1,340 results on '"NUCLEAR EXPORT"'
Search Results
2. Nuclear export of PML promotes p53-mediated apoptosis and ferroptosis
- Author
-
Ni, Yue, Chen, Hongce, Zhan, Qiuqiang, and Zhuang, Zhengfei
- Published
- 2024
- Full Text
- View/download PDF
3. Mutations of Key Functional Residues in CRM1/XPO1 Differently Alter Its Intranuclear Localization and the Nuclear Export of Endogenous Cargos.
- Author
-
Omaetxebarria, Miren Josu, Sendino, Maria, Arrizabalaga, Liher, Mota, Irune, Zubiaga, Ana Maria, and Rodríguez, José Antonio
- Subjects
- *
MUTAGENESIS , *NUCLEOLUS , *FREIGHT & freightage , *CYTOPLASM , *PHOSPHORYLATION - Abstract
CRM1 (XPO1) has been well-characterized as a shuttling receptor that mediates the export of protein and RNA cargos to the cytoplasm, and previous analyses have pinpointed several key residues (A541, F572, K568, S1055, and Q742) that modulate CRM1 export activity. CRM1 also has a less studied nuclear function in RNA biogenesis, which is reflected by its localization to the Cajal body and the nucleolus. Here, we have investigated how the mutation of these key residues affects the intranuclear localization of CRM1 and its ability to mediate export of endogenous cargos. We identify A541K as a separation-of-function mutant that reveals the independent nature of the Cajal body and nucleolar localizations of CRM1. We also show that the F572A mutation may have strikingly opposite effects on the export of specific cargos. Importantly, and in contrast to previous claims, our findings indicate that S1055 phosphorylation is not generally required for CRM1 function and that the Q742 is not a function-defining residue in human CRM1. Collectively, our findings provide new insights into an understudied aspect of CRM1 biology and highlight several important issues related to CRM1 function and regulation that need to be re-evaluated and addressed in more detail. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Peptidylprolyl isomerase D circular RNA sensitizes breast cancer to trastuzumab through remodeling HER2 N4-acetylcytidine modification.
- Author
-
Wang, Shengting, Li, Qian, Wang, Yufang, Li, Xiaoming, Feng, Xinghua, Wei, Yuxuan, Wang, Jiaman, and Zhou, Xin
- Abstract
Human epidermal growth factor receptor 2 (HER2) overexpression and activation are crucial for trastuzumab resistance in HER2-positive breast cancer; however, the potential regulatory mechanism of HER2 is still largely undetermined. In this study, a novel circular RNA derived from peptidylprolyl isomerase D (PPID) is identified as a negative regulator of trastuzumab resistance. Circ-PPID is highly stable and significantly downregulated in trastuzumab-resistant cells and tissues. Restoration of circ-PPID markedly enhances HER2-positive breast cell sensitivity to trastuzumab in vitro and in vivo. Circ-PPID directly binds to N-acetyltransferase 10 (NAT10) in the nucleus and blocks the interaction between NAT10 and HER2 mRNA, reducing N4-acetylcytidine (ac4C) modification on HER2 exon 25, leading to HER2 mRNA decay. Intriguingly, the subcellular localization of circ-PPID differs between trastuzumab-sensitive and -resistant cells. Circ-PPID in trastuzumab-resistant cells is located more in the cytoplasm, mainly due to the upregulation of Exportin 4 (XPO4), which results in the loss of spatial conditions for circ-PPID to bind to nuclear NAT10. Taken together, our data suggest that circ-PPID is a previously unappreciated ac4C-dependent HER2 epigenetic regulator, providing a promising therapeutic direction for overcoming trastuzumab resistance in clinical setting. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Dual modes of ZFC3H1 confer selectivity in nuclear RNA sorting.
- Author
-
Fan, Jing, Wang, Yimin, Wen, Miaomiao, Tong, Deng, Wu, Kai, Yan, Kunming, Jia, Peixuan, Zhu, Yi, Liu, Qinyu, Zou, Hecun, Zhao, Peng, Lu, Falong, Yun, Caihong, Xue, Yuanchao, Zhou, Yu, and Cheng, Hong
- Subjects
- *
GENETIC transcription , *GENETIC translation , *EXOSOMES , *RNA , *MESSENGER RNA - Abstract
The export and degradation pathways compete to sort nuclear RNAs, yet the default pathway remains unclear. Sorting of mature RNAs to degradation, facilitated by the exosome co-factor poly(A) exosome targeting (PAXT), is particularly challenging for their resemblance to mRNAs intended for translation. Here, we unveil that ZFC3H1, a core PAXT component, is co-transcriptionally loaded onto the first exon/intron of RNA precursors (pre-RNAs). Interestingly, this initial loading does not lead to pre-RNA degradation, as ZFC3H1 adopts a "closed" conformation, effectively blocking exosome recruitment. As processing progresses, RNA fate can be reshaped. Longer RNAs with more exons are allowed for nuclear export. By contrast, short RNAs with fewer exons preferentially recruit transient PAXT components ZC3H3 and RBM26/27 to the 3′ end, triggering ZFC3H1 "opening" and subsequent exosomal degradation. Together, the decoupled loading and activation of ZFC3H1 pre-configures RNA fate for decay while still allowing a switch to nuclear export, depending on mature RNA features. [Display omitted] • ZFC3H1 is prevalently loaded onto the 5′ end of pre-RNAs in an inactive mode • ZFC3H1 loaded on pre-RNAs prevents dysregulated mRNA export • Long multi-exonic RNAs are sorted for export, while short intron-few RNAs for decay • Short RNAs favorably recruit ZC3H3 and RBM26/27 to the 3′ end, activating ZFC3H1 Fan et al. report that poly(A) exosome targeting (PAXT) presets decay as the default RNA fate at an early stage of transcription. This fate can be reshaped to export or remain as decay based on exon number and mature transcript length. This degradation-centric model ensures both the rapid degradation of unwanted RNAs and the efficient export of functional mRNAs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. DDX19A promotes gastric cancer cell proliferation and migration by activating the PI3K/AKT pathway
- Author
-
Yu Cheng, Yanjie Lu, Jing Xue, Xuemei Wang, Lili Zhou, Yu Luo, and Yuhong Li
- Subjects
Gastric cancer ,DDX19A ,PIK3CA ,PI3K/AKT ,Nuclear export ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Background DEAD-box RNA helicase 19 A (DDX19A) is overexpressed in cervical squamous cell carcinoma. However, its role in gastric cancer remains unclear. The present study aimed to explore the role and underlying mechanism of DDX19A in the development of gastric cancer. Methods The expression of DDX19A in gastric cancer and paracancerous tissues was evaluated through quantitative polymerase chain reaction, western blotting, and immunohistochemical staining. The biological functions of DDX19A in gastric cancer were determined using CCK8, plate colony-forming, and Transwell migration assays. The specific mechanism of DDX19A in gastric cancer cells was studied using western blotting, RNA-binding protein immunoprecipitation, mRNA half-life detection, and nuclear and cytoplasmic RNA isolation. Results DDX19A was highly expressed in gastric cancer and positively associated with malignant clinicopathological features and poor prognosis. Additionally, DDX19A promoted gastric cancer cell proliferation, migration, and epithelial–mesenchymal transition phenotypes. Mechanistically, DDX19A activated the PI3K/AKT pathway by upregulating phosphatidylinositol-3-kinase (PIK3CA) expression. Furthermore, DDX19A interacted with PIK3CA mRNA, stabilized it, and facilitated its export from the nucleus. Conclusions Our study reveals a novel mechanism whereby DDX19A promotes the proliferation and migration of gastric cancer cells by enhancing the stability and nuclear export of PIK3CA mRNA, thereby activating the PI3K/AKT pathway.
- Published
- 2024
- Full Text
- View/download PDF
7. Novel mechanisms of MITF regulation identified in a mouse suppressor screen.
- Author
-
Vu, Hong Nhung, Valdimarsson, Matti Már, Sigurbjörnsdóttir, Sara, Bergsteinsdóttir, Kristín, Debbache, Julien, Bismuth, Keren, Swing, Deborah A, Hallsson, Jón H, Larue, Lionel, Arnheiter, Heinz, Copeland, Neal G, Jenkins, Nancy A, Heidarsson, Petur O, and Steingrímsson, Eiríkur
- Abstract
MITF, a basic Helix-Loop-Helix Zipper (bHLHZip) transcription factor, plays vital roles in melanocyte development and functions as an oncogene. We perform a genetic screen for suppressors of the Mitf-associated pigmentation phenotype in mice and identify an intragenic Mitf mutation that terminates MITF at the K316 SUMOylation site, leading to loss of the C-end intrinsically disordered region (IDR). The resulting protein is more nuclear but less stable than wild-type MITF and retains DNA-binding ability. As a dimer, it can translocate wild-type and mutant MITF partners into the nucleus, improving its own stability thus ensuring nuclear MITF supply. smFRET analysis shows interactions between K316 SUMOylation and S409 phosphorylation sites across monomers; these interactions largely explain the observed effects. The recurrent melanoma-associated E318K mutation in MITF, which affects K316 SUMOylation, also alters protein regulation in concert with S409. This suggests that residues K316 and S409 of MITF are impacted by SUMOylation and phosphorylation, respectively, mediating effects on nuclear localization and stability through conformational changes. Our work provides a novel mechanism of genetic suppression, and an example of how apparently deleterious mutations lead to normal phenotypes. Synopsis: An intragenic Mitf suppressor mutation was identified that terminates MITF at the K316 SUMOylation site, leading to loss of the C-end intrinsically disordered region (IDR). This mutation provides novel information on how the dynamic IDR mediates MITF localization and stability. The MITF suppressor mutation uncovers a novel mechanism of genetic suppression that opens unexpected insights into MITF subcellular localization and stability. The MITF suppressor mutation provides one explanation for how humans with knockout mutations in essential genes are viable and normal. An intragenic Mitf suppressor mutation was identified that terminates MITF at the K316 SUMOylation site, leading to loss of the C-end intrinsically disordered region (IDR). This mutation provides novel information on how the dynamic IDR mediates MITF localization and stability. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Novel--and Not So Novel--Inhibitors of the Multifunctional CRM1 Protein.
- Author
-
Aumann, Waitman K., Kazi, Rafi, Harrington, Amanda M., and Wechsler, Daniel S.
- Subjects
- *
NUCLEAR pore complex , *CARRIER proteins , *SMALL molecules , *BRAIN tumors , *PROTEINS - Abstract
Chromosome Region Maintenance 1 (CRM1), also known as Exportin 1 (XPO1), is a protein that is critical for transport of proteins and RNA to the cytoplasm through the nuclear pore complex. CRM1 inhibition with small molecule inhibitors is currently being studied in many cancers, including leukemias, solid organ malignancies and brain tumors. We review the structure of CRM1, its role in nuclear export, the current availability of CRM1 inhibitors, and the role of CRM1 in a number of distinct cellular processes. A deeper understanding of how CRM1 functions in nuclear export as well as other cellular processes may allow for the development of additional novel CRM1 inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. RNAi‐mediated knockdown of exportin 1 negatively affected ovary development, survival and maize mosaic virus accumulation in its insect vector Peregrinus maidis.
- Author
-
Xavier, Cesar A. D., Tyson, Clara, Kerner, Leo M., and Whitfield, Anna E.
- Subjects
- *
LIFE cycles (Biology) , *INSECT physiology , *NUCLEOCYTOPLASMIC interactions , *NUCLEAR transport , *OVARIES , *OVIPARITY , *MOSAIC viruses , *BRAIN physiology - Abstract
Exportin 1 (XPO1) is the major karyopherin‐β nuclear receptor mediating the nuclear export of hundreds of proteins and some classes of RNA and regulates several critical processes in the cell, including cell‐cycle progression, transcription and translation. Viruses have co‐opted XPO1 to promote nucleocytoplasmic transport of viral proteins and RNA. Maize mosaic virus (MMV) is a plant‐infecting rhabdovirus transmitted in a circulative propagative manner by the corn planthopper, Peregrinus maidis. MMV replicates in the nucleus of plant and insect hosts, and it remains unknown whether MMV co‐opts P. maidis XPO1 (PmXPO1) to complete its life cycle. Because XPO1 plays multiple regulatory roles in cell functions and virus infection, we hypothesized that RNAi‐mediated silencing of XPO1 would negatively affect MMV accumulation and insect physiology. Although PmXPO1 expression was not modulated during MMV infection, PmXPO1 knockdown negatively affected MMV accumulation in P. maidis at 12 and 15 days after microinjection. Likewise, PmXPO1 knockdown negatively affected P. maidis survival and reproduction. PmXPO1 exhibited tissue‐specific expression patterns with higher expression in the ovaries compared with the guts of adult females. Survival rate was significantly lower for PmXPO1 knockdown females, compared with controls, but no effect was observed for males. PmXPO1 knockdown experiments revealed a role for PmXPO1 in ovary function and egg production. Oviposition and egg hatch on plants were dramatically reduced in females treated with dsRNA PmXPO1. These results suggest that PmXPO1 is a positive regulator of P. maidis reproduction and that it plays a proviral role in the insect vector supporting MMV infection. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Inhibition of RAN attenuates influenza a virus replication and nucleoprotein nuclear export
- Author
-
Lei Cao, Ziwei She, Ya Zhao, Chuxing Cheng, Yaqin Li, Ting Xu, Haiying Mao, Yumei Zhang, Xianfeng Hui, Xian Lin, Ting Wang, Xiaomei Sun, Kun Huang, Lianzhong Zhao, and Meilin Jin
- Subjects
Influenza A virus ,nuclear export ,viral ribonucleoprotein ,RAN ,bepotastine ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
Nuclear export of the viral ribonucleoprotein (vRNP) is a critical step in the influenza A virus (IAV) life cycle and may be an effective target for the development of anti-IAV drugs. The host factor ras-related nuclear protein (RAN) is known to participate in the life cycle of several viruses, but its role in influenza virus replication remains unknown. In the present study, we aimed to determine the function of RAN in influenza virus replication using different cell lines and subtype strains. We found that RAN is essential for the nuclear export of vRNP, as it enhances the binding affinity of XPO1 toward the viral nuclear export protein NS2. Depletion of RAN constrained the vRNP complex in the nucleus and attenuated the replication of various subtypes of influenza virus. Using in silico compound screening, we identified that bepotastine could dissociate the RAN-XPO1-vRNP trimeric complex and exhibit potent antiviral activity against influenza virus both in vitro and in vivo. This study demonstrates the important role of RAN in IAV replication and suggests its potential use as an antiviral target.
- Published
- 2024
- Full Text
- View/download PDF
11. Antitumor activity and low gastrointestinal toxicity of a novel selective inhibitor of nuclear export, SZJK-0421, in multiple myeloma
- Author
-
Jing Wang, Hang Miao, Xuxing Shen, Lin Yang, Yongqiang Zhu, and Lijuan Chen
- Subjects
Multiple myeloma ,Selective inhibitor ,Nuclear export ,Cytotoxicity ,Gastrointestinal toxicity ,Therapeutics. Pharmacology ,RM1-950 - Published
- 2024
- Full Text
- View/download PDF
12. Antitumor activity and low gastrointestinal toxicity of a novel selective inhibitor of nuclear export, SZJK-0421, in multiple myeloma.
- Author
-
Wang, Jing, Miao, Hang, Shen, Xuxing, Yang, Lin, Zhu, Yongqiang, and Chen, Lijuan
- Subjects
MULTIPLE myeloma ,ANTINEOPLASTIC agents ,CYTOTOXINS - Published
- 2024
- Full Text
- View/download PDF
13. The mRNA-capping enzyme localizes to stress granules in the cytoplasm and maintains cap homeostasis of target mRNAs.
- Author
-
Gayen, Anakshi, Mukherjee, Avik, Kumar, Krishna, Majumder, Shubhra, Chakrabarti, Saikat, and Mukherjee, Chandrama
- Subjects
- *
STRESS granules , *HOMEOSTASIS , *CYTOPLASM , *ENZYMES , *MOLECULAR docking - Abstract
The model of RNA stability has undergone a transformative shift with the revelation of a cytoplasmic capping activity that means a subset of transcripts are recapped autonomously of their nuclear counterparts. The present study demonstrates nucleo-cytoplasmic shuttling of the mRNA-capping enzyme (CE, also known as RNA guanylyltransferase and 5'-phosphatase; RNGTT), traditionally acknowledged for its nuclear localization and functions, elucidating its contribution to cytoplasmic capping activities. A unique nuclear export sequence in CE mediates XPO1-dependent nuclear export of CE. Notably, during sodium arsenite-induced oxidative stress, cytoplasmic CE (cCE) congregates within stress granules (SGs). Through an integrated approach involving molecular docking and subsequent co-immunoprecipitation, we identify eIF3b, a constituent of SGs, as an interactive associate of CE, implying that it has a potential role in guiding cCE to SGs. We measured the cap status of specific mRNA transcripts from U2OS cells that were non-stressed, stressed and recovered from stress, which indicated that cCE-target transcripts lost their caps during stress but remarkably regained cap stability during the recovery phase. This comprehensive study thus uncovers a novel facet of cytoplasmic CE, which facilitates cellular recovery from stress by maintaining cap homeostasis of target mRNAs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Exporting nuclear export inhibitors from hematologic to solid tumour malignancies
- Author
-
Alexandra Chirino, Alyssa Maye, and Justin Taylor
- Subjects
cancer ,nuclear export ,solid tumors ,selinexor ,XPO1 ,Therapeutics. Pharmacology ,RM1-950 - Published
- 2024
- Full Text
- View/download PDF
15. Exporting nuclear export inhibitors from hematologic to solid tumour malignancies.
- Author
-
Chirino, Alexandra, Maye, Alyssa, and Taylor, Justin
- Subjects
BORTEZOMIB ,APOPTIN ,NUCLEAR pore complex ,TUMORS ,DIFFUSE large B-cell lymphomas - Abstract
The article offers information on the potential expansion of nuclear export inhibitors, specifically Exportin 1 (XPO1) inhibitors, from treating hematologic malignancies to solid tumors. Topics discussed include the challenges and successes of selinexor in clinical trials for solid tumors; the need for targeted patient populations in future trials; and the exploration of second-generation XPO1 inhibitors like eltanexor and felezonexor.
- Published
- 2024
- Full Text
- View/download PDF
16. Selinexor: Targeting a novel pathway in multiple myeloma
- Author
-
Clifton C. Mo, Andrew J. Yee, Shonali Midha, Monique A. Hartley‐Brown, Omar Nadeem, Elizabeth K. O'Donnell, Giada Bianchi, Adam S. Sperling, Jacob P. Laubach, and Paul G. Richardson
- Subjects
multiple myeloma ,nuclear export ,refractory ,relapsed ,selective inhibitor of nuclear export ,targeted therapy ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Selinexor is an orally bioavailable selective inhibitor of nuclear export compound that inhibits exportin‐1 (XPO1), a novel therapeutic target that is overexpressed in multiple myeloma (MM) and is responsible for the transport of ∼220 nuclear proteins to the cytoplasm, including tumour suppressor proteins. Inhibition of this process has demonstrated substantial antimyeloma activity in preclinical studies, both alone and in combination with established MM therapeutics. Based on a clinical trial programme encompassing multiple combination regimens, selinexor‐based therapy has been approved for the treatment of relapsed/refractory MM (RRMM), with selinexor‐dexamethasone approved in the later‐relapse setting for penta‐refractory patients and selinexor‐bortezomib‐dexamethasone approved for patients who have received ≥1 prior therapy. Here, we provide a comprehensive review of the clinical data on selinexor‐based regimens, including recent updates from the 2022 American Society of Hematology annual meeting, and summarise ongoing studies of this novel targeted agent in newly diagnosed MM and RRMM.
- Published
- 2023
- Full Text
- View/download PDF
17. Structural and computational studies of HIV-1 RNA.
- Author
-
Levintov, Lev and Vashisth, Harish
- Subjects
HIV ,RNA ,VIRAL DNA ,GENETIC transcription ,DNA replication - Abstract
Viruses remain a global threat to animals, plants, and humans. The type 1 human immunodeficiency virus (HIV-1) is a member of the retrovirus family and carries an RNA genome, which is reverse transcribed into viral DNA and further integrated into the host-cell DNA for viral replication and proliferation. The RNA structures from the HIV-1 genome provide valuable insights into the mechanisms underlying the viral replication cycle. Moreover, these structures serve as models for designing novel therapeutic approaches. Here, we review structural data on RNA from the HIV-1 genome as well as computational studies based on these structural data. The review is organized according to the type of structured RNA element which contributes to different steps in the viral replication cycle. This is followed by an overview of the HIV-1 transactivation response element (TAR) RNA as a model system for understanding dynamics and interactions in the viral RNA systems. The review concludes with a description of computational studies, highlighting the impact of biomolecular simulations in elucidating the mechanistic details of various steps in the HIV-1's replication cycle. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Nuclear envelope budding and its cellular functions.
- Author
-
Keuenhof, Katharina S., Kohler, Verena, Broeskamp, Filomena, Panagaki, Dimitra, Speese, Sean D., Büttner, Sabrina, and Höög, Johanna L.
- Subjects
- *
NUCLEAR membranes , *CELL physiology , *NUCLEAR pore complex , *HOMEOSTASIS , *NUCLEAR proteins , *MEMBRANE proteins - Abstract
The nuclear pore complex (NPC) has long been assumed to be the sole route across the nuclear envelope, and under normal homeostatic conditions it is indeed the main mechanism of nucleo-cytoplasmic transport. However, it has also been known that e.g. herpesviruses cross the nuclear envelope utilizing a pathway entitled nuclear egress or envelopment/de-envelopment. Despite this, a thread of observations suggests that mechanisms similar to viral egress may be transiently used also in healthy cells. It has since been proposed that mechanisms like nuclear envelope budding (NEB) can facilitate the transport of RNA granules, aggregated proteins, inner nuclear membrane proteins, and mis-assembled NPCs. Herein, we will summarize the known roles of NEB as a physiological and intrinsic cellular feature and highlight the many unanswered questions surrounding these intriguing nuclear events. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
19. Viral Subversion of the Chromosome Region Maintenance 1 Export Pathway and Its Consequences for the Cell Host.
- Author
-
Mghezzi-Habellah, Makram, Prochasson, Léa, Jalinot, Pierre, and Mocquet, Vincent
- Subjects
- *
NUCLEOCYTOPLASMIC interactions , *BIOLOGICAL transport , *CARRIER proteins , *PROTEIN transport , *CHROMOSOMES , *HOMEOSTASIS , *CYTOPLASM - Abstract
In eukaryotic cells, the spatial distribution between cytoplasm and nucleus is essential for cell homeostasis. This dynamic distribution is selectively regulated by the nuclear pore complex (NPC), which allows the passive or energy-dependent transport of proteins between these two compartments. Viruses possess many strategies to hijack nucleocytoplasmic shuttling for the benefit of their viral replication. Here, we review how viruses interfere with the karyopherin CRM1 that controls the nuclear export of protein cargoes. We analyze the fact that the viral hijacking of CRM1 provokes are-localization of numerous cellular factors in a suitable place for specific steps of viral replication. While CRM1 emerges as a critical partner for viruses, it also takes part in antiviral and inflammatory response regulation. This review also addresses how CRM1 hijacking affects it and the benefits of CRM1 inhibitors as antiviral treatments. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. CK2 Inhibits TIMELESS Nuclear Export and Modulates CLOCK Transcriptional Activity to Regulate Circadian Rhythms
- Author
-
Cai, Yao D, Xue, Yongbo, Truong, Cindy C, Del Carmen-Li, Jose, Ochoa, Christopher, Vanselow, Jens T, Murphy, Katherine A, Li, Ying H, Liu, Xianhui, Kunimoto, Ben L, Zheng, Haiyan, Zhao, Caifeng, Zhang, Yong, Schlosser, Andreas, and Chiu, Joanna C
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Sleep Research ,Genetics ,Neurosciences ,1.1 Normal biological development and functioning ,Generic health relevance ,Active Transport ,Cell Nucleus ,Animals ,CLOCK Proteins ,Circadian Rhythm ,Drosophila Proteins ,Drosophila melanogaster ,Humans ,Sleep Disorders ,Circadian Rhythm ,CK2 ,CLOCK ,Drosophila ,FASPS ,PERIOD ,TIMELESS ,XPO1 ,circadian clock ,nuclear export ,phosphorylation ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Psychology - Abstract
Circadian clocks orchestrate daily rhythms in organismal physiology and behavior to promote optimal performance and fitness. In Drosophila, key pacemaker proteins PERIOD (PER) and TIMELESS (TIM) are progressively phosphorylated to perform phase-specific functions. Whereas PER phosphorylation has been extensively studied, systematic analysis of site-specific TIM phosphorylation is lacking. Here, we identified phosphorylation sites of PER-bound TIM by mass spectrometry, given the importance of TIM as a modulator of PER function in the pacemaker. Among the 12 TIM phosphorylation sites we identified, at least two of them are critical for circadian timekeeping as mutants expressing non-phosphorylatable mutations exhibit altered behavioral rhythms. In particular, we observed that CK2-dependent phosphorylation of TIM(S1404) promotes nuclear accumulation of PER-TIM heterodimers by inhibiting the interaction of TIM and nuclear export component, Exportin 1 (XPO1). We propose that proper level of nuclear PER-TIM accumulation is necessary to facilitate kinase recruitment for the regulation of daily phosphorylation rhythm and phase-specific transcriptional activity of CLOCK (CLK). Our results highlight the contribution of phosphorylation-dependent nuclear export of PER-TIM heterodimers to the maintenance of circadian periodicity and identify a new mechanism by which the negative elements of the circadian clock (PER-TIM) regulate the positive elements (CLK-CYC). Finally, because the molecular phenotype of tim(S1404A) non-phosphorylatable mutant exhibits remarkable similarity to that of a mutation in human timeless that underlies familial advanced sleep phase syndrome (FASPS), our results revealed an unexpected parallel between the functions of Drosophila and human TIM and may provide new insights into the molecular mechanisms underlying human FASPS.
- Published
- 2021
21. Retroviral hijacking of host transport pathways for genome nuclear export
- Author
-
Ryan T. Behrens and Nathan M. Sherer
- Subjects
virus ,nuclear export ,nuclear pore complex ,nuclear envelope ,subcellular trafficking ,nuclear membrane budding ,Microbiology ,QR1-502 - Abstract
ABSTRACTRecent advances in the study of virus-cell interactions have improved our understanding of how viruses that replicate their genomes in the nucleus (e.g., retroviruses, hepadnaviruses, herpesviruses, and a subset of RNA viruses) hijack cellular pathways to export these genomes to the cytoplasm where they access virion egress pathways. These findings shed light on novel aspects of viral life cycles relevant to the development of new antiviral strategies and can yield new tractable, virus-based tools for exposing additional secrets of the cell. The goal of this review is to summarize defined and emerging modes of virus-host interactions that drive the transit of viral genomes out of the nucleus across the nuclear envelope barrier, with an emphasis on retroviruses that are most extensively studied. In this context, we prioritize discussion of recent progress in understanding the trafficking and function of the human immunodeficiency virus type 1 Rev protein, exemplifying a relatively refined example of stepwise, cooperativity-driven viral subversion of multi-subunit host transport receptor complexes.
- Published
- 2023
- Full Text
- View/download PDF
22. Nuclear Import and Export of YAP and TAZ.
- Author
-
Kofler, Michael and Kapus, András
- Subjects
- *
TUMOR treatment , *GROWTH factors , *BIOLOGICAL transport , *FIBROSIS , *CELLULAR signal transduction , *CELL nuclei , *MEMBRANE proteins , *CARRIER proteins - Abstract
Simple Summary: YAP and TAZ are transcriptional activators, which play critical roles in the regulation of cell/organ growth and regeneration. Accordingly, they are important mediators and modulators of cancer formation and spread, impacting a wide range of processes from cancer stem cell differentiation through the control of proliferation, phenotype determination, and migration, till the induction of antitumor drug resistance. These widespread activities are due to their profound effect on gene expression. Importantly, to exert these physiological and pathological functions, they must enter the nucleus, while for the termination of their transcriptional effects, they must exit. However, the critical structural requirements (nuclear localization signal and nuclear export signal) as well as their passageway(s) and mechanism(s) for crossing the nuclear pore are only beginning to emerge. This review summarizes the current knowledge about the nuclear traffic of YAP and TAZ, a fundamental, novel, and potentially exploitable aspect of their biology. Yes-associated Protein (YAP) and its paralog Transcriptional Coactivator with PDZ-binding Motif (TAZ) are major regulators of gene transcription/expression, primarily controlled by the Hippo pathway and the cytoskeleton. Integrating an array of chemical and mechanical signals, they impact growth, differentiation, and regeneration. Accordingly, they also play key roles in tumorigenesis and metastasis formation. Their activity is primarily regulated by their localization, that is, Hippo pathway- and/or cytoskeleton-controlled cytosolic or nuclear sequestration. While many details of such prevailing retention models have been elucidated, much less is known about their actual nuclear traffic: import and export. Although their size is not far from the cutoff for passive diffusion through the nuclear pore complex (NPC), and they do not contain any classic nuclear localization (NLS) or nuclear export signal (NES), evidence has been accumulating that their shuttling involves mediated and thus regulatable/targetable processes. The aim of this review is to summarize emerging information/concepts about their nucleocytoplasmic shuttling, encompassing the relevant structural requirements (NLS, NES), nuclear transport receptors (NTRs, karyophererins), and NPC components, along with the potential transport mechanisms and their regulation. While dissecting retention vs. transport is often challenging, the emerging picture suggests that YAP/TAZ shuttles across the NPC via multiple, non-exclusive, mediated mechanisms, constituting a novel and intriguing facet of YAP/TAZ biology. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
23. Exploiting a rodent cell block for intrinsic resistance to HIV-1 gene expression in human T cells
- Author
-
Ryan T. Behrens, Jyothi Krishnaswamy Rajashekar, James W. Bruce, Edward L. Evans, Amelia M. Hansen, Natalia Salazar-Quiroz, Lacy M. Simons, Paul Ahlquist, Judd F. Hultquist, Priti Kumar, and Nathan M. Sherer
- Subjects
human immunodeficiency virus ,lentiviruses ,transcription ,nuclear export ,Cyclin T1 ,CCNT1 ,Microbiology ,QR1-502 - Abstract
ABSTRACT HIV-1 virion production is inefficient in cells derived from mice and other rodents reflecting cell-intrinsic defects to interactions between the HIV-1 auxiliary proteins Tat and Rev and host dependency factors CCNT1 (Cyclin T1) and XPO1 (exportin-1, also known as CRM1), respectively. In human cells, Tat binds CCNT1 to enhance viral RNA transcription and Rev recruits XPO1 to mediate the nuclear export of intron-containing viral RNA. In mouse cells, Tat’s interactions with CCNT1 are inefficient, mapped to a single species-specific residue Y261 instead of C261 in humans. Rev interacts poorly with murine XPO1, mapped to a trio of amino acids T411/V412/S414 instead of P411/M412/F414 in humans. To determine if these discrete species-specific regions of otherwise conserved housekeeping proteins represent viable targets for inhibiting HIV-1 replication in humans, herein, we employed CRISPR/Cas9 to recode the relevant regions of CCNT1 and XPO1 in human CD4+ T cells. While efforts to modify XPO1 were inconclusive, we generated isogenic CCNT1.C261Y cell lines exhibiting remarkable resistance to HIV-1 Tat, exhibiting near total inactivation of viral gene expression for all X4- and R5-tropic HIV-1 strains tested, as well as the more distantly related primate lentiviruses HIV-2 and SIVagm. Induction of viral reactivation using latency reversal agents (LRAs) was also restricted in CCNT1.C261Y cells. These studies validate a minor and naturally occurring, species-specific difference in a conserved human host factor as a compelling potential target for achieving broad-acting cell-intrinsic resistance to HIV’s post-integration phases. Importance Unlike humans, mice are unable to support HIV-1 infection. This is due, in part, to a constellation of defined minor, species-specific differences in conserved host proteins needed for viral gene expression. Here, we used precision CRISPR/Cas9 gene editing to engineer a “mousified” version of one such host protein, cyclin T1 (CCNT1), in human T cells. CCNT1 is essential for efficient HIV-1 transcription, making it an intriguing target for gene-based inactivation of virus replication. We show that isogenic cell lines engineered to encode CCNT1 bearing a single mouse-informed amino acid change (tyrosine in place of cysteine at position 261) exhibit potent, durable, and broad-spectrum resistance to HIV-1 and other pathogenic lentiviruses, and with no discernible impact on host cell biology. These results provide proof of concept for targeting CCNT1 in the context of one or more functional HIV-1 cure strategies.
- Published
- 2023
- Full Text
- View/download PDF
24. The CRM1‐mediated nuclear export effectuates the role conversion of tumour suppressor gene BATF2 in colorectal cancer.
- Author
-
Yu, Peiyao, Chen, Bonan, Xie, Fuda, Yu, Jun, To, Ka Fai, and Kang, Wei
- Subjects
- *
TUMOR suppressor genes , *NUCLEAR factor of activated T-cells , *COLORECTAL cancer , *CHRONIC leukemia , *CELL cycle proteins , *NUCLEAR transport , *HEREDITARY nonpolyposis colorectal cancer - Abstract
This article discusses the role of CRM1-mediated nuclear export in the conversion of the tumor suppressor gene BATF2 in colorectal cancer (CRC). The dysregulation of nuclear-cytoplasmic translocation (NCT) has been linked to the initiation and progression of CRC. The study demonstrates that CRM1 is responsible for the NCT of BATF2, leading to its cytoplasmic translocation and activation of the AP-1/cyclin D1/pRb signaling pathway, which promotes tumor growth and progression. Inhibiting the nuclear export of BATF2 through targeting CRM1 may be a promising therapeutic strategy for CRC. [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
25. Geminivirus satellite-encoded βC1 activates UPR, induces bZIP60 nuclear export, and manipulates the expression of bZIP60 downstream genes to benefit virus infection.
- Author
-
Zhang, Mingzhen, Cao, Buwei, Zhang, Hui, Fan, Zaifeng, Zhou, Xueping, and Li, Fangfang
- Abstract
UPR is a conserved response in eukaryotes and can alleviate endoplasmic reticulum (ER) stresses induced by abiotic and biotic stresses. The interactions between UPR and plant RNA viruses have been documented, while the interplays between UPR and plant DNA viruses remain unknown. Using tomato yellow leaf curl China virus (TYLCCNV) and its associated betasatellite (TYLCCNB) as a model, we indicate that TYLCCNB βC1 is a major inducer of UPR and can upregulate the expression of bZIP60, a transcription factor in Nicotiana benthamiana plants. Treatment using ER stress inducers or overexpression of NbbZIP60 increases βC1 accumulation and benefits TYLCCNV/TYLCCNB infection in N. benthamiana plants, and vice versa. In the TYLCCNV/TYLCCNB-infected or the βC1-expressing cells, NbbZIP60 is exported from the nucleus to the nuclear periphery via the XPO1 pathway, and blocking the XPO1 pathway inhibited TYLCCNV/TYLCCNB infection. We have found that the NbbZIP60-regulated pro-survival genes could promote virus infection, and the pro-death gene plays a contrasting role in virus infection. This study reveals that geminivirus infection activates UPR and utilizes the up-regulated molecular chaperons to promote viral infection, and then induces the nuclear export of NbbZIP60 to evade plant defense response, which is a distinct virulence strategy exploited by plant pathogens. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
26. Nuclear export of BATF2 enhances colorectal cancer proliferation through binding to CRM1.
- Author
-
Zhou, Jie, Lei, Zengjie, Chen, Jianfang, Liao, Shengbo, Chen, Yanrong, Liu, Chengxiang, Huang, Shuo, Li, Liuli, Zhang, Yan, Wang, Pei, Huang, Yinghui, Li, Jianjun, and Liang, Houjie
- Subjects
- *
TANDEM mass spectrometry , *COLORECTAL cancer , *AP-1 transcription factor , *LIQUID chromatography-mass spectrometry , *LEUCINE zippers , *RETINOBLASTOMA protein , *TUMOR suppressor genes , *IMMUNOPRECIPITATION - Abstract
Background: During the tumourigenesis and development of colorectal cancer (CRC), the inactivation of tumour suppressor genes is closely involved, although detailed molecular mechanisms remain elusive. Accumulating studies, including ours, have demonstrated that basic leucine zipper transcription factor ATF (activating transcription factor)‐like 2 (BATF2) is a capable tumour suppressor that localises in the nucleus. However, its different subcellular localisation, potential functions and underlying mechanisms are unclear. Methods: The translocation of BATF2 and its clinical relevance were detected using CRC samples, cell lines and xenograft nude mice. Candidate BATF2‐binding proteins were screened using co‐immunoprecipitation, quantitative label‐free liquid chromatography–tandem mass spectrometry proteomic analysis, Western blotting and immunofluorescence. Recombinant plasmids, point mutations and siRNAs were applied to clarify the binding sites between BATF2 and chromosome region maintenance 1 (CRM1). Results: The present study found that BATF2 was mainly localised in the cytoplasm, rather than nucleus, of CRC cells in vitro and in vivo, while cytoplasmic BATF2 expression was inversely correlated with the prognosis of CRC patients. Furthermore, we identified the nuclear export and subsequent ubiquitin‐mediated degradation of BATF2 in CRC cells. Mechanistically, a functional nuclear export sequence (any amino acid) was characterised in BATF2 protein, through which BATF2 bound to CRM1 and translocated out of nucleus, ultimately enhancing CRC growth via inducing activator protein 1 (AP‐1)/cyclin D1/phosphorylated retinoblastoma protein (pRb) signalling pathway. Additionally, nuclear export of BATF2 can be retarded by the mutation of NES in BATF2 or the knockdown of CRM1, whereas CRM1 expression was negatively associated with nuclear BATF2 expression and the prognosis of CRC patients. Conclusion: These findings revealed the biological effects and underlying mechanisms of cytoplasmic localisation of BATF2. Furthermore, suppressing nuclear export of BATF2 via mutating its NES region or inhibiting CRM1 expression may serve as a promising therapeutic strategy against CRC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
27. Intramolecular interaction of NEP regulated by CRM1 ensures the unidirectional transport of M1 for the nuclear export of influenza viral ribonucleoprotein
- Author
-
Mikako Hirohama, Shun Yamashita, Masamitsu N. Asaka, Takahiro Kuroki, and Atsushi Kawaguchi
- Subjects
CRM1 ,nuclear export ,NEP ,intramolecular interaction ,influenza A virus ,Microbiology ,QR1-502 - Abstract
IntroductionThe influenza virus genome consists of single-stranded RNAs and forms viral ribonucleoprotein (RNP) complexes. After viral genome replication in the nucleus, the viral RNP interacts with viral protein M1. The M1-viral RNP complex is exported to the cytoplasm via the CRM1-dependent pathway using NS2/NEP as an export adaptor protein. NEP is a 14 kDa protein and diffusely localizes in the nucleus and cytoplasm. Upon binding to the NLS motif of M1, NEP inhibits the nuclear accumulation of M1 and promotes the nuclear export of M1-viral RNP complex. However, the detail mechanism by which NEP binds to M1 only in the nucleus remains unclear.MethodsTo visualize the interaction of NEP with M1 in the formation of vRNP export complexes, we performed in situ proximity ligation assays. The close proximity of N-terminal and C-terminal domains of NEP was tested by split Renilla luciferase complementation assays in which the N-terminal and C-terminal fragments of Renilla luciferase were fused to the N-terminus and C-terminus of NEP, respectively.Results and discussionWe found that the intramolecular interaction of NEP inhibits the interaction of NEP with M1. The intramolecular interaction of NEP was mediated through the interaction of the N-terminal NES motif with the M1-binding domain at the C-terminus. By adding leptomycin B, a potent inhibitor of CRM1, the interaction of NEP with M1 was impaired. These results suggest that CRM1 disrupts the intramolecular interaction of NEP by recognizing the NES motif at the N-terminus of NEP, thereby promoting the interaction of NEP with M1. We also found that NEP mutant deficient in the intramolecular interaction was co-localized with M1 at the plasma membrane and did not show nuclear localization with M1. Based on these results, we propose that the intramolecular interaction of NEP regulated by CRM1 ensures the unidirectional transport of M1.
- Published
- 2023
- Full Text
- View/download PDF
28. The pseudokinase TRIB1 toggles an intramolecular switch to regulate COP1 nuclear export
- Author
-
Kung, Jennifer E and Jura, Natalia
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Active Transport ,Cell Nucleus ,Amino Acid Sequence ,Cell Nucleus ,HEK293 Cells ,Humans ,Intracellular Signaling Peptides and Proteins ,Karyopherins ,Protein Binding ,Protein Serine-Threonine Kinases ,Receptors ,Cytoplasmic and Nuclear ,Sequence Homology ,Ubiquitin ,Ubiquitin-Protein Ligases ,Ubiquitination ,WD40 Repeats ,COP1 ,E3 ubiquitin ligase ,nuclear export ,pseudokinase ,Tribbles ,Information and Computing Sciences ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
COP1 is a highly conserved ubiquitin ligase that regulates diverse cellular processes in plants and metazoans. Tribbles pseudokinases, which only exist in metazoans, act as scaffolds that interact with COP1 and its substrates to facilitate ubiquitination. Here, we report that, in addition to this scaffolding role, TRIB1 promotes nuclear localization of COP1 by disrupting an intramolecular interaction between the WD40 domain and a previously uncharacterized regulatory site within COP1. This site, which we have termed the pseudosubstrate latch (PSL), resembles the consensus COP1-binding motif present in known COP1 substrates. Our findings support a model in which binding of the PSL to the WD40 domain stabilizes a conformation of COP1 that is conducive to CRM1-mediated nuclear export, and TRIB1 displaces this intramolecular interaction to induce nuclear retention of COP1. Coevolution of Tribbles and the PSL in metazoans further underscores the importance of this role of Tribbles in regulating COP1 function.
- Published
- 2019
29. SUMOylation mediates the disassembly of the Smad4 nuclear export complex via RanGAP1 in KELOIDS.
- Author
-
Lin, Xiaohu, Pang, Qianqian, Hu, Jie, Sun, Jiaqi, Dai, Siya, Yu, Yijia, and Xu, Jinghong
- Subjects
SMAD proteins ,KELOIDS ,NUCLEAR transport (Cytology) ,NUCLEOCYTOPLASMIC interactions ,NEUROLOGICAL disorders - Abstract
Sentrin/small ubiquitin‐like modifier (SUMO) has emerged as a powerful mediator regulating biological processes and participating in pathophysiological processes that cause human diseases, such as cancer, myocardial fibrosis and neurological disorders. Sumoylation has been shown to play a positive regulatory role in keloids. However, the sumoylation mechanism in keloids remains understudied. We proposed that sumoylation regulates keloids via a complex. RanGAP1 acted as a synergistic, functional partner of SUMOs in keloids. Nuclear accumulation of Smad4, a TGF‐β/Smad pathway member, was associated with RanGAP1 after SUMO1 inhibition. RanGAP1*SUMO1 mediated the nuclear accumulation of Smad4 due to its impact on nuclear export and reduction in the dissociation of Smad4 and CRM1. We clarified a novel mechanism of positive regulation of sumoylation in keloids and demonstrated the function of sumoylation in Smad4 nuclear export. The NPC‐associated RanGAP1*SUMO1 complex functions as a disassembly machine for the export receptor CRM1 and Smad4. Our research provides new perspectives for the mechanisms of keloids and nucleocytoplasmic transport. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
30. Protein biomarkers for response to XPO1 inhibition in haematologic malignancies.
- Author
-
Totiger, Tulasigeri M., Chaudhry, Sana, Musi, Elgilda, Afaghani, Jumana, Montoya, Skye, Owusu‐Ansah, Frank, Lee, Stanley, Schwartz, Gary, Klimek, Virginia, and Taylor, Justin
- Subjects
NUCLEOCYTOPLASMIC interactions ,RESPONSE inhibition ,DIFFUSE large B-cell lymphomas ,NUCLEAR proteins ,MULTIPLE myeloma ,PROTEINS - Abstract
XPO1 (Exportin‐1) is the nuclear export protein responsible for the normal shuttling of several proteins and RNA species between the nucleocytoplasmic compartment of eukaryotic cells. XPO1 recognizes the nuclear export signal (NES) of its cargo proteins to facilitate its export. Alterations of nuclear export have been shown to play a role in oncogenesis in several types of solid tumour and haematologic cancers. Over more than a decade, there has been substantial progress in targeting nuclear export in cancer using selective XPO1 inhibitors. This has resulted in recent approval for the first‐in‐class drug selinexor for use in relapsed, refractory multiple myeloma and diffuse large B‐cell lymphoma (DLBCL). Despite these successes, not all patients respond effectively to XPO1 inhibition and there has been lack of biomarkers for response to XPO1 inhibitors in the clinic. Using haematologic malignancy cell lines and samples from patients with myelodysplastic neoplasms treated with selinexor, we have identified XPO1, NF‐κB(p65), MCL‐1 and p53 protein levels as protein markers of response to XPO1 inhibitor therapy. These markers could lead to the identification of response upon XPO1 inhibition for more accurate decision‐making in the personalized treatment of cancer patients undergoing treatment with selinexor. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
31. Inhibition of influenza A virus replication by a marine derived quinolone alkaloid targeting virus nucleoprotein.
- Author
-
Zhang, Yang, Xu, Wei‐Feng, Yu, Yunjia, Zhang, Qun, Huang, Lianghao, Hao, Cui, Shao, Chang‐Lun, and Wang, Wei
- Subjects
INFLUENZA A virus ,VIRAL replication ,INFLUENZA viruses ,ALKALOIDS ,ORAL drug administration - Abstract
Owing to the emergence of drug resistance and high morbidity and mortality, the need for novel anti‐influenza A virus (IAV) drugs with divergent targets is highly sought after. Herein, a novel quinolone alkaloid (QLA) derived from marine fungus was discovered with broad‐spectrum anti‐IAV activities with low toxicity. Distinct from current anti‐IAV drugs, QLA may block virus replication and viral RNA (vRNA) export from the nucleus by targeting virus nucleoprotein (NP). QLA can block the binding of chromosome region maintenance 1 to nuclear export signal 3 of NP to inhibit the nuclear export of NP and vRNP. QLA may also affect vRNP assembly by interfering with the binding of NP to RNA rather than NP oligomerization. Arg305 and Phe488‐Gly490 may be required for the interaction between QLA and NP, and the binding pocket around these amino acids may be a promising target for anti‐IAV drugs. Importantly, oral administration of QLA can protect the mice against IAV‐induced death and weight loss, superior to the effects of the clinical drug oseltamivir. In summary, the marine derived compound QLA has the potential to be developed into a novel anti‐IAV agent targeting virus NP protein in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
32. mRNA export factors store nascent transcripts within nuclear speckles as an adaptive response to transient global inhibition of transcription.
- Author
-
Williams, Tobias D., Michalak, Ewa M., Carey, Kirstyn.T., Lam, Enid Y.N., Anderson, Ashley, Griesbach, Esther, Chan, Yih-Chih, Papasaikas, Panagiotis, Tan, Vicky W.T., Ngo, Linh, MacPherson, Laura, Gilan, Omer, Rucinski, Amber, Rutkowska-Klute, Anna, Zinn, Nico, Grandi, Paola, Bantscheff, Marcus, Prinjha, Rab K., Dawson, Sarah-Jane, and Chao, Jeffrey A.
- Subjects
- *
GENETIC transcription , *RESPONSE inhibition , *EPIGENETICS , *MESSENGER RNA , *INVECTIVE - Abstract
Several transcription inhibitors have been developed as cancer therapies. However, they show modest clinical activity, highlighting that our understanding of the cellular response to transcriptional inhibition remains incomplete. Here we report that potent inhibitors of transcription not only impact mRNA output but also markedly impair mRNA transcript localization and nuclear export. We demonstrate that retention of newly transcribed mRNA in nuclear speckles is an adaptive response to chemically distinct transcriptional inhibitors. Retained transcripts are fully processed and accumulate in proportion to the expression level of the genes from which they emanate. The TREX mRNA export complex plays an integral role in directing nascent transcripts to nuclear speckles where they are bound to NXF1, protected from degradation, and poised for rapid export following re-initiation of transcription. Our findings provide new insights into the crosstalk between transcription and mRNA export with important implications for drugs aiming to inhibit transcription for therapeutic gain. [Display omitted] • Global inhibition of transcription impairs mRNA localization and nuclear export • TREX directs mRNAs to the nuclear speckle where they are bound by NXF1 • Mature, export-competent mRNAs are retained and protected in nuclear speckles • Reinitiation of transcription leads to rapid mRNA export preserving cell identity Transcription inhibitors also disrupt nuclear export. Here, Williams et al. reveal that mRNA export factors sense transcription inhibition and adapt by storing mature export-competent mRNA in nuclear speckles. This enables rapid release when transcription resumes and ensures retention of cellular identity and viability during a transient global transcription insult. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
33. RCC1 regulation of subcellular protein localization via Ran GTPase drives pancreatic ductal adenocarcinoma growth.
- Author
-
Bannoura, Sahar F., Aboukameel, Amro, Khan, Husain Yar, Uddin, Md Hafiz, Jang, Hyejeong, Beal, Eliza W, Thangasamy, Amalraj, Shi, Yang, Kim, Seongho, Wagner, Kay-Uwe, Beydoun, Rafic, El-Rayes, Bassel F., Philip, Philip A., Mohammad, Ramzi M., Saif, Muhammad Wasif, Al-Hallak, Mohammed Najeeb, Pasche, Boris C., and Azmi, Asfar S.
- Subjects
- *
GENE expression , *NUCLEOCYTOPLASMIC interactions , *NUCLEAR transport (Cytology) , *HUMAN cell cycle , *NUCLEAR proteins , *PANCREATIC tumors - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy, with limited therapeutic options. Here, we evaluated the role of regulator of chromosome condensation 1 (RCC1) in PDAC. RCC1 functions as a guanine exchange factor for GTP-binding nuclear protein Ran (Ran) GTPase and is involved in nucleocytoplasmic transport. RCC1 RNA expression is elevated in PDAC tissues compared to normal pancreatic tissues and correlates with poor prognosis. RCC1 silencing by RNAi and CRISPR-Cas9 knockout (KO) results in reduced proliferation in 2-D and 3-D cell cultures. RCC1 knockdown (KD) reduced migration and clonogenicity, enhanced apoptosis, and altered cell cycle progression in human PDAC and murine cells from LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx1-Cre (KPC) tumors. Mechanistically, RCC1 KO shows widespread transcriptomic alterations including regulation of PTK7, a co-receptor of the Wnt signaling pathway. RCC1 KD disrupted subcellular Ran localization and the Ran gradient. Nuclear and cytosolic proteomics revealed altered subcellular proteome localization in Rcc1 KD KPC-tumor-derived cells and several altered metabolic biosynthesis pathways. In vivo, RCC1 KO cells show reduced tumor growth potential when injected as sub-cutaneous xenografts. Finally, RCC1 KD sensitized PDAC cells to gemcitabine chemotherapy treatment. This study reveals the role of RCC1 in pancreatic cancer as a novel molecular vulnerability that could be exploited to enhance therapeutic response. • RCC1 is overexpressed in PDAC thus driving proliferation and progression. • Silencing of RCC1 reduced viability and enhanced apoptosis of PDAC cells. • Ran is regulated by RCC1 which is essential for subcellular proteome organization. • Tumor growth was arrested in RCC1 knockout PDAC xenograft mouse models. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. Live Cell Imaging Reveals HBV Capsid Translocation from the Nucleus To the Cytoplasm Enabled by Cell Division
- Author
-
Sofia Romero, Nuruddin Unchwaniwala, Edward L. Evans, Kevin W. Eliceiri, Daniel D. Loeb, and Nathan M. Sherer
- Subjects
core protein ,hepatitis B virus ,live cell imaging ,nuclear export ,subcellular trafficking ,virus assembly ,Microbiology ,QR1-502 - Abstract
ABSTRACT Hepatitis B virus (HBV) capsid assembly is traditionally thought to occur predominantly in the cytoplasm, where the virus gains access to the virion egress pathway. To better define sites of HBV capsid assembly, we carried out single cell imaging of HBV Core protein (Cp) subcellular trafficking over time under conditions supporting genome packaging and reverse transcription in Huh7 hepatocellular carcinoma cells. Time-course analyses including live cell imaging of fluorescently tagged Cp derivatives showed Cp to accumulate in the nucleus at early time points (~24 h), followed by a marked re-distribution to the cytoplasm at 48 to 72 h. Nucleus-associated Cp was confirmed to be capsid and/or high-order assemblages using a novel dual label immunofluorescence strategy. Nuclear-to-cytoplasmic re-localization of Cp occurred predominantly during nuclear envelope breakdown in conjunction with cell division, followed by strong cytoplasmic retention of Cp. Blocking cell division resulted in strong nuclear entrapment of high-order assemblages. A Cp mutant, Cp-V124W, predicted to exhibit enhanced assembly kinetics, also first trafficked to the nucleus to accumulate at nucleoli, consistent with the hypothesis that Cp’s transit to the nucleus is a strong and constitutive process. Taken together, these results provide support for the nucleus as an early-stage site of HBV capsid assembly, and provide the first dynamic evidence of cytoplasmic retention after cell division as a mechanism underpinning capsid nucleus-to-cytoplasm relocalization. IMPORTANCE Hepatitis B virus (HBV) is an enveloped, reverse-transcribing DNA virus that is a major cause of liver disease and hepatocellular carcinoma. Subcellular trafficking events underpinning HBV capsid assembly and virion egress remain poorly characterized. Here, we developed a combination of fixed and long-term (>24 h) live cell imaging technologies to study the single cell trafficking dynamics of the HBV Core Protein (Cp). We demonstrate that Cp first accumulates in the nucleus, and forms high-order structures consistent with capsids, with the predominant route of nuclear egress being relocalization to the cytoplasm during cell division in conjunction with nuclear membrane breakdown. Single cell video microscopy demonstrated unequivocally that Cp's localization to the nucleus is constitutive. This study represents a pioneering application of live cell imaging to study HBV subcellular transport, and demonstrates links between HBV Cp and the cell cycle.
- Published
- 2023
- Full Text
- View/download PDF
35. Nuclear export inhibition jumbles epithelial–mesenchymal states and gives rise to migratory disorder in healthy epithelia
- Author
-
Carly M Krull, Haiyi Li, and Amit Pathak
- Subjects
epithelial cells ,nuclear export ,nucleocytoplasmic transport ,epithelial–mesenchymal transition ,EMT ,mechanobiology ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Dynamic nucleocytoplasmic transport of E-M factors regulates cellular E-M states; yet, it remains unknown how simultaneously trapping these factors affects epithelia at the macroscale. To explore this question, we performed nuclear export inhibition (NEI) via leptomycin B and Selinexor treatment, which biases nuclear localization of CRM1-associated E-M factors. We examined changes in collective cellular phenotypes across a range of substrate stiffnesses. Following NEI, soft substrates elevate collective migration of MCF10A cells for up to 24 hr, while stiffer substrates reduce migration at all time points. Our results suggest that NEI disrupts migration through competition between intercellular adhesions and mechanoactivation, generally causing loss of cell–cell coordination. Specifically, across substrate stiffnesses, NEI fosters an atypical E-M state wherein MCF10A cells become both more epithelial and more mesenchymal. We observe that NEI fosters a range of these concurrent phenotypes, from more epithelial shYAP MCF10A cells to more mesenchymal MDCK II cells. α-Catenin emerges as a potential link between E-M states, where it maintains normal levels of intercellular adhesion and transmits mechanoactive characteristics to collective behavior. Ultimately, to accommodate the concurrent states observed here, we propose an expanded E-M model, which may help further understand fundamental biological phenomena and inform pathological treatments.
- Published
- 2023
- Full Text
- View/download PDF
36. Cytoplasmic Amplification of Transcriptional Noise Generates Substantial Cell-to-Cell Variability
- Author
-
Hansen, Maike MK, Desai, Ravi V, Simpson, Michael L, and Weinberger, Leor S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,Human Genome ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,Biological Variation ,Population ,Cytoplasm ,Embryonic Stem Cells ,HEK293 Cells ,Humans ,Jurkat Cells ,Mice ,Models ,Theoretical ,RNA Processing ,Post-Transcriptional ,RNA ,Messenger ,Single Molecule Imaging ,Single-Cell Analysis ,Transcriptional Activation ,bursting ,mRNA degradation ,mathematical modeling ,noise amplification ,noise attenuation ,nuclear export ,single molecule RNA FISH ,stochastic noise ,transcription ,translation ,Biochemistry and cell biology - Abstract
Transcription is an episodic process characterized by probabilistic bursts, but how the transcriptional noise from these bursts is modulated by cellular physiology remains unclear. Using simulations and single-molecule RNA counting, we examined how cellular processes influence cell-to-cell variability (noise). The results show that RNA noise is higher in the cytoplasm than the nucleus in ∼85% of genes across diverse promoters, genomic loci, and cell types (human and mouse). Measurements show further amplification of RNA noise in the cytoplasm, fitting a model of biphasic mRNA conversion between translation- and degradation-competent states. This multi-state translation-degradation of mRNA also causes substantial noise amplification in protein levels, ultimately accounting for ∼74% of intrinsic protein variability in cell populations. Overall, the results demonstrate how noise from transcriptional bursts is intrinsically amplified by mRNA processing, leading to a large super-Poissonian variability in protein levels.
- Published
- 2018
37. The THO Complex Coordinates Transcripts for Synapse Development and Dopamine Neuron Survival
- Author
-
Maeder, Celine I, Kim, Jae-Ick, Liang, Xing, Kaganovsky, Konstantin, Shen, Ao, Li, Qin, Li, Zhaoyu, Wang, Sui, Xu, XZ Shawn, Li, Jin Billy, Xiang, Yang Kevin, Ding, Jun B, and Shen, Kang
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Genetics ,Biological Sciences ,Brain Disorders ,Neurosciences ,1.1 Normal biological development and functioning ,Generic health relevance ,Neurological ,Active Transport ,Cell Nucleus ,Amyotrophic Lateral Sclerosis ,Animals ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Calcium Signaling ,Cell Nucleus ,Cyclic AMP Response Element-Binding Protein ,Dopaminergic Neurons ,Female ,Humans ,Male ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,Mutagenesis ,Mutation ,Missense ,Nuclear Proteins ,Protein Subunits ,Synapses ,CREB ,THO complex ,coordinated genetic program ,dopamine neurons ,neurodegeneration ,nuclear export ,presynapse assembly ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Synaptic vesicle and active zone proteins are required for synaptogenesis. The molecular mechanisms for coordinated synthesis of these proteins are not understood. Using forward genetic screens, we identified the conserved THO nuclear export complex (THOC) as an important regulator of presynapse development in C. elegans dopaminergic neurons. In THOC mutants, synaptic messenger RNAs are retained in the nucleus, resulting in dramatic decrease of synaptic protein expression, near complete loss of synapses, and compromised dopamine function. CRE binding protein (CREB) interacts with THOC to mark synaptic transcripts for efficient nuclear export. Deletion of Thoc5, a THOC subunit, in mouse dopaminergic neurons causes severe defects in synapse maintenance and subsequent neuronal death in the substantia nigra compacta. These cellular defects lead to abrogated dopamine release, ataxia, and animal death. Together, our results argue that nuclear export mechanisms can select specific mRNAs and be a rate-limiting step for neuronal differentiation and survival.
- Published
- 2018
38. Nuclear Export in Non-Hodgkin Lymphoma and Implications for Targeted XPO1 Inhibitors.
- Author
-
Trkulja, Kyla L., Manji, Farheen, Kuruvilla, John, and Laister, Rob C.
- Subjects
- *
NON-Hodgkin's lymphoma , *IMMUNOMODULATORS , *MESSENGER RNA , *TRANSCRIPTION factors , *GENE expression , *NON-coding RNA , *RITUXIMAB - Abstract
Exportin-1 (XPO1) is a key player in the nuclear export pathway and is overexpressed in almost all cancers. This is especially relevant for non-Hodgkin lymphoma (NHL), where high XPO1 expression is associated with poor prognosis due to its oncogenic role in exporting proteins and RNA that are involved in cancer progression and treatment resistance. Here, we discuss the proteins and RNA transcripts that have been identified as XPO1 cargo in NHL lymphoma including tumour suppressors, immune modulators, and transcription factors, and their implications for oncogenesis. We then highlight the research to date on XPO1 inhibitors such as selinexor and other selective inhibitors of nuclear export (SINEs), which are used to treat some cases of non-Hodgkin lymphoma. In vitro, in vivo, and clinical studies investigating the anti-cancer effects of SINEs from bench to bedside, both as a single agent and in combination, are also reported. Finally, we discuss the limitations of the current research landscape and future directions to better understand and improve the clinical utility of SINE compounds in NHL. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
39. Nucleo-Cytoplasmic Transport of ZIKV Non-Structural 3 Protein Is Mediated by Importin-α/β and Exportin CRM-1.
- Author
-
Adrián De Jesús-González, Luis, Noé Palacios-Rápalo, Selvin, Manuel Reyes-Ruiz, José, Fidel Osuna-Ramos, Juan, Noe Farfán-Morales, Carlos, Daniel Cordero-Rivera, Carlos, Cisneros, Bulmaro, Gutiérrez-Escolano, Ana Lorena, and María del Ángela, Rosa
- Subjects
- *
NUCLEAR membranes , *VIRAL proteins , *CELL nuclei , *CELL morphology , *NUCLEAR proteins - Abstract
Flaviviruses have a cytoplasmic replicative cycle, and crucial events, such as genome translation and replication, occur in the endoplasmic reticulum. However, some viral proteins, such as C, NS1, and NS5 from Zika virus (ZIKV) containing nuclear localization signals (NLSs) and nuclear export signals (NESs), are also located in the nucleus of Vero cells. The NS2A, NS3, and NS4A proteins from dengue virus (DENV) have also been reported to be in the nucleus of A549 cells, and our group recently reported that the NS3 protein is also located in the nucleus of Huh7 and C636 cells during DENV infection. However, the NS3 protease-helicase from ZIKV locates in the perinuclear region of infected cells and alters the morphology of the nuclear lamina, a component of the nuclear envelope. Furthermore, ZIKV NS3 has been reported to accumulate on the concave face of altered kidney-shaped nuclei and may be responsible for modifying other elements of the nuclear envelope. However, nuclear localization of NS3 from ZIKV has not been substantially investigated in human host cells. Our group has recently reported that DENV and ZIKV NS3 alter the nuclear pore complex (NPC) by cleaving some nucleoporins. Here, we demonstrate the presence of ZIKV NS3 in the nucleus of Huh7 cells early in infection and in the cytoplasm at later times postinfection. In addition, we found that ZIKV NS3 contains an NLS and a putative NES and uses the classic import (importin-a/b) and export pathway via CRM-1 to be transported between the cytoplasm and the nucleus. IMPORTANCE Flaviviruses have a cytoplasmic replication cycle, but recent evidence indicates that nuclear elements play a role in their viral replication. Viral proteins, such as NS5 and C, are imported into the nucleus, and blocking their import prevents replication. Because of the importance of the nucleus in viral replication and the role of NS3 in the modification of nuclear components, we investigated whether NS3 can be localized in the nucleus during ZIKV infection. We found that NS3 is imported into the nucleus via the importin pathway and exported to the cytoplasm via CRM-1. The significance of viral protein nuclear import and export and its relationship with infection establishment is highlighted, emphasizing the development of new host-directed antiviral therapeutic strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. ATM phosphorylates PP2A subunit A resulting in nuclear export and spatiotemporal regulation of the DNA damage response.
- Author
-
Sule, Amrita, Golding, Sarah E., Ahmad, Syed F., Watson, James, Ahmed, Mostafa H., Kellogg, Glen E., Bernas, Tytus, Koebley, Sean, Reed, Jason C., Povirk, Lawrence F., and Valerie, Kristoffer
- Abstract
Ataxia telangiectasia mutated (ATM) is a serine–threonine protein kinase and important regulator of the DNA damage response (DDR). One critical ATM target is the structural subunit A (PR65–S401) of protein phosphatase 2A (PP2A), known to regulate diverse cellular processes such as mitosis and cell growth as well as dephosphorylating many proteins during the recovery from the DDR. We generated mouse embryonic fibroblasts expressing PR65-WT, -S401A (cannot be phosphorylated), and -S401D (phospho-mimetic) transgenes. Significantly, S401 mutants exhibited extensive chromosomal aberrations, impaired DNA double-strand break (DSB) repair and underwent increased mitotic catastrophe after radiation. Both S401A and the S401D cells showed impaired DSB repair (nonhomologous end joining and homologous recombination repair) and exhibited delayed DNA damage recovery, which was reflected in reduced radiation survival. Furthermore, S401D cells displayed increased ERK and AKT signaling resulting in enhanced growth rate further underscoring the multiple roles ATM–PP2A signaling plays in regulating prosurvival responses. Time-lapse video and cellular localization experiments showed that PR65 was exported to the cytoplasm after radiation by CRM1, a nuclear export protein, in line with the very rapid pleiotropic effects observed. A putative nuclear export sequence (NES) close to S401 was identified and when mutated resulted in aberrant PR65 shuttling. Our study demonstrates that the phosphorylation of a single, critical PR65 amino acid (S401) by ATM fundamentally controls the DDR, and balances DSB repair quality, cell survival and growth by spatiotemporal PR65 nuclear–cytoplasmic shuttling mediated by the nuclear export receptor CRM1. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
41. Multiplexed cellular profiling identifies an organoselenium compound as an inhibitor of CRM1‐mediated nuclear export.
- Author
-
Jimenez, Lucia, Mayoral‐Varo, Victor, Amenábar, Carlos, Ortega, Judit, Sequeira, João G. N., Machuqueiro, Miguel, Mourato, Cristiana, Silvestri, Romano, Angeli, Andrea, Carta, Fabrizio, Supuran, Claudiu T., Megías, Diego, Ferreira, Bibiana I., and Link, Wolfgang
- Subjects
- *
ORGANOSELENIUM compounds , *GREEN fluorescent protein , *VIRAL proteins , *FLUORESCENT proteins , *CHIMERIC proteins , *SELENOPROTEINS - Abstract
Chromosomal region maintenance 1 (CRM1 also known as Xpo1 and exportin‐1) is the receptor for the nuclear export controlling the intracellular localization and function of many cellular and viral proteins that play a crucial role in viral infections and cancer. The inhibition of CRM1 has emerged as a promising therapeutic approach to interfere with the lifecycle of many viruses, for the treatment of cancer, and to overcome therapy resistance. Recently, selinexor has been approved as the first CRM1 inhibitor for the treatment of multiple myeloma, providing proof of concept for this therapeutic option with a new mode of action. However, selinexor is associated with dose‐limiting toxicity and hence, the discovery of alternative small molecule leads that could be developed as less toxic anticancer and antiviral therapeutics will have a significant impact in the clinic. Here, we report a CRM1 inhibitor discovery platform. The development of this platform includes reporter cell lines that monitor CRM1 activity by using red fluorescent protein or green fluorescent protein‐labeled HIV‐1 Rev protein with a strong heterologous nuclear export signal. Simultaneously, the intracellular localization of other proteins, to be interrogated for their capacity to undergo CRM1‐mediated export, can be followed by co‐culturing stable cell lines expressing fluorescent fusion proteins. We used this platform to interrogate the mode of nuclear export of several proteins, including PDK1, p110α, STAT5A, FOXO1, 3, 4 and TRIB2, and to screen a compound collection. We show that while p110α partially relies on CRM1‐dependent nuclear export, TRIB2 is exported from the nucleus in a CRM1‐independent manner. Compound screening revealed the striking activity of an organoselenium compound on the CRM1 nuclear export receptor. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
42. Nuclear transport and subcellular localization of the dystrophin Dp71 and Dp40 isoforms in the PC12 cell line.
- Author
-
Sánchez, Alberto, Aragón, Jorge, Ceja, Víctor, Rendon, Alvaro, and Montanez, Cecilia
- Subjects
- *
DYSTROPHIN , *CELL lines , *ALTERNATIVE RNA splicing , *CELL proliferation , *CELL culture , *CELL nuclei - Abstract
The shortest dystrophins, Dp71 and Dp40, are transcribed from the DMD gene through an internal promoter located in intron 62. These proteins are the main product of the DMD gene in the nervous system and have been involved in various functions related to cellular differentiation and proliferation as well as other cellular processes. Dp71 mRNA undergoes alternative splicing that results in different Dp71 protein isoforms. The subcellular localization of some of these isoforms in the PC12 cell line has been previously reported, and a differential subcellular distribution was observed, which suggests a particular role for each isoform. With the aim of obtaining information on their function, this study identified factors involved in the nuclear transport of Dp71 and Dp40 isoforms in the PC12 cell line. Cell cultures were treated with specific nuclear import/export inhibitors to determine the Dp71 isoform transport routes. The results showed that all isoforms of Dp71 and Dp40 included in the analysis have the ability to enter the cell nucleus through α/β importin, and the main route of nuclear export for Dp71 isoforms is through the exportin CRM1, which is not the case for Dp40. • All dystrophin Dp71 isoforms and Dp40 are transported to the nucleus. • Dp71 isoforms are mainly exported from the nucleus by the exportin CRM1. • Dp71 isoforms and Dp40 are imported to the nucleus by the importin α/β. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
43. Stimulus‐specific remodeling of the neuronal transcriptome through nuclear intron‐retaining transcripts.
- Author
-
Mazille, Maxime, Buczak, Katarzyna, Scheiffele, Peter, and Mauger, Oriane
- Subjects
- *
TRANSCRIPTOMES , *NUCLEAR membranes , *ALTERNATIVE RNA splicing , *RNA splicing , *GENE expression , *CYTOSOL , *CELLULAR signal transduction - Abstract
The nuclear envelope has long been considered primarily a physical barrier separating nuclear and cytosolic contents. More recently, nuclear compartmentalization has been shown to have additional regulatory functions in controlling gene expression. A sizeable proportion of protein‐coding mRNAs is more prevalent in the nucleus than in the cytosol, suggesting regulated mRNA trafficking to the cytosol, but the mechanisms underlying controlled nuclear mRNA retention remain unclear. Here, we provide a comprehensive map of the subcellular localization of mRNAs in mature mouse cortical neurons, and reveal that transcripts retained in the nucleus comprise the majority of stable intron‐retaining mRNAs. Systematically probing the fate of nuclear transcripts upon neuronal stimulation, we found opposite effects on sub‐populations of transcripts: while some are targeted for degradation, others complete splicing to generate fully mature mRNAs that are exported to the cytosol and mediate rapid increases in protein levels. Finally, different forms of stimulation mobilize distinct groups of intron‐retaining transcripts, with this selectivity arising from the activation of specific signaling pathways. Overall, our findings uncover a cue‐specific control of intron retention as a major regulator of acute remodeling of the neuronal transcriptome. Synopsis: Regulated intron retention represents a mechanism for the control of gene expression. In neurons, this is shown here to affect nuclear retention of mRNAs in response to specific neuronal stimuli. Subcellular mapping of transcripts shows that a large portion of stable intron‐retaining mRNAs is localized to the nucleus. Introns retained in nuclear‐stored transcripts resemble canonically spliced introns and their splicing can be regulated by neuronal stimuli.In response to neuronal stimulation, a subset of intron‐retaining transcripts complete their splicing, are exported to the cytosol, and are recruited for rapid protein synthesis.Distinct sub‐populations of intron‐retaining transcripts are regulated in response to specific neuronal cues.Distinct signaling pathways regulate neuronal stimulus‐dependent regulation of nuclear intron retention transcripts. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
44. Potential of PAR-4 as a Therapeutic Target for Pancreatic Cancer
- Author
-
Uddin, Md. Hafiz, Azmi, Asfar S., Mohammad, Ramzi M., and Rangnekar, Vivek M., editor
- Published
- 2021
- Full Text
- View/download PDF
45. Multifaceted Effects of Ligand on Nuclear Receptor Mobility
- Author
-
Allison, Lizabeth A., Roggero, Vincent R., and Badr, Mostafa Z., editor
- Published
- 2021
- Full Text
- View/download PDF
46. Multiplexed Dual-Color Fluorescence-Based Distinction Between Nuclear Trapping and Translocation of FOXO3.
- Author
-
Amenabar C, Jimenez L, Mourato C, Mayoral-Varo V, Megías D, Ferreira BI, and Link W
- Subjects
- Humans, Receptors, Cytoplasmic and Nuclear metabolism, Exportin 1 Protein, Karyopherins metabolism, rev Gene Products, Human Immunodeficiency Virus metabolism, rev Gene Products, Human Immunodeficiency Virus genetics, Protein Transport, Cell Line, Nuclear Localization Signals metabolism, Genes, Reporter, Luminescent Proteins metabolism, Luminescent Proteins genetics, Green Fluorescent Proteins metabolism, Green Fluorescent Proteins genetics, HEK293 Cells, Nuclear Export Signals, Forkhead Box Protein O3 metabolism, Forkhead Box Protein O3 genetics, Cell Nucleus metabolism, Active Transport, Cell Nucleus
- Abstract
FOXO3 is a transcription factor that mainly exerts its functions in the cell nucleus. The amino acid sequence of FOXO3 contains a nuclear localization sequence (NLS) and a nuclear export sequence (NES) allowing for nuclear/cytoplasmic shuttling that plays an important role in regulating FOXO3 activity. Nuclear accumulation of FOXO3 proteins can be the result of translocation to the nucleus triggered by upstream regulatory input or trapping of FOXO3 within the nucleus through the inhibition of its nuclear export via the receptor CRM1. In order to distinguish these two modes of FOXO3 activation, we have generated a multiplexed assay. The development of this platform includes a reporter cell line that monitors CRM1 activity by using RFP-labeled HIV-1 Rev. protein with a strong heterologous NES. Simultaneously, the intracellular localization of FOXO3 can be monitored by a second cell line stably expressing GFP-FOXO3. Here we describe a detailed protocol on how to co-culture these reporter cell lines and use them to interrogate compound-induced FOXO3 activation in order to understand the mode of action., (© 2025. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2025
- Full Text
- View/download PDF
47. Autophagy Modulates Glioblastoma Cell Sensitivity to Selinexor-mediated XPO1 inhibition.
- Author
-
Tang Y, Sprinzen L, Terada Y, Kiang KM, Li C, Zeng Y, Liu F, Zhou H, Liang X, Zhang J, Pieper RO, Chen B, and Zhang L
- Abstract
Background: Selinexor is a selective inhibitor of exportin-1 (XPO1), a key mediator of the nucleocytoplasmic transport for molecules critical to tumor cell survival. Selinexor's lethality is generally associated with the induction of apoptosis, and in some cases, with autophagy-induced apoptosis. We performed this study to determine Selinexor's action in glioblastoma (GBM) cells, which are notoriously resistant to apoptosis., Methods: Patient-derived GBM cells were treated with Selinexor, and drug response and autophagy levels were monitored. Homozygous C528S XPO1 mutant GBM43 cells were generated by CRISPR/Cas9 editing. Single Selinexor or combination treatment with autophagy inhibitors was evaluated. In addition, bulk-tissue, single-cell, and spatial transcriptome were analyzed, and molecular docking was performed., Results: Although all cell lines exhibited a dose- and time-dependent reduction of cell viability, the most profound molecular response to Selinexor was induction of autophagy instead of apoptosis. Selinexor-induced autophagy was an on-target consequence of XPO1 inhibition, and could be mitigated by expression of a mutant, Selinexor-resistant form of XPO1, and Selinexor-induced autophagy was related at least in part to nuclear trapping of the transcription factor TFEB. Furthermore, genetic or pharmacologic suppression of autophagy sensitized the cells to Selinexor-induced toxicity in association with the induction of apoptosis. Finally, in intracranial PDX studies, the combination of Selinexor with the autophagy inhibitor chloroquine significantly impeded tumor growth and extended mouse survival relative to single-agent treatment., Conclusion: These results suggest that activation of autophagy confers a protective mechanism against Selinexor in GBM cells, and that the combination of Selinexor with autophagy inhibitors may serve as a viable means to enhance Selinexor-induced cell death., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2024
- Full Text
- View/download PDF
48. Functional dissection of N-terminal nuclear trafficking signals of SETDB1
- Author
-
Jaemin Eom, Kyuheum Jeon, Jung Sun Park, and Yong-Kook Kang
- Subjects
NES ,NLS ,nuclear import ,nuclear export ,ATF7IP ,PML ,Biology (General) ,QH301-705.5 - Abstract
SETDB1 is a histone H3-lysine 9-specific methyltransferase that fulfills epigenetic functions inside the nucleus; however, when overexpressed, SETDB1 majorily localizes in the cytoplasm. SETDB1 has a single nuclear-localization-signal (NLS) motif and two successive nuclear-export-signal (NES1 and NES2) motifs in the N-terminus, suggesting that SETDB1 localization is the consequence of a balance between the two antithetic motifs. Here, we performed a series of motif deletions to characterize their effects on the cellular movement of SETDB1. Given the cytoplasmic localization of GFP-SETDB1 in the whole form, without the NES motifs, GFP-SETDB1 was not nuclear, and 3xNLS addition plus NES removal held the majority of GFP-SETDB1 within the nucleus. The results indicated that the cytoplasmic localization of GFP-SETDB1 is the combined result of weak NLS and robust NESs. In ATF7IP-overexpressing cells, GFP-SETDB1 entered the nucleus only in the presence of the NES1 motif; neither the NES2 nor NLS motif was necessary. Since subcellular fractionation results showed that ATF7IP was nuclear-only, an intermediary protein may interact specifically with the NES1 motif after stimulation by ATF7IP. When GFP-SETDB1 had either NES1 or NES2, it was precipitated (in immunoprecipitation) and colocalized (in immunofluorescence) with ATF7IP, indicating that GFP-SETDB1 interacts with ATF7IP through the NES motifs in the nucleus. The regulated nuclear entry of SETDB1 is assumed to set a tight restriction on its abundance within the nucleus, thereby ensuring balanced nuclear SETDB1 levels.
- Published
- 2022
- Full Text
- View/download PDF
49. Investigation of Reversible Covalent Inhibitors of CRM1
- Author
-
Vo, Duy Anh
- Subjects
Organic chemistry ,Biochemistry ,covalent inhibitor ,CRM1 ,intrinsic reversibility ,Leptomycin B ,natural product ,nuclear export - Abstract
The Chromosome Region of Maintenance 1 or CRM1 protein is the master regulator that handles the export of hundreds of proteins and RNA molecules from eukaryotic cell nuclei. The imbalance of highly regulated cellular pathways due to overexpression of CRM1 is a distinct characteristic in forms of cancer. The first compound found to inhibit CRM1-dependent nuclear export was the natural drug Leptomycin B (LMB), which blocks export by competitively interacting with a highly conserved cleft on CRM1 required for nuclear export signal recognition. Previous work has clarified that CRM1 inhibition by LMB is through an irreversible covalent inhibition, while second-generation inhibitors were designed through a slowly reversible covalent inhibition, leading to compounds with greatly improved tolerability that are in Phase I/II clinical trials. Chemically, the mechanism of action can be attributed to the Michael Addition between a nucleophilic cysteine residue and an electrophilic inhibitor. The work in this dissertation demonstrates the predictability and tunable of the reversibility of CRM1 inhibition. In particular, third-generation CRM1 inhibitors were designed with specific electronic nature at the C-α position in order to increase the acidity of the α-proton and potentially enhance the reversible Michael Addition and deconjugation from CRM1. We anticipate that the computational and experimental methods described in this dissertation will improve understanding of the therapeutic target, CRM1, and can be applied to similar targets.
- Published
- 2023
50. Pepper SUMO E3 ligase CaDSIZ1 enhances drought tolerance by stabilizing the transcription factor CaDRHB1.
- Author
-
Joo, Hyunhee, Lim, Chae Woo, and Lee, Sung Chul
- Subjects
- *
UBIQUITIN ligases , *DROUGHT tolerance , *TRANSCRIPTION factors , *UBIQUITINATION , *POST-translational modification , *CAPSICUM annuum , *PEPPERS - Abstract
Summary: Small ubiquitin‐like modifier (SUMO) conjugation (SUMOylation) is a reversible post‐translational modification associated with protein stability and activity, and modulates hormone signaling and stress responses in plants. Previously, we reported that the pepper dehydration‐responsive homeobox domain transcription factor CaDRHB1 acts as a positive modulator of drought response.Here, we show that CaDRHB1 protein stability is enhanced by SUMO E3 ligase Capsicum annuum DRHB1‐interacting SAP and Miz domain (SIZ1) (CaDSIZ1)‐mediated SUMOylation in response to drought, thereby positively modulating abscisic acid (ABA) signaling and drought responses. Substituting lysine (K) 138 of CaDRHB1 with arginine reduced CaDSIZ1‐mediated SUMOylation, indicating that K138 is the principal site for SUMO conjugation.Virus‐induced silencing of CaDSIZ1 promoted CaDRHB1 degradation, suggesting that CaDSIZ1 is involved in drought‐induced SUMOylation of CaDRHB1. CaDSIZ1 interacted with and facilitated SUMO conjugation of CaDRHB1. CaDRHB1, mainly localized in the nucleus, but also in the cytoplasm in the SUMOylation mimic state, suggesting that SUMOylation of CaDRHB1 promotes its nuclear export, leading to cytoplasmic accumulation.Moreover, CaDSIZ1‐silenced pepper plants were less sensitive to ABA and considerably sensitive to drought stress, whereas CaDSIZ1‐overexpressing plants displayed ABA‐hypersensitive and drought‐tolerant phenotypes. Collectively, our data indicate that CaDSIZ1‐mediated SUMOylation of CaDRHB1 functions in ABA‐mediated drought tolerance. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.