1
|
Secco I, Backovic A, Tomczyk M, Mura A, Li G, Bortolotti F, Vodret S, Dal Ferro M, Chiavacci E, Zentilin L, Sinagra G, Zacchigna S, Mano M, Giacca M. Genetic tracing and topography of spontaneous and stimulated cardiac regeneration in mice. NATURE CARDIOVASCULAR RESEARCH 2025; 4:397-411. [PMID: 40055464 PMCID: PMC11994457 DOI: 10.1038/s44161-025-00623-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 02/06/2025] [Indexed: 04/15/2025]
Abstract
Despite recent efforts to stimulate endogenous cardiomyocyte proliferation for cardiac regeneration, the lack of reliable in vivo methods for monitoring cardiomyocyte replication has hindered our understanding of its mechanisms. Thymidine analogs, used to label proliferating cells, are unsuitable for long-term cardiac regeneration studies as their DNA incorporation elicits a damage response, leading to their elimination. Here we present CycleTrack, a genetic strategy based on the transcriptional activation of Cre recombinase from a temporally regulated cyclin B2 promoter segment, for permanent labeling of cardiomyocytes passing through the G2/M phase. Using CycleTrack, we visualized cardiomyocyte turnover in neonatal and adult mice under various conditions, including pregnancy, increased ventricular afterload, and myocardial infarction. CycleTrack also provided visual and quantitative evidence of ventricular remuscularization following treatment with pro-regenerative microRNAs. We identify the subendocardium as a key site of mitotic activity and provide a mode of cardiomyocyte division along their short axis. CycleTrack is a powerful tool to monitor cardiomyocyte renewal during regenerative interventions.
Collapse
Affiliation(s)
- Ilaria Secco
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
- MRC/BHF Centre of Research Excellence in Advanced Cardiac Therapies (REACT), King's College London, London, UK
| | - Ana Backovic
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Mateusz Tomczyk
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
- MRC/BHF Centre of Research Excellence in Advanced Cardiac Therapies (REACT), King's College London, London, UK
| | - Antonio Mura
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Gang Li
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
- MRC/BHF Centre of Research Excellence in Advanced Cardiac Therapies (REACT), King's College London, London, UK
| | - Francesca Bortolotti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Simone Vodret
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Matteo Dal Ferro
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano-Isontina, University of Trieste, Trieste, Italy
| | - Elena Chiavacci
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano-Isontina, University of Trieste, Trieste, Italy
| | - Serena Zacchigna
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Miguel Mano
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, UK.
- MRC/BHF Centre of Research Excellence in Advanced Cardiac Therapies (REACT), King's College London, London, UK.
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy.
| |
Collapse
|
2
|
Böttger EC, Santhosh Kumar H, Steiner A, Sotirakis E, Thiam K, Isnard Petit P, Seebeck P, Wolfer DP, Shcherbakov D, Akbergenov R. Translational error in mice increases with ageing in an organ-dependent manner. Nat Commun 2025; 16:2069. [PMID: 40021653 PMCID: PMC11871305 DOI: 10.1038/s41467-025-57203-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 02/10/2025] [Indexed: 03/03/2025] Open
Abstract
The accuracy of protein synthesis and its relation to ageing has been of long-standing interest. To study whether spontaneous changes in the rate of ribosomal error occur as a function of age, we first determined that stop-codon readthrough is a more sensitive read-out of mistranslation due to codon-anticodon mispairing than missense amino acid incorporation. Subsequently, we developed knock-in mice for in-vivo detection of stop-codon readthrough using a gain-of-function Kat2-TGA-Fluc readthrough reporter which combines fluorescent and sensitive bioluminescent imaging techniques. We followed expression of reporter proteins in-vivo over time, and assessed Kat2 and Fluc expression in tissue extracts and by whole organ ex-vivo imaging. Collectively, our results provide evidence for an organ-dependent, age-related increase in translational error: stop-codon readthrough increases with age in muscle (+ 75%, p < 0.001) and brain (+ 50%, p < 0.01), but not in liver (p > 0.5). Together with recent data demonstrating premature ageing in mice with an error-prone ram mutation, our findings highlight age-related decline of translation fidelity as a possible contributor to ageing.
Collapse
Affiliation(s)
- Erik C Böttger
- Institut für Medizinische Mikrobiologie, Universität Zürich, Zurich, Switzerland
| | | | - Adrian Steiner
- Anatomisches Institut, Universität Zürich, and Institut für Bewegungswissenschaften und Sport, ETH Zürich, Zurich, Switzerland
| | | | | | | | - Petra Seebeck
- Zurich Integrative Rodent Physiology (ZIRP), University of Zurich, Zurich, Switzerland
| | - David P Wolfer
- Anatomisches Institut, Universität Zürich, and Institut für Bewegungswissenschaften und Sport, ETH Zürich, Zurich, Switzerland
| | - Dimitri Shcherbakov
- Institut für Medizinische Mikrobiologie, Universität Zürich, Zurich, Switzerland
- Anatomisches Institut, Universität Zürich, and Institut für Bewegungswissenschaften und Sport, ETH Zürich, Zurich, Switzerland
| | - Rashid Akbergenov
- Institut für Medizinische Mikrobiologie, Universität Zürich, Zurich, Switzerland.
- Biozentrum University of Basel, Basel, Switzerland.
| |
Collapse
|
3
|
Ahmed MS, Nguyen NUN, Nakada Y, Hsu CC, Farag A, Lam NT, Wang P, Thet S, Menendez-Montes I, Elhelaly WM, Lou X, Secco I, Tomczyk M, Zentilin L, Pei J, Cui M, Dos Santos M, Liu X, Liu Y, Zaha D, Walcott G, Tomchick DR, Xing C, Zhang CC, Grishin NV, Giacca M, Zhang J, Sadek HA. Identification of FDA-approved drugs that induce heart regeneration in mammals. NATURE CARDIOVASCULAR RESEARCH 2024; 3:372-388. [PMID: 39183959 PMCID: PMC11343477 DOI: 10.1038/s44161-024-00450-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/06/2024] [Indexed: 08/27/2024]
Abstract
Targeting Meis1 and Hoxb13 transcriptional activity could be a viable therapeutic strategy for heart regeneration. In this study, we performd an in silico screening to identify FDA-approved drugs that can inhibit Meis1 and Hoxb13 transcriptional activity based on the resolved crystal structure of Meis1 and Hoxb13 bound to DNA. Paromomycin (Paro) and neomycin (Neo) induced proliferation of neonatal rat ventricular myocytes in vitro and displayed dose-dependent inhibition of Meis1 and Hoxb13 transcriptional activity by luciferase assay and disruption of DNA binding by electromobility shift assay. X-ray crystal structure revealed that both Paro and Neo bind to Meis1 near the Hoxb13-interacting domain. Administration of Paro-Neo combination in adult mice and in pigs after cardiac ischemia/reperfusion injury induced cardiomyocyte proliferation, improved left ventricular systolic function and decreased scar formation. Collectively, we identified FDA-approved drugs with therapeutic potential for induction of heart regeneration in mammals.
Collapse
Affiliation(s)
- Mahmoud Salama Ahmed
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- These authors contributed equally: Mahmoud Salama Ahmed, Ngoc Uyen Nhi Nguyen
| | - Ngoc Uyen Nhi Nguyen
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- These authors contributed equally: Mahmoud Salama Ahmed, Ngoc Uyen Nhi Nguyen
| | - Yuji Nakada
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ching-Cheng Hsu
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ayman Farag
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nicholas T. Lam
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ping Wang
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Suwannee Thet
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ivan Menendez-Montes
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Waleed M. Elhelaly
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xi Lou
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ilaria Secco
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King’s College London, London, UK
| | - Mateusz Tomczyk
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King’s College London, London, UK
| | - Lorena Zentilin
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Jimin Pei
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Miao Cui
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Matthieu Dos Santos
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaoye Liu
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yan Liu
- Eugene McDermott Center for Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David Zaha
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gregory Walcott
- Division of Cardiovascular Diseases, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Diana R. Tomchick
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cheng Cheng Zhang
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nick V. Grishin
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King’s College London, London, UK
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hesham A. Sadek
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
4
|
Dolfini D, Gnesutta N, Mantovani R. Expression and function of NF-Y subunits in cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189082. [PMID: 38309445 DOI: 10.1016/j.bbcan.2024.189082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024]
Abstract
NF-Y is a Transcription Factor (TF) targeting the CCAAT box regulatory element. It consists of the NF-YB/NF-YC heterodimer, each containing an Histone Fold Domain (HFD), and the sequence-specific subunit NF-YA. NF-YA expression is associated with cell proliferation and absent in some post-mitotic cells. The review summarizes recent findings impacting on cancer development. The logic of the NF-Y regulome points to pro-growth, oncogenic genes in the cell-cycle, metabolism and transcriptional regulation routes. NF-YA is involved in growth/differentiation decisions upon cell-cycle re-entry after mitosis and it is widely overexpressed in tumors, the HFD subunits in some tumor types or subtypes. Overexpression of NF-Y -mostly NF-YA- is oncogenic and decreases sensitivity to anti-neoplastic drugs. The specific roles of NF-YA and NF-YC isoforms generated by alternative splicing -AS- are discussed, including the prognostic value of their levels, although the specific molecular mechanisms of activity are still to be deciphered.
Collapse
Affiliation(s)
- Diletta Dolfini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, Milano 20133, Italy
| | - Nerina Gnesutta
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, Milano 20133, Italy
| | - Roberto Mantovani
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, Milano 20133, Italy.
| |
Collapse
|
5
|
Nakashiba T, Ogoh K, Iwano S, Sugiyama T, Mizuno-Iijima S, Nakashima K, Mizuno S, Sugiyama F, Yoshiki A, Miyawaki A, Abe K. Development of two mouse strains conditionally expressing bright luciferases with distinct emission spectra as new tools for in vivo imaging. Lab Anim (NY) 2023; 52:247-257. [PMID: 37679611 PMCID: PMC10533401 DOI: 10.1038/s41684-023-01238-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/01/2023] [Indexed: 09/09/2023]
Abstract
In vivo bioluminescence imaging (BLI) has been an invaluable noninvasive method to visualize molecular and cellular behaviors in laboratory animals. Bioluminescent reporter mice harboring luciferases for general use have been limited to a classical luciferase, Luc2, from Photinus pyralis, and have been extremely powerful for various in vivo studies. However, applicability of reporter mice for in vivo BLI could be further accelerated by increasing light intensity through the use of other luciferases and/or by improving the biodistribution of their substrates in the animal body. Here we created two Cre-dependent reporter mice incorporating luciferases oFluc derived from Pyrocoeli matsumurai and Akaluc, both of which had been reported previously to be brighter than Luc2 when using appropriate substrates; we then tested their bioluminescence in neural tissues and other organs in living mice. When expressed throughout the body, both luciferases emitted an intense yellow (oFluc) or far-red (Akaluc) light easily visible to the naked eye. oFluc and Akaluc were similarly bright in the pancreas for in vivo BLI; however, Akaluc was superior to oFluc for brain imaging, because its substrate, AkaLumine-HCl, was distributed to the brain more efficiently than the oFluc substrate, D-luciferin. We also demonstrated that the lights produced by oFluc and Akaluc were sufficiently spectrally distinct from each other for dual-color imaging in a single living mouse. Taken together, these novel bioluminescent reporter mice are an ideal source of cells with bright bioluminescence and may facilitate in vivo BLI of various tissues/organs for preclinical and biomedical research in combination with a wide variety of Cre-driver mice.
Collapse
Affiliation(s)
- Toshiaki Nakashiba
- Experimental Animal Division, RIKEN BioResource Research Center, Tsukuba, Japan.
| | - Katsunori Ogoh
- Corporate Research and Development Center, Olympus Corporation, Hachioji, Japan
| | - Satoshi Iwano
- Laboratory for Cell Function and Dynamics, RIKEN Center for Brain Science, Wako, Japan
- Institute for Tenure Track Promotion, University of Miyazaki, Miyazaki, Japan
| | - Takashi Sugiyama
- Corporate Research and Development Center, Olympus Corporation, Hachioji, Japan
- R&D Division, Evident Corporation, Hachioji, Japan
| | - Saori Mizuno-Iijima
- Experimental Animal Division, RIKEN BioResource Research Center, Tsukuba, Japan
| | - Kenichi Nakashima
- Gene Engineering Division, RIKEN BioResource Research Center, Tsukuba, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Atsushi Yoshiki
- Experimental Animal Division, RIKEN BioResource Research Center, Tsukuba, Japan
| | - Atsushi Miyawaki
- Laboratory for Cell Function and Dynamics, RIKEN Center for Brain Science, Wako, Japan
| | - Kuniya Abe
- Technology and Development Team for Mammalian Genome Dynamics, RIKEN BioResource Research Center, Tsukuba, Japan
| |
Collapse
|
6
|
Sroka MW, Skopelitis D, Vermunt MW, Preall JB, El Demerdash O, de Almeida LMN, Chang K, Utama R, Gryder B, Caligiuri G, Ren D, Nalbant B, Milazzo JP, Tuveson DA, Dobin A, Hiebert SW, Stengel KR, Mantovani R, Khan J, Kohli RM, Shi J, Blobel GA, Vakoc CR. Myo-differentiation reporter screen reveals NF-Y as an activator of PAX3-FOXO1 in rhabdomyosarcoma. Proc Natl Acad Sci U S A 2023; 120:e2303859120. [PMID: 37639593 PMCID: PMC10483665 DOI: 10.1073/pnas.2303859120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/11/2023] [Indexed: 08/31/2023] Open
Abstract
Recurrent chromosomal rearrangements found in rhabdomyosarcoma (RMS) produce the PAX3-FOXO1 fusion protein, which is an oncogenic driver and a dependency in this disease. One important function of PAX3-FOXO1 is to arrest myogenic differentiation, which is linked to the ability of RMS cells to gain an unlimited proliferation potential. Here, we developed a phenotypic screening strategy for identifying factors that collaborate with PAX3-FOXO1 to block myo-differentiation in RMS. Unlike most genes evaluated in our screen, we found that loss of any of the three subunits of the Nuclear Factor Y (NF-Y) complex leads to a myo-differentiation phenotype that resembles the effect of inactivating PAX3-FOXO1. While the transcriptomes of NF-Y- and PAX3-FOXO1-deficient RMS cells bear remarkable similarity to one another, we found that these two transcription factors occupy nonoverlapping sites along the genome: NF-Y preferentially occupies promoters, whereas PAX3-FOXO1 primarily binds to distal enhancers. By integrating multiple functional approaches, we map the PAX3 promoter as the point of intersection between these two regulators. We show that NF-Y occupies CCAAT motifs present upstream of PAX3 to function as a transcriptional activator of PAX3-FOXO1 expression in RMS. These findings reveal a critical upstream role of NF-Y in the oncogenic PAX3-FOXO1 pathway, highlighting how a broadly essential transcription factor can perform tumor-specific roles in governing cellular state.
Collapse
Affiliation(s)
| | | | - Marit W. Vermunt
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | | | | | | | - Kenneth Chang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY11724
| | - Raditya Utama
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY11724
| | - Berkley Gryder
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH44106
| | | | - Diqiu Ren
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Benan Nalbant
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY11724
| | | | | | | | - Scott W. Hiebert
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN37232
| | - Kristy R. Stengel
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY10461
| | - Roberto Mantovani
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20133Milano, Italy
| | - Javed Khan
- Genetics Branch, National Cancer Institute, NIH, Bethesda, MD20892
| | - Rahul M. Kohli
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA19104
| | - Junwei Shi
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Gerd A. Blobel
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | | |
Collapse
|
7
|
Caforio M, Tumino N, Sorino C, Manni I, Di Giovenale S, Piaggio G, Iezzi S, Strimpakos G, Mattei E, Moretta L, Fanciulli M, Vacca P, Locatelli F, Folgiero V. AATF/Che-1 RNA polymerase II binding protein overexpression reduces the anti-tumor NK-cell cytotoxicity through activating receptors modulation. Front Immunol 2023; 14:1191908. [PMID: 37435061 PMCID: PMC10332273 DOI: 10.3389/fimmu.2023.1191908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/06/2023] [Indexed: 07/13/2023] Open
Abstract
Introduction AATF/Che-1 over-expression in different tumors is well known and its effect on tumorigenicity is mainly due to its central role demonstrated in the oncogenic pathways of solid tumors, where it controls proliferation and viability. The effect exerted by tumors overexpressing Che-1 on the immune response has not yet been investigated. Methods Starting from ChIP-sequencing data we confirmed Che-1 enrichment on Nectin-1 promoter. Several co-cultures experiments between NK-cells and tumor cells transduced by lentiviral vectors carrying Che-1-interfering sequence, analyzed by flow-cytometry have allowed a detailed characterization of NK receptors and tumor ligands expression. Results Here, we show that Che-1 is able to modulate the expression of Nectin-1 ligand at the transcriptional level, leading to the impairment of killing activity of NK-cells. Nectin-1 down-modulation induces a modification in NK-cell ligands expression able to interact with activating receptors and to stimulate NK-cell function. In addition, NK-cells from Che-1 transgenic mice, confirming a reduced expression of activating receptors, exhibit impaired activation and a preferential immature status. Discussion The critical equilibrium between NK-cell ligand expression on tumor cells and the interaction with NK cell receptors is affected by Che-1 over-expression and partially restored by Che-1 interference. The evidence of a new role for Che-1 as regulator of anti-tumor immunity supports the necessity to develop approaches able to target this molecule which shows a dual tumorigenic function as cancer promoter and immune response modulator.
Collapse
Affiliation(s)
- Matteo Caforio
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Nicola Tumino
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children’s Hospital Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Cristina Sorino
- Stabilimento Allevamento Fornitore e Utilizzatore (SAFU) Laboratory, Department of Research, Advanced Diagnostic, Technological Innovation, Regina Elena National Cancer Institute Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Isabella Manni
- Stabilimento Allevamento Fornitore e Utilizzatore (SAFU) Laboratory, Department of Research, Advanced Diagnostic, Technological Innovation, Regina Elena National Cancer Institute Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Stefano Di Giovenale
- Stabilimento Allevamento Fornitore e Utilizzatore (SAFU) Laboratory, Department of Research, Advanced Diagnostic, Technological Innovation, Regina Elena National Cancer Institute Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Giulia Piaggio
- Stabilimento Allevamento Fornitore e Utilizzatore (SAFU) Laboratory, Department of Research, Advanced Diagnostic, Technological Innovation, Regina Elena National Cancer Institute Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Simona Iezzi
- Stabilimento Allevamento Fornitore e Utilizzatore (SAFU) Laboratory, Department of Research, Advanced Diagnostic, Technological Innovation, Regina Elena National Cancer Institute Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Georgios Strimpakos
- National Research Council (CNR), Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy
| | - Elisabetta Mattei
- Consiglio Nazionale delle Ricerche (CNR)-Institute of Cell Biology and Neurobiology, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit , Children Hospital Bambino Gesù, RomaLM, Rome, Italy
| | - M. Fanciulli
- Stabilimento Allevamento Fornitore e Utilizzatore (SAFU) Laboratory, Department of Research, Advanced Diagnostic, Technological Innovation, Regina Elena National Cancer Institute Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Paola Vacca
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children’s Hospital Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| | - Valentina Folgiero
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
8
|
Flori E, Mosca S, Cardinali G, Briganti S, Ottaviani M, Kovacs D, Manni I, Truglio M, Mastrofrancesco A, Zaccarini M, Cota C, Piaggio G, Picardo M. The Activation of PPARγ by (2Z,4E,6E)-2-methoxyocta-2,4,6-trienoic Acid Counteracts the Epithelial–Mesenchymal Transition Process in Skin Carcinogenesis. Cells 2023; 12:cells12071007. [PMID: 37048080 PMCID: PMC10093137 DOI: 10.3390/cells12071007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the most common UV-induced keratinocyte-derived cancer, and its progression is characterized by the epithelial–mesenchymal transition (EMT) process. We previously demonstrated that PPARγ activation by 2,4,6-octatrienoic acid (Octa) prevents cutaneous UV damage. We investigated the possible role of the PPARγ activators Octa and the new compound (2Z,4E,6E)-2-methoxyocta-2,4,6-trienoic acid (A02) in targeting keratinocyte-derived skin cancer. Like Octa, A02 exerted a protective effect against UVB-induced oxidative stress and DNA damage in NHKs. In the squamous cell carcinoma A431 cells, A02 inhibited cell proliferation and increased differentiation markers’ expression. Moreover, Octa and even more A02 counteracted the TGF-β1-dependent increase in mesenchymal markers, intracellular ROS, the activation of EMT-related signal transduction pathways, and cells’ migratory capacity. Both compounds, especially A02, counterbalanced the TGF-β1-induced cell membrane lipid remodeling and the release of bioactive lipids involved in EMT. In vivo experiments on a murine model useful to study cell proliferation in adult animals showed the reduction of areas characterized by active cell proliferation in response to A02 topical treatment. In conclusion, targeting PPARγ may be useful for the prevention and treatment of keratinocyte-derived skin cancer.
Collapse
Affiliation(s)
- Enrica Flori
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
- Correspondence: (E.F.); (M.P.)
| | - Sarah Mosca
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Giorgia Cardinali
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Stefania Briganti
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Monica Ottaviani
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Daniela Kovacs
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Isabella Manni
- SAFU Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Roma, Italy
| | - Mauro Truglio
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Arianna Mastrofrancesco
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Marco Zaccarini
- Genetic Research, Molecular Biology and Dermatopathology Unit, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Carlo Cota
- Genetic Research, Molecular Biology and Dermatopathology Unit, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Giulia Piaggio
- SAFU Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Roma, Italy
| | - Mauro Picardo
- Faculty of Medicine, Unicamillus International Medical University, 00131 Rome, Italy
- Correspondence: (E.F.); (M.P.)
| |
Collapse
|
9
|
Li Z, You C, Huang J, Zhang M. NFYAv1 is a Tumor-Promoting Transcript Associated with Poor Prognosis of Hepatocellular Carcinoma. Med Sci Monit 2023; 29:e938410. [PMID: 36680333 PMCID: PMC9875548 DOI: 10.12659/msm.938410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Nuclear Transcription Factor Y Subunit Alpha (NFYA), together with NFYB and NFYC, form a sequence-specific heterotrimeric nuclear transcription factor (NFY), but their functional role in hepatocellular carcinoma (HCC) is still unclear. In this study, we explored the association between the NFY subunit genes and the survival of primary hepatocellular carcinoma (HCC) patients in The Cancer Genome Atlas (TCGA). The transcript-specific effect on HCC cell growth was studied. MATERIAL AND METHODS RNA-seq data from the Genotype-Tissue Expression Project (GTEx) and TCGA were analyzed in combination. In vitro cellular and molecular studies were conducted using SK-Hep-1 and Hep3B cells. Pearson's correlation coefficients were calculated to assess correlations. Welch's unpaired t test and one-way ANOVA with post hoc Tukey's multiple comparisons were performed. Kaplan-Meier (K-M) survival curves were assessed by conducting log-rank (Mantel-Cox) test. RESULTS NFYA was the only prognosis-related gene. Among the 2 splicing transcripts of NFYA, the long isoform (NFYAv1, NM_002505.5) but not the short-form (NFYAv2, NM_021705.4) was significantly associated with worse progression-free survival (PFS) (high [n=179] vs low [n=179], HR: 1.657, 95% CI: 1.228-2.235, P<0.001) and disease-specific survival (DSS) (high [n=175] vs low [n=175], HR: 1.986, 95% CI: 1.269-3.108, P<0.001) in HCC patients. GO/KEGG analysis in TCGA confirmed that NFYAv1 and NFYAv2 co-expressed (|Pearson's r|≥0.6) genes in primary HCC patients were enriched in quite different GO/KEGG terms. NFYAv1 knockdown significantly decreased cell viability and increased G0/G1 cell cycle arrest. The shRNA only targeting NFYAv1 had a significantly stronger growth-inhibiting effect than the shRNA targeting both NFYAv1 and NFYAv2. CONCLUSIONS This study showed that NFYAv1 is a tumor-promoting transcript associated with poor prognosis of HCC.
Collapse
Affiliation(s)
- Zigang Li
- Department of Hepatobiliary Surgery, The First People’s Hospital of Yibin, Yibin, Sichuan, PR China
| | - Chengshan You
- Department of Endocrinology, The Second People’s Hospital of Yibin, Yibin, Sichuan, PR China
| | - Jun Huang
- Department of Hepatobiliary Surgery, The First People’s Hospital of Yibin, Yibin, Sichuan, PR China
| | - Ming Zhang
- Department of Hepatobiliary Surgery, The First People’s Hospital of Yibin, Yibin, Sichuan, PR China
| |
Collapse
|
10
|
Manni I, de Latouliere L, Piaggio G. Bioluminescence and Optical Imaging: Principles and Applications. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00105-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
11
|
Baldari S, Manni I, Di Rocco G, Paolini F, Palermo B, Piaggio G, Toietta G. Reduction of Cell Proliferation by Acute C 2H 6O Exposure. Cancers (Basel) 2021; 13:4999. [PMID: 34638483 PMCID: PMC8508324 DOI: 10.3390/cancers13194999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/17/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022] Open
Abstract
Endogenous acetaldehyde production from the metabolism of ingested alcohol exposes hematopoietic progenitor cells to increased genotoxic risk. To develop possible therapeutic strategies to prevent or reverse alcohol abuse effects, it would be critical to determine the temporal progression of acute ethanol toxicity on progenitor cell numbers and proliferative status. We followed the variation of the cell proliferation rate in bone marrow and spleen in response to acute ethanol intoxication in the MITO-Luc mouse, in which NF-Y-dependent cell proliferation can be assessed in vivo by non-invasive bioluminescent imaging. One week after ethanol administration, bioluminescent signals in bone marrow and spleen decreased below the level corresponding to physiological proliferation, and they progressively resumed to pre-treatment values in approximately 4 weeks. Boosting acetaldehyde catabolism by administration of an aldehyde dehydrogenase activity activator or administration of polyphenols with antioxidant activity partially restored bone marrow cells' physiological proliferation. These results indicate that in this mouse model, bioluminescent alteration reflects the reduction of the physiological proliferation rate of bone marrow progenitor cells due to the toxic effect of aldehydes generated by alcohol oxidation. In summary, this study presents a novel view of the impact of acute alcohol intake on bone marrow cell proliferation in vivo.
Collapse
Affiliation(s)
- Silvia Baldari
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (S.B.); (F.P.); (B.P.)
| | - Isabella Manni
- Stabilimento Allevatore Fornitore Utilizzatore (SAFU), IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (I.M.); (G.P.)
| | - Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Francesca Paolini
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (S.B.); (F.P.); (B.P.)
| | - Belinda Palermo
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (S.B.); (F.P.); (B.P.)
| | - Giulia Piaggio
- Stabilimento Allevatore Fornitore Utilizzatore (SAFU), IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (I.M.); (G.P.)
| | - Gabriele Toietta
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (S.B.); (F.P.); (B.P.)
| |
Collapse
|
12
|
Panzeri V, Manni I, Capone A, Naro C, Sacconi A, Di Agostino S, de Latouliere L, Montori A, Pilozzi E, Piaggio G, Capurso G, Sette C. The RNA-binding protein MEX3A is a prognostic factor and regulator of resistance to gemcitabine in pancreatic ductal adenocarcinoma. Mol Oncol 2021; 15:579-595. [PMID: 33159833 PMCID: PMC7858117 DOI: 10.1002/1878-0261.12847] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/16/2020] [Accepted: 11/05/2020] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer. Most patients present with advanced disease at diagnosis, which only permits palliative chemotherapeutic treatments. RNA dysregulation is a hallmark of most human cancers, including PDAC. To test the impact of RNA processing dysregulation on PDAC pathology, we performed a bioinformatics analysis to identify RNA-binding proteins (RBPs) associated with prognosis. Among the 12 RBPs associated with progression-free survival, we focused on MEX3A because it was recently shown to mark an intestinal stem cell population that is refractory to chemotherapeutic treatments, a typical feature of PDAC. Increased expression of MEX3A was correlated with higher disease stage in PDAC patients and with tumor development in a mouse model of PDAC. Depletion of MEX3A in PDAC cells enhanced sensitivity to chemotherapeutic treatment with gemcitabine, whereas its expression was increased in PDAC cells selected upon chronic exposure to the drug. RNA-sequencing analyses highlighted hundreds of genes whose expression is sensitive to MEX3A expression, with significant enrichment in cell cycle genes. MEX3A binds to its target mRNAs, like cyclin-dependent kinase 6 (CDK6), and promotes their stability. Accordingly, knockdown of MEX3A caused a significant reduction in PDAC cell proliferation and in progression to the S phase of the cell cycle. These findings uncover a novel role for MEX3A in the acquisition and maintenance of chemoresistance by PDAC cells, suggesting that it may represent a novel therapeutic target for PDAC.
Collapse
MESH Headings
- Animals
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Cell Line, Tumor
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Humans
- Mice
- Mice, Knockout
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplasms, Experimental/diagnosis
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- Prognosis
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Gemcitabine
Collapse
Affiliation(s)
- Valentina Panzeri
- Department of Science Medical/Chirurgic and Translational MedicineUniversity of Rome “Sapienza”Italy
- Department of NeuroscienceSection of Human AnatomyCatholic University of the Sacred HeartRomeItaly
| | - Isabella Manni
- UOSD SAFUDepartment of Research, Diagnosis and Innovative TechnologiesIRCCS Regina Elena National Cancer InstituteRomeItaly
| | | | - Chiara Naro
- Department of NeuroscienceSection of Human AnatomyCatholic University of the Sacred HeartRomeItaly
- IRCCS Fondazione Policlinico Agostino GemelliRomeItaly
| | - Andrea Sacconi
- Clinical Trial Center, Biostatistics and Bioinformatics UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Silvia Di Agostino
- Oncogenomic and Epigenetic UnitDepartment of Diagnostic Research and Technological InnovationIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Luisa de Latouliere
- UOSD SAFUDepartment of Research, Diagnosis and Innovative TechnologiesIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Andrea Montori
- Department of Clinical and Molecular MedicineUOC Anatomia PatologicaSant' Andrea HospitalSapienza University of RomeItaly
| | - Emanuela Pilozzi
- Department of Clinical and Molecular MedicineUOC Anatomia PatologicaSant' Andrea HospitalSapienza University of RomeItaly
| | - Giulia Piaggio
- UOSD SAFUDepartment of Research, Diagnosis and Innovative TechnologiesIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Gabriele Capurso
- PancreatoBiliary Endoscopy and EUS DivisionPancreas Translational and Clinical Research CenterSan Raffaele Scientific Institute IRCCSMilanItaly
| | - Claudio Sette
- Department of NeuroscienceSection of Human AnatomyCatholic University of the Sacred HeartRomeItaly
- Fondazione Santa Lucia IRCCSRomeItaly
| |
Collapse
|
13
|
de Latouliere L, Manni I, Ferrari L, Pisati F, Totaro MG, Gurtner A, Marra E, Pacello L, Pozzoli O, Aurisicchio L, Capogrossi MC, Deflorian G, Piaggio G. MITO-Luc/GFP zebrafish model to assess spatial and temporal evolution of cell proliferation in vivo. Sci Rep 2021; 11:671. [PMID: 33436662 PMCID: PMC7804000 DOI: 10.1038/s41598-020-79530-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 12/09/2020] [Indexed: 01/05/2023] Open
Abstract
We developed a novel reporter transgenic zebrafish model called MITO-Luc/GFP zebrafish in which GFP and luciferase expression are under the control of the master regulator of proliferation NF-Y. In MITO-Luc/GFP zebrafish it is possible to visualize cell proliferation in vivo by fluorescence and bioluminescence. In this animal model, GFP and luciferase expression occur in early living embryos, becoming tissue specific in juvenile and adult zebrafish. By in vitro and ex vivo experiments we demonstrate that luciferase activity in adult animals occurs in intestine, kidney and gonads, where detectable proliferating cells are located. Further, by time lapse experiments in live embryos, we observed a wave of GFP positive cells following fin clip. In adult zebrafish, in addition to a bright bioluminescence signal on the regenerating tail, an early unexpected signal coming from the kidney occurs indicating not only a fin cell proliferation, but also a systemic response to tissue damage. Finally, we observed that luciferase activity was inhibited by anti-proliferative interventions, i.e. 5FU, cell cycle inhibitors and X-Rays. In conclusion, MITO-Luc/GFP zebrafish is a novel animal model that may be crucial to assess the spatial and temporal evolution of cell proliferation in vivo.
Collapse
Affiliation(s)
- Luisa de Latouliere
- UOSD SAFU, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Isabella Manni
- UOSD SAFU, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Laura Ferrari
- IFOM - FIRC Institute of Molecular Oncology, Milan, Italy
| | - Federica Pisati
- Histopathology Unit, Cogentech S.C.a.R.L, 20139, Milan, Italy
| | | | - Aymone Gurtner
- UOSD SAFU, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.,Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Emanuele Marra
- Takis s.r.l., via Castel Romano 100, 00128, Rome, Italy.,VITARES -APS, via Castel Romano 100, 00128, Rome, Italy
| | | | - Ombretta Pozzoli
- Laboratorio Di Biologia Vascolare e Medicina Rigenerativa - Centro Cardiologico Monzino - IRCCS (Istituto Di Ricovero E Cura a Carattere Scientifico), Milan, Italy.,Pfizer Italia, Via A.M. Mozzoni 12, 20152, Milan, Italy
| | - Luigi Aurisicchio
- Takis s.r.l., via Castel Romano 100, 00128, Rome, Italy.,VITARES -APS, via Castel Romano 100, 00128, Rome, Italy
| | - Maurizio C Capogrossi
- Johns Hopkins University School of Medicine, Division of Cardiology, 301 Building, Suite 2400, 4940 Eastern Avenue, Baltimore, MD, 21224, USA.,Laboratory of Cardiovascular Sciences, National Institute on Aging/National Institutes of Health, Baltimore, MD, 21224, USA
| | - Gianluca Deflorian
- IFOM - FIRC Institute of Molecular Oncology, Milan, Italy.,Cogentech SRL - Benefit Corporation, Milan, Italy
| | - Giulia Piaggio
- UOSD SAFU, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| |
Collapse
|
14
|
Bezzecchi E, Ronzio M, Mantovani R, Dolfini D. NF-Y Overexpression in Liver Hepatocellular Carcinoma (HCC). Int J Mol Sci 2020; 21:E9157. [PMID: 33271832 PMCID: PMC7731131 DOI: 10.3390/ijms21239157] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 12/24/2022] Open
Abstract
NF-Y is a pioneer trimeric transcription factor formed by the Histone Fold Domain (HFD) NF-YB/NF-YC subunits and NF-YA. Three subunits are required for DNA binding. CCAAT-specificity resides in NF-YA and transactivation resides in Q-rich domains of NF-YA and NF-YC. They are involved in alternative splicing (AS). We recently showed that NF-YA is overexpressed in breast and lung carcinomas. We report here on the overexpression of all subunits in the liver hepatocellular carcinoma (HCC) TCGA database, specifically the short NF-YAs and NF-YC2 (37 kDa) isoforms. This is observed at all tumor stages, in viral-infected samples and independently from the inflammatory status. Up-regulation of NF-YAs and NF-YC, but not NF-YB, is associated to tumors with mutant p53. We used a deep-learning-based method (DeepCC) to extend the partitioning of the three molecular clusters to all HCC TCGA tumors. In iCluster3, CCAAT is a primary matrix found in promoters of up-regulated genes, and cell-cycle pathways are enriched. Finally, clinical data indicate that, globally, only NF-YAs, but not HFD subunits, correlate with the worst prognosis; in iCluster1 patients, however, all subunits correlate. The data show a difference with other epithelial cancers, in that global overexpression of the three subunits is reported and clinically relevant in a subset of patients; yet, they further reinstate the regulatory role of the sequence-specific subunit.
Collapse
Affiliation(s)
| | | | | | - Diletta Dolfini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy; (E.B.); (M.R.); (R.M.)
| |
Collapse
|
15
|
Infante P, Malfanti A, Quaglio D, Balducci S, De Martin S, Bufalieri F, Mastrotto F, Basili I, Garofalo M, Lospinoso Severini L, Mori M, Manni I, Moretti M, Nicoletti C, Piaggio G, Caliceti P, Botta B, Ghirga F, Salmaso S, Di Marcotullio L. Glabrescione B delivery by self-assembling micelles efficiently inhibits tumor growth in preclinical models of Hedgehog-dependent medulloblastoma. Cancer Lett 2020; 499:220-231. [PMID: 33249196 DOI: 10.1016/j.canlet.2020.11.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/25/2022]
Abstract
Aberrant activation of the Hedgehog (Hh) pathway leads to the development of several tumors, including medulloblastoma (MB), the most common pediatric brain malignancy. Hh inhibitors acting on GLI1, the final effector of Hh signaling, offer a valuable opportunity to overcome the pitfalls of the existing therapies to treat Hh-driven cancers. In this study, the toxicity, delivery, biodistribution, and anticancer efficacy of Glabrescione B (GlaB), a selective GLI1 inhibitor, were investigated in preclinical models of Hh-dependent MB. To overcome its poor water solubility, GlaB was formulated with a self-assembling amphiphilic polymer forming micelles, called mPEG5kDa-cholane. mPEG5kDa-cholane/GlaB showed high drug loading and stability, low cytotoxicity, and long permanence in the bloodstream. We found that mPEG5kDa-cholane efficiently enhanced the solubility of GlaB, thus avoiding the use of organic solvents. mPEG5kDa-cholane/GlaB possesses favorable pharmacokinetics and negligible toxicity. Remarkably, GlaB encapsulated in mPEG5kDa-cholane micelles was delivered through the blood-brain barrier and drastically inhibited tumor growth in both allograft and orthotopic models of Hh-dependent MB. Our findings reveal that mPEG5kDa-cholane/GlaB is a good candidate for the treatment of Hh-driven tumors and provide relevant implications for the translation of GlaB into clinical practice.
Collapse
Affiliation(s)
- Paola Infante
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Roma, Italy
| | - Alessio Malfanti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs, University La Sapienza, Roma, Italy
| | - Silvia Balducci
- Department of Chemistry and Technology of Drugs, University La Sapienza, Roma, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Irene Basili
- Department of Molecular Medicine, University La Sapienza, Roma, Italy
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Isabella Manni
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Roma, Italy
| | - Marta Moretti
- Department of Experimental Medicine, University La Sapienza, Roma, Italy
| | - Carmine Nicoletti
- DAHFMO-Unit of Histology and Medical Embryology, University La Sapienza, Roma, Italy; Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, University La Sapienza, Roma, Italy
| | - Giulia Piaggio
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Roma, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Bruno Botta
- Department of Chemistry and Technology of Drugs, University La Sapienza, Roma, Italy
| | - Francesca Ghirga
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Roma, Italy.
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, University La Sapienza, Roma, Italy; Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, University La Sapienza, Roma, Italy.
| |
Collapse
|
16
|
Menini S, Iacobini C, de Latouliere L, Manni I, Vitale M, Pilozzi E, Pesce C, Cappello P, Novelli F, Piaggio G, Pugliese G. Diabetes promotes invasive pancreatic cancer by increasing systemic and tumour carbonyl stress in Kras G12D/+ mice. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:152. [PMID: 32778157 PMCID: PMC7418209 DOI: 10.1186/s13046-020-01665-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 08/04/2020] [Indexed: 01/13/2023]
Abstract
Background Type 1 and 2 diabetes confer an increased risk of pancreatic cancer (PaC) of similar magnitude, suggesting a common mechanism. The recent finding that PaC incidence increases linearly with increasing fasting glucose levels supports a central role for hyperglycaemia, which is known to cause carbonyl stress and advanced glycation end-product (AGE) accumulation through increased glycolytic activity and non-enzymatic reactions. This study investigated the impact of hyperglycaemia on invasive tumour development and the underlying mechanisms involved. Methods Pdx1-Cre;LSL-KrasG12D/+ mice were interbred with mitosis luciferase reporter mice, rendered diabetic with streptozotocin and treated or not with carnosinol (FL-926-16), a selective scavenger of reactive carbonyl species (RCS) and, as such, an inhibitor of AGE formation. Mice were monitored for tumour development by in vivo bioluminescence imaging. At the end of the study, pancreatic tissue was collected for histology/immunohistochemistry and molecular analyses. Mechanistic studies were performed in pancreatic ductal adenocarcinoma cell lines challenged with high glucose, glycolysis- and glycoxidation-derived RCS, their protein adducts AGEs and sera from diabetic patients. Results Cumulative incidence of invasive PaC at 22 weeks of age was 75% in untreated diabetic vs 25% in FL-926-16-gtreated diabetic and 8.3% in non-diabetic mice. FL-926-16 treatment suppressed systemic and pancreatic carbonyl stress, extracellular signal-regulated kinases (ERK) 1/2 activation, and nuclear translocation of Yes-associated protein (YAP) in pancreas. In vitro, RCS scavenging and AGE elimination completely inhibited cell proliferation stimulated by high glucose, and YAP proved essential in mediating the effects of both glucose-derived RCS and their protein adducts AGEs. However, RCS and AGEs induced YAP activity through distinct pathways, causing reduction of Large Tumour Suppressor Kinase 1 and activation of the Epidermal Growth Factor Receptor/ERK signalling pathway, respectively. Conclusions An RCS scavenger and AGE inhibitor prevented the accelerating effect of diabetes on PainINs progression to invasive PaC, showing that hyperglycaemia promotes PaC mainly through increased carbonyl stress. In vitro experiments demonstrated that both circulating RCS/AGEs and tumour cell-derived carbonyl stress generated by excess glucose metabolism induce proliferation by YAP activation, hence providing a molecular mechanism underlying the link between diabetes and PaC (and cancer in general).
Collapse
Affiliation(s)
- Stefano Menini
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy
| | - Carla Iacobini
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy
| | - Luisa de Latouliere
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy.,SAFU-unit, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Isabella Manni
- SAFU-unit, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Martina Vitale
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy.,Pathology Unit, University "La Sapienza", Sant'Andrea Hospital, Rome, Italy
| | - Carlo Pesce
- DINOGMI, University of Genoa Medical School, Genoa, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Giulia Piaggio
- SAFU-unit, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy.
| |
Collapse
|
17
|
Carpenter RS, Marbourg JM, Brennan FH, Mifflin KA, Hall JCE, Jiang RR, Mo XM, Karunasiri M, Burke MH, Dorrance AM, Popovich PG. Spinal cord injury causes chronic bone marrow failure. Nat Commun 2020; 11:3702. [PMID: 32710081 PMCID: PMC7382469 DOI: 10.1038/s41467-020-17564-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 07/01/2020] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) causes immune dysfunction, increasing the risk of infectious morbidity and mortality. Since bone marrow hematopoiesis is essential for proper immune function, we hypothesize that SCI disrupts bone marrow hematopoiesis. Indeed, SCI causes excessive proliferation of bone marrow hematopoietic stem and progenitor cells (HSPC), but these cells cannot leave the bone marrow, even after challenging the host with a potent inflammatory stimulus. Sequestration of HSPCs in bone marrow after SCI is linked to aberrant chemotactic signaling that can be reversed by post-injury injections of Plerixafor (AMD3100), a small molecule inhibitor of CXCR4. Even though Plerixafor liberates HSPCs and mature immune cells from bone marrow, competitive repopulation assays show that the intrinsic long-term functional capacity of HSPCs is still impaired in SCI mice. Together, our data suggest that SCI causes an acquired bone marrow failure syndrome that may contribute to chronic immune dysfunction.
Collapse
Affiliation(s)
- Randall S Carpenter
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Jessica M Marbourg
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Faith H Brennan
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Katherine A Mifflin
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Jodie C E Hall
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Roselyn R Jiang
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Xiaokui M Mo
- Center for Biostatistics and Bioinformatics, The Ohio State University, Columbus, OH, USA
| | - Malith Karunasiri
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Matthew H Burke
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Adrienne M Dorrance
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA.
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
18
|
Libetti D, Bernardini A, Sertic S, Messina G, Dolfini D, Mantovani R. The Switch from NF-YAl to NF-YAs Isoform Impairs Myotubes Formation. Cells 2020; 9:cells9030789. [PMID: 32214056 PMCID: PMC7140862 DOI: 10.3390/cells9030789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/19/2020] [Accepted: 03/21/2020] [Indexed: 12/19/2022] Open
Abstract
NF-YA, the regulatory subunit of the trimeric transcription factor (TF) NF-Y, is regulated by alternative splicing (AS) generating two major isoforms, “long” (NF-YAl) and “short” (NF-YAs). Muscle cells express NF-YAl. We ablated exon 3 in mouse C2C12 cells by a four-guide CRISPR/Cas9n strategy, obtaining clones expressing exclusively NF-YAs (C2-YAl-KO). C2-YAl-KO cells grow normally, but are unable to differentiate. Myogenin and—to a lesser extent, MyoD— levels are substantially lower in C2-YAl-KO, before and after differentiation. Expression of the fusogenic Myomaker and Myomixer genes, crucial for the early phases of the process, is not induced. Myomaker and Myomixer promoters are bound by MyoD and Myogenin, and Myogenin overexpression induces their expression in C2-YAl-KO. NF-Y inactivation reduces MyoD and Myogenin, but not directly: the Myogenin promoter is CCAAT-less, and the canonical CCAAT of the MyoD promoter is not bound by NF-Y in vivo. We propose that NF-YAl, but not NF-YAs, maintains muscle commitment by indirectly regulating Myogenin and MyoD expression in C2C12 cells. These experiments are the first genetic evidence that the two NF-YA isoforms have functionally distinct roles.
Collapse
|
19
|
Manni I, de Latouliere L, Gurtner A, Piaggio G. Transgenic Animal Models to Visualize Cancer-Related Cellular Processes by Bioluminescence Imaging. Front Pharmacol 2019; 10:235. [PMID: 30930779 PMCID: PMC6428995 DOI: 10.3389/fphar.2019.00235] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/25/2019] [Indexed: 12/21/2022] Open
Abstract
Preclinical animal models are valuable tools to improve treatments of malignant diseases, being an intermediate step of experimentation between cell culture and human clinical trials. Among different animal models frequently used in cancer research are mouse and, more recently, zebrafish models. Indeed, most of the cellular pathways are highly conserved between human, mouse and zebrafish, thus rendering these models very attractive. Recently, several transgenic reporter mice and zebrafishes have been generated in which the luciferase reporter gene are placed under the control of a promoter whose activity is strictly related to specific cancer cellular processes. Other mouse models have been generated by the cDNA luciferase knockin in the locus of a gene whose expression/activity has increased in cancer. Using BioLuminescence Imaging (BLI), we have now the opportunity to spatiotemporal visualize cell behaviors, among which proliferation, apoptosis, migration and immune responses, in any body district in living animal in a time frame process. We provide here a review of the available models to visualized cancer and cancer-associated events in living animals by BLI and as they have been successful in identifying new stages of early tumor progression, new interactions between different tissues and new therapeutic responsiveness.
Collapse
Affiliation(s)
- Isabella Manni
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Luisa de Latouliere
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Aymone Gurtner
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Piaggio
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
20
|
Inhibition of SIRT1 deacetylase and p53 activation uncouples the anti-inflammatory and chemopreventive actions of NSAIDs. Br J Cancer 2019; 120:537-546. [PMID: 30739913 PMCID: PMC6461760 DOI: 10.1038/s41416-018-0372-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 11/28/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022] Open
Abstract
Background Nonsteroidal anti-inflammatory drugs (NSAIDs) have been proposed as chemopreventive agents for many tumours; however, the mechanism responsible for their anti-neoplastic activity remains elusive and the side effects due to cyclooxygenase (COX) inhibition prevent this clinical application. Methods Molecular biology, in silico, cellular and in vivo tools, including innovative in vivo imaging and classical biochemical assays, were applied to identify and characterise the COX-independent anti-cancer mechanism of NSAIDs. Results Here, we show that tumour-protective functions of NSAIDs and exisulind (a sulindac metabolite lacking anti-inflammatory activity) occur through a COX-independent mechanism. We demonstrate these NSAIDs counteract carcinogen-induced proliferation by inhibiting the sirtuin 1 (SIRT1) deacetylase activity, augmenting acetylation and activity of the tumour suppressor p53 and increasing the expression of the antiproliferative gene p21. These properties are shared by all NSAIDs except for ketoprofen lacking anti-cancer properties. The clinical interest of the mechanism identified is underlined by our finding that p53 is activated in mastectomy patients undergoing intraoperative ketorolac, a treatment associated with decreased relapse risk and increased survival. Conclusion Our study, for the first-time, links NSAID chemopreventive activity with direct SIRT1 inhibition and activation of the p53/p21 anti-oncogenic pathway, suggesting a novel strategy for the design of tumour-protective drugs.
Collapse
|
21
|
Menini S, Iacobini C, de Latouliere L, Manni I, Ionta V, Blasetti Fantauzzi C, Pesce C, Cappello P, Novelli F, Piaggio G, Pugliese G. The advanced glycation end-product N ϵ -carboxymethyllysine promotes progression of pancreatic cancer: implications for diabetes-associated risk and its prevention. J Pathol 2018. [PMID: 29533466 DOI: 10.1002/path.5072] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Diabetes is an established risk factor for pancreatic cancer (PaC), together with obesity, a Western diet, and tobacco smoking. The common mechanistic link might be the accumulation of advanced glycation end-products (AGEs), which characterizes all of the above disease conditions and unhealthy habits. Surprisingly, however, the role of AGEs in PaC has not been examined yet, despite the evidence of a tumour-promoting role of receptor for advanced glycation end-products (RAGE), the receptor for AGEs. Here, we tested the hypothesis that AGEs promote PaC through RAGE activation. To this end, we investigated the effects of the AGE Nϵ -carboxymethyllysine (CML) in human pancreatic ductal adenocarcinoma (PDA) cell lines and in a mouse model of Kras-driven PaC interbred with a bioluminescent model of proliferation. Tumour growth was monitored in vivo by bioluminescence imaging and confirmed by histology. CML promoted PDA cell growth and RAGE expression, in a concentration-dependent and time-dependent manner, and activated downstream tumourigenic signalling pathways. These effects were counteracted by RAGE antagonist peptide (RAP). Exogenous AGE administration to PaC-prone mice induced RAGE upregulation in pancreatic intraepithelial neoplasias (PanINs) and markedly accelerated progression to invasive PaC. At 11 weeks of age (6 weeks of CML treatment), PaC was observed in eight of 11 (72.7%) CML-treated versus one of 11 (9.1%) vehicle-treated [control (Ctr)] mice. RAP delayed PanIN development in Ctr mice but failed to prevent PaC promotion in CML-treated mice, probably because of competition with soluble RAGE for binding to AGEs and/or compensatory upregulation of the RAGE homologue CD166/ activated leukocyte cell adhesion molecule, which also favoured tumour spread. These findings indicate that AGEs modulate the development and progression of PaC through receptor-mediated mechanisms, and might be responsible for the additional risk conferred by diabetes and other conditions characterized by increased AGE accumulation. Finally, our data suggest that an AGE reduction strategy, instead of RAGE inhibition, might be suitable for the risk management and prevention of PaC. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Stefano Menini
- Department of Clinical and Molecular Medicine, 'La Sapienza' University, Rome, Italy
| | - Carla Iacobini
- Department of Clinical and Molecular Medicine, 'La Sapienza' University, Rome, Italy
| | - Luisa de Latouliere
- Department of Clinical and Molecular Medicine, 'La Sapienza' University, Rome, Italy
| | - Isabella Manni
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Vittoria Ionta
- Department of Clinical and Molecular Medicine, 'La Sapienza' University, Rome, Italy
| | | | - Carlo Pesce
- DINOGMI, University of Genoa Medical School, Genoa, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Giulia Piaggio
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, 'La Sapienza' University, Rome, Italy
| |
Collapse
|
22
|
Carafa V, Nebbioso A, Cuomo F, Rotili D, Cobellis G, Bontempo P, Baldi A, Spugnini EP, Citro G, Chambery A, Russo R, Ruvo M, Ciana P, Maravigna L, Shaik J, Radaelli E, De Antonellis P, Tarantino D, Pirolli A, Ragno R, Zollo M, Stunnenberg HG, Mai A, Altucci L. RIP1–HAT1–SIRT Complex Identification and Targeting in Treatment and Prevention of Cancer. Clin Cancer Res 2018. [DOI: 10.1158/1078-0432.ccr-17-3081] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
23
|
Cicchillitti L, Manni I, Mancone C, Regazzo G, Spagnuolo M, Alonzi T, Carlomosti F, Dell'Anna ML, Dell'Omo G, Picardo M, Ciana P, Capogrossi MC, Tripodi M, Magenta A, Rizzo MG, Gurtner A, Piaggio G. The laminA/NF-Y protein complex reveals an unknown transcriptional mechanism on cell proliferation. Oncotarget 2018; 8:2628-2646. [PMID: 27793050 PMCID: PMC5356829 DOI: 10.18632/oncotarget.12914] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 10/10/2016] [Indexed: 12/02/2022] Open
Abstract
Lamin A is a component of the nuclear matrix that also controls proliferation by largely unknown mechanisms. NF-Y is a ubiquitous protein involved in cell proliferation composed of three subunits (-YA -YB -YC) all required for the DNA binding and transactivation activity. To get clues on new NF-Y partner(s) we performed a mass spectrometry screening of proteins that co-precipitate with the regulatory subunit of the complex, NF-YA. By this screening we identified lamin A as a novel putative NF-Y interactor. Co-immunoprecipitation experiments and confocal analysis confirmed the interaction between the two endogenous proteins. Interestingly, this association occurs on euchromatin regions, too. ChIP experiments demonstrate lamin A enrichment in several promoter regions of cell cycle related genes in a NF-Y dependent manner. Gain and loss of function experiments reveal that lamin A counteracts NF-Y transcriptional activity. Taking advantage of a recently generated transgenic reporter mouse, called MITO-Luc, in which an NF-Y–dependent promoter controls luciferase expression, we demonstrate that lamin A counteracts NF-Y transcriptional activity not only in culture cells but also in living animals. Altogether, our data demonstrate the occurrence of lamin A/NF-Y interaction and suggest a possible role of this protein complex in regulation of NF-Y function in cell proliferation.
Collapse
Affiliation(s)
- Lucia Cicchillitti
- Department of Research, Advanced Diagnostics and Technological Innovation, SAFU Unit, Translational Research Area, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Isabella Manni
- Department of Research, Advanced Diagnostics and Technological Innovation, SAFU Unit, Translational Research Area, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Carmine Mancone
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Department of Epidemiology and Preclinical Research, 00149 Rome, Italy.,Department of Cellular Biotechnologies and Haematology, Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy
| | - Giulia Regazzo
- Department of Research, Advanced Diagnostics and Technological Innovation, Genomic and Epigenetic Unit, Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy
| | - Manuela Spagnuolo
- Department of Research, Advanced Diagnostics and Technological Innovation, Genomic and Epigenetic Unit, Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy
| | - Tonino Alonzi
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Department of Epidemiology and Preclinical Research, 00149 Rome, Italy
| | - Fabrizio Carlomosti
- Fondazione Luigi Maria Monti, Istituto Dermopatico dell'Immacolata-IRCCS, Laboratorio di Patologia Vascolare, 00167 Rome, Italy
| | - Maria Lucia Dell'Anna
- Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute, IRCCS, 00144 Rome, Italy
| | - Giulia Dell'Omo
- Department of Oncology and Hemato-Oncology and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Mauro Picardo
- Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute, IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Paolo Ciana
- Center of Excellence on Neurodegenerative Diseases, Department of Oncology and Hemato-Oncology, University of Milan, 20133 Milan, Italy
| | - Maurizio C Capogrossi
- Fondazione Luigi Maria Monti, Istituto Dermopatico dell'Immacolata-IRCCS, Laboratorio di Patologia Vascolare, Via dei Monti di Creta 104, Rome 00167, Italy Rome, Italy
| | - Marco Tripodi
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Department of Epidemiology and Preclinical Research, 00149 Rome, Italy.,Department of Cellular Biotechnologies and Haematology, Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessandra Magenta
- Fondazione Luigi Maria Monti, Istituto Dermopatico dell'Immacolata-IRCCS, Laboratorio di Patologia Vascolare, Via dei Monti di Creta 104, Rome 00167, Italy Rome, Italy
| | - Maria Giulia Rizzo
- Department of Research, Advanced Diagnostics and Technological Innovation, Genomic and Epigenetic Unit, Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy
| | - Aymone Gurtner
- Department of Research, Advanced Diagnostics and Technological Innovation, SAFU Unit, Translational Research Area, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Giulia Piaggio
- Department of Research, Advanced Diagnostics and Technological Innovation, SAFU Unit, Translational Research Area, Regina Elena National Cancer Institute, 00144 Rome, Italy
| |
Collapse
|
24
|
Rizzi N, Rebecchi M, Levandis G, Ciana P, Maggi A. Identification of novel loci for the generation of reporter mice. Nucleic Acids Res 2017; 45:e37. [PMID: 27899606 PMCID: PMC5389565 DOI: 10.1093/nar/gkw1142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/08/2016] [Indexed: 12/25/2022] Open
Abstract
Deciphering the etiology of complex pathologies at molecular level requires longitudinal studies encompassing multiple biochemical pathways (apoptosis, proliferation, inflammation, oxidative stress). In vivo imaging of current reporter animals enabled the spatio-temporal analysis of specific molecular events, however, the lack of a multiplicity of loci for the generalized and regulated expression of the integrated transgenes hampers the creation of systems for the simultaneous analysis of more than a biochemical pathways at the time. We here developed and tested an in vivo-based methodology for the identification of multiple insertional loci suitable for the generation of reliable reporter mice. The validity of the methodology was tested with the generation of novel mice useful to report on inflammation and oxidative stress.
Collapse
Affiliation(s)
- Nicoletta Rizzi
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9 20133 Milan, Italy
| | - Monica Rebecchi
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9 20133 Milan, Italy
| | - Giovanna Levandis
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9 20133 Milan, Italy
| | - Paolo Ciana
- Center of Excellence on Neurodegenerative Diseases and Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Via Balzaretti 9 20133 Milan, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9 20133 Milan, Italy
| |
Collapse
|
25
|
Novel Locally Active Estrogens Accelerate Cutaneous Wound Healing-Part 2. Sci Rep 2017; 7:2510. [PMID: 28566747 PMCID: PMC5451472 DOI: 10.1038/s41598-017-02820-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/19/2017] [Indexed: 11/19/2022] Open
Abstract
Estrogen deprivation is associated with delayed healing, while estrogen replacement therapy (ERT) accelerates acute wound healing and protects against development of chronic wounds. However, current estrogenic molecules have undesired systemic effects, thus the aim of our studies is to generate new molecules for topic administration that are devoid of systemic effects. Following a preliminary study, the new 17β-estradiol derivatives 1 were synthesized. The estrogenic activity of these novel compounds was evaluated in vitro using the cell line ERE-Luc B17 stably transfected with an ERE-Luc reporter. Among the 17β-estradiol derivatives synthesized, compounds 1e and 1f showed the highest transactivation potency and were therefore selected for the study of their systemic estrogenic activity. The study of these compounds in the ERE-Luc mouse model demonstrated that both compounds lack systemic effects when administered in the wound area. Furthermore, wound-healing experiments showed that 1e displays a significant regenerative and anti-inflammatory activity. It is therefore confirmed that this class of compounds are suitable for topical administration and have a clear beneficial effect on wound healing.
Collapse
|
26
|
Manni I, Di Rocco G, Fusco S, Leone L, Barbati SA, Carapella CM, Grassi C, Piaggio G, Toietta G. Monitoring the Response of Hyperbilirubinemia in the Mouse Brain by In Vivo Bioluminescence Imaging. Int J Mol Sci 2016; 18:50. [PMID: 28036021 PMCID: PMC5297685 DOI: 10.3390/ijms18010050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 12/18/2016] [Accepted: 12/22/2016] [Indexed: 01/02/2023] Open
Abstract
Increased levels of unconjugated bilirubin are neurotoxic, but the mechanism leading to neurological damage has not been completely elucidated. Innovative strategies of investigation are needed to more precisely define this pathological process. By longitudinal in vivo bioluminescence imaging, we noninvasively visualized the brain response to hyperbilirubinemia in the MITO-Luc mouse, in which light emission is restricted to the regions of active cell proliferation. We assessed that acute hyperbilirubinemia promotes bioluminescence in the brain region, indicating an increment in the cell proliferation rate. Immunohistochemical detection in brain sections of cells positive for both luciferase and the microglial marker allograft inflammatory factor 1 suggests proliferation of microglial cells. In addition, we demonstrated that brain induction of bioluminescence was altered by pharmacological displacement of bilirubin from its albumin binding sites and by modulation of the blood-brain barrier permeability, all pivotal factors in the development of bilirubin-induced neurologic dysfunction. We also determined that treatment with minocycline, an antibiotic with anti-inflammatory and neuroprotective properties, or administration of bevacizumab, an anti-vascular endothelial growth factor antibody, blunts bilirubin-induced bioluminescence. Overall the study supports the use of the MITO-Luc mouse as a valuable tool for the rapid response monitoring of drugs aiming at preventing acute bilirubin-induced neurological dysfunction.
Collapse
Affiliation(s)
- Isabella Manni
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy.
| | - Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy.
| | - Salvatore Fusco
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Lucia Leone
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Saviana Antonella Barbati
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | | | - Claudio Grassi
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Giulia Piaggio
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy.
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy.
| |
Collapse
|
27
|
Gurtner A, Manni I, Piaggio G. NF-Y in cancer: Impact on cell transformation of a gene essential for proliferation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1860:604-616. [PMID: 27939755 DOI: 10.1016/j.bbagrm.2016.12.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 12/17/2022]
Abstract
NF-Y is a ubiquitous heterotrimeric transcription factor with a binding affinity for the CCAAT consensus motif, one of the most common cis-acting element in the promoter and enhancer regions of eukaryote genes in direct (CCAAT) or reverse (ATTGG) orientation. NF-Y consists of three subunits, NF-YA, the regulatory subunit of the trimer, NF-YB, and NF-YC, all required for CCAAT binding. Growing evidence in cells and animal models support the notion that NF-Y, driving transcription of a plethora of cell cycle regulatory genes, is a key player in the regulation of proliferation. Proper control of cellular growth is critical for cancer prevention and uncontrolled proliferation is a hallmark of cancer cells. Indeed, during cell transformation aberrant molecular pathways disrupt mechanisms controlling proliferation and many growth regulatory genes are altered in tumors. Here, we review bioinformatics, molecular and functional evidence indicating the involvement of the cell cycle regulator NF-Y in cancer-associated pathways. This article is part of a Special Issue entitled: Nuclear Factor Y in Development and Disease, edited by Prof. Roberto Mantovani.
Collapse
Affiliation(s)
- Aymone Gurtner
- Department of Research, Advanced Diagnostics and Technological Innovation, UOSD SAFU, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Isabella Manni
- Department of Research, Advanced Diagnostics and Technological Innovation, UOSD SAFU, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Giulia Piaggio
- Department of Research, Advanced Diagnostics and Technological Innovation, UOSD SAFU, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| |
Collapse
|
28
|
de Latouliere L, Manni I, Iacobini C, Pugliese G, Grazi GL, Perri P, Cappello P, Novelli F, Menini S, Piaggio G. A bioluminescent mouse model of proliferation to highlight early stages of pancreatic cancer: A suitable tool for preclinical studies. Ann Anat 2016; 207:2-8. [DOI: 10.1016/j.aanat.2015.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/16/2015] [Accepted: 11/17/2015] [Indexed: 01/23/2023]
|
29
|
Principi E, Girardello R, Bruno A, Manni I, Gini E, Pagani A, Grimaldi A, Ivaldi F, Congiu T, De Stefano D, Piaggio G, de Eguileor M, Noonan DM, Albini A. Systemic distribution of single-walled carbon nanotubes in a novel model: alteration of biochemical parameters, metabolic functions, liver accumulation, and inflammation in vivo. Int J Nanomedicine 2016; 11:4299-316. [PMID: 27621623 PMCID: PMC5012628 DOI: 10.2147/ijn.s109950] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The increasing use of carbon nanotubes (CNTs) in several industrial applications raises concerns on their potential toxicity due to factors such as tissue penetrance, small dimensions, and biopersistence. Using an in vivo model for CNT environmental exposure, mimicking CNT exposition at the workplace, we previously found that CNTs rapidly enter and disseminate in the organism, initially accumulating in the lungs and brain and later reaching the liver and kidneys via the bloodstream in CD1 mice. Here, we monitored and traced the accumulation of single-walled CNTs (SWCNTs), administered systemically in mice, in different organs and the subsequent biological responses. Using the novel in vivo model, MITO-Luc bioluminescence reporter mice, we found that SWCNTs induce systemic cell proliferation, indicating a dynamic response of cells of both bone marrow and the immune system. We then examined metabolic (water/food consumption and dejections), functional (serum enzymes), and morphological (organs and tissues) alterations in CD1 mice treated with SWCNTs, using metabolic cages, performing serum analyses, and applying histological, immunohistochemical, and ultrastructural (transmission electron microscopy) methods. We observed a transient accumulation of SWCNTs in the lungs, spleen, and kidneys of CD1 mice exposed to SWCNTs. A dose- and time-dependent accumulation was found in the liver, associated with increases in levels of aspartate aminotransferase, alanine aminotransferase and bilirubinemia, which are metabolic markers associated with liver damage. Our data suggest that hepatic accumulation of SWCNTs associated with liver damage results in an M1 macrophage-driven inflammation.
Collapse
Affiliation(s)
- Elisa Principi
- Vascular Biology and Angiogenesis, Scientific and Technology Pole, IRCCS MultiMedica, Milano
| | - Rossana Girardello
- Department of Biotechnology and Life Sciences, University of Insubria, Varese
| | - Antonino Bruno
- Vascular Biology and Angiogenesis, Scientific and Technology Pole, IRCCS MultiMedica, Milano
| | - Isabella Manni
- Department of Research, Advanced Diagnosis and Innovation, Regina Elena National Cancer Institute, Rome
| | - Elisabetta Gini
- Department of Biotechnology and Life Sciences, University of Insubria, Varese
| | - Arianna Pagani
- Vascular Biology and Angiogenesis, Scientific and Technology Pole, IRCCS MultiMedica, Milano
| | - Annalisa Grimaldi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese
| | - Federico Ivaldi
- Department of Neuroscience, Ophthalmology and Genetics, University of Genoa, Genoa
| | - Terenzio Congiu
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Daniela De Stefano
- Vascular Biology and Angiogenesis, Scientific and Technology Pole, IRCCS MultiMedica, Milano
| | - Giulia Piaggio
- Department of Research, Advanced Diagnosis and Innovation, Regina Elena National Cancer Institute, Rome
| | - Magda de Eguileor
- Department of Biotechnology and Life Sciences, University of Insubria, Varese
| | - Douglas M Noonan
- Vascular Biology and Angiogenesis, Scientific and Technology Pole, IRCCS MultiMedica, Milano; Department of Biotechnology and Life Sciences, University of Insubria, Varese
| | - Adriana Albini
- Vascular Biology and Angiogenesis, Scientific and Technology Pole, IRCCS MultiMedica, Milano
| |
Collapse
|
30
|
Basile V, Baruffaldi F, Dolfini D, Belluti S, Benatti P, Ricci L, Artusi V, Tagliafico E, Mantovani R, Molinari S, Imbriano C. NF-YA splice variants have different roles on muscle differentiation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:627-38. [PMID: 26921500 DOI: 10.1016/j.bbagrm.2016.02.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 11/19/2022]
Abstract
The heterotrimeric CCAAT-binding factor NF-Y controls the expression of a multitude of genes involved in cell cycle progression. NF-YA is present in two alternatively spliced isoforms, NF-YAs and NF-YAl, differing in 28 aminoacids in the N-terminal Q-rich activation domain. NF-YAs has been identified as a regulator of stemness and proliferation in mouse embryonic cells (mESCs) and human hematopoietic stem cells (hHSCs), whereas the role of NF-YAl is not clear. In the muscle system, NF-YA expression is observed in proliferating cells, but barely detectable in terminally differentiated cells in vitro and adult skeletal muscle in vivo. Here, we show that NF-YA inactivation in mouse myoblasts impairs both proliferation and differentiation. The overexpression of the two NF-YA isoforms differentially affects myoblasts fate: NF-YAs enhance cell proliferation, while NF-YAl boosts differentiation. The molecular mechanisms were investigated by expression profilings, detailing the opposite programs of the two isoforms. Bioinformatic analysis of the regulated promoters failed to detect a significant presence of CCAAT boxes in the regulated genes. NF-YAl activates directly Mef2D, Six genes, and p57kip2 (Cdkn1c), and indirectly the myogenic regulatory factors (MRFs). Specifically, Cdkn1c activation is induced by NF-Y binding to its CCAAT promoter and by reducing the expression of the lncRNA Kcnq1ot1, a negative regulator of Cdkn1c transcription. Overall, our results indicate that NF-YA alternative splicing is an influential muscle cell determinant, through direct regulation of selected cell cycle blocking genes, and, directly and indirectly, of muscle-specific transcription factors.
Collapse
Affiliation(s)
- Valentina Basile
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Via Campi 213/D e Via Campi 287, Modena, Italy
| | - Fiorenza Baruffaldi
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Via Campi 213/D, 41125 Modena, Italy
| | - Diletta Dolfini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy
| | - Silvia Belluti
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Via Campi 213/D e Via Campi 287, Modena, Italy
| | - Paolo Benatti
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Via Campi 213/D e Via Campi 287, Modena, Italy
| | - Laura Ricci
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Via Campi 213/D e Via Campi 287, Modena, Italy
| | - Valentina Artusi
- Centro di Ricerche Genomiche, Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| | - Enrico Tagliafico
- Centro di Ricerche Genomiche, Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| | - Roberto Mantovani
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy
| | - Susanna Molinari
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Via Campi 213/D, 41125 Modena, Italy.
| | - Carol Imbriano
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Via Campi 213/D e Via Campi 287, Modena, Italy.
| |
Collapse
|
31
|
Rizzi N, Manni I, Vantaggiato C, Delledonne GA, Gentileschi MP, Maggi A, Piaggio G, Ciana P. In VivoImaging of Cell Proliferation for a Dynamic, Whole Body, Analysis of Undesired Drug Effects. Toxicol Sci 2015; 145:296-306. [DOI: 10.1093/toxsci/kfv056] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
32
|
Lu YH, Dallner OS, Birsoy K, Fayzikhodjaeva G, Friedman JM. Nuclear Factor-Y is an adipogenic factor that regulates leptin gene expression. Mol Metab 2015; 4:392-405. [PMID: 25973387 PMCID: PMC4420997 DOI: 10.1016/j.molmet.2015.02.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/05/2015] [Indexed: 12/22/2022] Open
Abstract
Objective Leptin gene expression is highly correlated with cellular lipid content in adipocytes but the transcriptional mechanisms controlling leptin expression in vivo are poorly understood. In this report, we set out to identify cis- and trans-regulatory elements controlling leptin expression. Methods Leptin-BAC luciferase transgenic mice combining with other computational and molecular techniques were used to identify transcription regulatory elements including a CCAAT-binding protein Nuclear Factor Y (NF-Y). The function of NF-Y in adipocyte was studied in vitro with 3T3-L1 cells and in vivo with adipocyte-specific knockout of NF-Y. Results Using Leptin-BAC luciferase mice, we showed that DNA sequences between −22 kb and +8.8 kb can confer quantitative expression of a leptin reporter. Computational analysis of sequences and gel shift assays identified a 32 bp sequence (chr6: 28993820–2899385) consisting a CCAAT binding site for Nuclear Factor Y (NF-Y) and this was confirmed by a ChIP assay in vivo. A deletion of this 32 bp sequence in the −22 kb to +8.8 kb leptin-luciferase BAC reporter completely abrogates luciferase reporter activity in vivo. RNAi mediated knockdown of NF-Y interfered with adipogenesis in vitro and adipocyte-specific knockout of NF-Y in mice reduced expression of leptin and other fat specific genes in vivo. Further analyses of the fat specific NF-Y knockout revealed that these animals develop a moderately severe lipodystrophy that is remediable with leptin therapy. Conclusions These studies advance our understanding of leptin gene expression and show that NF-Y controls the expression of leptin and other adipocyte genes and identifies a new form of lipodystrophy.
Collapse
Affiliation(s)
- Yi-Hsueh Lu
- Laboratory of Molecular Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Olof Stefan Dallner
- Laboratory of Molecular Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Kivanc Birsoy
- Laboratory of Molecular Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Gulya Fayzikhodjaeva
- Laboratory of Molecular Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA ; Howard Hughes Medical Institute, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| |
Collapse
|
33
|
Courties G, Herisson F, Sager HB, Heidt T, Ye Y, Wei Y, Sun Y, Severe N, Dutta P, Scharff J, Scadden DT, Weissleder R, Swirski FK, Moskowitz MA, Nahrendorf M. Ischemic stroke activates hematopoietic bone marrow stem cells. Circ Res 2015; 116:407-17. [PMID: 25362208 PMCID: PMC4312511 DOI: 10.1161/circresaha.116.305207] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 10/30/2014] [Indexed: 01/07/2023]
Abstract
RATIONALE The mechanisms leading to an expanded neutrophil and monocyte supply after stroke are incompletely understood. OBJECTIVE To test the hypothesis that transient middle cerebral artery occlusion (tMCAO) in mice leads to activation of hematopoietic bone marrow stem cells. METHODS AND RESULTS Serial in vivo bioluminescence reporter gene imaging in mice with tMCAO revealed that bone marrow cell cycling peaked 4 days after stroke (P<0.05 versus pre tMCAO). Flow cytometry and cell cycle analysis showed activation of the entire hematopoietic tree, including myeloid progenitors. The cycling fraction of the most upstream hematopoietic stem cells increased from 3.34%±0.19% to 7.32%±0.52% after tMCAO (P<0.05). In vivo microscopy corroborated proliferation of adoptively transferred hematopoietic progenitors in the bone marrow of mice with stroke. The hematopoietic system's myeloid bias was reflected by increased expression of myeloid transcription factors, including PU.1 (P<0.05), and by a decline in lymphocyte precursors. In mice after tMCAO, tyrosine hydroxylase levels in sympathetic fibers and bone marrow noradrenaline levels rose (P<0.05, respectively), associated with a decrease of hematopoietic niche factors that promote stem cell quiescence. In mice with genetic deficiency of the β3 adrenergic receptor, hematopoietic stem cells did not enter the cell cycle in increased numbers after tMCAO (naive control, 3.23±0.22; tMCAO, 3.74±0.33, P=0.51). CONCLUSIONS Ischemic stroke activates hematopoietic stem cells via increased sympathetic tone, leading to a myeloid bias of hematopoiesis and higher bone marrow output of inflammatory Ly6C(high) monocytes and neutrophils.
Collapse
Affiliation(s)
- Gabriel Courties
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Fanny Herisson
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Hendrik B Sager
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Timo Heidt
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Yuxiang Ye
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Ying Wei
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Yuan Sun
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Nicolas Severe
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Partha Dutta
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Jennifer Scharff
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - David T Scadden
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Ralph Weissleder
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Filip K Swirski
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Michael A Moskowitz
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Matthias Nahrendorf
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (G.C., H.B.S., T.H., Y.Y., Y.S., P.D., J.S., R.W., F.K.S., M.N.); Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown (F.H., Y.W., M.A.M.); Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.).
| |
Collapse
|
34
|
Spallotta F, Cencioni C, Straino S, Nanni S, Rosati J, Artuso S, Manni I, Colussi C, Piaggio G, Martelli F, Valente S, Mai A, Capogrossi MC, Farsetti A, Gaetano C. A nitric oxide-dependent cross-talk between class I and III histone deacetylases accelerates skin repair. J Biol Chem 2013; 288:11004-12. [PMID: 23463510 DOI: 10.1074/jbc.m112.441816] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In a mouse model of skin repair we found that the class I-IIa histone deacetylase inhibitor trichostatin A accelerated tissue regeneration. Unexpectedly, this effect was suppressed by Sirtinol, a class III histone deacetylase (HDAC) (sirtuin)-selective inhibitor. The role of sirtuins (SIRTs) was then investigated by using resveratrol and a novel SIRT1-2-3 activator, the MC2562 compound we synthesized recently. Both resveratrol and MC2562 were effective in accelerating wound repair. The local administration of natural or synthetic SIRT activators, in fact, significantly accelerated skin regeneration by increasing keratinocyte proliferation. In vitro experiments revealed that the activation of SIRTs stimulated keratinocyte proliferation via endothelial NO synthase phosphorylation and NO production. In this condition, the class I member HDAC2 was found S-nitrosylated on cysteine, a post-transduction modification associated with loss of activity and DNA binding capacity. After deacetylase inhibitor or SIRT activator treatment, ChIP showed, in fact, a significant HDAC2 detachment from the promoter region of insulin growth factor I (IGF-I), fibroblast growth factor 10 (FGF-10), and Epithelial Growth Factor (EGF), which may be the final recipients and effectors of the SIRT-NO-HDAC signaling cascade. Consistently, the effect of SIRT activators was reduced in the presence of NG-nitro-L-arginine methyl ester (L-NAME), a general inhibitor of NO synthesis. In conclusion, the NO-dependent cross-talk among class III and I histone deacetylases suggests an unprecedented signaling pathway important for skin repair.
Collapse
Affiliation(s)
- Francesco Spallotta
- Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino, Istituto di Ricerca e Cura a Carattere Scientifico, 20138 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Oliva P, Roncoroni C, Radaelli E, Brunialti E, Rizzi N, De Maglie M, Scanziani E, Piaggio G, Ciana P, Komm B, Maggi A. Global Profiling of TSEC Proliferative Potential by the Use of a Reporter Mouse for Proliferation. Reprod Sci 2012; 20:119-28. [DOI: 10.1177/1933719111431002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Paolo Oliva
- TOP srl, Transgenic Operative Products, Lodi, Italy
| | | | - Enrico Radaelli
- DIPAV, Faculty of Veterinary Medicine, University of Milan, Milan, Italy
| | | | | | - Marcella De Maglie
- TOP srl, Transgenic Operative Products, Lodi, Italy
- DIPAV, Faculty of Veterinary Medicine, University of Milan, Milan, Italy
| | | | - Giulia Piaggio
- Experimental Oncology Department, Istituto Regina Elena, IRCCS, Rome, Italy
| | - Paolo Ciana
- Department of Pharmacological Sciences, University of Milan, Milan, Italy
| | - Barry Komm
- Pfizer Inc. (B.K.), Collegeville, Pennsylvania 19426, USA
| | - Adriana Maggi
- Department of Pharmacological Sciences, University of Milan, Milan, Italy
| |
Collapse
|