726 results on '"Ambesi-Impiombato A"'
Search Results
2. Computational framework for the prediction of transcription factor binding sites by multiple data integration
- Author
-
Bansal Mukesh, Ambesi-Impiombato Alberto, Liò Pietro, and di Bernardo Diego
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurophysiology and neuropsychology ,QP351-495 - Abstract
Abstract Control of gene expression is essential to the establishment and maintenance of all cell types, and its dysregulation is involved in pathogenesis of several diseases. Accurate computational predictions of transcription factor regulation may thus help in understanding complex diseases, including mental disorders in which dysregulation of neural gene expression is thought to play a key role. However, biological mechanisms underlying the regulation of gene expression are not completely understood, and predictions via bioinformatics tools are typically poorly specific. We developed a bioinformatics workflow for the prediction of transcription factor binding sites from several independent datasets. We show the advantages of integrating information based on evolutionary conservation and gene expression, when tackling the problem of binding site prediction. Consistent results were obtained on a large simulated dataset consisting of 13050 in silico promoter sequences, on a set of 161 human gene promoters for which binding sites are known, and on a smaller set of promoters of Myc target genes. Our computational framework for binding site prediction can integrate multiple sources of data, and its performance was tested on different datasets. Our results show that integrating information from multiple data sources, such as genomic sequence of genes' promoters, conservation over multiple species, and gene expression data, indeed improves the accuracy of computational predictions.
- Published
- 2006
- Full Text
- View/download PDF
3. Enrichment analysis of phenotypic data for drug repurposing in rare diseases
- Author
-
Alberto Ambesi-Impiombato, Kimberly Cox, Sylvie Ramboz, Daniela Brunner, Mukesh Bansal, and Emer Leahy
- Subjects
drug discovery ,phenotypic screening ,tianeptine ,huntington (disease) ,animal models ,smartcube ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Drug-induced Behavioral Signature Analysis (DBSA), is a machine learning (ML) method for in silico screening of compounds, inspired by analytical methods quantifying gene enrichment in genomic analyses. When applied to behavioral data it can identify drugs that can potentially reverse in vivo behavioral symptoms in animal models of human disease and suggest new hypotheses for drug discovery and repurposing. We present a proof-of-concept study aiming to assess Drug-induced Behavioral Signature Analysis (DBSA) as a systematic approach for drug discovery for rare disorders. We applied Drug-induced Behavioral Signature Analysis to high-content behavioral data obtained with SmartCube®, an automated in vivo phenotyping platform. The therapeutic potential of several dozen approved drugs was assessed for phenotypic reversal of the behavioral profile of a Huntington’s Disease (HD) murine model, the Q175 heterozygous knock-in mice. The in silico Drug-induced Behavioral Signature Analysis predictions were enriched for drugs known to be effective in the symptomatic treatment of Huntington’s Disease, including bupropion, modafinil, methylphenidate, and several SSRIs, as well as the atypical antidepressant tianeptine. To validate the method, we tested acute and chronic effects of tianeptine (20 mg/kg, i. p.) in vivo, using Q175 mice and wild type controls. In both experiments, tianeptine significantly rescued the behavioral phenotype assessed with the SmartCube® platform. Our target-agnostic method thus showed promise for identification of symptomatic relief treatments for rare disorders, providing an alternative method for hypothesis generation and drug discovery for disorders with huge disease burden and unmet medical needs.
- Published
- 2023
- Full Text
- View/download PDF
4. Clonal evolution mechanisms in NT5C2 mutant-relapsed acute lymphoblastic leukaemia.
- Author
-
Tzoneva, Gannie, Dieck, Chelsea L, Oshima, Koichi, Ambesi-Impiombato, Alberto, Sánchez-Martín, Marta, Madubata, Chioma J, Khiabanian, Hossein, Yu, Jiangyan, Waanders, Esme, Iacobucci, Ilaria, Sulis, Maria Luisa, Kato, Motohiro, Koh, Katsuyoshi, Paganin, Maddalena, Basso, Giuseppe, Gastier-Foster, Julie M, Loh, Mignon L, Kirschner-Schwabe, Renate, Mullighan, Charles G, Rabadan, Raul, and Ferrando, Adolfo A
- Subjects
Animals ,Humans ,Mice ,Disease Models ,Animal ,Recurrence ,Purines ,5'-Nucleotidase ,IMP Dehydrogenase ,Guanosine ,Xenograft Model Antitumor Assays ,Cell Proliferation ,Drug Resistance ,Neoplasm ,Mutation ,Female ,Male ,Receptor ,Notch1 ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,HEK293 Cells ,Clonal Evolution ,Gain of Function Mutation ,Mercaptopurine ,5-Nucleotidase ,Disease Models ,Animal ,Drug Resistance ,Neoplasm ,Receptor ,Notch1 ,General Science & Technology - Abstract
Relapsed acute lymphoblastic leukaemia (ALL) is associated with resistance to chemotherapy and poor prognosis. Gain-of-function mutations in the 5'-nucleotidase, cytosolic II (NT5C2) gene induce resistance to 6-mercaptopurine and are selectively present in relapsed ALL. Yet, the mechanisms involved in NT5C2 mutation-driven clonal evolution during the initiation of leukaemia, disease progression and relapse remain unknown. Here we use a conditional-and-inducible leukaemia model to demonstrate that expression of NT5C2(R367Q), a highly prevalent relapsed-ALL NT5C2 mutation, induces resistance to chemotherapy with 6-mercaptopurine at the cost of impaired leukaemia cell growth and leukaemia-initiating cell activity. The loss-of-fitness phenotype of NT5C2+/R367Q mutant cells is associated with excess export of purines to the extracellular space and depletion of the intracellular purine-nucleotide pool. Consequently, blocking guanosine synthesis by inhibition of inosine-5'-monophosphate dehydrogenase (IMPDH) induced increased cytotoxicity against NT5C2-mutant leukaemia lymphoblasts. These results identify the fitness cost of NT5C2 mutation and resistance to chemotherapy as key evolutionary drivers that shape clonal evolution in relapsed ALL and support a role for IMPDH inhibition in the treatment of ALL.
- Published
- 2018
5. Benefits of global mutant huntingtin lowering diminish over time in a Huntington’s disease mouse model
- Author
-
Deanna M. Marchionini, Jeh-Ping Liu, Alberto Ambesi-Impiombato, Kimberly Kerker, Kim Cirillo, Mukesh Bansal, Rich Mushlin, Daniela Brunner, Sylvie Ramboz, Mei Kwan, Kirsten Kuhlbrodt, Karsten Tillack, Finn Peters, Leena Rauhala, John Obenauer, Jonathan R. Greene, Christopher Hartl, Vinod Khetarpal, Brenda Lager, Jim Rosinski, Jeff Aaronson, Morshed Alam, Ethan Signer, Ignacio Muñoz-Sanjuán, David Howland, and Scott O. Zeitlin
- Subjects
Neuroscience ,Medicine - Abstract
We have developed an inducible Huntington’s disease (HD) mouse model that allows temporal control of whole-body allele-specific mutant huntingtin (mHtt) expression. We asked whether moderate global lowering of mHtt (~50%) was sufficient for long-term amelioration of HD-related deficits and, if so, whether early mHtt lowering (before measurable deficits) was required. Both early and late mHtt lowering delayed behavioral dysfunction and mHTT protein aggregation, as measured biochemically. However, long-term follow-up revealed that the benefits, in all mHtt-lowering groups, attenuated by 12 months of age. While early mHtt lowering attenuated cortical and striatal transcriptional dysregulation evaluated at 6 months of age, the benefits diminished by 12 months of age, and late mHtt lowering did not ameliorate striatal transcriptional dysregulation at 12 months of age. Only early mHtt lowering delayed the elevation in cerebrospinal fluid neurofilament light chain that we observed in our model starting at 9 months of age. As small-molecule HTT-lowering therapeutics progress to the clinic, our findings suggest that moderate mHtt lowering allows disease progression to continue, albeit at a slower rate, and could be relevant to the degree of mHTT lowering required to sustain long-term benefits in humans.
- Published
- 2022
- Full Text
- View/download PDF
6. A Quantitative Analysis of Localized Robustness of MYCN in Neuroblastoma
- Author
-
Sandhu, Romeil, Tannenbaum, Sarah, Diolaiti, Daniel, Ambesi-Impiombato, Alberto, Kung, Andrew, and Tannenbaum, Allen
- Subjects
Quantitative Biology - Molecular Networks - Abstract
The amplification of the gene MYCN (V-myc myelocytomatosis viral-valeted oncogene, neuroblastoma derived) has been a well-documented indicator for poor prognosis in neuroblastoma, a childhood cancer. Unfortunately, there has been limited success in understanding MYCN functionality in the landscape of neuroblastoma and more importantly, given that MYCN has been deemed undruggable, the need to potentially illuminate key opportunities that indirectly target MYCN is of great interest. To this end, this work employs an emerging quantitative technique from network science, namely network curvature, to quantify the biological robustness of MYCN and its surrounding neighborhood. In particular, when amplified in Stage IV cancer, MYCN exhibits higher curvature (more robust) than those samples with under expressed MYCN levels. When examining the surrounding neighborhood, the above argument still holds for network curvature, but is lost when only analyzing differential expression - a common technique amongst oncologists and computational/molecular biologists. This finding points to the problem (and possible solution) of drug targeting in the context of complexity and indirect cell signaling affects that have often been obfuscated through traditional techniques.
- Published
- 2015
7. Mutational and functional genetics mapping of chemotherapy resistance mechanisms in relapsed acute lymphoblastic leukemia
- Author
-
Oshima, Koichi, Zhao, Junfei, Pérez-Durán, Pablo, Brown, Jessie A., Patiño-Galindo, Juan Angel, Chu, Timothy, Quinn, Aidan, Gunning, Thomas, Belver, Laura, Ambesi-Impiombato, Alberto, Tosello, Valeria, Wang, Zhengqiang, Sulis, Maria Luisa, Kato, Motohiro, Koh, Katsuyoshi, Paganin, Maddalena, Basso, Giuseppe, Balbin, Milagros, Nicolas, Concepcion, Gastier-Foster, Julie M., Devidas, Meenakshi, Loh, Mignon L., Paietta, Elisabeth, Tallman, Martin S., Rowe, Jacob M., Litzow, Mark, Minden, Mark D., Meijerink, Jules, Rabadan, Raul, and Ferrando, Adolfo
- Published
- 2020
- Full Text
- View/download PDF
8. Spaceflight Induced Disorders: Potential Nutritional Countermeasures
- Author
-
Fabio Costa, Francesco Saverio Ambesi-Impiombato, Tommaso Beccari, Carmela Conte, Samuela Cataldi, Francesco Curcio, and Elisabetta Albi
- Subjects
microgravity ,energy intake ,life-style ,nutrition ,spaceflight ,Biotechnology ,TP248.13-248.65 - Abstract
Space travel is an extreme experience even for the astronaut who has received extensive basic training in various fields, from aeronautics to engineering, from medicine to physics and biology. Microgravity puts a strain on members of space crews, both physically and mentally: short-term or long-term travel in orbit the International Space Station may have serious repercussions on the human body, which may undergo physiological changes affecting almost all organs and systems, particularly at the muscular, cardiovascular and bone compartments. This review aims to highlight recent studies describing damages of human body induced by the space environment for microgravity, and radiation. All novel conditions, to ally unknown to the Darwinian selection strategies on Earth, to which we should add the psychological stress that astronauts suffer due to the inevitable forced cohabitation in claustrophobic environments, the deprivation from their affections and the need to adapt to a new lifestyle with molecular changes due to the confinement. In this context, significant nutritional deficiencies with consequent molecular mechanism changes in the cells that induce to the onset of physiological and cognitive impairment have been considered.
- Published
- 2021
- Full Text
- View/download PDF
9. The Transcriptional Landscape of the Mammalian Genome
- Author
-
Carninci, P., Kasukawa, T., Katayama, S., Gough, J., Frith, M. C., Maeda, N., Oyama, R., Ravasi, T., Lenhard, B., Wells, C., Kodzius, R., Shimokawa, K., Bajic, V. B., Brenner, S. E., Batalov, S., Forrest, A. R. R., Zavolan, M., Davis, M. J., Wilming, L. G., Aidinis, V., Allen, J. E., Ambesi-Impiombato, A., Apweiler, R., Aturaliya, R. N., Bailey, T. L., Bansal, M., Baxter, L., Beisel, K. W., Bersano, T., Bono, H., Chalk, A. M., Chiu, K. P., Choudhary, V., Christoffels, A., Clutterbuck, D. R., Crowe, M. L., Dalla, E., Dalrymple, B. P., de Bono, B., Della Gatta, G., di Bernardo, D., Down, T., Engstrom, P., Fagiolini, M., Faulkner, G., Fletcher, C. F., Fukushima, T., Furuno, M., Futaki, S., Gariboldi, M., Georgii-Hemming, P., Gingeras, T. R., Gojobori, T., Green, R. E., Gustincich, S., Harbers, M., Hayashi, Y., Hensch, T. K., Hirokawa, N., Hill, D., Huminiecki, L., Iacono, M., Ikeo, K., Iwama, A., Ishikawa, T., Jakt, M., Kanapin, A., Katoh, M., Kawasawa, Y., Kelso, J., Kitamura, H., Kitano, H., Kollias, G., Krishnan, S. P. T., Kruger, A., Kummerfeld, S. K., Kurochkin, I. V., Lareau, L. F., Lazarevic, D., Lipovich, L., Liu, J., Liuni, S., McWilliam, S., Babu, M. Madan, Madera, M., Marchionni, L., Matsuda, H., Matsuzawa, S., Miki, H., Mignone, F., Miyake, S., Morris, K., Mottagui-Tabar, S., Mulder, N., Nakano, N., Nakauchi, H., Ng, P., Nilsson, R., Nishiguchi, S., Nishikawa, S., Nori, F., Ohara, O., Okazaki, Y., Orlando, V., Pang, K. C., Pavan, W. J., Pavesi, G., Pesole, G., Petrovsky, N., Piazza, S., Reed, J., Reid, J. F., Ring, B. Z., Ringwald, M., Rost, B., Ruan, Y., Salzberg, S. L., Sandelin, A., Schneider, C., Schöbach, C., Sekiguchi, K., Semple, C. A. M., Seno, S., Sessa, L., Sheng, Y., Shibata, Y., Shimada, H., Shimada, K., Silva, D., Sinclair, B., Sperling, S., Stupka, E., Sugiura, K., Sultana, R., Takenaka, Y., Taki, K., Tammoja, K., Tan, S. L., Tang, S., Taylor, M. S., Tegner, J., Teichmann, S. A., Ueda, H. R., van Nimwegen, E., Verardo, R., Wei, C. L., Yagi, K., Yamanishi, H., Zabarovsky, E., Zhu, S., Zimmer, A., Hide, W., Bult, C., Grimmond, S. M., Teasdale, R. D., Liu, E. T., Brusic, V., Quackenbush, J., Wahlestedt, C., Mattick, J. S., Hume, D. A., Kai, C., Sasaki, D., Tomaru, Y., Fukuda, S., Kanamori-Katayama, M., Suzuki, M., Aoki, J., Arakawa, T., Iida, J., Imamura, K., Itoh, M., Kato, T., Kawaji, H., Kawagashira, N., Kawashima, T., Kojima, M., Kondo, S., Konno, H., Nakano, K., Ninomiya, N., Nishio, T., Okada, M., Plessy, C., Shibata, K., Shiraki, T., Suzuki, S., Tagami, M., Waki, K., Watahiki, A., Okamura-Oho, Y., Suzuki, H., and Kawai, J.
- Published
- 2005
10. Synergistic antileukemic therapies in NOTCH1 -induced T-ALL
- Author
-
Sanchez-Martin, Marta, Ambesi-Impiombato, Alberto, Qin, Yue, Herranz, Daniel, Bansal, Mukesh, Girardi, Tiziana, Paietta, Elisabeth, Tallman, Martin S., Rowe, Jacob M., De Keersmaecker, Kim, Califano, Andrea, and Ferrando, Adolfo A.
- Published
- 2017
11. Growth Factor-Dependent Activation of αvβ3 Integrin in Normal Epithelial Cells: Implications for Tumor Invasion
- Author
-
Trusolino, Livio, Serini, Guido, Cecchini, Germana, Besati, Cristina, Ambesi-Impiombato, Francesco Saverio, Marchisio, Pier Carlo, and De Filippi, Rosaria
- Published
- 1998
12. Oncogenic hijacking of the stress response machinery in T cell acute lymphoblastic leukemia
- Author
-
Kourtis, Nikos, Lazaris, Charalampos, Hockemeyer, Kathryn, Balandrán, Juan Carlos, Jimenez, Alejandra R., Mullenders, Jasper, Gong, Yixiao, Trimarchi, Thomas, Bhatt, Kamala, Hu, Hai, Shrestha, Liza, Ambesi-Impiombato, Alberto, Kelliher, Michelle, Paietta, Elisabeth, Chiosis, Gabriela, Guzman, Monica L., Ferrando, Adolfo A., Tsirigos, Aristotelis, and Aifantis, Iannis
- Published
- 2018
- Full Text
- View/download PDF
13. Pharmacological inhibition of the transcription factor PU.1 in leukemia
- Author
-
Antony-Debre, Ileana, Paul, Ananya, Leite, Joana, Mitchell, Kelly, Kim, Hye Mi, Carvajal, Luis A., Todorova, Tihomira I., Huang, Kenneth, Kumar, Arvind, Farahat, Abdelbasset A., Bartholdy, Boris, Narayanagari, Swathi-Rao, Chen, Jiahao, Ambesi-Impiombato, Alberto, Ferrando, Adolfo A., Mantzaris, loannis, Gavathiotis, Evripidis, Verma, Amit, Will, Britta, Boykin, David W., Wilson, W. David, Poon, Gregory M.K., and Steidl, Ulrich
- Subjects
Mutation -- Research ,Transcription factors -- Genetic aspects ,Acute myelocytic leukemia -- Patient outcomes -- Genetic aspects -- Care and treatment ,Health care industry - Abstract
The transcription factor PU.1 is often impaired in patients with acute myeloid leukemia (AML). Here, we used AML cells that already had low PU.1 levels and further inhibited PU.1 using either RNA interference or, to our knowledge, first-in-class smallmolecule inhibitors of PU.1 that we developed specifically to allosterically interfere with PU.1-chromatin binding through interaction with the DNA minor groove that flanks PU.1-binding motifs. These small molecules of the heterocyclic diamidine family disrupted the interaction of PU.1 with target gene promoters and led to downregulation of canonical PU.1 transcriptional targets. shRNA or small-molecule inhibition of PU.1 in AML cells from either [PU.1.sup.lo] mutant mice or human patients with AML-inhibited cell growth and clonogenicity and induced apoptosis. In murine and human AML (xeno)transplantation models, treatment with our PU.1 inhibitors decreased tumor burden and resulted in increased survival. Thus, our study provides proof of concept that PU.1 inhibition has potential as a therapeutic strategy for the treatment of AML and for the development of small-molecule inhibitors of PU.1., Introduction Acute myeloid leukemia (AML) is a cancer of the hematopoietic system characterized by the abnormal clonal proliferation of immature cells following various genetic and epigenetic alterations. Despite efforts to [...]
- Published
- 2017
- Full Text
- View/download PDF
14. Localization of nuclear actin in nuclear lipid microdomains of liver and hepatoma cells: Possible involvement of sphingomyelin metabolism
- Author
-
Cataldi Samuela, Lazzarini Andrea, Codini Michela, Cascianelli Giacomo, Floridi Alessandro, Bartoccini Elisa, Ceccarini Maria Rachele, Ambesi-Impiombato Francesco Saverio, Beccari Tommaso, Curcio Francesco, and Albi Elisabetta
- Subjects
Biotechnology ,TP248.13-248.65 - Abstract
Nuclear actin has been implicated in different nuclear functions. In this work, its localization in nuclear membrane, chromatin and nuclear lipid microdomains was investigated. The implication of sphingomyelin metabolism was studied. Nuclear membrane, chromatin and nuclear lipid microdomains were purified from hepatocyte nuclei and H35 human hepatoma cell nuclei. The presence of β-actin was analyzed with immunoblotting by using specific antibodies. Sphingomyelinase, sphingomyelin-synthase, and phosphatidylcholine-specific phospholipase C activities were assayed by using radioactivity sphingomyelin and phosphatidylcholine as substrate. The results showed that β-actin is localized in nuclear lipid microdomains and it increases in cancer cells. Evidence is provided to the difference of phosphatidylcholine and sphingomyelin metabolism in various subnuclear fractions of cancer cell nuclei compared with normal cells. Our findings show increase of sphingomyelin-synthase and inhibition of sphingomyelinase activity only in nuclear lipid microdomains. Nuclear lipid microdomains, constituted by phosphatidylcholine, sphingomyelin and cholesterol, play a role as platform for β-actin anchoring. Possible role of sphingomyelin metabolism in cancer cells is discussed.
- Published
- 2017
- Full Text
- View/download PDF
15. Supplementary Data from Phf6 Loss Enhances HSC Self-Renewal Driving Tumor Initiation and Leukemia Stem Cell Activity in T-ALL
- Author
-
Wendorff, Agnieszka A., primary, Quinn, S. Aidan, primary, Rashkovan, Marissa, primary, Madubata, Chioma J., primary, Ambesi-Impiombato, Alberto, primary, Litzow, Mark R., primary, Tallman, Martin S., primary, Paietta, Elisabeth, primary, Paganin, Maddalena, primary, Basso, Giuseppe, primary, Gastier-Foster, Julie M., primary, Loh, Mignon L., primary, Rabadan, Raul, primary, Van Vlierberghe, Pieter, primary, and Ferrando, Adolfo A., primary
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Methods Tables from Phf6 Loss Enhances HSC Self-Renewal Driving Tumor Initiation and Leukemia Stem Cell Activity in T-ALL
- Author
-
Wendorff, Agnieszka A., primary, Quinn, S. Aidan, primary, Rashkovan, Marissa, primary, Madubata, Chioma J., primary, Ambesi-Impiombato, Alberto, primary, Litzow, Mark R., primary, Tallman, Martin S., primary, Paietta, Elisabeth, primary, Paganin, Maddalena, primary, Basso, Giuseppe, primary, Gastier-Foster, Julie M., primary, Loh, Mignon L., primary, Rabadan, Raul, primary, Van Vlierberghe, Pieter, primary, and Ferrando, Adolfo A., primary
- Published
- 2023
- Full Text
- View/download PDF
17. Data from Phf6 Loss Enhances HSC Self-Renewal Driving Tumor Initiation and Leukemia Stem Cell Activity in T-ALL
- Author
-
Wendorff, Agnieszka A., primary, Quinn, S. Aidan, primary, Rashkovan, Marissa, primary, Madubata, Chioma J., primary, Ambesi-Impiombato, Alberto, primary, Litzow, Mark R., primary, Tallman, Martin S., primary, Paietta, Elisabeth, primary, Paganin, Maddalena, primary, Basso, Giuseppe, primary, Gastier-Foster, Julie M., primary, Loh, Mignon L., primary, Rabadan, Raul, primary, Van Vlierberghe, Pieter, primary, and Ferrando, Adolfo A., primary
- Published
- 2023
- Full Text
- View/download PDF
18. Mutational landscape, clonal evolution patterns, and role of RAS mutations in relapsed acute lymphoblastic leukemia
- Author
-
Oshima, Koichi, Khiabanian, Hossein, da Silva-Almeida, Ana C., Tzoneva, Gannie, Abate, Francesco, Ambesi-Impiombato, Alberto, Sanchez-Martin, Marta, Carpenter, Zachary, Penson, Alex, Perez-Garcia, Arianne, Eckert, Cornelia, Nicolas, Concepción, Balbin, Milagros, Sulis, Maria Luisa, Kato, Motohiro, Koh, Katsuyoshi, Paganin, Maddalena, Basso, Giuseppe, Gastier-Foster, Julie M., Devidas, Meenakshi, Loh, Mignon L., Kirschner-Schwabe, Renate, Palomero, Teresa, Rabadan, Raul, and Ferrando, Adolfo A.
- Published
- 2016
19. Supplementary Data from Phf6 Loss Enhances HSC Self-Renewal Driving Tumor Initiation and Leukemia Stem Cell Activity in T-ALL
- Author
-
Adolfo A. Ferrando, Pieter Van Vlierberghe, Raul Rabadan, Mignon L. Loh, Julie M. Gastier-Foster, Giuseppe Basso, Maddalena Paganin, Elisabeth Paietta, Martin S. Tallman, Mark R. Litzow, Alberto Ambesi-Impiombato, Chioma J. Madubata, Marissa Rashkovan, S. Aidan Quinn, and Agnieszka A. Wendorff
- Abstract
Supplementary Figures 1-8
- Published
- 2023
- Full Text
- View/download PDF
20. Data from Phf6 Loss Enhances HSC Self-Renewal Driving Tumor Initiation and Leukemia Stem Cell Activity in T-ALL
- Author
-
Adolfo A. Ferrando, Pieter Van Vlierberghe, Raul Rabadan, Mignon L. Loh, Julie M. Gastier-Foster, Giuseppe Basso, Maddalena Paganin, Elisabeth Paietta, Martin S. Tallman, Mark R. Litzow, Alberto Ambesi-Impiombato, Chioma J. Madubata, Marissa Rashkovan, S. Aidan Quinn, and Agnieszka A. Wendorff
- Abstract
The plant homeodomain 6 gene (PHF6) is frequently mutated in human T-cell acute lymphoblastic leukemia (T-ALL); however, its specific functional role in leukemia development remains to be established. Here, we show that loss of PHF6 is an early mutational event in leukemia transformation. Mechanistically, genetic inactivation of Phf6 in the hematopoietic system enhances hematopoietic stem cell (HSC) long-term self-renewal and hematopoietic recovery after chemotherapy by rendering Phf6 knockout HSCs more quiescent and less prone to stress-induced activation. Consistent with a leukemia-initiating tumor suppressor role, inactivation of Phf6 in hematopoietic progenitors lowers the threshold for the development of NOTCH1-induced T-ALL. Moreover, loss of Phf6 in leukemia lymphoblasts activates a leukemia stem cell transcriptional program and drives enhanced T-ALL leukemia-initiating cell activity. These results implicate Phf6 in the control of HSC homeostasis and long-term self-renewal and support a role for PHF6 loss as a driver of leukemia-initiating cell activity in T-ALL.Significance:Phf6 controls HSC homeostasis, leukemia initiation, and T-ALL leukemia-initiating cell self-renewal. These results substantiate a role for PHF6 mutations as early events and drivers of leukemia stem cell activity in the pathogenesis of T-ALL.This article is highlighted in the In This Issue feature, p. 305
- Published
- 2023
- Full Text
- View/download PDF
21. Therapeutic targeting of HES1 transcriptional programs in T-ALL
- Author
-
Schnell, Stephanie A., Ambesi-Impiombato, Alberto, Sanchez-Martin, Marta, Belver, Laura, Xu, Luyao, Qin, Yue, Kageyama, Ryoichiro, and Ferrando, Adolfo A.
- Published
- 2015
- Full Text
- View/download PDF
22. WT1 loss attenuates the TP53-induced DNA damage response in T-cell acute lymphoblastic leukemia
- Author
-
Fulvio Bordin, Erich Piovan, Elena Masiero, Alberto Ambesi-Impiombato, Sonia Minuzzo, Roberta Bertorelle, Valeria Sacchetto, Giorgia Pilotto, Giuseppe Basso, Paola Zanovello, Alberto Amadori, and Valeria Tosello
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Loss-of-function mutations and deletions in Wilms tumor 1 (WT1) gene are present in approximately 10% of T-cell acute lymphoblastic leukemia. Clinically, WT1 mutations are enriched in relapsed series and are associated to inferior relapse-free survival in thymic T-cell acute lymphoblastic leukemia cases. Here we demonstrate that WT1 plays a critical role in the response to DNA damage in T-cell leukemia. WT1 loss conferred resistance to DNA damaging agents and attenuated the transcriptional activation of important apoptotic regulators downstream of TP53 in TP53-competent MOLT4 T-leukemia cells but not in TP53-mutant T-cell acute lymphoblastic leukemia cell lines. Notably, WT1 loss positively affected the expression of the X-linked inhibitor of apoptosis protein, XIAP, and genetic or chemical inhibition with embelin (a XIAP inhibitor) significantly restored sensitivity to γ-radiation in both T-cell acute lymphoblastic leukemia cell lines and patient-derived xenografts. These results reveal an important role for the WT1 tumor suppressor gene in the response to DNA damage, and support the view that anti-XIAP targeted therapies could have a role in the treatment of WT1-mutant T-cell leukemia.
- Published
- 2018
- Full Text
- View/download PDF
23. Benefits of global mutant huntingtin lowering diminish over time in a Huntington’s disease mouse model
- Author
-
Marchionini, Deanna M., primary, Liu, Jeh-Ping, additional, Ambesi-Impiombato, Alberto, additional, Kerker, Kimberly, additional, Cirillo, Kim, additional, Bansal, Mukesh, additional, Mushlin, Rich, additional, Brunner, Daniela, additional, Ramboz, Sylvie, additional, Kwan, Mei, additional, Kuhlbrodt, Kirsten, additional, Tillack, Karsten, additional, Peters, Finn, additional, Rauhala, Leena, additional, Obenauer, John, additional, Greene, Jonathan R., additional, Hartl, Christopher, additional, Khetarpal, Vinod, additional, Lager, Brenda, additional, Rosinski, Jim, additional, Aaronson, Jeff, additional, Alam, Morshed, additional, Signer, Ethan, additional, Muñoz-Sanjuán, Ignacio, additional, Howland, David, additional, and Zeitlin, Scott O., additional
- Published
- 2022
- Full Text
- View/download PDF
24. Enrichment analysis of phenotypic data for drug repurposing in rare diseases.
- Author
-
Ambesi-Impiombato, Alberto, Cox, Kimberly, Ramboz, Sylvie, Brunner, Daniela, Bansal, Mukesh, and Leahy, Emer
- Subjects
DRUG repositioning ,DRUG discovery ,HUNTINGTON disease ,RARE diseases ,BEHAVIORAL assessment ,MACHINE learning ,ANTIDEPRESSANTS - Abstract
Drug-induced Behavioral Signature Analysis (DBSA), is a machine learning (ML) method for in silico screening of compounds, inspired by analytical methods quantifying gene enrichment in genomic analyses. When applied to behavioral data it can identify drugs that can potentially reverse in vivo behavioral symptoms in animal models of human disease and suggest new hypotheses for drug discovery and repurposing. We present a proof-of-concept study aiming to assess Drug-induced Behavioral Signature Analysis (DBSA) as a systematic approach for drug discovery for rare disorders. We applied Drug-induced Behavioral Signature Analysis to high-content behavioral data obtained with SmartCube®, an automated in vivo phenotyping platform. The therapeutic potential of several dozen approved drugs was assessed for phenotypic reversal of the behavioral profile of a Huntington's Disease (HD) murine model, the Q175 heterozygous knock-in mice. The in silico Drug-induced Behavioral Signature Analysis predictions were enriched for drugs known to be effective in the symptomatic treatment of Huntington's Disease, including bupropion, modafinil, methylphenidate, and several SSRIs, as well as the atypical antidepressant tianeptine. To validate the method, we tested acute and chronic effects of tianeptine (20 mg/kg, i. p.) in vivo, using Q175 mice and wild type controls. In both experiments, tianeptine significantly rescued the behavioral phenotype assessed with the SmartCube® platform. Our target-agnostic method thus showed promise for identification of symptomatic relief treatments for rare disorders, providing an alternative method for hypothesis generation and drug discovery for disorders with huge disease burden and unmet medical needs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
25. Genetic loss of SH2B3 in acute lymphoblastic leukemia
- Author
-
Perez-Garcia, Arianne, Ambesi-Impiombato, Alberto, Hadler, Michael, Rigo, Isaura, LeDuc, Charles A., Kelly, Kara, Jalas, Chaim, Paietta, Elisabeth, Racevskis, Janis, Rowe, Jacob M., Tallman, Martin S., Paganin, Maddalena, Basso, Giuseppe, Tong, Wei, Chung, Wendy K., and Ferrando, Adolfo A.
- Published
- 2013
- Full Text
- View/download PDF
26. Prognostic relevance of integrated genetic profiling in adult T-cell acute lymphoblastic leukemia
- Author
-
Van Vlierberghe, Pieter, Ambesi-Impiombato, Alberto, De Keersmaecker, Kim, Hadler, Michael, Paietta, Elisabeth, Tallman, Martin S., Rowe, Jacob M., Forne, Carles, Rue, Montserrat, and Ferrando, Adolfo A.
- Published
- 2013
- Full Text
- View/download PDF
27. Metabolic reprogramming induces resistance to anti-NOTCH1 therapies in T cell acute lymphoblastic leukemia
- Author
-
Herranz, Daniel, Ambesi-Impiombato, Alberto, Sudderth, Jessica, Sanchez-Martin, Marta, Belver, Laura, Tosello, Valeria, Xu, Luyao, Wendorff, Agnieszka A., Castillo, Mireia, Haydu, J. Erika, Marquez, Javier, Mates, Jose M., Kung, Andrew L., Rayport, Stephen, Cordon-Cardo, Carlos, DeBerardinis, Ralph J., and Ferrando, Adolfo A.
- Subjects
T cells ,Acute lymphocytic leukemia ,Biological sciences ,Health - Abstract
Activating mutations in NOTCH1 are common in T cell acute lymphoblastic leukemia (T-ALL). Here we identify glutaminolysis as a critical pathway for leukemia cell growth downstream of NOTCH1 and a key determinant of the response to anti-NOTCH1 therapies in vivo. Mechanistically, inhibition of NOTCH1 signaling in T-ALL induces a metabolic shutdown, with prominent inhibition of glutaminolysis and triggers autophagy as a salvage pathway supporting leukemia cell metabolism. Consequently, inhibition of glutaminolysis and inhibition of autophagy strongly and synergistically enhance the antileukemic effects of anti-NOTCH1 therapy in mice harboring T-ALL. Moreover, we demonstrate that Pten loss upregulates glycolysis and consequently rescues leukemic cell metabolism, thereby abrogating the antileukemic effects of NOTCH1 inhibition. Overall, these results identify glutaminolysis as a major node in cancer metabolism controlled by NOTCH1 and as therapeutic target for the treatment of T-ALL., NOTCH1 signaling is a conserved signal transduction pathway with a prominent role in cell differentiation and tissue patterning during development (1). In the hematopoietic system, NOTCH1 has been implicated in [...]
- Published
- 2015
- Full Text
- View/download PDF
28. Benefits of global mutant huntingtin lowering diminish over time in a Huntington's disease mouse model
- Author
-
Marchionini, Deanna M, primary, Liu, Jeh-Ping, additional, Ambesi-Impiombato, Alberto, additional, Cox, Kimberly, additional, Cirillo, Kim, additional, Bansal, Mukesh, additional, Mushlin, Rich, additional, Brunner, Daniela, additional, Ramboz, Sylvie, additional, Kwan, Mei, additional, Kuhlbrodt, Kirsten, additional, Tillack, Karsten, additional, Peters, Finn, additional, Rauhala, Leena, additional, Obenauer, John, additional, Greene, Jonathan R, additional, Hartle, Christopher, additional, Khetarpal, Vinod, additional, Lager, Brenda, additional, Rosinski, Jim, additional, Aaronson, Jeff, additional, Alam, Morshed, additional, Signer, Ethan, additional, Muñoz-Sanjuán, Ignacio, additional, Howland, David, additional, and Zeitlin, Scott O, additional
- Published
- 2022
- Full Text
- View/download PDF
29. Hypovitaminosis D3, Leukopenia, and Human Serotonin Transporter Polymorphism in Anorexia Nervosa and Bulimia Nervosa
- Author
-
Anna Tasegian, Francesco Curcio, Laura Dalla Ragione, Francesca Rossetti, Samuela Cataldi, Michela Codini, Francesco Saverio Ambesi-Impiombato, Tommaso Beccari, and Elisabetta Albi
- Subjects
Pathology ,RB1-214 - Abstract
Vitamin D3 has been described to have different extraskeletal roles by acting as parahormone in obesity, diabetes, cancer, cognitive impairment, and dementia and to have important regulatory functions in innate immunity. There are no studies showing extraskeletal changes associated with hypovitaminosis D3 in eating disorders. Methods. We have analyzed the blood of 18 patients affected by anorexia nervosa and bulimia nervosa collected over a 15-month period. We performed a panel of chemical and clinical analyses: the assay of vitamin D3, the immunoblotting of vitamin D receptor and peroxisome proliferator-activated receptor gamma, and the genotyping of 5-hydroxytryptamine transporter linked polymorphic region. Results. We choose 18 patients with a normal blood test profile such as thyroid hormones, hepatic and renal parameters, triglycerides, proteins, vitamin B12, and folic acid. Among these emerged the case of a woman with long-term anorexia nervosa and the case of a woman with long-term bulimia nervosa both complicated by anxiety and depression, severe hypovitaminosis D3, decrease of vitamin D receptor, leukopenia, and 5-hydroxytryptamine transporter linked polymorphic region short allele. Conclusion. The results induce hypothesising that the severe hypovitaminosis D3 might be responsible for the lack of the inflammatory response and the depressive symptoms in patients with long-term eating disorders.
- Published
- 2016
- Full Text
- View/download PDF
30. e-Cadherin in 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-Induced Parkinson Disease
- Author
-
Samuela Cataldi, Michela Codini, Stéphane Hunot, François-Pierre Légeron, Ivana Ferri, Paola Siccu, Angelo Sidoni, Francesco Saverio Ambesi-Impiombato, Tommaso Beccari, Francesco Curcio, and Elisabetta Albi
- Subjects
Pathology ,RB1-214 - Abstract
Today a large number of studies are focused on clarifying the complexity and diversity of the pathogenetic mechanisms inducing Parkinson disease. We used 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a neurotoxin that induces Parkinson disease, to evaluate the change of midbrain structure and the behavior of the anti-inflammatory factor e-cadherin, interleukin-6, tyrosine hydroxylase, phosphatase and tensin homolog, and caveolin-1. The results showed a strong expression of e-cadherin, variation of length and thickness of the heavy neurofilaments, increase of interleukin-6, and reduction of tyrosine hydroxylase known to be expression of dopamine cell loss, reduction of phosphatase and tensin homolog described to impair responses to dopamine, and reduction of caveolin-1 known to be expression of epithelial-mesenchymal transition and fibrosis. The possibility that the overexpression of the e-cadherin might be implicated in the anti-inflammatory reaction to MPTP treatment by influencing the behavior of the other analyzed molecules is discussed.
- Published
- 2016
- Full Text
- View/download PDF
31. Combinatorial ETS1-Dependent Control of Oncogenic NOTCH1 Enhancers in T-cell Leukemia
- Author
-
Yiran Liu, Cher Sha, Alberto Ambesi-Impiombato, Michael C. Ostrowski, Erin Kim, Theresa M. Keeley, Jahnavi K. Nalamolu, Qing Wang, Mark Y. Chiang, Adolfo A. Ferrando, Ran Yan, Rohan Kodgule, Russell J.H. Ryan, Arvind Rao, Nicholas Kunnath, Linda C. Samuelson, Barbara L. Kee, Giusy Della Gatta, Anna C. McCarter, Rork Kuick, Ashley Melnick, and Mengxi Sun
- Subjects
Leukemia, T-Cell ,Carcinogenesis ,Effector ,Lymphoblastic Leukemia ,T-cell leukemia ,General Medicine ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,Chromatin ,Proto-Oncogene Protein c-ets-1 ,Mice ,ETS1 ,Antineoplastic Combined Chemotherapy Protocols ,Cancer research ,Animals ,Receptor, Notch1 ,Enhancer ,Gene ,Transcription factor ,Signal Transduction - Abstract
Notch activation is highly prevalent among cancers, in particular T-cell acute lymphoblastic leukemia (T-ALL). However, the use of pan-Notch inhibitors to treat cancers has been hampered by adverse effects, particularly intestinal toxicities. To circumvent this barrier in T-ALL, we aimed to inhibit ETS1, a developmentally important T-cell transcription factor previously shown to cobind Notch response elements. Using complementary genetic approaches in mouse models, we show that ablation of Ets1 leads to strong Notch-mediated suppressive effects on T-cell development and leukemogenesis but milder intestinal effects than pan-Notch inhibitors. Mechanistically, genome-wide chromatin profiling studies demonstrate that Ets1 inactivation impairs recruitment of multiple Notch-associated factors and Notch-dependent activation of transcriptional elements controlling major Notch-driven oncogenic effector pathways. These results uncover previously unrecognized hierarchical heterogeneity of Notch-controlled genes and point to Ets1-mediated enucleation of Notch–Rbpj transcriptional complexes as a target for developing specific anti-Notch therapies in T-ALL that circumvent the barriers of pan-Notch inhibition. Significance: Notch signaling controls developmentally important and tissue-specific activities, raising barriers for developing anti-Notch therapies. Pivoting away from pan-Notch inhibitors, we show antileukemic but less toxic effects of targeting ETS1, a T-cell NOTCH1 cofactor. These results demonstrate the feasibility of context-dependent suppression of NOTCH1 programs for the treatment of T-ALL. This article is highlighted in the In This Issue feature, p. 127
- Published
- 2020
- Full Text
- View/download PDF
32. Adipose Tissue-Derived Stem Cell in Vitro Differentiation in a Three-Dimensional Dental Bud Structure
- Author
-
Ferro, Federico, Spelat, Renza, Falini, Giuseppe, Gallelli, Annarita, D'Aurizio, Federica, Puppato, Elisa, Pandolfi, Maura, Beltrami, Antonio Paolo, Cesselli, Daniela, Beltrami, Carlo Alberto, Ambesi-Impiombato, Francesco Saverio, and Curcio, Francesco
- Published
- 2011
- Full Text
- View/download PDF
33. A NOTCH1-driven MYC enhancer promotes T cell development, transformation and acute lymphoblastic leukemia
- Author
-
Herranz, Daniel, Ambesi-Impiombato, Alberto, Palomero, Teresa, Schnell, Stephanie A., Belver, Laura, Wendorff, Agnieszka A., Xu, Luyao, Castillo-Martin, Mireia, Llobet-Navas, David, Cordon-Cardo, Carlos, Clappier, Emmanuelle, Soulier, Jean, and Ferrando, Adolfo A.
- Subjects
Gene mutations -- Research -- Analysis ,T cells -- Analysis -- Research -- Physiological aspects ,Acute lymphocytic leukemia -- Analysis -- Research ,Biological sciences ,Health - Abstract
Efforts to identify and annotate cancer driver genetic lesions have been focused primarily on the analysis of protein- coding genes; however, most genetic abnormalities found in human cancer are located in intergenic regions. Here we identify a new long range-acting MYC enhancer controlled by NOTCH 1 that is targeted by recurrent chromosomal duplications in human T cell acute lymphoblastic leukemia (T-ALL). This highly conserved regulatory element, hereby named N-Me for NOTCH MYC enhancer, is located within a broad super-enhancer region +1.47 Mb from the MYC transcription initiating site, interacts with the MYC proximal promoter and induces orientation-independent MYC expression in reporter assays. Moreover, analysis of N-Me knockout mice demonstrates a selective and essential role of this regulatoiy element during thymocyte development and in NOTCH 1-induced T-ALL. Together these results identify N-Me as a long-range oncogenic enhancer implicated directly in the pathogenesis of human leukemia and highlight the importance of the NOTCH 1-MYC regulatoiy axis in T cell transformation and as a therapeutic target in T-ALL., T-ALL is an aggressive hematologic malignancy resulting from the transformation of immature T cell progenitor cells (1) in which activation of transcription factor oncogenes and deregulation of transcriptional regulatory networks [...]
- Published
- 2014
- Full Text
- View/download PDF
34. The DLEU2/ miR-15a/ 16-1 Cluster Controls B Cell Proliferation and Its Deletion Leads to Chronic Lymphocytic Leukemia
- Author
-
Klein, Ulf, Lia, Marie, Crespo, Marta, Siegel, Rachael, Shen, Qiong, Mo, Tongwei, Ambesi-Impiombato, Alberto, Califano, Andrea, Migliazza, Anna, Bhagat, Govind, and Dalla-Favera, Riccardo
- Published
- 2010
- Full Text
- View/download PDF
35. Spaceflight Induced Disorders: Potential Nutritional Countermeasures
- Author
-
Samuela Cataldi, Elisabetta Albi, Francesco Curcio, Carmela Conte, Tommaso Beccari, Fabio Costa, and Francesco Saverio Ambesi-Impiombato
- Subjects
0301 basic medicine ,life-style ,Histology ,Biomedical Engineering ,Context (language use) ,Bioengineering ,Review ,Spaceflight ,medicine.disease_cause ,law.invention ,spaceflight ,03 medical and health sciences ,0302 clinical medicine ,law ,medicine ,Psychological stress ,Cognitive impairment ,Life style ,Bioengineering and Biotechnology ,microgravity ,030104 developmental biology ,nutrition ,energy intake ,Molecular mechanism ,Darwinian selection ,TP248.13-248.65 ,030217 neurology & neurosurgery ,Cognitive psychology ,Biotechnology - Abstract
Space travel is an extreme experience even for the astronaut who has received extensive basic training in various fields, from aeronautics to engineering, from medicine to physics and biology. Microgravity puts a strain on members of space crews, both physically and mentally: short-term or long-term travel in orbit the International Space Station may have serious repercussions on the human body, which may undergo physiological changes affecting almost all organs and systems, particularly at the muscular, cardiovascular and bone compartments. This review aims to highlight recent studies describing damages of human body induced by the space environment for microgravity, and radiation. All novel conditions, to ally unknown to the Darwinian selection strategies on Earth, to which we should add the psychological stress that astronauts suffer due to the inevitable forced cohabitation in claustrophobic environments, the deprivation from their affections and the need to adapt to a new lifestyle with molecular changes due to the confinement. In this context, significant nutritional deficiencies with consequent molecular mechanism changes in the cells that induce to the onset of physiological and cognitive impairment have been considered.
- Published
- 2021
- Full Text
- View/download PDF
36. Mutational and functional genetics mapping of chemotherapy resistance mechanisms in relapsed acute lymphoblastic leukemia
- Author
-
Martin S. Tallman, Junfei Zhao, Mignon L. Loh, Giuseppe Basso, Motohiro Kato, Meenakshi Devidas, Pablo Pérez-Durán, Timothy Chu, Julie M. Gastier-Foster, Maddalena Paganin, Alberto Ambesi-Impiombato, Katsuyoshi Koh, Zhengqiang Wang, Adolfo A. Ferrando, Laura Belver, Mark R. Litzow, Jessie A. Brown, Raul Rabadan, Concepcion Nicolas, Jules P.P. Meijerink, Elisabeth Paietta, Thomas Gunning, Aidan Quinn, Maria Luisa Sulis, Valeria Tosello, Juan Ángel Patiño-Galindo, Mark D. Minden, Jacob M. Rowe, Koichi Oshima, and Milagros Balbín
- Subjects
Drug ,Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,Lymphoblastic Leukemia ,media_common.quotation_subject ,Article ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,Internal medicine ,Acute lymphocytic leukemia ,Medicine ,CRISPR ,Humans ,Child ,media_common ,business.industry ,Cancer ,Combination chemotherapy ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Prognosis ,Leukemia ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,business ,Chemotherapy resistance - Abstract
Multiagent combination chemotherapy can be curative in acute lymphoblastic leukemia (ALL). Still, patients with primary refractory disease or with relapsed leukemia have a very poor prognosis. Here we integrate an in-depth dissection of the mutational landscape across diagnostic and relapsed pediatric and adult ALL samples with genome-wide CRISPR screen analysis of gene–drug interactions across seven ALL chemotherapy drugs. By combining these analyses, we uncover diagnostic and relapse-specific mutational mechanisms as well as genetic drivers of chemoresistance. Functionally, our data identify common and drug-specific pathways modulating chemotherapy response and underscore the effect of drug combinations in restricting the selection of resistance-driving genetic lesions. In addition, by identifying actionable targets for the reversal of chemotherapy resistance, these analyses open therapeutic opportunities for the treatment of relapse and refractory disease. Ferrando and colleagues analyze matched diagnostic and relapsed acute lymphocytic leukemia by whole-genome sequencing, and perform in vitro genome-wide CRISPR screens, to examine alterations associated with chemotherapy resistance.
- Published
- 2021
37. Effects of Thyrotropin on the Thyroid Cell Membrane: Hyperpolarization Induced by Hormone-Receptor Interaction
- Author
-
Grollman, Evelyn F., Lee, George, Ambesi-Impiombato, F. S., Meldolesi, Maria F., Aloj, Salvatore M., Coon, Hayden G., Kaback, H. Ronald, and Kohn, Leonard D.
- Published
- 1977
38. Long-Term Culture and Functional Characterization of Follicular Cells from Adult Normal Human Thyroids
- Author
-
Curcio, Francesco, Ambesi-Impiombato, Francesco Saverio, Perrella, Giuseppina, and Coon, Hayden G.
- Published
- 1994
39. Culture of Hormone-Dependent Functional Epithelial Cells from Rat Thyroids
- Author
-
Ambesi-Impiombato, F. S., Parks, L. A. M., and Coon, H. G.
- Published
- 1980
40. Reverse engineering of TLX oncogenic transcriptional networks identifies RUNX1 as tumor suppressor in T-ALL
- Author
-
Gatta, Giusy Della, Palomero, Teresa, Perez-Garcia, Arianne, Ambesi-Impiombato, Alberto, Bansal, Mukesh, Carpenter, Zachary W., Keersmaecker, Kim De, Sole, Xavier, Xu, Luyao, Paietta, Elisabeth, Racevskis, Janis, Wiernik, Peter H., Rowe, Jacob M., Meijerink, Jules P., Califano, Andrea, and Ferrando, Adolfo A.
- Subjects
Tumor suppressor genes -- Physiological aspects -- Research ,Acute lymphocytic leukemia -- Risk factors -- Genetic aspects -- Research ,Genetic transcription -- Research ,Biological sciences ,Health - Abstract
The TLX1 and TLX3 transcription factor oncogenes have a key role in the pathogenesis of T cell acute lymphoblastic leukemia (T-ALL) (1.2). Here we used reverse engineering of global transcriptional networks to decipher the oncogenic regulatory circuit controlled by TLX1 and TLX3. This systems biology analysis defined T cell leukemia homeobox 1 (TLX1) and TLX3 as master regulators of an oncogenic transcriptional circuit governing T-ALL. Notably, a network structure analysis of this hierarchical network identified RUNX1 as a key mediator of the T-ALL induced by TLX1 and TLX3 and predicted a tumor-suppressor role for RUNX1 in T cell transformation. Consistent with these results, we identified recurrent somatic loss-of-function mutations in RUNX1 in human T-ALL. Overall, these results place TLX1 and TLX3 at the top of an oncogenic transcriptional network controlling leukemia development, show the power of network analyses to identify key elements in the regulatory circuits governing human cancer and identify RUNX1 as a tumor-suppressor gene in T-ALL., TLX1 and TLX3 encode highly related homeobox transcription factor oncogenes frequently activated by chromosomal translocations in T-ALL(3-5). To interrogate the transcriptional programs associated with aberrant expression of TLX1 and TLX3, [...]
- Published
- 2012
- Full Text
- View/download PDF
41. Computational Analysis of Multidimensional Behavioral Alterations After Chronic Social Defeat Stress
- Author
-
Lorsch, Zachary S., primary, Ambesi-Impiombato, Alberto, additional, Zenowich, Rebecca, additional, Morganstern, Irene, additional, Leahy, Emer, additional, Bansal, Mukesh, additional, Nestler, Eric J., additional, and Hanania, Taleen, additional
- Published
- 2021
- Full Text
- View/download PDF
42. Spaceflight Induced Disorders: Potential Nutritional Countermeasures
- Author
-
Costa, Fabio, primary, Ambesi-Impiombato, Francesco Saverio, additional, Beccari, Tommaso, additional, Conte, Carmela, additional, Cataldi, Samuela, additional, Curcio, Francesco, additional, and Albi, Elisabetta, additional
- Published
- 2021
- Full Text
- View/download PDF
43. FRTL-5 experiment during ENEIDE mission
- Author
-
Meli A., Perrella G., Toller M., Zambito A. M., Spelat R., Moretti M., Ferro F., Curcio F., and Ambesi-Impiombato F. S.
- Published
- 2007
- Full Text
- View/download PDF
44. Computational Analysis of Multidimensional Behavioral Alterations After Chronic Social Defeat Stress
- Author
-
Irene Morganstern, Zachary S. Lorsch, Alberto Ambesi-Impiombato, Emer Leahy, Taleen Hanania, Rebecca Zenowich, Mukesh Bansal, and Eric J. Nestler
- Subjects
0301 basic medicine ,Imipramine ,Social defeat ,Social Defeat ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Animal model ,Group differences ,Medicine ,Animals ,Computational analysis ,Social Behavior ,Biological Psychiatry ,Depression (differential diagnoses) ,Psychiatric Disease ,Behavior, Animal ,business.industry ,Mice, Inbred C57BL ,Antidepressant medication ,Disease Models, Animal ,030104 developmental biology ,business ,Neuroscience ,030217 neurology & neurosurgery ,Stress, Psychological ,medicine.drug - Abstract
Background The study of depression in humans depends on animal models that attempt to mimic specific features of the human syndrome. Most studies focus on one or a few behavioral domains, with time and practical considerations prohibiting a comprehensive evaluation. Although machine learning has enabled unbiased analysis of behavior in animals, this has not yet been applied to animal models of psychiatric disease. Methods We performed chronic social defeat stress (CSDS) in mice and evaluated behavior with PsychoGenics’ SmartCube, a high-throughput unbiased automated phenotyping platform that collects >2000 behavioral features based on machine learning. We evaluated group differences at several times post-CSDS and after administration of the antidepressant medication imipramine. Results SmartCube analysis after CSDS successfully separated control and defeated-susceptible mice, and defeated-resilient mice more resembled control mice. We observed a potentiation of CSDS effects over time. Treatment of susceptible mice with imipramine induced a 40.2% recovery of the defeated-susceptible phenotype as assessed by SmartCube. Conclusions High-throughput analysis can simultaneously evaluate multiple behavioral alterations in an animal model for the study of depression, which provides a more unbiased and holistic approach to evaluating group differences after CSDS and perhaps can be applied to other mouse models of psychiatric disease.
- Published
- 2020
45. Acid and Neutral Sphingomyelinase Behavior in Radiation-Induced Liver Pyroptosis and in the Protective/Preventive Role of rMnSOD
- Author
-
Eugene Krasavin, Elisabetta Albi, Francesco Curcio, Federica Filomena Patria, Carmela Conte, Aldo Mancini, Michela Codini, Irina Nakashidze, Francesco Saverio Ambesi-Impiombato, Tommaso Beccari, Ivana Ferri, Samuela Cataldi, Maria Rachele Ceccarini, Antonella Borrelli, Alexander Ivanov, and Oleg Belov
- Subjects
Programmed cell death ,Radiation-Protective Agents ,liver ,Exosome ,Article ,Catalysis ,lcsh:Chemistry ,Inorganic Chemistry ,Mice ,neutral sphingomyelinase ,Gene expression ,Pyroptosis ,medicine ,Animals ,Physical and Theoretical Chemistry ,acid sphingomyelinase ,SOD ,Radiation Injuries ,lcsh:QH301-705.5 ,Molecular Biology ,Spectroscopy ,Chemistry ,Endoplasmic reticulum ,Caspase 1 ,Organic Chemistry ,General Medicine ,Acid sphingomyelinase ,Liver ,Neutral sphingomyelinase ,Radiation ,Sphingomyelins ,Computer Science Applications ,Cell biology ,radiation ,Sphingomyelin Phosphodiesterase ,lcsh:Biology (General) ,lcsh:QD1-999 ,Female ,Sphingomyelin ,Immunostaining ,medicine.drug - Abstract
Sphingomyelins (SMs) are a class of relevant bioactive molecules that act as key modulators of different cellular processes, such as growth arrest, exosome formation, and the inflammatory response influenced by many environmental conditions, leading to pyroptosis, a form of programmed cell death due to Caspase-1 involvement. To study liver pyroptosis and hepatic SM metabolism via both lysosomal acid SMase (aSMase) and endoplasmic reticulum/nucleus neutral SMase (nSMase) during the exposure of mice to radiation and to ascertain if this process can be modulated by protective molecules, we used an experimental design (previously used by us) to evaluate the effects of both ionizing radiation and a specific protective molecule (rMnSOD) in the brain in collaboration with the Joint Institute for Nuclear Research, Dubna (Russia). As shown by the Caspase-1 immunostaining of the liver sections, the radiation resulted in the loss of the normal cell structure alongside a progressive and dose-dependent increase of the labelling, treatment, and pretreatment with rMnSOD, which had a significant protective effect on the livers. SM metabolic analyses, performed on aSMase and nSMase gene expression, as well as protein content and activity, proved that rMnSOD was able to significantly reduce radiation-induced damage by playing both a protective role via aSMase and a preventive role via nSMase.
- Published
- 2020
- Full Text
- View/download PDF
46. The impact of long-term exposure to space environment on adult mammalian organisms: a study on mouse thyroid and testis.
- Author
-
Maria Angela Masini, Elisabetta Albi, Cristina Barmo, Tommaso Bonfiglio, Lara Bruni, Laura Canesi, Samuela Cataldi, Francesco Curcio, Marta D'Amora, Ivana Ferri, Katsumasa Goto, Fuminori Kawano, Remo Lazzarini, Elisabetta Loreti, Naoya Nakai, Takashi Ohira, Yoshinobu Ohira, Silvio Palmero, Paola Prato, Franco Ricci, Linda Scarabelli, Tsubasa Shibaguchi, Renza Spelat, Felice Strollo, and Francesco Saverio Ambesi-Impiombato
- Subjects
Medicine ,Science - Abstract
Hormonal changes in humans during spaceflight have been demonstrated but the underlying mechanisms are still unknown. To clarify this point thyroid and testis/epididymis, both regulated by anterior pituitary gland, have been analyzed on long-term space-exposed male C57BL/10 mice, either wild type or pleiotrophin transgenic, overexpressing osteoblast stimulating factor-1. Glands were submitted to morphological and functional analysis.In thyroids, volumetric ratios between thyrocytes and colloid were measured. cAMP production in 10(-7)M and 10(-8)M thyrotropin-treated samples was studied. Thyrotropin receptor and caveolin-1 were quantitized by immunoblotting and localized by immunofluorescence. In space-exposed animals, both basal and thyrotropin-stimulated cAMP production were always higher. Also, the structure of thyroid follicles appeared more organized, while thyrotropin receptor and caveolin-1 were overexpressed. Unlike the control samples, in the space samples thyrotropin receptor and caveolin-1 were both observed at the intracellular junctions, suggesting their interaction in specific cell membrane microdomains.In testes, immunofluorescent reaction for 3β- steroid dehydrogenase was performed and the relative expressions of hormone receptors and interleukin-1β were quantified by RT-PCR. Epididymal sperm number was counted. In space-exposed animals, the presence of 3β and 17β steroid dehydrogenase was reduced. Also, the expression of androgen and follicle stimulating hormone receptors increased while lutenizing hormone receptor levels were not affected. The interleukin 1 β expression was upregulated. The tubular architecture was altered and the sperm cell number was significantly reduced in spaceflight mouse epididymis (approx. -90% vs. laboratory and ground controls), indicating that the space environment may lead to degenerative changes in seminiferous tubules.Space-induced changes of structure and function of thyroid and testis/epididymis could be responsible for variations of hormone levels in human during space missions. More research, hopefully a reflight of MDS, would be needed to establish whether the space environment acts directly on the peripheral glands or induces changes in the hypotalamus-pituitary-glandular axis.
- Published
- 2012
- Full Text
- View/download PDF
47. Fresh New Air in Space? ‘MoMa’ on the A.S.I. Launch Pad
- Author
-
Ambesi Impiombato, Francesco Saverio, Curcio, Francesco, Meli, Antonella, and Zambito, Anna Maria
- Published
- 2006
- Full Text
- View/download PDF
48. Oncogenic hijacking of the stress response machinery in T cell acute lymphoblastic leukemia
- Author
-
Iannis Aifantis, Adolfo A. Ferrando, Hai Hu, Thomas Trimarchi, Alberto Ambesi-Impiombato, Kathryn Hockemeyer, Juan Carlos Balandrán, Alejandra R. Jimenez, Monica L. Guzman, Liza Shrestha, Gabriela Chiosis, Yixiao Gong, Elisabeth Paietta, Michelle A. Kelliher, Nikos Kourtis, Jasper Mullenders, Kamala Bhatt, Charalampos Lazaris, and Aristotelis Tsirigos
- Subjects
0301 basic medicine ,T cell ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Biochemistry ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Heat Shock Transcription Factors ,Stress, Physiological ,Cellular stress response ,Cell Line, Tumor ,medicine ,Animals ,Humans ,HSP90 Heat-Shock Proteins ,Heat shock ,HSF1 ,Receptors, Notch ,Biochemistry, Genetics and Molecular Biology(all) ,Gene Expression Regulation, Leukemic ,fungi ,General Medicine ,Oncogenes ,medicine.disease ,Hsp90 ,Cell biology ,Hematopoiesis ,Heat shock factor ,Mice, Inbred C57BL ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Chaperone (protein) ,biology.protein ,Heat-Shock Response ,Genetics and Molecular Biology(all) ,Signal Transduction - Abstract
Cellular transformation is accompanied by extensive re-wiring of many biological processes leading to augmented levels of distinct types of cellular stress, including proteotoxic stress. Cancer cells critically depend on stress-relief pathways for their survival. However, the mechanisms underlying the transcriptional initiation and maintenance of the oncogenic stress response remain elusive. Here, we show that the expression of heat shock transcription factor 1 (HSF1) and the downstream mediators of the heat shock response is transcriptionally upregulated in T-cell acute lymphoblastic leukemia (T-ALL). Hsf1 ablation suppresses the growth of human T-ALL and eradicates leukemia in mouse models of T-ALL, while sparing normal hematopoiesis. HSF1 drives a compact transcriptional program and among the direct HSF1 targets, specific chaperones and co-chaperones mediate its critical role in T-ALL. Notably, we demonstrate that the central T-ALL oncogene NOTCH1 hijacks the cellular stress response machinery by inducing the expression of HSF1 and its downstream effectors. The NOTCH1 signaling status controls the levels of chaperone/co-chaperone complexes and predicts the response of T-ALL patient samples to HSP90 inhibition. Our data demonstrate an integral crosstalk between mediators of oncogene and non-oncogene addiction and reveal critical nodes of the heat shock response pathway that can be targeted therapeutically.
- Published
- 2018
49. VDR independent induction of acid-sphingomyelinase by 1,23(OH)2 D3 in gastric cancer cells: Impact on apoptosis and cell morphology
- Author
-
Angelo Sidoni, Ivana Ferri, Samuela Cataldi, Michela Codini, Andrea Lazzarini, Elisabetta Albi, Giovanna Traina, Maria Rachele Ceccarini, Katia Fettucciari, Francesco Saverio Ambesi-Impiombato, Francesco Curcio, and Tommaso Beccari
- Subjects
0301 basic medicine ,PTEN ,medicine.medical_specialty ,Acid sphingomyelinase ,Gastric cancer cells ,Vitamin D ,Vitamin D receptor ,medicine.disease_cause ,Cell morphology ,Biochemistry ,Calcitriol receptor ,03 medical and health sciences ,Internal medicine ,medicine ,Tensin ,biology ,General Medicine ,Molecular biology ,030104 developmental biology ,Endocrinology ,Cancer cell ,biology.protein ,Sphingomyelin ,Carcinogenesis ,medicine.drug - Abstract
1 alpha,25-dihydroxyvitamin D3 (1,23(OH)2 D3) is known to play a dual role in cancer, by promoting or inhibiting carcinogenesis via 1,23(OH)2 D3 receptor (VDR) and phosphatase and tensin homolog deleted on chromosome 10 (PTEN). Fok I polymorphism of VDR may indirectly influence the receptor levels through autoregulation. The involvement of neutral sphingomyelinase in the non-classic VDR-mediated genomic pathway response to 1,23(OH)2 D3 treatment has been reported. Until now no information were reported about Fok I polymorphism of VDR in NCI-N87 human gastric cancer cells and the relation between acid sphingomyelinase and 1,23(OH)2 D3. Herein, we showed that NCI-N87 human gastric cancer cells are homozygous for the Fok I 'C' allele; resulting in a three amino acid-truncated protein form of the VDR. Surprisingly 1,23(OH)2 D3 treatments strongly down-regulated the expression of VDR whereas acid sphingomyelinase and PTEN expression were upregulated. No changes of neutral sphingomyelinase expression were observed after 1,23(OH)2 D3 treatment, whereas acid sphingomyelinase activity increased. Furthermore 1,23(OH)2 D3 induced over-expression of caspase 8, CDKN2B, MAP3K5, cytochrome C apoptotic genes. Morphological analysis highlighted some very large round or oval cells and small cells with angular or fusiform extensions, confirmed by MIB-1 immunodetection and Hercep test. Taken together our results indicated that the action of 1,23(OH)2 D3 in gastric cancer cells was independent on 1,23(OH)2 D3 receptor and suggested the acid sphingomyelinase as a possible target to induce molecular events.
- Published
- 2018
- Full Text
- View/download PDF
50. Clonal evolution mechanisms in NT5C2 mutant relapsed acute lymphoblastic leukemia
- Author
-
Alberto Ambesi-Impiombato, Hossein Khiabanian, Mignon L. Loh, Gannie Tzoneva, Koichi Oshima, Chelsea L. Dieck, Giuseppe Basso, Maria Luisa Sulis, Julie M. Gastier-Foster, Maddalena Paganin, Ilaria Iacobucci, Chioma J. Madubata, Adolfo A. Ferrando, Esmé Waanders, Jiangyan Yu, Raul Rabadan, Charles G. Mullighan, Marta Sanchez-Martin, Motohiro Kato, Katsuyoshi Koh, and Renate Kirschner-Schwabe
- Subjects
0301 basic medicine ,Male ,medicine.medical_treatment ,Mutant ,Drug Resistance ,medicine.disease_cause ,Somatic evolution in cancer ,Mice ,0302 clinical medicine ,IMP Dehydrogenase ,Recurrence ,hemic and lymphatic diseases ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Receptor, Notch1 ,Cytotoxicity ,5-Nucleotidase ,5'-Nucleotidase ,Cancer ,Pediatric ,Mutation ,Multidisciplinary ,Guanosine ,Mercaptopurine ,Lymphoblast ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Phenotype ,3. Good health ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Gain of Function Mutation ,Female ,Development of treatments and therapeutic interventions ,Receptor ,Pediatric Cancer ,Childhood Leukemia ,General Science & Technology ,Biology ,Article ,Clonal Evolution ,03 medical and health sciences ,Rare Diseases ,All institutes and research themes of the Radboud University Medical Center ,Nucleotidase ,medicine ,Genetics ,Animals ,Humans ,Cell Proliferation ,Chemotherapy ,Notch1 ,Animal ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,030104 developmental biology ,HEK293 Cells ,Drug Resistance, Neoplasm ,Purines ,Disease Models ,Cancer research ,Neoplasm - Abstract
Relapsed acute lymphoblastic leukaemia (ALL) is associated with resistance to chemotherapy and poor prognosis. Gain-of-function mutations in the 5'-nucleotidase, cytosolic II (NT5C2) gene induce resistance to 6-mercaptopurine and are selectively present in relapsed ALL. Yet, the mechanisms involved in NT5C2 mutation-driven clonal evolution during the initiation of leukaemia, disease progression and relapse remain unknown. Here we use a conditional-and-inducible leukaemia model to demonstrate that expression of NT5C2(R367Q), a highly prevalent relapsed-ALL NT5C2 mutation, induces resistance to chemotherapy with 6-mercaptopurine at the cost of impaired leukaemia cell growth and leukaemia-initiating cell activity. The loss-of-fitness phenotype of NT5C2+/R367Q mutant cells is associated with excess export of purines to the extracellular space and depletion of the intracellular purine-nucleotide pool. Consequently, blocking guanosine synthesis by inhibition of inosine-5'-monophosphate dehydrogenase (IMPDH) induced increased cytotoxicity against NT5C2-mutant leukaemia lymphoblasts. These results identify the fitness cost of NT5C2 mutation and resistance to chemotherapy as key evolutionary drivers that shape clonal evolution in relapsed ALL and support a role for IMPDH inhibition in the treatment of ALL.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.