhy/+ > Pten+/− > Ptenhy/− (mutants in which we have rescued the embryonic lethality due to complete Pten inactivation) > Pten prostate conditional knockout (Ptenpc) mutants. In addition, we have generated and comparatively analyzed two distinct Ptenpc mutants in which Pten is inactivated focally or throughout the entire prostatic epithelium. We find that the extent of Pten inactivation dictate in an exquisite dose-dependent fashion CaP progression, its incidence, latency, and biology. The dose of Pten affects key downstream targets such as Akt, p27Kip1, mTOR, and FOXO3. Our results provide conclusive genetic support for the notion that PTEN is haploinsufficient in tumor suppression and that its dose is a key determinant in cancer progression.


Cell | 2007

Ubiquitination Regulates PTEN Nuclear Import and Tumor Suppression

Lloyd C. Trotman; Xinjiang Wang; Andrea Alimonti; Zhenbang Chen; Julie Teruya-Feldstein; Haijuan Yang; Nikola P. Pavletich; Brett S. Carver; Carlos Cordon-Cardo; Hediye Erdjument-Bromage; Paul Tempst; Sung Gil Chi; Hyo Jong Kim; Tom Misteli; Xuejun Jiang; Pier Paolo Pandolfi

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.


Cell | 2007

NEDD4-1 Is a Proto-Oncogenic Ubiquitin Ligase for PTEN

Xinjiang Wang; Lloyd C. Trotman; Theresa M. Koppie; Andrea Alimonti; Zhenbang Chen; Zhonghua Gao; Junru Wang; Hediye Erdjument-Bromage; Paul Tempst; Carlos Cordon-Cardo; Pier Paolo Pandolfi; Xuejun Jiang

The tumor suppressor PTEN, a critical regulator for multiple cellular processes, is mutated or deleted frequently in various human cancers. Subtle reductions in PTEN expression levels have profound impacts on carcinogenesis. Here we show that PTEN level is regulated by ubiquitin-mediated proteasomal degradation, and purified its ubiquitin ligase as HECT-domain protein NEDD4-1. In cells NEDD4-1 negatively regulates PTEN stability by catalyzing PTEN polyubiquitination. Consistent with the tumor-suppressive role of PTEN, overexpression of NEDD4-1 potentiated cellular transformation. Strikingly, in a mouse cancer model and multiple human cancer samples where the genetic background of PTEN was normal but its protein levels were low, NEDD4-1 was highly expressed, suggesting that aberrant upregulation of NEDD4-1 can posttranslationally suppress PTEN in cancers. Elimination of NEDD4-1 expression inhibited xenotransplanted tumor growth in a PTEN-dependent manner. Therefore, NEDD4-1 is a potential proto-oncogene that negatively regulates PTEN via ubiquitination, a paradigm analogous to that of Mdm2 and p53.


Nature Genetics | 2010

Subtle variations in Pten dose determine cancer susceptibility

Andrea Alimonti; Arkaitz Carracedo; John G. Clohessy; Lloyd C. Trotman; Caterina Nardella; Ainara Egia; Leonardo Salmena; Katia Sampieri; William J. Haveman; Edi Brogi; Andrea L. Richardson; Jiangwen Zhang; Pier Paolo Pandolfi

Cancer susceptibility has been attributed to at least one heterozygous genetic alteration in a tumor suppressor gene (TSG). It has been hypothesized that subtle variations in TSG expression can promote cancer development. However, this hypothesis has not yet been definitively supported in vivo. Pten is a TSG frequently lost in human cancer and mutated in inherited cancer-predisposition syndromes. Here we analyze Pten hypermorphic mice (Ptenhy/+), expressing 80% normal levels of Pten. Ptenhy/+ mice develop a spectrum of tumors, with breast tumors occurring at the highest penetrance. All breast tumors analyzed here retained two intact copies of Pten and maintained Pten levels above heterozygosity. Notably, subtle downregulation of Pten altered the steady-state biology of the mammary tissues and the expression profiles of genes involved in cancer cell proliferation. We present an alterative working model for cancer development in which subtle reductions in the dose of TSGs predispose to tumorigenesis in a tissue-specific manner.


Nature | 2006

Identification of a tumour suppressor network opposing nuclear Akt function

Lloyd C. Trotman; Andrea Alimonti; Pier Paolo Scaglioni; Jason A. Koutcher; Carlos Cordon-Cardo; Pier Paolo Pandolfi

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 p27kip1. 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.


Nature Cell Biology | 2001

Import of adenovirus DNA involves the nuclear pore complex receptor CAN/Nup214 and histone H1

Lloyd C. Trotman; Nicole Mosberger; Maarten Fornerod; Robert P. Stidwill; Urs F. Greber

Adenovirus type 2 (Ad2) imports its DNA genome through the nuclear pore complex (NPC) of cells in interphase for viral production. Here we identify the NPC-filament protein CAN/Nup214 as a docking site for incoming Ad2 capsids. Binding to CAN is independent of cytosolic factors. Capsids disassemble at NPCs to free their DNA for import. This process requires binding of nuclear histone H1 to the stably docked capsids and involves H1-import factors, restricting this irreversible process to the proximity of the nucleus. Our results provide a molecular mechanism for disassembly of Ad2 and reveal an unexpected function of histone H1 in virus-mediated DNA import.


Journal of Cell Biology | 2012

Ndfip1 regulates nuclear Pten import in vivo to promote neuronal survival following cerebral ischemia

Jason Howitt; Jenny Lackovic; Ley-Hian Low; Adam Naguib; Alison Macintyre; Choo-Peng Goh; Jennifer K. Callaway; Vicki E. Hammond; Tim Thomas; Matthew Dixon; Ulrich Putz; John Silke; Perry F. Bartlett; Baoli Yang; Sharad Kumar; Lloyd C. Trotman; Seong-Seng Tan

PTEN nuclear entry driven by ubiquitination is mediated by the ligase-interacting protein Ndfip1 and is essential for neuronal survival in mice after cerebral ischemia.


Cancer Cell | 2003

PTEN and p53: Who will get the upper hand?

Lloyd C. Trotman; Pier Paolo Pandolfi

Mutations of PTEN and p53 are very frequent, yet often mutually exclusive due to functional interdependence of the proteins and, according to a new study, the most intimate possible interaction: direct binding.


Annual Review of Pharmacology and Toxicology | 2014

Turning off AKT: PHLPP as a drug target.

Alexandra C. Newton; Lloyd C. Trotman

Precise control of the balance between protein phosphorylation, catalyzed by protein kinases, and protein dephosphorylation, catalyzed by protein phosphatases, is essential for cellular homeostasis. Dysregulation of this balance leads to pathophysiological states, driving diseases such as cancer, heart disease, and diabetes. Aberrant phosphorylation of components of the pathways that control cell growth and cell survival are particularly prevalent in cancer. One of the most studied tumor suppressors in these pathways is the lipid phosphatase PTEN (phosphatase and tensin homolog deleted on chromosome ten), which dephosphorylates the lipid second messenger phosphatidylinositol 3,4,5-trisphosphate (PIP3), thus preventing activation of the oncogenic kinase AKT (v-akt murine thymoma viral oncogene homolog). In 2005, the discovery of a family of protein phosphatases whose members directly dephosphorylate and inactivate AKT introduced a new negative regulator of the phosphoinositide 3-kinase (PI3K) oncogenic pathway. Pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP) isozymes comprise a novel tumor suppressor family whose two members, PHLPP1 and PHLPP2, are deleted as frequently as PTEN in cancers such as those of the prostate. PHLPP is thus a novel therapeutic target to suppress oncogenic pathways and is a potential candidate biomarker to stratify patients for the appropriate targeted therapeutics. This review discusses the role of PHLPP in terminating AKT signaling and how pharmacological intervention would impact this pathway.

Collaboration


Dive into the Lloyd C. Trotman's collaboration.

Top Co-Authors

Avatar

Carlos Cordon-Cardo

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Pier Paolo Pandolfi

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dawid G. Nowak

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Muhan Chen

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Adam Naguib

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Tali Herzka

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Hyejin Cho

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Mireia Castillo-Martin

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Kaitlin Watrud

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Wu Zheng

Cold Spring Harbor Laboratory

View shared research outputs
Researchain Logo
Decentralizing Knowledge