Network


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

Hotspot


Dive into the research topics where Adam Naguib is active.

Publication


Featured researches published by Adam Naguib.


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.


Trends in Cell Biology | 2013

PTEN plasticity: how the taming of a lethal gene can go too far.

Adam Naguib; Lloyd C. Trotman

PTEN loss drives many cancers and recent genetic studies reveal that often PTEN is antagonised at the protein level without alteration of DNA or RNA expression. This scenario can already cause malignancy, because PTEN is haploinsufficient. We here review normally occurring mechanisms of PTEN protein regulation and discuss three processes where PTEN plasticity is needed: ischaemia, development, and wound healing. These situations demand transient PTEN suppression, whereas cancer exploits them for continuous proliferation and survival advantages. Therefore, increased understanding of PTEN plasticity may help us better interpret tumour development and ultimately lead to drug targets for PTEN supporting cancer therapy.


Cell Reports | 2018

Mitochondrial Complex I Inhibitors Expose a Vulnerability for Selective Killing of Pten-Null Cells

Adam Naguib; Grinu Mathew; Colleen R. Reczek; Kaitlin Watrud; Alexandra Ambrico; Tali Herzka; Irene Casanova Salas; Matthew F. Lee; Nour El-Amine; Wu Zheng; M. Emilia Di Francesco; Joseph R. Marszalek; Darryl Pappin; Navdeep S. Chandel; Lloyd C. Trotman

SUMMARY A hallmark of advanced prostate cancer (PC) is the concomitant loss of PTEN and p53 function. To selectively eliminate such cells, we screened cytotoxic compounds on Pten−/−;Trp53−/− fibroblasts and their Pten-WT reference. Highly selective killing of Pten-null cells can be achieved by deguelin, a natural insecticide. Deguelin eliminates Pten-deficient cells through inhibition of mitochondrial complex I (CI). Five hundred-fold higher drug doses are needed to obtain the same killing of Pten-WT cells, even though deguelin blocks their electron transport chain equally well. Selectivity arises because mitochondria of Pten-null cells consume ATP through complex V, instead of producing it. The resulting glucose dependency can be exploited to selectively kill Pten-null cells with clinically relevant CI inhibitors, especially if they are lipophilic. In vivo, deguelin suppressed disease in our genetically engineered mouse model for metastatic PC. Our data thus introduce a vulnerability for highly selective targeting of incurable PC with inhibitors of CI.


Cancer Research | 2011

Abstract 2405: Identification of PHLPP as a tumour suppressor reveals the role of pathway feedback compensation in PTEN-mutant prostate cancer progression

Muhan Chen; Christopher P. Pratt; Martha E. Zeeman; Nicholaus Schultz; Barry S. Taylor; Audrey O'Neill; Mireia Castillo-Martin; Dawid G. Nowak; Adam Naguib; Danielle M. Grace; Jernej Murn; Chris Sander; William L. Gerald; Carlos Cordon-Cardo; Alexandra C. Newton; Brett S. Carver; Lloyd C. Trotman

Hyper-activation of the PI 3-Kinase/ AKT pathway is common in many cancer types. Tumourigenesis through this pathway is prevented by concerted action of multiple tumour suppressor genes. Most notably, PTEN reverts PI 3-Kinase activity whereas excessive pathway activation triggers the p53-mediated senescence arrest. However, it remains ill defined if and at what stage this response acts in human prostate cancer. Here we identify the AKT-inactivating phosphatase PHLPP as a tumour suppressor and demonstrate how the p53-response can antagonise co-deletion of PTEN and PHLPP to form a barrier against prostate cancer progression. We show that Phlpp-loss causes neoplasia and upon partial Pten-loss, carcinoma in mouse prostate. In this setting, Phlpp-deficiency triggers growth arrest via mTorC1-dependent activation of p53 and we find that co-deletion of Pten and Phlpp selects for spontaneous inactivation of p53 in prostate. Validating this conditional gene inactivation scheme in a comprehensive genomic patient data set we find that co-deletion of PTEN and PHLPP is almost exclusively observed in metastatic prostate cancer and tightly correlated to deletion of TP53. Furthermore, PTEN/ PHLPP expression can be used to predict disease outcome in these patients, comparable to the standard histology based method, but adding actionable information on pathway status. Finally, we show that both known PHLPP isoforms compensate for PTEN-suppression in a novel pathway feedback explaining their co-deletion with PTEN in the metastatic samples. Surprisingly, we find that the feedback surge of these genes is sensitive to some pharmacological inhibitors of the PI 3-Kinase pathway. Collectively, our findings emphasise the need for careful evaluation of PI 3-Kinase target therapy effects in prostate cancer and highlight the value of genetically engineered mouse models in this process. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2405. doi:10.1158/1538-7445.AM2011-2405


Cancer Research | 2013

Abstract 5156: Upstream activation of PTEN.

Adam Naguib; Gyula Bencze; Christopher R. Faehnle; Thomas Schalch; Zsolt Lazar; Cristian Ruse; Leemor Joshua-Tor; Darryl Pappin; Lloyd C. Trotman

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC The PTEN tumor suppressor is among the most frequently altered genes of cancer. PTEN suppression at the protein level is critically associated with disease since PTEN is haploinsufficient in many cancer types, including prostate. We have recently shown strong cooperation between protein phosphatases and PTEN to suppress PI 3-Kinase and AKT signaling in prostate cancer. Intriguingly, our findings showed that PTEN status orchestrates a PHLPP2 response by controlling the protein levels of this phosphatase. Yet, little is known about genes that control the levels and activity of PTEN to a degree that they critically maintain its function in disease. Therefore, it is assumed that PTEN is constitutively active in normal cells. Here we identify the upstream activation mechanism of PTEN and discuss the consequences for cancer diagnosis and therapy with PI 3-Kinase pathway inhibitors. Citation Format: Adam Naguib, Gyula Bencze, Christopher R. Faehnle, Thomas Schalch, Zsolt Lazar, Cristian I. Ruse, Leemor Joshua-Tor, Darryl J. Pappin, Lloyd C. Trotman. Upstream activation of PTEN. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5156. doi:10.1158/1538-7445.AM2013-5156


Cancer Research | 2011

Abstract 1269: Identifying Pten-sensitive drug therapy through metabolic phenotype-arrays

Adam Naguib; Dawid G. Nowak; Muhan Chen; Lloyd C. Trotman

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL The loss or mutation of tumor suppressor genes is a predominant event in the initiation, progression and metastatic development of cancer. PTEN and p53 are frequently inactivated in lethal metastatic prostate cancer, as they provide a critical growth, proliferation and anti-apoptotic advantage to cells in which their activity is diminished. In mouse prostate cancer models, loss of p53 alone in the prostate does not result in noticeable neoplasia. However, subsequent additional deletion of Pten causes lethal prostatic adenocarcinoma within 6 months, illustrating the need to combat the cooperative power of these two genetic lesions. Although inactivation of these tumor suppressor genes has been widely described at the level of their cancer phenotype, it has recently emerged that one understudied route to combat lesions harboring these alterations is to understand and exploit critical underlying changes in their metabolic makeup. In the present study, we used an array platform to determine the growth phenotype of p53 null Mouse Embryonic Fibroblast (MEFs) under close to 1200 conditions involving different sources of energy, amino acids, hormones, growth factors, chemical ions and responses to chemotherapy agents. with subsequent comparison to the response in p53/Pten double-null MEFs. Our analysis showed that loss of Pten critically altered response, utilization and sensitivity especially to specific hormones, ions and chemotherapy agents. Since this approach revealed Pten-status specific cell sensitivities, we are utilizing this information to establish selective targeting of cells as a precursor to therapeutic intervention in our genetically engineered mouse models of prostate cancer, which harbor the identical tumor suppressor lesions. Collectively, our results establish a rapid screening platform for identification of genotype-specific anti-cancer agents. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 1269. doi:10.1158/1538-7445.AM2011-1269


Cancer Cell | 2011

Identification of PHLPP1 as a tumor suppressor reveals the role of feedback activation in PTEN-mutant prostate cancer progression

Muhan Chen; Christopher P. Pratt; Martha E. Zeeman; Nikolaus Schultz; Barry S. Taylor; Audrey O'Neill; Mireia Castillo-Martin; Dawid G. Nowak; Adam Naguib; Danielle M. Grace; Jernej Murn; Nicholas Navin; Gurinder Singh Atwal; Chris Sander; William L. Gerald; Carlos Cordon-Cardo; Alexandra C. Newton; Brett S. Carver; Lloyd C. Trotman


Molecular Cell | 2015

PTEN Functions by Recruitment to Cytoplasmic Vesicles

Adam Naguib; Gyula Bencze; Hyejin Cho; Wu Zheng; Ante Tocilj; Elad Elkayam; Christopher R. Faehnle; Nadia Jaber; Christopher P. Pratt; Muhan Chen; Wei-Xing Zong; Michael S. Marks; Leemor Joshua-Tor; Darryl Pappin; Lloyd C. Trotman


Cell Reports | 2015

P53 Mutations Change Phosphatidylinositol Acyl Chain Composition

Adam Naguib; Gyula Bencze; Dannielle D. Engle; Iok In Christine Chio; Tali Herzka; Kaitlin Watrud; Szilvia Bencze; David A. Tuveson; Darryl Pappin; Lloyd C. Trotman


Archive | 2017

Endocytosis and PTEN in Tumor Suppression

M. Lee; Adam Naguib; Lloyd C. Trotman; Nour El-Amine

Collaboration


Dive into the Adam Naguib's collaboration.

Top Co-Authors

Avatar

Lloyd C. Trotman

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Darryl Pappin

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

Christopher P. Pratt

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gyula Bencze

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Leemor Joshua-Tor

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Dawid G. Nowak

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Wu Zheng

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge