Network


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

Hotspot


Dive into the research topics where Harilaos Filippakis is active.

Publication


Featured researches published by Harilaos Filippakis.


Journal of Virology | 2013

Replication Stress and Mitotic Dysfunction in Cells Expressing Simian Virus 40 Large T Antigen

Liang Hu; Harilaos Filippakis; Haomin Huang; Tim J. Yen; Ole Gjoerup

ABSTRACT We previously demonstrated that simian virus 40 (SV40) large T antigen (LT) binds to the Bub1 kinase, a key regulator of the spindle checkpoint and chromosome segregation. Bub1 mutations or altered expression patterns are linked to chromosome missegregation and are considered to be a driving force in some human cancers. Here we report that LT, dependent on Bub1 binding, causes micronuclei, lagging chromatin, and anaphase bridges, which are hallmarks of chromosomal instability (CIN) and Bub1 insufficiency. Using time-lapse microscopy, we demonstrate that LT imposes a Bub1 binding-dependent delay in the metaphase-to-anaphase transition. Kinetochore fibers reveal that LT, via Bub1 binding, causes aberrant kinetochore (KT)-microtubule (MT) attachments and a shortened interkinetochore distance, consistent with a lack of tension. Previously, we showed that LT also induces the DNA damage response (DDR) via Bub1 binding. Using inducible LT cell lines, we show that an activated DDR was observed before the appearance of anaphase bridges and micronuclei. Furthermore, LT induction in serum-starved cells demonstrated γ-H2AX accumulation in cells that had not yet entered mitosis. Thus, DDR activation can occur independently of chromosome segregation defects. Replication stress pathways may be responsible, because signatures of replication stress were observed, which were attenuated by exogenous supplementation with nucleosides. Our observations allow us to propose a model that explains and integrates the diverse manifestations of genomic instability induced by LT.


American Journal of Respiratory Cell and Molecular Biology | 2015

Tuberous Sclerosis Complex 2 Loss Increases Lysophosphatidylcholine Synthesis in Lymphangioleiomyomatosis

Carmen Priolo; Stéphane J. H. Ricoult; Damir Khabibullin; Harilaos Filippakis; Jane Yu; Brendan D. Manning; Clary B. Clish; Elizabeth P. Henske

Lymphangioleiomyomatosis (LAM) is a destructive lung disease affecting women. LAM is caused by mutations in the tuberous sclerosis complex (TSC) genes. The TSC protein complex inhibits the mechanistic/mammalian target of rapamycin complex 1 (mTORC1), which is a master regulator of cellular metabolism. Using mass spectrometry-based lipid profiling, we analyzed plasma from patients with LAM and discovered elevated levels of four lysophosphatidylcholine (LPC) species (C16:0, C18:0, C18:1, and C20:4) compared with those in healthy control women. To investigate whether these lipids are generated in a TSC2-dependent manner, we profiled in vitro preclinical models of TSC/LAM and found significant LPC accumulation in TSC2-deficient cells relative to TSC2-expressing control cells. These lysoglycerophospholipid changes occurred alongside changes in other phospholipid and neutral lipid species. Treatment with rapamycin or torin1 or down-regulation of sterol regulatory element-binding protein (SREBP), a lipogenic transcription factor, did not suppress LPC in TSC2-deficient cells. Inhibition of distinct isoforms of phospholipase A2 decreased the proliferation of TSC2-deficient cells. Collectively, these results demonstrate that TSC2-deficient cells have enhanced choline phospholipid metabolism and reveal a novel function of the TSC proteins in choline lysoglycerophospholipid metabolism, with implications for disease pathogenesis and targeted therapeutic strategies.


Current Cancer Drug Targets | 2015

Autophagy : Moving Benchside Promises to Patient Bedsides

Amine Belaid; Papa Diogop Ndiaye; Harilaos Filippakis; Jérémie Roux; Eric Rottinger; Yacine Graba; Patrick Brest; Paul Hofman; Baharia Mograbi

Survival rates of patients with metastatic or recurrent cancers have remained virtually unchanged during the past 30 years. This fact makes the need for new therapeutic options even more urgent. An attractive option would be to target autophagy, an essential quality control process that degrades toxic aggregates, damaged organelles, and signaling proteins, and acts as a tumor suppressor pathway of tumor initiation. Conversely, other fascinating observations suggest that autophagy supports cancer progression, relapse, metastasis, dormancy and resistance to therapy. This review provides an overview of the contradictory roles that autophagy plays in cancer initiation and progression and discusses the promises and challenges of current strategies that target autophagy for cancer therapy.


Oncotarget | 2017

Rapamycin-induced miR-21 promotes mitochondrial homeostasis and adaptation in mTORC1 activated cells

Hilaire C. Lam; Heng Jia Liu; Christian V. Baglini; Harilaos Filippakis; Nicola Alesi; Julie Nijmeh; Heng Du; Alicia Llorente Lope; Katherine A. Cottrill; Adam Handen; John M. Asara; David J. Kwiatkowski; Issam Ben-Sahra; William M. Oldham; Stephen Y. Chan; Elizabeth P. Henske

mTORC1 hyperactivation drives the multi-organ hamartomatous disease tuberous sclerosis complex (TSC). Rapamycin inhibits mTORC1, inducing partial tumor responses; however, the tumors regrow following treatment cessation. We discovered that the oncogenic miRNA, miR-21, is increased in Tsc2-deficient cells and, surprisingly, further increased by rapamycin. To determine the impact of miR-21 in TSC, we inhibited miR-21 in vitro. miR-21 inhibition significantly repressed the tumorigenic potential of Tsc2-deficient cells and increased apoptosis sensitivity. Tsc2-deficient cells’ clonogenic and anchorage independent growth were reduced by ∼50% (p<0.01) and ∼75% (p<0.0001), respectively, and combined rapamycin treatment decreased soft agar growth by ∼90% (p<0.0001). miR-21 inhibition also increased sensitivity to apoptosis. Through a network biology-driven integration of RNAseq data, we discovered that miR-21 promotes mitochondrial adaptation and homeostasis in Tsc2-deficient cells. miR-21 inhibition reduced mitochondrial polarization and function in Tsc2-deficient cells, with and without co-treatment with rapamycin. Importantly, miR-21 inhibition limited Tsc2-deficient tumor growth in vivo, reducing tumor size by approximately 3-fold (p<0.0001). When combined with rapamcyin, miR-21 inhibition showed even more striking efficacy, both during treatment and after treatment cessation, with a 4-fold increase in median survival following rapamycin cessation (p=0.0008). We conclude that miR-21 promotes mTORC1-driven tumorigenesis via a mechanism that involves the mitochondria, and that miR-21 is a potential therapeutic target for TSC-associated hamartomas and other mTORC1-driven tumors, with the potential for synergistic efficacy when combined with rapalogs.


Oncotarget | 2017

Lysosomal regulation of cholesterol homeostasis in tuberous sclerosis complex is mediated via NPC1 and LDL-R

Harilaos Filippakis; Nicola Alesi; Barbara Ogorek; Julie Nijmeh; Damir Khabibullin; Catherine Gutierrez; Alexander J. Valvezan; James Cunningham; Carmen Priolo; Elizabeth P. Henske

Tuberous sclerosis complex (TSC) is a multisystem disease associated with hyperactive mTORC1. The impact of TSC1/2 deficiency on lysosome-mediated processes is not fully understood. We report here that inhibition of lysosomal function using chloroquine (CQ) upregulates cholesterol homeostasis genes in TSC2-deficient cells. This TSC2-dependent transcriptional signature is associated with increased accumulation and intracellular levels of both total cholesterol and cholesterol esters. Unexpectedly, engaging this CQ-induced cholesterol uptake pathway together with inhibition of de novo cholesterol synthesis allows survival of TSC2-deficient, but not TSC2-expressing cells. The underlying mechanism of TSC2-deficient cell survival is dependent on exogenous cholesterol uptake via LDL-R, and endosomal trafficking mediated by Vps34. Simultaneous inhibition of lysosomal and endosomal trafficking inhibits uptake of esterified cholesterol and cell growth in TSC2-deficient, but not TSC2-expressing cells, highlighting the TSC-dependent lysosome-mediated regulation of cholesterol homeostasis and pointing toward the translational potential of these pathways for the therapy of TSC.


Biochimica et Biophysica Acta | 2011

The enhanced host-cell permissiveness of human cytomegalovirus is mediated by the Ras signaling pathway

Harilaos Filippakis; Panagiota Dimitropoulou; Aristides G. Eliopoulos; Demetrios A. Spandidos; George Sourvinos

Human cytomegalovirus utilizes cellular signal transduction pathways to activate viral or cellular transcription factors involved in the control of viral gene expression and DNA replication. In the present study, we demonstrate that Harvey-ras-transformed cells show increased permissiveness to human cytomegalovirus when compared to their parental non-transformed cells. Both the progeny viral yield and the protein levels were elevated in the human cytomegalovirus-infected Harvey-ras-transformed cells requiring active viral gene replication, as shown by the infection with UV-inactivated human cytomegalovirus. Inhibition of Ras or of key molecules of the Ras pathway, effectively suppressed viral infection in the Harvey-ras-transformed cells. On a cellular level, the human cytomegalovirus-infected Harvey-ras-transformed cells formed larger cellular foci, which were significantly higher in number, compared to the uninfected cells and preferentially recruited human cytomegalovirus virions, thereby incriminating human cytomegalovirus infection for the increased transformation of these cells. Furthermore, proliferation assays revealed a higher rate for the human cytomegalovirus-infected Harvey-ras-transformed cells compared to mock-infected cells, whereas human cytomegalovirus infection had no considerable effect on the proliferation of the non-transformed cells. Higher susceptibility to apoptosis was also detected in the human cytomegalovirus-infected ras-transformed cells, which in combination with the higher progeny virus reveals a mode by which human cytomegalovirus achieves efficient spread of infection in the cells expressing the oncogenic Harvey-ras (12V) gene. Collectively, our data suggest that human cytomegalovirus employs the host-cell Ras signaling pathway to ensue viral expression and ultimately successful propagation. Transformed cells with an activated Ras signaling pathway are therefore particularly susceptible to human cytomegalovirus infection.


Scientific Reports | 2018

Vps34-mediated macropinocytosis in Tuberous Sclerosis Complex 2-deficient cells supports tumorigenesis

Harilaos Filippakis; Amine Belaid; Brian J. Siroky; Constance Wu; Nicola Alesi; Thomas Hougard; Julie Nijmeh; Hilaire C. Lam; Elizabeth P. Henske

Tuberous Sclerosis Complex (TSC), a rare genetic disorder with mechanistic target of rapamycin complex 1 (mTORC1) hyperactivation, is characterized by multi-organ hamartomatous benign tumors including brain, skin, kidney, and lung (Lymphangioleiomyomatosis). mTORC1 hyperactivation drives metabolic reprogramming including glucose and glutamine utilization, protein, nucleic acid and lipid synthesis. To investigate the mechanisms of exogenous nutrients uptake in Tsc2-deficient cells, we measured dextran uptake, a polysaccharide internalized via macropinocytosis. Tsc2-deficient cells showed a striking increase in dextran uptake (3-fold, p < 0.0001) relative to Tsc2-expressing cells, which was decreased (3-fold, p < 0.0001) with mTOR inhibitor, Torin1. Pharmacologic and genetic inhibition of the lipid kinase Vps34 markedly abrogated uptake of Dextran in Tsc2-deficient cells. Macropinocytosis was further increased in Tsc2-deficient cells that lack autophagic mechanisms, suggesting that autophagy inhibition leads to dependence on exogenous nutrient uptake in Tsc2-deficient cells. Treatment with a macropinocytosis inhibitor, ethylisopropylamiloride (EIPA), resulted in selective growth inhibition of Atg5-deficient, Tsc2-deficient cells (50%, p < 0.0001). Genetic inhibition of autophagy (Atg5−/− MEFs) sensitized cells with Tsc2 downregulation to the Vps34 inhibitor, SAR405, resulting in growth inhibition (75%, p < 0.0001). Finally, genetic downregulation of Vps34 inhibited tumor growth and increased tumor latency in an in vivo xenograft model of TSC. Our findings show that macropinocytosis is upregulated with Tsc2-deficiency via a Vps34-dependent mechanism to support their anabolic state. The dependence of Tsc2-deficient cells on exogenous nutrients may provide novel approaches for the treatment of TSC.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Impairment of gamma-glutamyl transferase 1 activity in the metabolic pathogenesis of chromophobe renal cell carcinoma

Carmen Priolo; Damir Khabibullin; Ed Reznik; Harilaos Filippakis; Barbara Ogorek; Taylor R. Kavanagh; Julie Nijmeh; Zachary T. Herbert; John M. Asara; David J. Kwiatkowski; Chin-Lee Wu; Elizabeth P. Henske

Significance The mechanisms of chromophobe renal cell carcinoma (ChRCC) pathogenesis remain a key knowledge gap. Through metabolomics, this study uncovered a fundamental metabolic mechanism underlying the pathogenesis of ChRCC, with key therapeutic implications for this rare tumor type, for which there are currently no specific targeted therapies. Further understanding of the impact of glutathione salvage pathway on mitochondrial function, tumor progression, and targeted therapy can provide insight into other cancers characterized by aberrant glutathione salvage pathway. Chromophobe renal cell carcinoma (ChRCC) accounts for 5% of all sporadic renal cancers and can also occur in genetic syndromes including Birt–Hogg–Dube (BHD) and tuberous sclerosis complex (TSC). ChRCC has a distinct accumulation of abnormal mitochondria, accompanied by characteristic chromosomal imbalances and relatively few “driver” mutations. Metabolomic profiling of ChRCC and oncocytomas (benign renal tumors that share pathological features with ChRCC) revealed both similarities and differences between these tumor types, with principal component analysis (PCA) showing a distinct separation. ChRCC have a striking decrease in intermediates of the glutathione salvage pathway (also known as the gamma-glutamyl cycle) compared with adjacent normal kidney, as well as significant changes in glycolytic and pentose phosphate pathway intermediates. We also found that gamma glutamyl transferase 1 (GGT1), the key enzyme of the gamma-glutamyl cycle, is expressed at ∼100-fold lower levels in ChRCC compared with normal kidney, while no change in GGT1 expression was found in clear cell RCC (ccRCC). Significant differences in specific metabolite abundance were found in ChRCC vs. ccRCC, including the oxidative stress marker ophthalmate. Down-regulation of GGT1 enhanced the sensitivity to oxidative stress and treatment with buthionine sulfoximine (BSO), which was associated with changes in glutathione-pathway metabolites. These data indicate that impairment of the glutathione salvage pathway, associated with enhanced oxidative stress, may have key therapeutic implications for this rare tumor type for which there are currently no specific targeted therapies.


Archive | 2018

Autophagy-Driven Cancer Drug Development

Amine Belaid; Barnabé Roméo; Harilaos Filippakis; Mickael Meyer; Iris Grosjean; Nathalie Yazbeck; Marie Angela Domdom; Guillemette Crépeaux; Romain K. Gherardi; Dominique Lagadic-Gossmann; Abderrahman Chargui; Eric Gilson; Delphine Benarroch-Popivker; Patrick Brest; Paul Hofman; Baharia Mograbi

Survival rates of patients with metastatic or recurrent cancers have remained virtually unchanged during the past 30 years. This fact makes the need for new therapeutic options even more urgent. An attractive option would be to target autophagy, an essential quality control process that degrades toxic aggregates, damaged organelles, and signaling proteins, and acts as a tumor suppressor pathway of tumor initiation. Conversely, other fascinating observations suggest that autophagy supports cancer progression, relapse, metastasis, dormancy, and resistance to therapy. This chapter provides an overview of the contradictory roles that autophagy plays in cancer initiation and progression and discusses the promises and challenges of current strategies that target autophagy for cancer therapy.


Clinical Cancer Research | 2017

Abstract A09: Therapeutic targeting of TSC2-deficient cells with Methotrexate: Results of a drug repurposing screen

Amine Belaid; Harilaos Filippakis; Hilaire C. Lam; Alexander J. Valvezan; Ruili Huang; Menghang Xia; Tianmu Wen; Christian V. Baglini; Srilatha Sakamuru; John M. Asara; Nguyen Truong Sinh; Jeffrey J. Heard; Fuyuhiko Tamanoi; Christopher M. Austin; Brendan D. Manning; Elizabeth P. Henske

Tuberous sclerosis complex (TSC) is a multisystem disorder that affects multiple organ systems, including tumors of the brain, heart, kidney, skin and lung. TSC is caused by germline loss-of-function gene mutations in TSC1 or TSC2, which inhibit the mammalian target of rapamycin (mTOR) signaling pathway. Rapalogs are effective cytostatic agents for the treatment of TSC, but continual lifelong therapy is needed. Therapies for TSC that induce a selective cytocidal response in TSC-deficient cells are not currently available, and could have a major clinical impact for TSC patients. We performed a quantitative high-throughput screen (qHTS) of the NCATS Pharmaceutical Collection (NPC) of 2816 compounds, many of which are drugs already approved for other diseases, for their effect on the proliferation of TSC2-deficient ELT3 cells vs. ELT3 cells in which TSC2 was re-expressed. Agents were tested at fifteen concentrations, from 0.6 nM to 46 µM. Six agents selectively inhibited the growth of the TSC2-deficient cells relative to the TSC2-reexpressing cells. One of top inhibitory agents, methotrexate (MTX), a dihydrofolate reductase inhibitor, is FDA-approved, making it a candidate for “repurposing”. In our studies, we confirmed the selective inhibitory effect of MTX on patient-derived TSC2-deficient cells. Flow cytometry analysis demonstrated that MTX (5 µM, 24 h) arrested 60.8% of the TSC2-deficient cells in S-phase, vs. 26.6% of the TSC2-expressing cells. The accumulation of S-phase arrested cells ultimately leads to apoptosis selectively in the TSC2-deficient cells, which was detected by flow cytometry of Propidium Iodide/Annexin V stained cells (52.3% of apoptosis in TSC2-deficient cells vs. 11% in TSC2-reexpressing cells) and immunoblotting for cleaved Caspase3 and cleaved PARP. Co-treatment with the mTOR inhibitor Rapamycin and MTX prevented MTX-induced apoptosis (52.3% apoptotic cells with MTX alone vs. 11.8% with MTX plus Rapamycin). In vivo, mice bearing TSC2-deficient ELT3 cell xenograft tumors were treated with MTX (25mg/kg or 50mg/kg) or rapamycin (1mg/kg) by intraperitoneal injection every other day over 3 weeks. We found that 50mg/kg of MTX significantly decreased the volume of tumors within two weeks of treatment, to a similar extent as Rapamycin treatment did. We conclude that Methotrexate acts as a selective cytotoxic agent in TSC2-deficient cells. This is consistent with recent discoveries that mTOR regulates the de novo purine and pyrimidine synthesis pathways. As an FDA-approved agent, Methotrexate has potential for clinical “repurposing” for the treatment of TSC-deficient tumors. Citation Format: Amine Belaid, Harilaos Filippakis, Hilaire Lam, Alexander Valvezan, Ruili Huang, Menghang Xia, Tianmu Wen, Christian Baglini, Srilatha Sakamuru, John Asara, Nguyen Truong Sinh, Jeffrey J. Heard, Fuyuhiko Tamanoi, Christopher Austin, Brendan Manning, Elizabeth P. Henske. Therapeutic targeting of TSC2-deficient cells with Methotrexate: Results of a drug repurposing screen. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Targeting the Vulnerabilities of Cancer; May 16-19, 2016; Miami, FL. Philadelphia (PA): AACR; Clin Cancer Res 2017;23(1_Suppl):Abstract nr A09.

Collaboration


Dive into the Harilaos Filippakis's collaboration.

Top Co-Authors

Avatar

Elizabeth P. Henske

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Carmen Priolo

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Damir Khabibullin

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Hilaire C. Lam

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Barbara Ogorek

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Julie Nijmeh

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Nicola Alesi

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amine Belaid

University of Nice Sophia Antipolis

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge