Network


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

Hotspot


Dive into the research topics where Benjamin D. Hopkins is active.

Publication


Featured researches published by Benjamin D. Hopkins.


Science | 2013

A Secreted PTEN Phosphatase That Enters Cells to Alter Signaling and Survival

Benjamin D. Hopkins; Barry Fine; Nicole Steinbach; Meaghan Dendy; Zachary Rapp; Jacquelyn Shaw; Kyrie Pappas; Jennifer S. Yu; Cindy Hodakoski; Sarah M. Mense; Joshua U. Klein; Sarah Pegno; Maria Luisa Sulis; Hannah Goldstein; Benjamin Amendolara; Liang Lei; Matthew Maurer; Jeffrey N. Bruce; Peter Canoll; Hanina Hibshoosh; Ramon Parsons

PTEN Variations The product of the tumor suppressor gene phosphate and tensin homolog on chromosome ten (PTEN) is a lipid and protein phosphatase that regulates important cellular processes, including growth, survival, and metabolism (see the Perspective by Leslie and Brunton). Though PTEN is best known for effects on the phosphatidylnositol 3-kinase (PI3K) signaling pathway, the PTEN protein is also found in the nucleus. Bassi et al. (p. 395) found that PTENs presence in the nucleus was regulated in response to covalent modification of the protein by SUMOylation and phosphorylation. Cells lacking nuclear PTEN showed increased sensitivity to DNA damage and underwent cell death if the PI3K pathway was also inhibited. Hopkins et al. (p. 399, published online 6 June) discovered an alternative translation start site in human PTEN messenger RNA that allowed expression of a protein, PTEN-Long, with about 170 extra amino acids. The unusual enzyme was released from cells and then taken up into other cells. In a mouse tumor model, uptake of the enzyme inhibited the PI3K pathway and inhibited tumor growth. An alternative translation start site produces an elongated PTEN that can enter tumor cells and kill them. [Also see Perspective by Leslie and Brunton] Phosphatase and tensin homolog on chromosome ten (PTEN) is a tumor suppressor and an antagonist of the phosphoinositide-3 kinase (PI3K) pathway. We identified a 576–amino acid translational variant of PTEN, termed PTEN-Long, that arises from an alternative translation start site 519 base pairs upstream of the ATG initiation sequence, adding 173 N-terminal amino acids to the normal PTEN open reading frame. PTEN-Long is a membrane-permeable lipid phosphatase that is secreted from cells and can enter other cells. As an exogenous agent, PTEN-Long antagonized PI3K signaling and induced tumor cell death in vitro and in vivo. By providing a means to restore a functional tumor-suppressor protein to tumor cells, PTEN-Long may have therapeutic uses.


Science | 2009

Activation of the PI3K Pathway in Cancer through Inhibition of PTEN by Exchange Factor P-REX2a

Barry Fine; Cindy Hodakoski; Susan Koujak; Tao Su; Lao H. Saal; Matthew J. Maurer; Benjamin D. Hopkins; Megan Keniry; Maria Luisa Sulis; Sarah M. Mense; Hanina Hibshoosh; Ramon Parsons

Reigning In Tumor Suppression Mitogenic signaling through phosphoinositide-3 kinase generates the lipid second messenger phosphatidyl inositol 3,4,5-trisphosphate (PIP3). The tumor suppressor gene product and lipid phosphatase PTEN (phosphatase and tensin homolog on chromosome 10) opposes such mitogenic signaling by dephosphorylating PIP3. In a screen for proteins that interact with PTEN, Fine et al. (p. 1261) identified P-REX2a, a guanine nucleotide exchange factor (GEF) for the RAC small guanosine triphosphatase. Endogenous P-REX2a and PTEN interacted in human embryonic kidney 293 cells, and P-REX2a inhibited catalytic activity of PTEN. Thus, like that of many protein phosphatases, the activity of PTEN is kept in check by an interacting protein inhibitor. P-REX2a thus provides a mechanism through which tumor cells may inactivate PTEN. Cancer cell growth is stimulated by the inhibition of a previously unknown step in cell signaling for tumor suppression. PTEN (phosphatase and tensin homolog on chromosome 10) is a tumor suppressor whose cellular regulation remains incompletely understood. We identified phosphatidylinositol 3,4,5-trisphosphate RAC exchanger 2a (P-REX2a) as a PTEN-interacting protein. P-REX2a mRNA was more abundant in human cancer cells and significantly increased in tumors with wild-type PTEN that expressed an activated mutant of PIK3CA encoding the p110 subunit of phosphoinositide 3-kinase subunit α (PI3Kα). P-REX2a inhibited PTEN lipid phosphatase activity and stimulated the PI3K pathway only in the presence of PTEN. P-REX2a stimulated cell growth and cooperated with a PIK3CA mutant to promote growth factor–independent proliferation and transformation. Depletion of P-REX2a reduced amounts of phosphorylated AKT and growth in human cell lines with intact PTEN. Thus, P-REX2a is a component of the PI3K pathway that can antagonize PTEN in cancer cells.


Cancer Research | 2009

3-Phosphoinositide-dependent kinase 1 potentiates upstream lesions on the phosphatidylinositol 3-kinase pathway in breast carcinoma.

Matthew Maurer; Tao Su; Lao H. Saal; Susan Koujak; Benjamin D. Hopkins; Christina R. Barkley; Jiaping Wu; Subhadra V. Nandula; Bhaskar Dutta; Yuli Xie; Y. Rebecca Chin; Da In Kim; Jennifer S. Ferris; Sofia K. Gruvberger-Saal; Mervi Laakso; Xiaomei Wang; Lorenzo Memeo; Albert Rojtman; Tulio Matos; Jennifer S. Yu; Carlos Cordon-Cardo; Jorma Isola; Mary Beth Terry; Alex Toker; Gordon B. Mills; Jean Zhao; Vundavalli V. Murty; Hanina Hibshoosh; Ramon Parsons

Lesions of ERBB2, PTEN, and PIK3CA activate the phosphatidylinositol 3-kinase (PI3K) pathway during cancer development by increasing levels of phosphatidylinositol-3,4,5-triphosphate (PIP(3)). 3-Phosphoinositide-dependent kinase 1 (PDK1) is the first node of the PI3K signal output and is required for activation of AKT. PIP(3) recruits PDK1 and AKT to the cell membrane through interactions with their pleckstrin homology domains, allowing PDK1 to activate AKT by phosphorylating it at residue threonine-308. We show that total PDK1 protein and mRNA were overexpressed in a majority of human breast cancers and that 21% of tumors had five or more copies of the gene encoding PDK1, PDPK1. We found that increased PDPK1 copy number was associated with upstream pathway lesions (ERBB2 amplification, PTEN loss, or PIK3CA mutation), as well as patient survival. Examination of an independent set of breast cancers and tumor cell lines derived from multiple forms of human cancers also found increased PDK1 protein levels associated with such upstream pathway lesions. In human mammary cells, PDK1 enhanced the ability of upstream lesions to signal to AKT, stimulate cell growth and migration, and rendered cells more resistant to PDK1 and PI3K inhibition. After orthotopic transplantation, PDK1 overexpression was not oncogenic but dramatically enhanced the ability of ERBB2 to form tumors. Our studies argue that PDK1 overexpression and increased PDPK1 copy number are common occurrences in cancer that potentiate the oncogenic effect of upstream lesions on the PI3K pathway. Therefore, we conclude that alteration of PDK1 is a critical component of oncogenic PI3K signaling in breast cancer.


Trends in Biochemical Sciences | 2014

PTEN function: the long and the short of it

Benjamin D. Hopkins; Cindy Hodakoski; Douglas Barrows; Sarah M. Mense; Ramon Parsons

Phosphatase and tensin homolog deleted on chromosome ten (PTEN) is a phosphatase that is frequently altered in cancer. PTEN has phosphatase-dependent and -independent roles, and genetic alterations in PTEN lead to deregulation of protein synthesis, the cell cycle, migration, growth, DNA repair, and survival signaling. PTEN localization, stability, conformation, and phosphatase activity are controlled by an array of protein-protein interactions and post-translational modifications. Thus, PTEN-interacting and -modifying proteins have profound effects on the tumor suppressive functions of PTEN. Moreover, recent studies identified mechanisms by which PTEN can exit cells, via either exosomal export or secretion, and act on neighboring cells. This review focuses on modes of PTEN protein regulation and ways in which perturbations in this regulation may lead to disease.


American Journal of Pathology | 2009

Gab2-Mediated Signaling Promotes Melanoma Metastasis

Basil Horst; Sofia K. Gruvberger-Saal; Benjamin D. Hopkins; Lindsey Bordone; Ying Yang; Karen A. Chernoff; Ijeoma Uzoma; Volker Schwipper; Jutta Liebau; Norma J. Nowak; Georg Brunner; David F. Owens; David L. Rimm; Ramon Parsons; Julide Tok Celebi

Metastatic melanoma is a disease with a poor prognosis that currently lacks effective treatments. Critical biological features of metastasis include acquisition of migratory competence, growth factor independence, and invasive potential. In an attempt to identify genes that contribute to melanoma pathogenesis, a genome-wide search using bacterial artificial chromosome array comparative genomic hybridization and single nucleotide polymorphism arrays in a series of 64 metastatic melanoma samples and 20 melanoma cell lines identified increased copy numbers of Gab2 located on 11q14.1. Gab2 is an adaptor protein that potentiates the activation of the Ras-Erk and PI3K-Akt pathways and has recently been implicated in human cancer; however, its role in melanoma has not been explored. In this study, we found that Gab2 was either amplified (approximately 11%) and/or overexpressed (approximately 50%) in melanoma. Gab2 protein expression correlated with clinical melanoma progression, and higher levels of expression were seen in metastatic melanomas compared with primary melanoma and melanocytic nevi. We found that overexpression of Gab2 potentiates, whereas silencing of Gab2 reduces, migration and invasion of melanoma cells. Gab2 mediated the hyperactivation of Akt signaling in the absence of growth factors, whereas inhibition of the PI3K-Akt pathway decreased Gab2-mediated tumor cell migration and invasive potential. Gab2 overexpression resulted in enhanced tumor growth and metastatic potential in vivo. These studies demonstrate a previously undefined role for Gab2 in melanoma tumor progression and metastasis.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Regulation of PTEN inhibition by the pleckstrin homology domain of P-REX2 during insulin signaling and glucose homeostasis

Cindy Hodakoski; Benjamin D. Hopkins; Douglas Barrows; Sarah M. Mense; Megan Keniry; Karen E. Anderson; Philip A. Kern; Phillip T. Hawkins; Len R. Stephens; Ramon Parsons

Significance Proper insulin signaling is necessary for regulating glucose metabolism and is often deregulated in cancer. Insulin activation of the phosphoinositide-3 kinase (PI3K) pathway is negatively regulated by the tumor suppressor phosphatase and tensin homolog deleted on chromosome 10 (PTEN). We previously described a novel inhibitor of PTEN activity, phosphatidylinositol 3,4,5-trisphosphate-dependent Rac exchanger 2 (P-REX2). Here, we show that P-REX2 inhibition of PTEN occurs through the pleckstrin homology (PH) domain and requires PTEN C-terminal tail phosphorylation. Furthermore, loss of Prex2 results in reduced insulin signaling and increased Pten activity, and mice lacking P-rex2 display impaired glucose uptake and insulin resistance. This reveals a crucial role for the PH domain of P-REX2 in the regulation of glucose metabolism through the inhibition of PTEN. Insulin activation of phosphoinositide 3-kinase (PI3K) signaling regulates glucose homeostasis through the production of phosphatidylinositol 3,4,5-trisphosphate (PIP3). The dual-specificity phosphatase and tensin homolog deleted on chromosome 10 (PTEN) blocks PI3K signaling by dephosphorylating PIP3, and is inhibited through its interaction with phosphatidylinositol 3,4,5-trisphosphate-dependent Rac exchanger 2 (P-REX2). The mechanism of inhibition and its physiological significance are not known. Here, we report that P-REX2 interacts with PTEN via two interfaces. The pleckstrin homology (PH) domain of P-REX2 inhibits PTEN by interacting with the catalytic region of PTEN, and the inositol polyphosphate 4-phosphatase domain of P-REX2 provides high-affinity binding to the postsynaptic density-95/Discs large/zona occludens-1-binding domain of PTEN. P-REX2 inhibition of PTEN requires C-terminal phosphorylation of PTEN to release the P-REX2 PH domain from its neighboring diffuse B-cell lymphoma homology domain. Consistent with its function as a PTEN inhibitor, deletion of Prex2 in fibroblasts and mice results in increased Pten activity and decreased insulin signaling in liver and adipose tissue. Prex2 deletion also leads to reduced glucose uptake and insulin resistance. In human adipose tissue, P-REX2 protein expression is decreased and PTEN activity is increased in insulin-resistant human subjects. Taken together, these results indicate a functional role for P-REX2 PH-domain–mediated inhibition of PTEN in regulating insulin sensitivity and glucose homeostasis and suggest that loss of P-REX2 expression may cause insulin resistance.


Science Signaling | 2014

A Unified Nomenclature and Amino Acid Numbering for Human PTEN

Rafael Pulido; Suzanne J. Baker; João T. Barata; Arkaitz Carracedo; Víctor J. Cid; Ian D. Chin-Sang; Vrushank Davé; Jeroen den Hertog; Peter N. Devreotes; Charis Eng; Frank B. Furnari; Maria Magdalena Georgescu; Arne Gericke; Benjamin D. Hopkins; Xeujun Jiang; Seung Rock Lee; Mathias Lösche; Prerna Malaney; Xavier Matias-Guiu; María Molina; Pier Paolo Pandolfi; Ramon Parsons; Paolo Pinton; Carmen Rivas; Rafael M. Rocha; Manuel Sánchez Rodríguez; Alonzo H. Ross; Manuel Serrano; Vuk Stambolic; Bangyan L. Stiles

With the discovery of an isoform based on an alternative translation start site, PTEN nomenclature needs an update. The tumor suppressor PTEN is a major brake for cell transformation, mainly due to its phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3] phosphatase activity that directly counteracts the oncogenicity of phosphoinositide 3-kinase (PI3K). PTEN mutations are frequent in tumors and in the germ line of patients with tumor predisposition or with neurological or cognitive disorders, which makes the PTEN gene and protein a major focus of interest in current biomedical research. After almost two decades of intense investigation on the 403-residue-long PTEN protein, a previously uncharacterized form of PTEN has been discovered that contains 173 amino-terminal extra amino acids, as a result of an alternate translation initiation site. To facilitate research in the field and to avoid ambiguities in the naming and identification of PTEN amino acids from publications and databases, we propose here a unifying nomenclature and amino acid numbering for this longer form of PTEN.


Science Signaling | 2015

PTEN inhibits PREX2-catalyzed activation of RAC1 to restrain tumor cell invasion

Sarah M. Mense; Douglas Barrows; Cindy Hodakoski; Nicole Steinbach; David Schoenfeld; William Su; Benjamin D. Hopkins; Tao Su; Barry Fine; Hanina Hibshoosh; Ramon Parsons

Cancer-associated mutations in PREX2 enable cells to overcome the tumor-suppressive effects of PTEN. Turning the tables on an inhibitor Loss-of-function mutations are commonly detected in the tumor suppressor PTEN in various cancers. The lipid phosphatase activity of PTEN inhibits cell proliferation. In cancer, enhanced cell proliferation and migration often go hand in hand. Indeed, PTEN is inhibited by PREX2, a protein that promotes cell migration. Mense et al. found that the inhibition was reciprocal: Independently from its lipid phosphatase activity, PTEN suppressed the activity of PREX2. Forms of PREX2 with cancer-associated mutations were not inhibited by PTEN, inhibited the lipid phosphatase activity of PTEN, and promoted cancer cell invasion. Analysis of human tumors revealed a correlation between PREX2 mutation and high PTEN expression, suggesting that tumors select for PREX2 mutants with attenuated PTEN inhibition. The tumor suppressor PTEN restrains cell migration and invasion by a mechanism that is independent of inhibition of the PI3K pathway and decreased activation of the kinase AKT. PREX2, a widely distributed GEF that activates the GTPase RAC1, binds to and inhibits PTEN. We used mouse embryonic fibroblasts and breast cancer cell lines to show that PTEN suppresses cell migration and invasion by blocking PREX2 activity. In addition to metabolizing the phosphoinositide PIP3, PTEN inhibited PREX2-induced invasion by a mechanism that required the tail domain of PTEN, but not its lipid phosphatase activity. Fluorescent nucleotide exchange assays revealed that PTEN inhibited the GEF activity of PREX2 toward RAC1. PREX2 is a frequently mutated GEF in cancer, and examination of human tumor data showed that PREX2 mutation was associated with high PTEN expression. Therefore, we tested whether cancer-derived somatic PREX2 mutants, which accelerate tumor formation of immortalized melanocytes, were inhibited by PTEN. The three stably expressed, somatic PREX2 cancer mutants that we tested were resistant to PTEN-mediated inhibition of invasion but retained the ability to inhibit the lipid phosphatase activity of PTEN. In vitro analysis showed that PTEN did not block the GEF activity of two PREX2 cancer mutants and had a reduced binding affinity for the third. Thus, PTEN antagonized migration and invasion by restraining PREX2 GEF activity, and PREX2 mutants are likely selected in cancer to escape PTEN-mediated inhibition of invasion.


Cancer Biology & Therapy | 2013

Alterations of EGFR, p53 and PTEN that mimic changes found in basal-like breast cancer promote transformation of human mammary epithelial cells

Maira M. Pires; Benjamin D. Hopkins; Lao H. Saal; Ramon Parsons

Breast cancer can be classified into different molecular subtypes with varying clinical and pathological characteristics. The basal-like breast cancer subtype represents one of the most aggressive and lethal types of breast cancer, and due to poor mechanistic understanding, it lacks targeted therapy. Many basal-like breast cancer patient samples display alterations of established drivers of cancer development, including elevated expression of EGFR, p53 inactivating mutations and loss of expression of the tumor suppressor PTEN; however, their contribution to human basal-like breast cancer pathogenesis remains ill-defined. Using non-transformed human mammary epithelial cells, we set out to determine whether altering EGFR, p53 and PTEN in different combinations could contribute to basal-like breast cancer progression through transformation of cells. Altering PTEN in combination with either p53 or EGFR in contrast to any of the single alterations caused increased growth of transformed colonies in soft agar. Concomitantly modifying all three genes led to the highest rate of cellular proliferation and the greatest degree of anchorage-independent colony formation. Results from our effort to engineer a model of BBC expressing alterations of EGFR, p53 and PTEN suggest that these changes are cooperative and likely play a causal role in basal-like breast cancer pathogenesis. Consideration should be given to targeting EGFR and restoring p53 and PTEN signaling simultaneously as a strategy for treatment of this subtype of breast cancer.


Clinical Cancer Research | 2014

Molecular Pathways: Intercellular PTEN and the Potential of PTEN Restoration Therapy

Benjamin D. Hopkins; Ramon Parsons

Phosphatase and Tensin homolog deleted on chromosome Ten (PTEN) acts as a tumor suppressor through both PI3K-dependent and -independent mechanisms. Reduced PTEN activity has been shown to affect not only tumor cell proliferation and survival but also the microenvironmental context in which nascent tumors develop. As a result of the multifaceted tumor-suppressive roles of PTEN, tumors evolve by selecting for clones in which PTEN activity is lost. PTEN activity within tumors can be modulated in numerous ways, including direct mutation, epigenetic regulation, and amplification or mutation of other proteins that can regulate or degrade PTEN. These events functionally prevent PTEN protein from acting within tumor cells. Paracrine roles for PTEN gene products (exosomal PTEN and PTEN-L) have recently been identified, through which PTEN gene products produced in one cell are able to enter recipient cells and contribute to PTEN functions. In preclinical models purified PTEN-L protein was able to enter tumor xenografts and downregulate PI3K signaling as well as cause tumor cell death. Here, we review the role of PTEN as a multifaceted tumor suppressor and reflect upon the potential for PTEN restoration therapy. Clin Cancer Res; 20(21); 5379–83. ©2014 AACR.

Collaboration


Dive into the Benjamin D. Hopkins's collaboration.

Top Co-Authors

Avatar

Ramon Parsons

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Cindy Hodakoski

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Sarah M. Mense

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Hanina Hibshoosh

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Douglas Barrows

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tao Su

Columbia University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge