Network


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

Hotspot


Dive into the research topics where Nicola Alesi is active.

Publication


Featured researches published by Nicola Alesi.


Current Drug Metabolism | 2011

Clinical pharmacogenetics of methotrexate.

Pasqualina Castaldo; Simona Magi; Annamaria Assunta Nasti; Sara Arcangeli; Vincenzo Lariccia; Nicola Alesi; Massimo Tocchini; Salvatore Amoroso

It is well known that interindividual variability can affect the response to many drugs in relation to age, gender, diet, and organ function. Pharmacogenomic studies have also documented that genetic polymorphisms can exert clinically significant effects in terms of drug resistance, efficacy and toxicity by modifying the expression of critical gene products (drug-metabolizing enzymes, transporters, and target molecules) as well as pharmacokinetic and pharmacodynamic parameters. A growing body of in vitro and clinical evidence suggests that common polymorphisms in the folate gene pathway are associated with an altered response to methotrexate (MTX) in patients with malignancy and autoimmune disease. Such polymorphisms may also induce significant MTX toxicity requiring expensive monitoring and treatment. Although the available data are not conclusive, they suggest that in the future MTX pharmacogenetics could play a key role in clinical practice by improving and tailoring treatment. This review describes the genetic polymorphisms that significantly influence MTX resistance, efficacy, and toxicity.


Cancer Research | 2017

p62/SQSTM1 cooperates with hyperactive mTORC1 to regulate glutathione production, maintain mitochondrial integrity and promote tumorigenesis

Hilaire C. Lam; Christian V. Baglini; Alicia Llorente Lope; Andrey Parkhitko; Heng-Jia Liu; Nicola Alesi; Izabela A. Malinowska; Darius Ebrahimi-Fakhari; Afshin Saffari; Jane Yu; Ana Pereira; Damir Khabibullin; Barbara Ogorek; Julie Nijmeh; Taylor R. Kavanagh; Adam Handen; Stephen Y. Chan; John M. Asara; William M. Oldham; Maria T. Diaz-Meco; Jorge Moscat; Mustafa Sahin; Carmen Priolo; Elizabeth P. Henske

p62/sequestosome-1 (SQSTM1) is a multifunctional adaptor protein and autophagic substrate that accumulates in cells with hyperactive mTORC1, such as kidney cells with mutations in the tumor suppressor genes tuberous sclerosis complex (TSC)1 or TSC2. Here we report that p62 is a critical mediator of TSC2-driven tumorigenesis, as Tsc2+/- and Tsc2f/f Ksp-CreERT2+ mice crossed to p62-/- mice were protected from renal tumor development. Metabolic profiling revealed that depletion of p62 in Tsc2-null cells decreased intracellular glutamine, glutamate, and glutathione (GSH). p62 positively regulated the glutamine transporter Slc1a5 and increased glutamine uptake in Tsc2-null cells. We also observed p62-dependent changes in Gcl, Gsr, Nqo1, and Srxn1, which were decreased by p62 attenuation and implicated in GSH production and utilization. p62 attenuation altered mitochondrial morphology, reduced mitochondrial membrane polarization and maximal respiration, and increased mitochondrial reactive oxygen species and mitophagy marker PINK1. These mitochondrial phenotypes were rescued by addition of exogenous GSH and overexpression of Sod2, which suppressed indices of mitochondrial damage and promoted growth of Tsc2-null cells. Finally, p62 depletion sensitized Tsc2-null cells to both oxidative stress and direct inhibition of GSH biosynthesis by buthionine sulfoximine. Our findings show how p62 helps maintain intracellular pools of GSH needed to limit mitochondrial dysfunction in tumor cells with elevated mTORC1, highlighting p62 and redox homeostasis as nodal vulnerabilities for therapeutic targeting in these tumors. Cancer Res; 77(12); 3255-67. ©2017 AACR.


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.


JCI insight | 2018

TSC2-deficient tumors have evidence of T cell exhaustion and respond to anti–PD-1/anti–CTLA-4 immunotherapy

Heng-Jia Liu; Patrick H. Lizotte; Heng Du; Maria C. Speranza; Hilaire C. Lam; Spencer Vaughan; Nicola Alesi; Kwok-Kin Wong; Gordon J. Freeman; Arlene H. Sharpe; Elizabeth P. Henske

Tuberous sclerosis complex (TSC) is an incurable multisystem disease characterized by mTORC1-hyperactive tumors. TSC1/2 mutations also occur in other neoplastic disorders, including lymphangioleiomyomatosis (LAM) and bladder cancer. Whether TSC-associated tumors will respond to immunotherapy is unknown. We report here that the programmed death 1 coinhibitory receptor (PD-1) is upregulated on T cells in renal angiomyolipomas (AML) and pulmonary lymphangioleiomyomatosis (LAM). In C57BL/6J mice injected with syngeneic TSC2-deficient cells, anti-PD-1 alone decreased 105K tumor growth by 67% (P < 0.0001); the combination of PD-1 and CTLA-4 blockade was even more effective in suppressing tumor growth. Anti-PD-1 induced complete rejection of TSC2-deficient 105K tumors in 37% of mice (P < 0.05). Double blockade of PD-1 and CTLA-4 induced rejection in 62% of mice (P < 0.01). TSC2 reexpression in TSC2-deficient TMKOC cells enhanced antitumor immunity by increasing T cell infiltration and production of IFN-γ/TNF-α by T cells, suggesting that TSC2 and mTORC1 play specific roles in the induction of antitumor immunity. Finally, 1 month of anti-PD-1 blockade reduced renal tumor burden by 53% (P < 0.01) in genetically engineered Tsc2+/- mice. Taken together, these data demonstrate for the first time to our knowledge that checkpoint blockade may have clinical efficacy for TSC and LAM, and possibly other benign tumor syndromes, potentially yielding complete and durable clinical responses.


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.


Cancer Research | 2016

Abstract A35: Regulation of the tumor microenvironment by oncogenic p62 in mTORC1-hyperactive diseases: Tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM)

Hilaire C. Lam; Andrey Parkhitko; Alicia Llorente Lope; Nicola Alesi; Damir Khabibullin; Harilaos Filippakis; Ana Pereira; Barbara Ogorek; Erik Zhang; Jane Yu; Carmen Priolo; Elizabeth P. Henske

Tuberous sclerosis complex (TSC) is a multisystem disease associated with tumors of the brain, skin and kidney as well as progressive cystic lung destruction characteristic of lymphangioleiomyomatosis (LAM). TSC and LAM are caused by loss-of-function mutations in TSC1 or TSC2, resulting in hyperactive mechanistic Target of Rapamycin complex 1 (mTORC1) signaling. We previously discovered that mTORC1 hyperactivation in TSC inhibits autophagy and promotes the accumulation of oncogenic p62/sequestosome 1 (PNAS, 2011). p62 exerts a pleiotropic role in tumor progression, including the regulation of NRF2 to combat oxidative stress, NFκB to promote cell survival and the degradation of ubiquitinated proteins to efficiently recycle organelles and proteins by autophagy. Knockdown of p62 in Tsc2-null cells inhibited tumorigenesis in xenografts in vivo, but did not impact proliferation in vitro, leading us to hypothesize that p62 exerts non-cell autonomous functions influencing the tumor microenvironment to promote Tsc2-/- cell tumorigenic potential. In unpublished work, we explored the molecular mechanisms by which p62 promotes tumorigenesis in TSC/LAM. First, we analyzed renal tumor burden in Tsc2+/-; p62-/- mice. Compared to littermate control Tsc2+/-; p62+/+ mice, Tsc2+/-; p62-/- mice have a 2-fold (p Our data point toward a model in which p62 and mTORC1 cooperatively regulate cellular IL6 in Tsc2-null cells to promote tumorigenesis in TSC. IL6 is correlated with poor prognosis, advanced disease and metastatic potential in numerous cancers. Rapamycin dramatically decreases p62 levels in TSC-deficient cells. Therefore, the induction of IL6 by rapamycin through decreased p62 expression may represent one of the first non-cell autonomous mechanisms by which mTORC1 inhibition impacts tumorigenesis. Since mTORC1 is hyperactivated in the majority of malignant tumors and more than 100 cancer clinical trials are ongoing with mTORC1 inhibitors, our findings may yield insights with broad therapeutic potential. Citation Format: Hilaire C. Lam, Andrey Parkhitko, Alicia Llorente Lope, Nicola Alesi, Damir Khabibullin, Harilaos Filippakis, Ana Pereira, Barbara Ogorek, Erik Zhang, Jane Yu, Carmen Priolo, Elizabeth P. Henske. Regulation of the tumor microenvironment by oncogenic p62 in mTORC1-hyperactive diseases: Tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM). [abstract]. In: Proceedings of the AACR Special Conference: Function of Tumor Microenvironment in Cancer Progression; 2016 Jan 7–10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2016;76(15 Suppl):Abstract nr A35.


Cancer Research | 2018

Abstract 1686: TSC2 enhances antitumor immunity and potentiates PD-1 and CTLA-4 blockade

Heng-Jia Liu; Patrick Lizotte; Heng Du; Maria C. Speranza; Spencer Vaughan; Nicola Alesi; Kwok-Kin Wong; Gordon J. Freeman; Arlene H. Sharpe; Elizabeth P. Henske


PLOS | 2016

Whole Exome Sequencing Identifies TSC1/TSC2 Biallelic Loss as the Primary and Sufficient Driver Event for Renal Angiomyolipoma Development

Krinio Giannikou; Izabela A. Malinowska; Trevor J. Pugh; Rachel Yan; Yuen-Yi Tseng; Jaegil Kim; Magdalena E. Tyburczy; Yvonne Chekaluk; Yang Liu; Nicola Alesi; Geraldine A. Finlay; Chin-Lee Wu; Sabina Signoretti; Elizabeth P. Henske; David J. Kwiatkowski; Coyin Oh; Matthew Meyerson; Gad Getz; Jesse S. Boehm


Circulation | 2014

Abstract 17264: β-Adrenergic Signaling Promotes Calcium Oscillations in Human Cardiac Progenitor Cells

Nicola Alesi; Chiara Mangiaracina; Polina Goichberg; Giulia Borghetti; Sergio Signore; Silvana Bardelli; Marco Moccetti; Andrea Sorrentino; Ewa Wybieralska; Laura Graciotti; Antonio Cannata; Kanako Waight; Sang Hun Shin; Jung Kim; Annarosa Leri; Piero Anversa; Tiziano Moccetti; Marcello Rota

Collaboration


Dive into the Nicola Alesi's collaboration.

Top Co-Authors

Avatar

Elizabeth P. Henske

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

Harilaos Filippakis

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
Top Co-Authors

Avatar

Andrea Sorrentino

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Annarosa Leri

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Antonio Cannata

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Carmen Priolo

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge