1
|
Kong W, Biswas A, Zhou D, Fiches G, Fujinaga K, Santoso N, Zhu J. Nucleolar protein NOP2/NSUN1 suppresses HIV-1 transcription and promotes viral latency by competing with Tat for TAR binding and methylation. PLoS Pathog 2020; 16:e1008430. [PMID: 32176734 PMCID: PMC7098636 DOI: 10.1371/journal.ppat.1008430] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/26/2020] [Accepted: 02/24/2020] [Indexed: 01/11/2023] Open
Abstract
Recent efforts have been paid to identify previously unrecognized HIV-1 latency-promoting genes (LPGs) that can potentially be targeted for eradication of HIV-1 latent reservoirs. From our earlier orthologous RNAi screens of host factors regulating HIV-1 replication, we identified that the nucleolar protein NOP2/NSUN1, a m5C RNA methyltransferase (MTase), is an HIV-1 restriction factor. Loss- and gain-of-function analyses confirmed that NOP2 restricts HIV-1 replication. Depletion of NOP2 promotes the reactivation of latently infected HIV-1 proviruses in multiple cell lines as well as primary CD4+ T cells, alone or in combination with latency-reversing agents (LRAs). Mechanistically, NOP2 associates with HIV-1 5' LTR, interacts with HIV-1 TAR RNA by competing with HIV-1 Tat protein, as well as contributes to TAR m5C methylation. RNA MTase catalytic domain (MTD) of NOP2 mediates its competition with Tat and binding with TAR. Overall, these findings verified that NOP2 suppresses HIV-1 transcription and promotes viral latency.
Collapse
Affiliation(s)
- Weili Kong
- Department of Pathology, Ohio State University College of Medicine, Columbus, Ohio, United States of America
- Gladstone Institute of Virology and Immunology, University of California, San Francisco, California, United States of America
| | - Ayan Biswas
- Department of Pathology, Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Dawei Zhou
- Department of Pathology, Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Guillaume Fiches
- Department of Pathology, Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Koh Fujinaga
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Netty Santoso
- Department of Pathology, Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Jian Zhu
- Department of Pathology, Ohio State University College of Medicine, Columbus, Ohio, United States of America
| |
Collapse
|
2
|
|
3
|
Ma Y, Zhao W, Shi C, Wang N, Fan T. Effects of HIV on metabolic and biological pathways of CD4 + T lymphocytes. Exp Ther Med 2018; 15:2946-2950. [PMID: 29456700 PMCID: PMC5795520 DOI: 10.3892/etm.2018.5749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/12/2017] [Indexed: 11/23/2022] Open
Abstract
The effects of human immunodeficiency virus (HIV) on the metabolic and biological pathways of cluster of differentiation (CD)4+ T lymphocytes were investigated. A total of 150 patients with acquired immune deficiency syndrome (AIDS) and 50 healthy individuals who were admitted to hospital for physical examination during the period of June 2016 to January 2017, were selected as subjects in the present study. According to the virus load, 150 AIDS patients were divided into three groups: i) Viral load >106 copies/ml (group A, n=39), ii) 104 copies/ml < viral load <105 copies/ml (group B, n=76), and iii) viral load <104 copies/ml (group C, n=35). The relationship between viral loads in the three groups and CD4+ T lymphocyte counts was assessed. Active lymphocytes were isolated from T lymphocytes in the subjects, and the ratio of Th1 to Th2 was measured by flow cytometry. Effects of HIV on human T-lymphocyte differentiation were observed. Differences in T-lymphocyte metabolites were detected by proton nuclear magnetic resonance and their biological pathways analyzed. The results showed that CD4+ T-cell counts were decreased with the increase of the viral loads of patients. The viral loads of AIDS patients differentiated T lymphocytes. In other words, high viral loads accelerated the differentiation of T lymphocytes into Th1 cells. In the high HIV viral load group, the levels of glycerol phosphodiesterase, 7-dehydrocholesterol, p-hydroxyphenylacetic acid, cholesterol and deoxyuridine were increased, but the levels of 3-methoxytyramine, cytidine deaminase, deoxycorticosterone and 3-hydroxybutyric acid were decreased. The viral loads of AIDS patients are associated with CD4+ T-cell counts and the ratio of CD4+ T to CD8+ T cells. At the same time, HIV viral loads can affect the lipid biosynthesis of T-lymphocyte membranes, thus affecting the differentiation and proliferation of T lymphocytes and finally intervening its mediated immune responses.
Collapse
Affiliation(s)
- Yanli Ma
- Department of Infectious Diseases, Qingdao Sixth People's Hospital, Qingdao, Shandong 266033, P.R. China
| | - Wenge Zhao
- Department of Liver Diseases, Qingdao Sixth People's Hospital, Qingdao, Shandong 266033, P.R. China
| | - Changhe Shi
- Department of Liver Diseases, Qingdao Sixth People's Hospital, Qingdao, Shandong 266033, P.R. China
| | - Ning Wang
- Department of Internal Medicine, Postgraduate School of Qingdao University, Qingdao, Shandong 266033, P.R. China
| | - Tianli Fan
- Department of Infectious Diseases, Qingdao Sixth People's Hospital, Qingdao, Shandong 266033, P.R. China
| |
Collapse
|
4
|
Ne E, Palstra RJ, Mahmoudi T. Transcription: Insights From the HIV-1 Promoter. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 335:191-243. [DOI: 10.1016/bs.ircmb.2017.07.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
5
|
Ao M, Pan Z, Qian Y, Tang B, Feng Z, Fang H, Wu Z, Chen J, Xue Y, Fang M. Design, synthesis, and biological evaluation of AV6 derivatives as novel dual reactivators of latent HIV-1. RSC Adv 2018; 8:17279-17292. [PMID: 35539279 PMCID: PMC9080425 DOI: 10.1039/c8ra01216d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/24/2018] [Indexed: 01/31/2023] Open
Abstract
The “shock and kill” strategy might be a promising therapeutic approach for HIV/AIDS due to the existence of latent viral reservoirs. A major challenge of the “shock and kill” strategy arises from the general lack of clinically effective latency-reversing agents (LRAs). The 2-methylquinoline derivative, antiviral 6 (AV6) has been reported to induce latent HIV-1 expression and act synergistically with a HDAC inhibitor VA to reverse HIV latency. We report herein the design and identification of AV6 analogues which possess the zinc-binding group of HDAC inhibitors and have dual acting mechanism for the reactivation of HIV-1 from latency. Evaluation of compounds for the reactivation of HIV-1 latency identified two excellent active compounds 12c and 12d. Further bioassays revealed that these two compounds reactivated latent HIV-1 through dual mechanism, the inhibition of HDACs and NFAT-required for early HIV-1 gene expression. Additionally, it was found that 12c and 12d could reactivate HIV-1 transcription by releasing P-TEFb from the inactive complex 7SK snRNP. At last, molecular docking identified their orientation and binding interactions at the active site of HDAC2. This experimental data suggests that 12c and 12d can be served as effective HIV-1 LRAs which can be taken up for further studies. As dual-acting HIV LRAs, compounds 12c and 12d could activate latent HIV-1 via the NFAT-required mechanism and as histone deacetylase (HDAC) inhibitors.![]()
Collapse
|
6
|
The Multifaceted Contributions of Chromatin to HIV-1 Integration, Transcription, and Latency. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 328:197-252. [PMID: 28069134 DOI: 10.1016/bs.ircmb.2016.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The capacity of the human immunodeficiency virus (HIV-1) to establish latent infections constitutes a major barrier to the development of a cure for HIV-1. In latent infection, replication competent HIV-1 provirus is integrated within the host genome but remains silent, masking the infected cells from the activity of the host immune response. Despite the progress in elucidating the molecular players that regulate HIV-1 gene expression, the mechanisms driving the establishment and maintenance of latency are still not fully understood. Transcription from the HIV-1 genome occurs in the context of chromatin and is subjected to the same regulatory mechanisms that drive cellular gene expression. Much like in eukaryotic genes, the nucleosomal landscape of the HIV-1 promoter and its position within genomic chromatin are determinants of its transcriptional activity. Understanding the multilayered chromatin-mediated mechanisms that underpin HIV-1 integration and expression is of utmost importance for the development of therapeutic strategies aimed at reducing the pool of latently infected cells. In this review, we discuss the impact of chromatin structure on viral integration, transcriptional regulation and latency, and the host factors that influence HIV-1 replication by regulating chromatin organization. Finally, we describe therapeutic strategies under development to target the chromatin-HIV-1 interplay.
Collapse
|
7
|
White CH, Moesker B, Ciuffi A, Beliakova-Bethell N. Systems biology applications to study mechanisms of human immunodeficiency virus latency and reactivation. World J Clin Infect Dis 2016; 6:6-21. [DOI: 10.5495/wjcid.v6.i2.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/15/2016] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Eradication of human immunodeficiency virus (HIV) in infected individuals is currently not possible because of the presence of the persistent cellular reservoir of latent infection. The identification of HIV latency biomarkers and a better understanding of the molecular mechanisms contributing to regulation of HIV expression might provide essential tools to eliminate these latently infected cells. This review aims at summarizing gene expression profiling and systems biology applications to studies of HIV latency and eradication. Studies comparing gene expression in latently infected and uninfected cells identify candidate latency biomarkers and novel mechanisms of latency control. Studies that profiled gene expression changes induced by existing latency reversing agents (LRAs) highlight uniting themes driving HIV reactivation and novel mechanisms that contribute to regulation of HIV expression by different LRAs. Among the reviewed gene expression studies, the common approaches included identification of differentially expressed genes and gene functional category assessment. Integration of transcriptomic data with other biological data types is presently scarce, and the field would benefit from increased adoption of these methods in future studies. In addition, designing prospective studies that use the same methods of data acquisition and statistical analyses will facilitate a more reliable identification of latency biomarkers using different model systems and the comparison of the effects of different LRAs on host factors with a role in HIV reactivation. The results from such studies would have the potential to significantly impact the process by which candidate drugs are selected and combined for future evaluations and advancement to clinical trials.
Collapse
|
8
|
Vernon LT, Jayashantha P, Chidzonga MM, Komesu MC, Nair RG, Johnson NW. Comorbidities associated with HIV and antiretroviral therapy (clinical sciences): a workshop report. Oral Dis 2016; 22 Suppl 1:135-48. [PMID: 27109282 PMCID: PMC5986297 DOI: 10.1111/odi.12412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 10/23/2015] [Accepted: 10/24/2015] [Indexed: 12/23/2022]
Abstract
In the era of combination antiretroviral therapy (ART), parsing out the effects of HIV vs ART on health outcomes is challenging. Nadir CD4 count, a marker of the extent of immunosuppression, has significant long-term impact on an array of disease states in HIV+ persons; however, in the dental literature, reporting of pre-ART exposure to immunosuppression has largely been ignored and this limits the validity of previous studies. In Workshop A1, we explain fully the importance of nadir CD4, pre-ART immunosuppression, and identify a need to include specific variables in future research. The questions posed herein are challenging, typically not neatly addressed by any one study and require integration of the latest evidence from the wider medical literature. We consider topics beyond the confines of the oral cavity and examine oral health in the complex context of ART era HIV immunopathophysiology. We depict how variability in geographic setting and time period (pre- and post-ART era) can impact oral conditions - influencing when HIV infection was detected (at what CD4 count), the type and timing of ART as well as social determinants such as strong stigma and limited access to care. We hope our Workshop will stir debate and energize a rigorous focus on relevant areas of future research in HIV/AIDS.
Collapse
Affiliation(s)
- L T Vernon
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA
| | - Plp Jayashantha
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Qld, Australia and Dental Hospital, and Sri Lanka Air Force Station Colombo, Sri Lanka, Australia
| | - M M Chidzonga
- College of Health Sciences, University of Zimbabwe, Avondale, Harare, Zimbabwe
| | - M C Komesu
- Department of Morphology, Stomatology Physiology, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - R G Nair
- Oral Medicine, School of Dentistry and Oral Health, Griffith University, Gold Coast, Qld, Australia and Cancer Services, Gold Coast University Hospital, Queensland Health, Qld, Autralia, Australia
| | - N W Johnson
- Menzies Health Institute, Griffith University, Gold Coast, Qld, Australia
| |
Collapse
|
9
|
Trends in Antiviral Strategies. VIRUS AS POPULATIONS 2016. [PMCID: PMC7149557 DOI: 10.1016/b978-0-12-800837-9.00009-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Viral populations are true moving targets regarding the genomic sequences to be targeted in antiviral designs. Experts from different fields have expressed the need of new paradigms for antiviral interventions and viral disease control. This chapter reviews several strategies that aim at counteracting the adaptive capacity of viral quasispecies. The proposed designs are based on combinations of different antiviral drugs and immune modulators, or in the administration of virus-specific mutagenic agents, in an approach termed lethal mutagenesis of viruses. It consists of decreasing viral fitness by an excess of mutations that render viral proteins sub-optimal or non-functional. Viral extinction by lethal mutagenesis involves several sequential, overlapping steps that recapitulate the major concepts of intra-population interactions and genetic information stability discussed in preceding chapters. Despite the magnitude of the challenge, the chapter closes with some optimistic prospects for an effective control of viruses displaying error-prone replication, based on the combined targeting of replication fidelity and the induction of the innate immune response.
Collapse
|
10
|
Small Molecule Inhibitors of BAF; A Promising Family of Compounds in HIV-1 Latency Reversal. EBioMedicine 2015; 3:108-121. [PMID: 26870822 PMCID: PMC4739437 DOI: 10.1016/j.ebiom.2015.11.047] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 11/19/2015] [Accepted: 11/25/2015] [Indexed: 11/23/2022] Open
Abstract
Persistence of latently infected cells in presence of Anti-Retroviral Therapy presents the main obstacle to HIV-1 eradication. Much effort is thus placed on identification of compounds capable of HIV-1 latency reversal in order to render infected cells susceptible to viral cytopathic effects and immune clearance. We identified the BAF chromatin remodeling complex as a key player required for maintenance of HIV-1 latency, highlighting its potential as a molecular target for inhibition in latency reversal. Here, we screened a recently identified panel of small molecule inhibitors of BAF (BAFi's) for potential to activate latent HIV-1. Latency reversal was strongly induced by BAFi's Caffeic Acid Phenethyl Ester and Pyrimethamine, two molecules previously characterized for clinical application. BAFi's reversed HIV-1 latency in cell line based latency models, in two ex vivo infected primary cell models of latency, as well as in HIV-1 infected patient's CD4 + T cells, without inducing T cell proliferation or activation. BAFi-induced HIV-1 latency reversal was synergistically enhanced upon PKC pathway activation and HDAC-inhibition. Therefore BAFi's constitute a promising family of molecules for inclusion in therapeutic combinatorial HIV-1 latency reversal. BAF complex inhibitors (BAFi's) activate latent HIV-1 in cell line models of latency. BAFi's in combination with HDAC inhibitors and PKC activators synergistically activate latent HIV-1. The BAFi's PYR and CAPE reverse HIV-1 latency in primary cell models of latency and in cells obtained from HIV-1 patients.
Access to Combination antiretroviral therapy (cART) has made HIV-1 infection a chronic disease. However, cART is not curative, as a small number of infected cells harboring silent virus with potential to renew the infection persist despite cART. Efforts to cure HIV-1 include activation of these latent cells, making them susceptible to immune clearance. Here we describe the activity of BAF inhibitors in HIV-1 activation in in vitro models of HIV-1 latency as well as in cells obtained from HIV-1 infected patient volunteers. Our data highlight the clinical potential of BAF inhibitors for inclusion in combinatorial therapy to reverse HIV-1 latency.
Collapse
Key Words
- BAF complex
- BAF, BRG-Brahma Associated Factors
- BAF250a, BAF Associated Factor 250 a
- BAFi, BAF inhibitor
- BRG-1, Brahma Related Gene 1
- CAPE, caffeic acid phenetyl esther
- ChIP, Chromatin Immunoprecipitation
- Chromatin remodeling
- CycA, Cyclophilin A
- DHS-1, DNase Hypersensitive Site 1
- ES cells, embryonic stem cells
- FAIRE, Formaldehyde Assisted Isolation of Regulatory Elements
- FBS, Fetal Bovine Serum
- GFP, Green Fluorescent Protein
- HDAC, histone deacetylase
- HIV
- HIV-1, human immunodeficiency virus type 1
- IFNß, Interferon beta
- IL10, Interleukin 10
- IL12, Interleukin 12
- IL4, Interleukin 4
- IL6, Interleukin 6
- INI-1, Integrase Interactor 1
- IRES, Internal Ribosome Entry Site
- IκB-α, Inhibitor of NF-κB – alpha
- LRA, latency reversal agent
- LTR, Long Terminal Repeat
- Latency
- MIP26, Major Intrinsic Protein
- MMP9, Matrix Metallopeptidase 9
- NF-κB, Nuclear Factor Kappa-light-chain-enhancer of activated B cells
- PBMC, peripheral blood mononuclear cell
- PBS, Phosphate Buffered Saline
- PKC, Protein Kinase C
- PYR, Pyrimethamine
- RT-qPCR, Reverse Transcriptase, quantitative Polymerase Chain Reaction
- SAHA, Suberoylanilide Hydroxamic Acid
- SD, Standard Deviation
- SOCS3, Suppressor Of Cytokine Signaling 3
- TGF-ß, Transforming Growth Factor beta
- TLR2, Toll-like Receptor 2
- bp, base pairs
- cART, combination Antiretroviral Therapy
- latency reversal agents
- nuc, nucleosome
- siRNA, small interfering RNA
Collapse
|
11
|
EZH2 phosphorylation regulates Tat-induced HIV-1 transactivation via ROS/Akt signaling pathway. FEBS Lett 2015; 589:4106-11. [DOI: 10.1016/j.febslet.2015.11.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 10/27/2015] [Accepted: 11/16/2015] [Indexed: 12/25/2022]
|
12
|
Gómez CE, Perdiguero B, García-Arriaza J, Cepeda V, Sánchez-Sorzano CÓ, Mothe B, Jiménez JL, Muñoz-Fernández MÁ, Gatell JM, López Bernaldo de Quirós JC, Brander C, García F, Esteban M. A Phase I Randomized Therapeutic MVA-B Vaccination Improves the Magnitude and Quality of the T Cell Immune Responses in HIV-1-Infected Subjects on HAART. PLoS One 2015; 10:e0141456. [PMID: 26544853 PMCID: PMC4636254 DOI: 10.1371/journal.pone.0141456] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/08/2015] [Indexed: 11/29/2022] Open
Abstract
Trial Design Previous studies suggested that poxvirus-based vaccines might be instrumental in the therapeutic HIV field. A phase I clinical trial was conducted in HIV-1-infected patients on highly active antiretroviral therapy (HAART), with CD4 T cell counts above 450 cells/mm3 and undetectable viremia. Thirty participants were randomized (2:1) to receive either 3 intramuscular injections of MVA-B vaccine (coding for clade B HIV-1 Env, Gag, Pol and Nef antigens) or placebo, followed by interruption of HAART. Methods The magnitude, breadth, quality and phenotype of the HIV-1-specific T cell response were assayed by intracellular cytokine staining (ICS) in 22 volunteers pre- and post-vaccination. Results MVA-B vaccine induced newly detected HIV-1-specific CD4 T cell responses and expanded pre-existing responses (mostly against Gag, Pol and Nef antigens) that were high in magnitude, broadly directed and showed an enhanced polyfunctionality with a T effector memory (TEM) phenotype, while maintaining the magnitude and quality of the pre-existing HIV-1-specific CD8 T cell responses. In addition, vaccination also triggered preferential CD8+ T cell polyfunctional responses to the MVA vector antigens that increase in magnitude after two and three booster doses. Conclusion MVA-B vaccination represents a feasible strategy to improve T cell responses in individuals with pre-existing HIV-1-specific immunity. Trial Registration ClinicalTrials.gov NCT01571466
Collapse
Affiliation(s)
- Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Victoria Cepeda
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carlos Óscar Sánchez-Sorzano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Beatriz Mothe
- IrsiCaixa-HIVACAT, Hospital Universitari Germans Trias i Pujol, Autonomous University of Barcelona, Badalona, Spain
| | | | | | | | | | - Christian Brander
- IrsiCaixa-HIVACAT, Hospital Universitari Germans Trias i Pujol, Autonomous University of Barcelona, Badalona, Spain
- Institució Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, and University of Vic and Central Catalonia, Vic, Spain
| | | | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- * E-mail:
| |
Collapse
|