1
|
Khan T, McFall DJ, Hussain AI, Frayser LA, Casilli TP, Steck MC, Sanchez-Brualla I, Kuehn NM, Cho M, Barnes JA, Harris BT, Vicini S, Forcelli PA. Senescent cell clearance ameliorates temporal lobe epilepsy and associated spatial memory deficits in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605784. [PMID: 39211239 PMCID: PMC11360968 DOI: 10.1101/2024.07.30.605784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Current therapies for the epilepsies only treat the symptoms, but do not prevent epileptogenesis (the process in which epilepsy develops). Many cellular responses during epileptogenesis are also common hallmarks of cellular senescence , which halts proliferation of damaged cells. Clearing senescent cells (SCs) restores function in several age-associated and neurodegenerative disease models. It is unknown whether SC accumulation contributes to epileptogenesis and associated cognitive impairments. To address this question, we used a mouse model of temporal lobe epilepsy (TLE) and characterized the senescence phenotype throughout epileptogenesis. SCs accumulated 2 weeks after SE and were predominantly microglia. We ablated SCs and reduced (and in some cases prevented) the emergence of spontaneous seizures and normalized cognitive function in mice. Suggesting that this is a translationally-relevant target we also found SC accumulation in resected hippocampi from patients with TLE. These findings indicate that SC ablation after an epileptogenic insult is a potential anti-epileptogenic therapy.
Collapse
|
2
|
Luo X, Ruan Z, Liu L. Causal relationship between telomere length and epilepsy: A bidirectional Mendelian randomization study. Epilepsia Open 2023; 8:1432-1439. [PMID: 37593897 PMCID: PMC10690705 DOI: 10.1002/epi4.12817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023] Open
Abstract
OBJECTIVE Observational studies have suggested a link between telomere length (TL) and epilepsy, but the direction of the effect and whether it is causal or not is still being debated. The objective of this study was to investigate the causal relationship between TL and epilepsy using Mendelian randomization (MR) analysis. METHODS We performed a bidirectional two-sample MR analysis using pooled statistics from genome-wide association studies (GWAS) of TL and epilepsy. Additionally, we conducted a replication analysis using data from another GWAS study on epilepsy to validate our findings. The final results were analyzed using five MR methods, with the inverse-variance weighted (IVW) method as the primary outcome. We applied methods such as radial MR, MR pleiotropy residual and outlier test and MR Steiger filters to exclude outliers. Sensitivity analyses were also conducted to assess heterogeneity and pleiotropy. RESULTS Our analysis found no evidence of a causal relationship between epilepsy and TL (all p-values >0.05). The sensitivity analysis confirms the robustness of these results. SIGNIFICANCE In summary, our study contradicts existing observational reports by not finding any evidence to support a causal relationship between epilepsy and TL. Further research is necessary to determine the underlying mechanism behind the association observed in observational studies.
Collapse
Affiliation(s)
- Xinxin Luo
- Jiangxi Provincial People's Hospital and The First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Zhichao Ruan
- Beijing University of Chinese MedicineBeijingChina
| | - Ling Liu
- Jiangxi Provincial People's Hospital and The First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| |
Collapse
|
3
|
La Torre M, Centofante E, Nicoletti C, Burla R, Giampietro A, Cannistrà F, Schirone L, Valenti V, Sciarretta S, Musarò A, Saggio I. Impact of diffused versus vasculature targeted DNA damage on the heart of mice depleted of telomeric factor Ft1. Aging Cell 2023; 22:e14022. [PMID: 37960940 PMCID: PMC10726857 DOI: 10.1111/acel.14022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/27/2023] [Accepted: 10/11/2023] [Indexed: 11/15/2023] Open
Abstract
DNA damage is emerging as a driver of heart disease, although the cascade of events, its timing, and the cell types involved are yet to be fully clarified. In this context, the implication of cardiomyocytes has been highlighted, while that of vasculature smooth muscle cells has been implicated but not explored exhaustively. In our previous work we characterized a factor called Ft1 in mice and AKTIP in humans whose depletion generates telomere instability and DNA damage. Herein, we explored the effect of the reduction of Ft1 on the heart with the goal of comparatively defining the impact of DNA damage targeted to vasculature smooth muscle cells to that of diffuse damage. Using two newly generated mouse models, Ft1 constitutively knocked out (Ft1ko) mice, and mice in which we targeted the Ft1 depletion to the smooth muscle cells (Ft1sm22ko), it is shown that both genetic models display cardiac defects but with differences. Both Ft1ko and Ft1sm22ko mice display hypertrophy, fibrosis, and functional heart defects. Interestingly, Ft1sm22ko mice have early milder pathological traits that become manifest with age. Significantly, the defects of Ft1ko mice, including the alteration of the left ventricle and functional heart defects, are rescued by depletion of the DNA damage sensor p53. These results point to Ft1 deficiency as a driver of cardiac disease and show that Ft1 deficiency targeted to vasculature smooth muscle cells generates a pre-pathological profile exacerbated by age.
Collapse
Affiliation(s)
- Mattia La Torre
- Department Biology and Biotechnologies “Charles Darwin”Sapienza UniversityRomeItaly
| | - Eleonora Centofante
- Department Biology and Biotechnologies “Charles Darwin”Sapienza UniversityRomeItaly
| | - Carmine Nicoletti
- DAHFMO‐Unit of Histology and Medical EmbryologySapienza UniversityRomeItaly
- Istituto Pasteur Fondazione Cenci BolognettiRomeItaly
| | - Romina Burla
- Department Biology and Biotechnologies “Charles Darwin”Sapienza UniversityRomeItaly
- CNR Institute of Molecular Biology and PathologyRomeItaly
| | | | - Federica Cannistrà
- Department Biology and Biotechnologies “Charles Darwin”Sapienza UniversityRomeItaly
| | | | | | - Sebastiano Sciarretta
- IRCCS NeuromedPozzilli ISItaly
- Department Medical and Surgical Sciences and BiotechnologiesSapienza UniversityRomeItaly
| | - Antonio Musarò
- DAHFMO‐Unit of Histology and Medical EmbryologySapienza UniversityRomeItaly
- Istituto Pasteur Fondazione Cenci BolognettiRomeItaly
| | - Isabella Saggio
- Department Biology and Biotechnologies “Charles Darwin”Sapienza UniversityRomeItaly
- Istituto Pasteur Fondazione Cenci BolognettiRomeItaly
- CNR Institute of Molecular Biology and PathologyRomeItaly
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
- NISB Institute of Structural BiologyNanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
4
|
Krishnamoorthy S, Muruganantham B, Yu JR, Park WY, Muthusami S. Exploring the utility of FTS as a bonafide binding partner for EGFR: A potential drug target for cervical cancer. Comput Biol Med 2023; 167:107592. [PMID: 37976824 DOI: 10.1016/j.compbiomed.2023.107592] [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: 07/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 11/19/2023]
Abstract
Establishment of human papilloma virus (HPV) infection and its progression to cervical cancer (CC) requires the participation of epidermal growth factor (EGF) receptor (EGFR) and fused toes homolog (FTS). This review is an attempt to understand the structure-function relationship between FTS and EGFR as a tool for the development of newer CC drugs. Motif analysis was performed using national center for biotechnology information (NCBI), kyoto encyclopedia of genes and genomes (KEGG), simple modular architecture research tool (SMART) and multiple expectation maximizations for motif elicitation (MEME) database. The secondary and tertiary structure prediction of FTS was performed using DISOPRED3 and threading assembly, respectively. A positive correlation was found between the transcript levels of FTS and EGFR. Amino acids responsible for interaction between EGFR and FTS were determined. The nine micro-RNAs (miRNAs) that regulates the expression of FTS were predicted using Network Analyst 3.0 database. hsa-miR-629-5p and hsa-miR-615-3p are identified as significant positive and negative regulators of FTS gene expression. This review opens up new avenues for the development of CC drugs which interfere with the interaction between FTS and EGFR.
Collapse
Affiliation(s)
- Sneha Krishnamoorthy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India
| | - Bharathi Muruganantham
- Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India
| | - Jae-Ran Yu
- Department of Environmental and Tropical Medicine, Konkuk University College of Medicine, Chungju, South Korea
| | - Woo-Yoon Park
- Department of Radiation Oncology Hospital, College of Medicine, Chungbuk National University, Cheongju, South Korea.
| | - Sridhar Muthusami
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India; Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India.
| |
Collapse
|
5
|
La Torre M, Merigliano C, Maccaroni K, Chojnowski A, Goh WI, Giubettini M, Vernì F, Capanni C, Rhodes D, Wright G, Burke B, Soddu S, Burla R, Saggio I. Combined alteration of lamin and nuclear morphology influences the localization of the tumor-associated factor AKTIP. J Exp Clin Cancer Res 2022; 41:273. [PMID: 36096808 PMCID: PMC9469526 DOI: 10.1186/s13046-022-02480-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022] Open
Abstract
Background Lamins, key nuclear lamina components, have been proposed as candidate risk biomarkers in different types of cancer but their accuracy is still debated. AKTIP is a telomeric protein with the property of being enriched at the nuclear lamina. AKTIP has similarity with the tumor susceptibility gene TSG101. AKTIP deficiency generates genome instability and, in p53−/− mice, the reduction of the mouse counterpart of AKTIP induces the exacerbation of lymphomas. Here, we asked whether the distribution of AKTIP is altered in cancer cells and whether this is associated with alterations of lamins. Methods We performed super-resolution imaging, quantification of lamin expression and nuclear morphology on HeLa, MCF7, and A549 tumor cells, and on non-transformed fibroblasts from healthy donor and HGPS (LMNA c.1824C > T p.Gly608Gly) and EDMD2 (LMNA c.775 T > G) patients. As proof of principle model combining a defined lamin alteration with a tumor cell setting, we produced HeLa cells exogenously expressing the HGPS lamin mutant progerin that alters nuclear morphology. Results In HeLa cells, AKTIP locates at less than 0.5 µm from the nuclear rim and co-localizes with lamin A/C. As compared to HeLa, there is a reduced co-localization of AKTIP with lamin A/C in both MCF7 and A549. Additionally, MCF7 display lower amounts of AKTIP at the rim. The analyses in non-transformed fibroblasts show that AKTIP mislocalizes in HGPS cells but not in EDMD2. The integrated analysis of lamin expression, nuclear morphology, and AKTIP topology shows that positioning of AKTIP is influenced not only by lamin expression, but also by nuclear morphology. This conclusion is validated by progerin-expressing HeLa cells in which nuclei are morphologically altered and AKTIP is mislocalized. Conclusions Our data show that the combined alteration of lamin and nuclear morphology influences the localization of the tumor-associated factor AKTIP. The results also point to the fact that lamin alterations per se are not predictive of AKTIP mislocalization, in both non-transformed and tumor cells. In more general terms, this study supports the thesis that a combined analytical approach should be preferred to predict lamin-associated changes in tumor cells. This paves the way of next translational evaluation to validate the use of this combined analytical approach as risk biomarker. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02480-5.
Collapse
|
6
|
Bazhanova E, Kozlov A. Mechanisms of apoptosis in drug-resistant epilepsy. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:43-50. [DOI: 10.17116/jnevro202212205143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
7
|
Merigliano C, Burla R, La Torre M, Del Giudice S, Teo H, Liew CW, Chojnowski A, Goh WI, Olmos Y, Maccaroni K, Giubettini M, Chiolo I, Carlton JG, Raimondo D, Vernì F, Stewart CL, Rhodes D, Wright GD, Burke BE, Saggio I. AKTIP interacts with ESCRT I and is needed for the recruitment of ESCRT III subunits to the midbody. PLoS Genet 2021; 17:e1009757. [PMID: 34449766 PMCID: PMC8428793 DOI: 10.1371/journal.pgen.1009757] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/09/2021] [Accepted: 08/04/2021] [Indexed: 11/18/2022] Open
Abstract
To complete mitosis, the bridge that links the two daughter cells needs to be cleaved. This step is carried out by the endosomal sorting complex required for transport (ESCRT) machinery. AKTIP, a protein discovered to be associated with telomeres and the nuclear membrane in interphase cells, shares sequence similarities with the ESCRT I component TSG101. Here we present evidence that during mitosis AKTIP is part of the ESCRT machinery at the midbody. AKTIP interacts with the ESCRT I subunit VPS28 and forms a circular supra-structure at the midbody, in close proximity with TSG101 and VPS28 and adjacent to the members of the ESCRT III module CHMP2A, CHMP4B and IST1. Mechanistically, the recruitment of AKTIP is dependent on MKLP1 and independent of CEP55. AKTIP and TSG101 are needed together for the recruitment of the ESCRT III subunit CHMP4B and in parallel for the recruitment of IST1. Alone, the reduction of AKTIP impinges on IST1 and causes multinucleation. Our data altogether reveal that AKTIP is a component of the ESCRT I module and functions in the recruitment of ESCRT III components required for abscission. To complete cell division, the bridge that links the two daughter cells needs to be cleaved. This step is carried out by a machinery named “endosomal sorting complex required for transport” (ESCRT). The dissection of this machinery is important in basic biology and for investigating diseases in which cell division is altered. AKTIP, a factor discovered to be needed for chromosome integrity, shares similarities with a component of the ESCRT machinery named TSG101. Here we present evidence that AKTIP is part of the ESCRT machinery, as TSG101. More specifically, we show that AKTIP physically interacts with members of the ESCRT machinery and forms a characteristic circular structure at the center of the bridge linking the daughter cells. We also show that the reduction of AKTIP levels causes defects in the assembly of the ESCRT machinery and in cell division. In future work, it will be interesting to investigate the association of AKTIP with cancer, because in tumorigenesis cell division is altered and since an implication in cancer has been described for TSG101 and other ESCRT factors.
Collapse
Affiliation(s)
| | - Romina Burla
- Sapienza University Dept. Biology and Biotechnology, Rome, Italy
- CNR Institute of Molecular Biology and Pathology, Rome, Italy
| | - Mattia La Torre
- Sapienza University Dept. Biology and Biotechnology, Rome, Italy
| | | | - Hsiangling Teo
- Institute of Structural Biology, Nanyang Technological University, Singapore
| | - Chong Wai Liew
- Institute of Structural Biology, Nanyang Technological University, Singapore
| | - Alexandre Chojnowski
- A*STAR, Developmental and Regenerative Biology, ASLR, Agency for Science, Technology and Research, Singapore
- A*STAR, Singapore Nuclear Dynamics and Architecture, ASLR Skin Research Labs, Agency for Science, Technology and Research, Singapore
| | - Wah Ing Goh
- A*STAR Microscopy Platform, Research Support Centre, Agency for Science, Technology and Research, Singapore
| | - Yolanda Olmos
- School of Cancer and Pharmaceutical Sciences, King’s College London, London, United Kingdom
- Organelle Dynamics Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Klizia Maccaroni
- Sapienza University Dept. Biology and Biotechnology, Rome, Italy
| | | | - Irene Chiolo
- University of Southern California, Molecular and Computational Biology Dept., Los Angeles, California, United States of America
| | - Jeremy G. Carlton
- School of Cancer and Pharmaceutical Sciences, King’s College London, London, United Kingdom
- Organelle Dynamics Laboratory, The Francis Crick Institute, London, United Kingdom
| | | | - Fiammetta Vernì
- Sapienza University Dept. Biology and Biotechnology, Rome, Italy
| | - Colin L. Stewart
- A*STAR, Developmental and Regenerative Biology, ASLR, Agency for Science, Technology and Research, Singapore
- Dept. of Physiology National University of Singapore, Singapore
| | - Daniela Rhodes
- Institute of Structural Biology, Nanyang Technological University, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Graham D. Wright
- A*STAR Microscopy Platform, Research Support Centre, Agency for Science, Technology and Research, Singapore
| | - Brian E. Burke
- A*STAR, Singapore Nuclear Dynamics and Architecture, ASLR Skin Research Labs, Agency for Science, Technology and Research, Singapore
| | - Isabella Saggio
- Sapienza University Dept. Biology and Biotechnology, Rome, Italy
- CNR Institute of Molecular Biology and Pathology, Rome, Italy
- Institute of Structural Biology, Nanyang Technological University, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
- * E-mail:
| |
Collapse
|
8
|
Sikora E, Bielak-Zmijewska A, Dudkowska M, Krzystyniak A, Mosieniak G, Wesierska M, Wlodarczyk J. Cellular Senescence in Brain Aging. Front Aging Neurosci 2021; 13:646924. [PMID: 33732142 PMCID: PMC7959760 DOI: 10.3389/fnagi.2021.646924] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/02/2021] [Indexed: 12/25/2022] Open
Abstract
Aging of the brain can manifest itself as a memory and cognitive decline, which has been shown to frequently coincide with changes in the structural plasticity of dendritic spines. Decreased number and maturity of spines in aged animals and humans, together with changes in synaptic transmission, may reflect aberrant neuronal plasticity directly associated with impaired brain functions. In extreme, a neurodegenerative disease, which completely devastates the basic functions of the brain, may develop. While cellular senescence in peripheral tissues has recently been linked to aging and a number of aging-related disorders, its involvement in brain aging is just beginning to be explored. However, accumulated evidence suggests that cell senescence may play a role in the aging of the brain, as it has been documented in other organs. Senescent cells stop dividing and shift their activity to strengthen the secretory function, which leads to the acquisition of the so called senescence-associated secretory phenotype (SASP). Senescent cells have also other characteristics, such as altered morphology and proteostasis, decreased propensity to undergo apoptosis, autophagy impairment, accumulation of lipid droplets, increased activity of senescence-associated-β-galactosidase (SA-β-gal), and epigenetic alterations, including DNA methylation, chromatin remodeling, and histone post-translational modifications that, in consequence, result in altered gene expression. Proliferation-competent glial cells can undergo senescence both in vitro and in vivo, and they likely participate in neuroinflammation, which is characteristic for the aging brain. However, apart from proliferation-competent glial cells, the brain consists of post-mitotic neurons. Interestingly, it has emerged recently, that non-proliferating neuronal cells present in the brain or cultivated in vitro can also have some hallmarks, including SASP, typical for senescent cells that ceased to divide. It has been documented that so called senolytics, which by definition, eliminate senescent cells, can improve cognitive ability in mice models. In this review, we ask questions about the role of senescent brain cells in brain plasticity and cognitive functions impairments and how senolytics can improve them. We will discuss whether neuronal plasticity, defined as morphological and functional changes at the level of neurons and dendritic spines, can be the hallmark of neuronal senescence susceptible to the effects of senolytics.
Collapse
Affiliation(s)
- Ewa Sikora
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Anna Bielak-Zmijewska
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Magdalena Dudkowska
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Adam Krzystyniak
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Grazyna Mosieniak
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Malgorzata Wesierska
- Laboratory of Neuropsychology, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| |
Collapse
|
9
|
Burla R, La Torre M, Maccaroni K, Verni F, Giunta S, Saggio I. Interplay of the nuclear envelope with chromatin in physiology and pathology. Nucleus 2020; 11:205-218. [PMID: 32835589 PMCID: PMC7529417 DOI: 10.1080/19491034.2020.1806661] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 12/21/2022] Open
Abstract
The nuclear envelope compartmentalizes chromatin in eukaryotic cells. The main nuclear envelope components are lamins that associate with a panoply of factors, including the LEM domain proteins. The nuclear envelope of mammalian cells opens up during cell division. It is reassembled and associated with chromatin at the end of mitosis when telomeres tether to the nuclear periphery. Lamins, LEM domain proteins, and DNA binding factors, as BAF, contribute to the reorganization of chromatin. In this context, an emerging role is that of the ESCRT complex, a machinery operating in multiple membrane assembly pathways, including nuclear envelope reformation. Research in this area is unraveling how, mechanistically, ESCRTs link to nuclear envelope associated factors as LEM domain proteins. Importantly, ESCRTs work also during interphase for repairing nuclear envelope ruptures. Altogether the advances in this field are giving new clues for the interpretation of diseases implicating nuclear envelope fragility, as laminopathies and cancer. ABBREVIATIONS na, not analyzed; ko, knockout; kd, knockdown; NE, nuclear envelope; LEM, LAP2-emerin-MAN1 (LEM)-domain containing proteins; LINC, linker of nucleoskeleton and cytoskeleton complexes; Cyt, cytoplasm; Chr, chromatin; MB, midbody; End, endosomes; Tel, telomeres; INM, inner nuclear membrane; NP, nucleoplasm; NPC, Nuclear Pore Complex; ER, Endoplasmic Reticulum; SPB, spindle pole body.
Collapse
Affiliation(s)
- Romina Burla
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
- CNR Institute of Molecular Biology and Pathology, Italy
| | - Mattia La Torre
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Klizia Maccaroni
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Fiammetta Verni
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Simona Giunta
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
- Rockefeller University, New York, NY, USA
| | - Isabella Saggio
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
- CNR Institute of Molecular Biology and Pathology, Italy
- Institute of Structural Biology, School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
10
|
Telomere shortening in patients with drug-resistant epilepsy. Epilepsy Res 2020; 166:106427. [DOI: 10.1016/j.eplepsyres.2020.106427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 11/21/2022]
|