Network


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

Hotspot


Dive into the research topics where Benjamin Amendolara is active.

Publication


Featured researches published by Benjamin Amendolara.


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.


The Journal of Neuroscience | 2014

Ribosome Profiling Reveals a Cell-Type-Specific Translational Landscape in Brain Tumors

Christian Gonzalez; Jennifer S. Sims; Nicholas Hornstein; Angeliki Mela; Franklin Garcia; X Liang Lei; David A. Gass; Benjamin Amendolara; Jeffrey N. Bruce; Peter Canoll; Peter A. Sims

Glioma growth is driven by signaling that ultimately regulates protein synthesis. Gliomas are also complex at the cellular level and involve multiple cell types, including transformed and reactive cells in the brain tumor microenvironment. The distinct functions of the various cell types likely lead to different requirements and regulatory paradigms for protein synthesis. Proneural gliomas can arise from transformation of glial progenitors that are driven to proliferate via mitogenic signaling that affects translation. To investigate translational regulation in this system, we developed a RiboTag glioma mouse model that enables cell-type-specific, genome-wide ribosome profiling of tumor tissue. Infecting glial progenitors with Cre-recombinant retrovirus simultaneously activates expression of tagged ribosomes and delivers a tumor-initiating mutation. Remarkably, we find that although genes specific to transformed cells are highly translated, their translation efficiencies are low compared with normal brain. Ribosome positioning reveals sequence-dependent regulation of ribosomal activity in 5′-leaders upstream of annotated start codons, leading to differential translation in glioma compared with normal brain. Additionally, although transformed cells express a proneural signature, untransformed tumor-associated cells, including reactive astrocytes and microglia, express a mesenchymal signature. Finally, we observe the same phenomena in human disease by combining ribosome profiling of human proneural tumor and non-neoplastic brain tissue with computational deconvolution to assess cell-type-specific translational regulation.


Cancer Research | 2014

The Transcriptional Regulatory Network of Proneural Glioma Determines the Genetic Alterations Selected During Tumor Progression

Adam M. Sonabend; Mukesh Bansal; Paolo Guarnieri; Liang Lei; Benjamin Amendolara; Craig Soderquist; Richard Leung; Jonathan Yun; Benjamin C. Kennedy; Julia Sisti; Samuel S. Bruce; Rachel Bruce; Reena Shakya; Thomas Ludwig; Steven S. Rosenfeld; Peter A. Sims; Jeffrey N. Bruce; Peter Canoll

Proneural glioblastoma is defined by an expression pattern resembling that of oligodendrocyte progenitor cells and carries a distinctive set of genetic alterations. Whether there is a functional relationship between the proneural phenotype and the associated genetic alterations is unknown. To evaluate this possible relationship, we performed a longitudinal molecular characterization of tumor progression in a mouse model of proneural glioma. In this setting, the tumors acquired remarkably consistent genetic deletions at late stages of progression, similar to those deleted in human proneural glioblastoma. Further investigations revealed that p53 is a master regulator of the transcriptional network underlying the proneural phenotype. This p53-centric transcriptional network and its associated phenotype were observed at both the early and late stages of progression, and preceded the proneural-specific deletions. Remarkably, deletion of p53 at the time of tumor initiation obviated the acquisition of later deletions, establishing a link between the proneural transcriptional network and the subtype-specific deletions selected during glioma progression.


Neuro-oncology | 2014

Convection-enhanced delivery of etoposide is effective against murine proneural glioblastoma

Adam M. Sonabend; Arthur S. Carminucci; Benjamin Amendolara; Mukesh Bansal; Richard Leung; Liang Lei; Ronald Realubit; Hai Li; Charles Karan; Jonathan Yun; Christopher Showers; Robert Rothcock; Jane O; Peter Canoll; Jeffrey N. Bruce

BACKGROUND Glioblastoma subtypes have been defined based on transcriptional profiling, yet personalized care based on molecular classification remains unexploited. Topoisomerase II (TOP2) contributes to the transcriptional signature of the proneural glioma subtype. Thus, we targeted TOP2 pharmacologically with etoposide in proneural glioma models. METHODS TOP2 gene expression was evaluated in mouse platelet derived growth factor (PDGF)(+)phosphatase and tensin homolog (PTEN)(-/-)p53(-/-) and PDGF(+)PTEN(-/-) proneural gliomas and cell lines, as well as human glioblastoma from The Cancer Genome Atlas. Correlation between TOP2 transcript levels and etoposide susceptibility was investigated in 139 human cancer cell lines from the Cancer Cell Line Encyclopedia public dataset and in mouse proneural glioma cell lines. Convection-enhanced delivery (CED) of etoposide was tested on cell-based PDGF(+)PTEN(-/-)p53(-/-) and retroviral-based PDGF(+)PTEN(-/-) mouse proneural glioma models. RESULTS TOP2 expression was significantly higher in human proneural glioblastoma and in mouse proneural tumors at early as well as late stages of development compared with normal brain. TOP2B transcript correlated with susceptibility to etoposide in mouse proneural cell lines and in 139 human cancer cell lines from the Cancer Cell Line Encyclopedia. Intracranial etoposide CED treatment (680 μM) was well tolerated by mice and led to a significant survival benefit in the PDGF(+)PTEN(-/-)p53(-/-) glioma model. Moreover, etoposide CED treatment at 80 μM but not 4 μM led to a significant survival advantage in the PDGF(+)PTEN(-/-) glioma model. CONCLUSIONS TOP2 is highly expressed in proneural gliomas, rendering its pharmacological targeting by intratumoral administration of etoposide by CED effective on murine proneural gliomas. We provide evidence supporting clinical testing of CED of etoposide with a molecular-based patient selection approach.


Journal for ImmunoTherapy of Cancer | 2013

Tumor-associated T cell receptor repertoires in low- and high-grade gliomas.

Jennifer S. Sims; Boris Grinshpun; Benjamin Amendolara; Yufeng Shen; Peter Canoll; Peter A. Sims; Jeffrey N. Bruce

Glioblastoma (GBM) remains prognostically dismal, with care centered on resection, motivating research into novel therapies. Although inducing anti-tumor immunity remains an attractive target for therapeutic and preventative intervention, the interplay between evolving dysregulation of the glioma microenvironment and T cell inefficacy remains poorly understood. In our murine model of proneural glioma, retroviral delivery of PDGF and cre-mediated knockout of PTEN in glial progenitors of adult C57BL/6 gives rise to slow-growing tumors, which were harvested at early- mid- and late-stage progression timepoints following induction, along with peripheral blood. From human patients, tissue from low- and high-grade glioma resections and corresponding peripheral lymphocytes were cryofrozen during surgery at New York Presbyterian-CUMC. For both species, we employed a commercially available primer set (iRepertoire) for nested PCR of the complementarity-determining region 3 (CDR3) of the TCR-alpha and TCR-beta chains from the T cell RNA, followed by next-generation sequencing on an Illumina MiSeq. We developed a computational pipeline for mapping TCR cassettes, in silico translation, pairwise analysis of tissue/periphery per subject, and error analysis. In the murine model, we observe that at late-stage, the intratumoral TCR repertoire diverges significantly from the peripheral, including dramatic expansion of single tumor-associated CDR3s, while the peripheral repertoire itself diverges from those of healthy mice. In both human patients and mice, we observed tumor-associated CDR3s, disproportionately abundant in tumor tissue compared to the corresponding peripheral blood, at both the amino acid and nucleotide level. In human samples we observed tumor-specific TCR expansions that were associated with particular functional subsets (CD8+, CD4+, Treg, NKT). Sequence-level study of the TCR repertoire promises new insight into the scope of glioma immunosuppression, especially systemic effects which remain elusive and the origins of intratumoral suppressive populations, and holds the potential for immunotherapeutic interventions, non-invasive diagnostics, and direct assessment of global responses to immunotherapy.


Columbia Undergraduate Science Journal | 2017

The Effect of Fasudil on Glioma Cell Migration

Athanassios Dovas; Benjamin Amendolara; Christine Wang; Peter Canoll


Neuro-oncology | 2014

IT-32DIVERGENCE OF INTRATUMORAL AND SYSTEMIC T CELL REPERTOIRES REFLECTS LOCAL MONOCYTE PROFILES DURING GLIOMA PROGRESSION

Jennifer S. Sims; Boris Grinshpun; Yaping Feng; Justin A. Neira; Jorge Samanamud; Benjamin Amendolara; Peter Canoll; Peter A. Sims; Yufeng Shen; Jeffrey N. Bruce


Neuro-oncology | 2014

MOLECULAR MOTORS AS NOVEL TARGETS TO BLOCK GLIOMA DISPERSION AND PROLIFERATION

Steven S. Rosenfeld; Monica Venere; Jan Lammerding; Sanja Ivkovic; Benjamin Amendolara; James Crish; Hannah Picariello; Jeremy N. Rich; Peter Canoll


Neuro-oncology | 2014

ET-57TOPOISOMERASE II IN PRONEURAL GLIOMAS, A MODULATOR OF TRANSCRIPTION AND A THERAPEUTIC TARGET.

Adam M. Sonabend; Arthur S. Carminucci; Benjamin Amendolara; Mukesh Bansal; Moshe Praver; Ronald Realubit; Hai Li; Charles Karan; Matei Banu; Liang Lei; Peter Canoll; Jeffrey N. Bruce


Neuro-oncology | 2013

HIGH-THROUGHPUT SEQUENCING OF TUMOR-ASSOCIATED T CELL RECEPTORS IN HUMAN AND MURINE GLIOMA

Jennifer S. Sims; Boris Grinshpun; Yaping Feng; Benjamin Amendolara; Yufeng Shen; Peter Canoll; Peter A. Sims; Jeffrey N. Bruce

Collaboration


Dive into the Benjamin Amendolara's collaboration.

Top Co-Authors

Avatar

Peter Canoll

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter A. Sims

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adam M. Sonabend

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge