3,039 results on '"Protein Translation"'
Search Results
2. Influence of 5′-UTR nucleotide composition on translation efficiency in Escherichiacoli
- Author
-
Li, Jinjin, Li, Jiaojiao, Li, Peixian, Zhang, Jie, Liu, Qian, and Qi, Hao
- Published
- 2025
- Full Text
- View/download PDF
3. Structural dynamics of human ribosomes in situ reconstructed by exhaustive high-resolution template matching
- Author
-
Rickgauer, J. Peter, Choi, Heejun, Moore, Andrew S., Denk, Winfried, and Lippincott-Schwartz, Jennifer
- Published
- 2024
- Full Text
- View/download PDF
4. Knotty is nice: Metabolite binding and RNA-mediated gene regulation by the preQ1 riboswitch family
- Author
-
Kiliushik, Daniil, Goenner, Coleman, Law, Matthew, Schroeder, Griffin M., Srivastava, Yoshita, Jenkins, Jermaine L., and Wedekind, Joseph E.
- Published
- 2024
- Full Text
- View/download PDF
5. IGF2BP1-mediated the stability and protein translation of FGFR1 mRNA regulates myogenesis through the ERK signaling pathway
- Author
-
Liu, Zhipeng, Deng, Kaiping, Su, Yalong, Zhang, Zhen, Shi, Chongyu, Wang, Jingang, Fan, Yixuan, Zhang, Guomin, and Wang, Feng
- Published
- 2024
- Full Text
- View/download PDF
6. Making Proteins with Electricity
- Author
-
McCaig, Colin D., Deshpande, R. D., Series Editor, and McCaig, Colin D.
- Published
- 2025
- Full Text
- View/download PDF
7. Targeting TUBB2B inhibits triple-negative breast cancer growth and brain-metastatic colonization.
- Author
-
He, Qingling, Hu, Jianyang, Ngo, Fung-Yin, Zhang, Huiqi, He, Lin, Huang, Hao, Wu, Tan, Pan, Yilin, Yang, Zihan, Jiang, Yuanyuan, Cho, William C., Cheuk, Wah, Tse, Gary M., Tsang, Julia Y., Yang, Mengsu, Zhang, Liang, Wang, Xin, Lo, Pui-Chi, Lau, C. Geoffrey, and Chin, Y. Rebecca
- Subjects
- *
TRIPLE-negative breast cancer , *ELONGATION factors (Biochemistry) , *MEDICAL sciences , *BRAIN metastasis , *BREAST cancer prognosis - Abstract
Background: The triple-negative subtype of breast cancer is particularly challenging to treat due to its aggressiveness with a high risk of brain metastasis, and the lack of effective targeted therapies. Tubulin beta 2B class IIb (TUBB2B), a β-tubulin isoform regulating axon guidance during embryonic development, was found to be overexpressed in various types of cancers including triple-negative breast cancer (TNBC). However, its functional roles in breast cancer or metastasis remain unclear. Methods: To identify TUBB2B as a novel molecular target in TNBC, we performed bioinformatics analysis to assess the association of TUBB2B expression and survival of patients. RNAscope in situ hybridization was used to examine TUBB2B expression in clinical breast tumor samples. The effect of TUBB2B knockdown on TNBC growth and brain metastasis colonization was evaluated by in vitro and in vivo assays. Mass spectrometry (MS) and biochemical experiments were performed to explore the underlying mechanisms. Preclinical efficacy of targeting TUBB2B was determined in xenograft studies using the siRNA-gold nanoparticle (siRNA-AuNP) approach. Results: TUBB2B, but not other β-tubulin isoforms, is frequently overexpressed in TNBC primary tumors as well as brain metastases. We also find that upregulation of TUBB2B is associated with poor prognosis in breast cancer patients. Silencing TUBB2B induces tumor cell death and inhibits the outgrowth of brain metastasis. Mechanistically, we identify eukaryotic translation elongation factor 1 alpha 1 (eEF1A1) as a novel interacting partner of TUBB2B, revealing a previously unexplored role of TUBB2B in translational regulation. In line with its neural-related functions, TUBB2B overexpression in TNBC cells activates astrocytes, which in turn upregulate TUBB2B in tumor cells. These findings suggest a feed-forward interaction between TUBB2B in TNBC cells and astrocytes that promotes brain metastatic colonization. Furthermore, we demonstrate the potent inhibition of TNBC xenograft growth as well as brain metastatic colonization using TUBB2B siRNA-AuNP treatment, indicating potential clinical applications of targeting TUBB2B for TNBC. Conclusions: TUBB2B is a novel TNBC gene that plays a key role in promoting tumor cell survival and brain metastatic colonization, and can be targeted by siRNA-AuNPs as a treatment strategy. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
8. Translation elongation defects activate the Caenorhabditis elegans ZIP-2 bZIP transcription factor-mediated toxin defense.
- Author
-
Kniazeva, Marina and Ruvkun, Gary
- Abstract
The Caenorhabditis elegans bZIP transcription factor ZIP-2 is activated by toxins or mutations that inhibit translational elongation. The zip-2 DNA-binding protein is encoded in a downstream main open reading frame (mORF), but under normal translation elongation conditions only an upstream overlapping oORF -1 frameshifted from mORF is translated. Mutations or toxins that slow translational elongation, but not inhibitors of translational initiation or termination, activate ZIP-2. An mORF initiation codon mutation does not disrupt the normal zip-2 response to translational elongation defects, suggesting that zip-2 activation does not depend on this ATG. An mORF early termination mutant can be activated by strong translation elongation inhibition, suggesting that translation initiated upstream on oORF +1 frameshifts when elongation is inhibited to the mORF reading frame downstream of the stop codon to activate a fused oORF/mORF ZIP-2 transcription factor. The protein and DNA sequences of zip-2 oORF and mORF are conserved across the Caenorhabditis, suggesting selection for particular codons sensitive to translational elongation defects. Mutations that disrupt the oORF initiation codon constitutively activate zip-2, but not if the mORF initiation codon is also mutant, showing that zip-2 oORF competes with mORF for translational initiation. oORF initiation codon mutation-activated zip-2 slows C. elegans growth, and this slow growth is suppressed by a zip-2 null mutation. A zip-2 null mutant also strongly suppresses the growth arrest caused by translational elongation inhibitors. Thus, ZIP-2 is both a sensor of translational elongation attack, and a defense regulatory output via its activation of response genes. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
9. Dopamine D1 and NMDA Receptor Co-Regulation of Protein Translation in Cultured Nucleus Accumbens Neurons.
- Author
-
Zimbelman, Alexa R., Wong, Benjamin, Murray, Conor H., Wolf, Marina E., and Stefanik, Michael T.
- Abstract
Protein translation is essential for some forms of synaptic plasticity. Here we used fluorescent noncanonical amino acid tagging (FUNCAT) to examine whether dopamine modulates protein translation in cultured nucleus accumbens (NAc) medium spiny neurons (MSN). These neurons were co-cultured with cortical neurons to restore excitatory synapses. We measured translation in MSNs under basal conditions and after disinhibiting excitatory transmission using the GABAA receptor antagonist bicuculline (2 h). Under basal conditions, translation was not altered by the D1-class receptor (D1R) agonist SKF81297 or the D2-class receptor (D2R) agonist quinpirole. Bicuculline alone robustly increased translation. This was reversed by quinpirole but not SKF81297. It was also reversed by co-incubation with the D1R antagonist SCH23390, but not the D2R antagonist eticlopride, suggesting dopaminergic tone at D1Rs. This was surprising because no dopamine neurons are present. An alternative explanation is that bicuculline activates translation by increasing glutamate tone at NMDA receptors (NMDAR) within D1R/NMDAR heteromers. Supporting this, immunocytochemistry and proximity ligation assays revealed D1R/NMDAR heteromers on NAc cells both in vitro and in vivo, confirming previous results. Furthermore, bicuculline’s effect was reversed to the same extent by SCH23390 alone, the NMDAR antagonist APV alone, or SCH23390 + APV. These results suggest that: (1) excitatory transmission stimulates translation in NAc MSNs, (2) this is opposed when glutamate activates D1R/NMDAR heteromers, even in the absence of dopamine, and (3) antagonist occupation of D1Rs within the heteromers prevents their activation. Our study is the first to suggest a role for D2 receptors and D1R/NMDAR heteromers in regulating protein translation. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
10. The translation initiation factor eIF2 is phosphorylated to inhibit protein translation through reactive oxygen species under nutrient deficiencies in Arabidopsis.
- Author
-
Cui, Xiaona, Cao, Yuanyuan, Lv, Mengyang, Zhou, Shuhao, Chen, Meijun, Li, Chengwei, and Zhang, Hairong
- Subjects
AMINO acid synthesis ,GENETIC translation ,REACTIVE oxygen species ,LIFE sciences ,DEFICIENCY diseases ,NITROGEN - Abstract
Nitrogen (N), phosphorus (P) or potassium (K) deficiency in plants can lead to a decrease in amino acid and protein synthesis. However, it is unknown how protein translation gets repressed during macronutrient deficiencies. Previous research has shown that general control non-depressible 1 (GCN1) cooperate with GCN2 to phosphorylate the alpha subunit of eukaryotic translation initiation factor (eIF2α). In this study, we observed phosphorylation of eIF2α under N, P, and K deficiencies, which was found to be lost in gcn1. Mutant gcn1 displayed higher sensitivity to macronutrient deficiencies compared to the wild-type (WT). The evidence of in situ reactive oxygen species (ROS) accumulation in leaves indicated that macronutrient starvation triggers ROS production. Treatment with Dimethylthiourea (DMTU), a ROS scavenger, eliminated ROS and reversed eIF2α phosphorylation induced by nutrient deficiency. Moreover, it was discovered that protein translation was reduced under N or K deficiency in the WT but not in gcn1, whereas under P deprivation, protein translation was reduced in both the WT and gcn1. We additionally found that DMTU can partially recover translation inhibition under N or K deprivation. Taken together, it is concluded that GCN1-GCN2-eIF2α pathway is regulated by ROS and is essential for plant survival under macronutrient starvation conditions. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
11. Efficient mRNA Delivery In Vitro and In Vivo Using a Polycharged Biodegradable Nanomaterial.
- Author
-
Yang, Xuejin, Xiao, Jingya, Staveness, Daryl, and Zang, Xiaoyu
- Subjects
- *
LIFE sciences , *BIOLOGICAL systems , *BIOMOLECULES , *RNA editing , *MESSENGER RNA - Abstract
As RNA rises as one of the most significant modalities for clinical applications and life science research, efficient tools for delivering and integrating RNA molecules into biological systems become essential. Herein, we report a formulation using a polycharged biodegradable nano-carrier, N1-501, which demonstrates superior efficiency and versatility in mRNA encapsulation and delivery in both cell and animal models. N1-501 is a polymeric material designed to function through a facile one-step formulation process suitable for various research settings. Its capability for mRNA transfection is investigated across a wide range of mRNA doses and in different biological models, including 18 tested cell lines and mouse models. This study also comprehensively analyzes N1-501's application for mRNA transfection by examining factors such as buffer composition and pH, incubation condition, and media type. Additionally, N1-501's superior in vivo mRNA transfection capability ensures its potential as an efficient and consistent tool for advancing mRNA-based therapies and genetic research. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. 蛋白质翻译重编程在肿瘤中的研究进展.
- Author
-
范颖, 刘洋, and 刘雁勇
- Abstract
Protein translational reprogramming is an important compensatory change made by cells in response to a variety of stimuli, resulting in rapid, specific changes to the cellular proteome. In tumor cells, this reprogramming is regulated through several mechanisms, including the internal ribosome entry site (IRES), cap-independent translational enhancers (CITE), and N6-methyladenosine (m6A) modifications. These processes play pivotal roles in controlling protein translational reprogramming, which is essential for tumorigenesis, progression, and treatment resistance. Further research into the function of protein translational reprogramming in tumors may reveal novel therapeutic targets and offer new avenues for cancer treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Ageing-associated long non-coding RNA extends lifespan and reduces translation in non-dividing cells.
- Author
-
Anver, Shajahan, Sumit, Ahmed Faisal, Sun, Xi-Ming, Hatimy, Abubakar, Thalassinos, Konstantinos, Marguerat, Samuel, Alic, Nazif, and Bähler, Jürg
- Abstract
Genomes produce widespread long non-coding RNAs (lncRNAs) of largely unknown functions. We characterize aal1 (ageing-associated lncRNA), which is induced in quiescent fission yeast cells. Deletion of aal1 shortens the chronological lifespan of non-dividing cells, while ectopic overexpression prolongs their lifespan, indicating that aal1 acts in trans. Overexpression of aal1 represses ribosomal-protein gene expression and inhibits cell growth, and aal1 genetically interacts with coding genes functioning in protein translation. The aal1 lncRNA localizes to the cytoplasm and associates with ribosomes. Notably, aal1 overexpression decreases the cellular ribosome content and inhibits protein translation. The aal1 lncRNA binds to the rpl1901 mRNA, encoding a ribosomal protein. The rpl1901 levels are reduced ~2-fold by aal1, which is sufficient to extend lifespan. Remarkably, the expression of the aal1 lncRNA in Drosophila boosts fly lifespan. We propose that aal1 reduces the ribosome content by decreasing Rpl1901 levels, thus attenuating the translational capacity and promoting longevity. Although aal1 is not conserved, its effect in flies suggests that animals feature related mechanisms that modulate ageing, based on the conserved translational machinery. Synopsis: The long-non-coding RNA aal1 (ageing-associated lncRNA) promotes the longevity of non-dividing cells likely by attenuating their translational capacity. aal1 localizes to the cytoplasm and binds to rpl1901 mRNA, which encodes a ribosomal protein, reducing its expression levels. aal1 decreases the cellular ribosome content and inhibits protein translation. Both aal1 expression or rpl1901 depletion prolong the chronological lifespan of non-dividing fission yeast cells. Ectopic expression of aal1 in Drosophila extends fly lifespan, suggesting a conserved mechanism. The long-non-coding RNA aal1 (ageing-associated lncRNA) promotes the longevity of non-dividing cells likely by attenuating their translational capacity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Proteomic characterization of regenerated cartilage following knee joint distraction; a human case-study.
- Author
-
Steijns, Jessica S. J. J., Green, Daniel, Peeters, Laura C. W., Emans, Pieter J., Boymans, Tim A., Stassen, Roderick H., van den Akker, Guus G. H., Cremers, Andy, Jutten, Liesbeth M. C., Anderson, James R., Peffers, Mandy J., Caron, Marjolein M. J., and Welting, Tim J. M.
- Subjects
- *
KNEE joint , *EXTRACELLULAR matrix proteins , *RNA polymerase II , *TOTAL knee replacement , *JOINTS (Anatomy) - Abstract
Purpose: Knee joint distraction is a surgical procedure with cartilage-regenerating properties. The composition of joint distraction-regenerated cartilage in human patients is poorly documented. In this case-study, provided a unique opportunity to biomolecularly characterize the regenerated tissue from a patient who underwent bilateral distraction and later knee replacements. Methods: Knee joint distraction was conducted using an external fixation frame and total knee arthroplasty was performed several years later. Radiographic imaging was performed to assess the status of the knee joint prior, during and after clinical interventions. Following total knee replacement, cartilage biopsies were collected and processed for tissue sectioning and histochemical staining. Tandem mass-spectrometry proteomics analysis was used to characterize and compare the proteomic composition. Results: Both knee joints showed joint-space improvement pre- and post-knee joint distraction. Regenerated cartilage was white with an irregular surface, while native (lateral) cartilage had a yellow appearance and smooth surface. Histochemical staining showed higher Safranin-O positivity in native cartilage compared to regenerated cartilage, and differences in collagen structure. Proteomic analysis did not reveal major differences in cartilage extracellular matrix protein abundance. Bioinformatic analyses revealed enrichment in ribosomal proteins (regenerated cartilage) and RNA Polymerase II Transcription Termination (native cartilage). Conclusion: Histologically, knee joint distraction-regenerated cartilage showed less glycosaminoglycans and disorganized collagen compared to native cartilage. However, mass-spectrometry has no major differences in extracellular matrix protein abundance, with proteomic clues suggesting protein translation regulation as a potential mechanism for regeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. AMBRA1 controls the translation of immune-specific genes in T lymphocytes.
- Author
-
Gottlieb, Simone, Wanjing Shang, Deji Ye, Satoshi Kubo, Ping Du Jiang, Shafer, Samantha, Leilei Xu, Lixin Zheng, Park, Ann Y., Jian Song, Waipan Chan, Zhiqin Zeng, Tingyan He, Schwarz, Benjamin, Häupl, Björn, Oellerich, Thomas, Lenardo, Michael J., and Yikun Yao
- Subjects
- *
CELL cycle regulation , *RNA regulation , *T cell receptors , *CELL cycle , *CELL death - Abstract
T cell receptor (TCR) engagement causes a global cellular response that entrains signaling pathways, cell cycle regulation, and cell death. The molecular regulation of mRNA translation in these processes is poorly understood. Using a whole-genome CRISPR screen for regulators of CD95 (FAS/APO-1)-mediated T cell death, we identified AMBRA1, a protein previously studied for its roles in autophagy, E3 ubiquitin ligase activity, and cyclin regulation. T cells lacking AMBRA1 resisted FAS-mediated cell death by down-regulating FAS expression at the translational level. We show that AMBRA1 is a vital regulator of ribosome protein biosynthesis and ribosome loading on select mRNAs, whereby it plays a key role in balancing TCR signaling with cell cycle regulation pathways. We also found that AMBRA1 itself is translationally controlled by TCR stimulation via the CD28-PI3K-mTORC1-EIF4F pathway. Together, these findings shed light on the molecular control of translation after T cell activation and implicate AMBRA1 as a translational regulator governing TCR signaling, cell cycle progression, and T cell death. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Cytosolic protein translation regulates cell asymmetry and function in early TCR activation of human CD8+ T lymphocytes.
- Author
-
Gomez-Moron, Alvaro, Tsukalov, Ilya, Scagnetti, Camila, Pertusa, Clara, Lozano-Prieto, Marta, Martınez-Fleta, Pedro, Requena, Silvia, Martın, Pilar, Alfranca, Aranzazu, Martin-Gayo, Enrique, and Martin-Cofreces, Noa B.
- Subjects
CYTOTOXIC T cells ,MITOCHONDRIAL proteins ,CELL physiology ,GENETIC translation ,PEPTIDES ,PERFORINS - Abstract
Introduction: CD8+ cytotoxic T lymphocytes (CTLs) are highly effective in defending against viral infections and tumours. They are activated through the recognition of peptide--MHC-I complex by the T-cell receptor (TCR) and costimulation. This cognate interaction promotes the organisation of intimate cell-- cell connections that involve cytoskeleton rearrangement to enable effector function and clearance of the target cell. This is key for the asymmetric transport and mobilisation of lytic granules to the cell--cell contact, promoting directed secretion of lytic mediators such as granzymes and perforin. Mitochondria play a role in regulating CTL function by controlling processes such as calcium flux, providing the necessary energy through oxidative phosphorylation, and its own protein translation on 55S ribosomes. However, the effect of acute inhibition of cytosolic translation in the rapid response after TCR has not been studied in mature CTLs. Methods: Here, we investigated the importance of cytosolic protein synthesis in human CTLs after early TCR activation and CD28 co-stimulation for the dynamic reorganisation of the cytoskeleton, mitochondria, and lytic granules through short-term chemical inhibition of 80S ribosomes by cycloheximide and 80S and 55S by puromycin. Results: We observed that eukaryotic ribosome function is required to allow proper asymmetric reorganisation of the tubulin cytoskeleton and mitochondria and mTOR pathway activation early upon TCR activation in human primary CTLs. Discussion: Cytosolic protein translation is required to increase glucose metabolism and degranulation capacity upon TCR activation and thus to regulate the full effector function of human CTLs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Pathogenic tau induces an adaptive elevation in mRNA translation rate at early stages of disease.
- Author
-
Zuniga, Gabrielle, Katsumura, Sakie, De Mange, Jasmine, Ramirez, Paulino, Atrian, Farzaneh, Morita, Masahiro, and Frost, Bess
- Subjects
- *
GENETIC translation , *ALZHEIMER'S disease , *NUCLEAR membranes , *TAUOPATHIES , *NEURODEGENERATION - Abstract
Alterations in the rate and accuracy of messenger RNA (mRNA) translation are associated with aging and several neurodegenerative disorders, including Alzheimer's disease and related tauopathies. We previously reported that error‐containing RNA that are normally cleared via nonsense‐mediated mRNA decay (NMD), a key RNA surveillance mechanism, are translated in the adult brain of a Drosophila model of tauopathy. In the current study, we find that newly‐synthesized peptides and translation machinery accumulate within nuclear envelope invaginations that occur as a consequence of tau pathology, and that the rate of mRNA translation is globally elevated in early stages of disease in adult brains of Drosophila models of tauopathy. Polysome profiling from adult heads of tau transgenic Drosophila reveals the preferential translation of specific mRNA that have been previously linked to neurodegeneration. Unexpectedly, we find that panneuronal elevation of NMD further elevates the global translation rate in tau transgenic Drosophila, as does treatment with rapamycin. As NMD activation and rapamycin both suppress tau‐induced neurodegeneration, their shared effect on translation suggests that elevated rates of mRNA translation are an early adaptive mechanism to limit neurodegeneration. Our work provides compelling evidence that tau‐induced deficits in NMD reshape the tau translatome by increasing translation of RNA that are normally repressed in healthy cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Lost in translation: How neurons cope with tRNA decoding.
- Author
-
Guo, Wei, Russo, Stefano, and Tuorto, Francesca
- Subjects
- *
UNFOLDED protein response , *GENETIC translation , *TRANSFER RNA , *DENATURATION of proteins , *CELL physiology , *NERVOUS system - Abstract
Post‐transcriptional tRNA modifications contribute to the decoding efficiency of tRNAs by supporting codon recognition and tRNA stability. Recent work shows that the molecular and cellular functions of tRNA modifications and tRNA‐modifying‐enzymes are linked to brain development and neurological disorders. Lack of these modifications affects codon recognition and decoding rate, promoting protein aggregation and translational stress response pathways with toxic consequences to the cell. In this review, we discuss the peculiarity of local translation in neurons, suggesting a role for fine‐tuning of translation performed by tRNA modifications. We provide several examples of tRNA modifications involved in physiology and pathology of the nervous system, highlighting their effects on protein translation and discussing underlying mechanisms, like the unfolded protein response (UPR), ribosome quality control (RQC), and no‐go mRNA decay (NGD), which could affect neuronal functions. We aim to deepen the understanding of the roles of tRNA modifications and the coordination of these modifications with the protein translation machinery in the nervous system. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. UTRs and Ago-2/miR-335 Complex Restricts Amylin Translation in Insulinoma and Human Pancreatic β-Cells.
- Author
-
Kudaibergenova, Zhanar, Pany, Satyabrata, Placheril, Elizabeth, and Jeremic, Aleksandar M.
- Subjects
- *
GENETIC regulation , *AMYLIN , *GENE expression , *PEPTIDES , *MICRORNA - Abstract
Amylin promoter and transcriptional factors are well-established, inducible factors in the production of the main amyloidogenic pancreatic hormone, human islet amyloid peptide (hIAPP) or amylin. However, posttranscriptional mechanisms driving hIAPP expression in pancreas remain enigmatic, and hence were explored here. The translational assay revealed that both 5′ and 3′ untranslated regions (UTRs) of hIAPP restricted expression of the luciferase constructs only in constructs driven by the hIAPP promoter. Bioinformatics analysis revealed several putative seed sequences for a dozen micro RNAs (miRNAs) in hIAPP's 3′ UTR. miR-182, miR-335, and miR-495 were the most downregulated miRNAs in stressed human islets exposed to endoplasmic reticulum (ER) or metabolic stressors, thapsigargin (TG) or high glucose (HG). Correspondingly, miR-335 mimics alone or in combination with miR-495 and miR-182 mimics significantly and potently (>3-fold) reduced hIAPP protein expression in HG-treated cultured human islets. siRNA-mediated silencing of Ago2 but not Ago1 significantly stimulated hIAPP expression and secretion from transfected, HG-treated human islets. Conversely, ectopic expression of Ago2 in hIAPP-expressing RIN-m5F cell line driven by CMV promoter reduced hIAPP intracellular protein levels. Collectively, the results point to a novel and synergistic role for hIAPP promoter, 5/3′ UTRs and Ago-2/miR-335 complex in post-transcriptional regulation of hIAPP gene expression in normal and metabolically active β-cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. Oncogenic Roles of Laminin Subunit Gamma‐2 in Intrahepatic Cholangiocarcinoma via Promoting EGFR Translation.
- Author
-
Zhang, Jianjuan, Ji, Fubo, Tan, Yaqi, Zhao, Lei, Zhao, Yongzhi, Liu, Jiaxin, Shao, Liyuan, Shi, Jiong, Ye, Meihua, He, Xianglei, Jin, Jianping, Zhao, Bin, Huang, Jun, Roessler, Stephanie, Zheng, Xin, and Ji, Junfang
- Subjects
- *
INTRAHEPATIC bile ducts , *EPIDERMAL growth factor receptors , *CHOLANGIOCARCINOMA , *PROTEIN-tyrosine kinase inhibitors , *LIVER cancer , *BILE ducts - Abstract
Intrahepatic cholangiocarcinoma (iCCA) is a highly lethal biliary epithelial cancer in the liver. Here, Laminin subunit gamma‐2 (LAMC2) with important oncogenic roles in iCCA is discovered. In a total of 231 cholangiocarcinoma patients (82% of iCCA patients) across four independent cohorts, LAMC2 is significantly more abundant in iCCA tumor tissue compared to normal bile duct and non‐tumor liver. Among 26.3% of iCCA patients, LAMC2 gene is amplified, contributing to its over‐expression. Functionally, silencing LAMC2 significantly blocks tumor formation in orthotopic iCCA mouse models. Mechanistically, it promotes EGFR protein translation via interacting with nascent unglycosylated EGFR in the endoplasmic reticulum (ER), resulting in activated EGFR signaling. LAMC2‐mediated EGFR translation also depends on its interaction with the ER chaperone BiP via their C‐terminus. Together LAMC2 and BiP generate a binding "pocket" of nascent EGFR and facilitate EGFR translation. Consistently, LAMC2‐high iCCA patients have poor prognosis in two iCCA cohorts. LAMC2‐high iCCA cells are highly sensitive to EGFR tyrosine kinase inhibitors (TKIs) treatment both in vitro and in vivo. Together, these data demonstrate LAMC2 as an oncogenic player in iCCA by promoting EGFR translation and an indicator to identify iCCA patients who may benefit from available EGFR‐targeted TKIs therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. GNN Codon Adjacency Tunes Protein Translation.
- Author
-
Sun, Joyce, Hwang, Pete, Sakkas, Eric D., Zhou, Yancheng, Perez, Luis, Dave, Ishani, Kwon, Jack B., McMahon, Audrey E., Wichman, Mia, Raval, Mitsu, Scopino, Kristen, Krizanc, Daniel, Thayer, Kelly M., and Weir, Michael P.
- Subjects
- *
PEPTIDES , *PROTEINS , *GENETIC translation , *GENETIC code , *AMINO acid sequence , *HYDROGEN bonding , *REQUESTS for proposals (Public contracts) - Abstract
The central dogma treats the ribosome as a molecular machine that reads one mRNA codon at a time as it adds each amino acid to its growing peptide chain. However, this and previous studies suggest that ribosomes actually perceive pairs of adjacent codons as they take three-nucleotide steps along the mRNA. We examined GNN codons, which we find are surprisingly overrepresented in eukaryote protein-coding open reading frames (ORFs), especially immediately after NNU codons. Ribosome profiling experiments in yeast revealed that ribosomes with NNU at their aminoacyl (A) site have particularly elevated densities when NNU is immediately followed (3′) by a GNN codon, indicating slower mRNA threading of the NNU codon from the ribosome's A to peptidyl (P) sites. Moreover, if the assessment was limited to ribosomes that have only recently arrived at the next codon, by examining 21-nucleotide ribosome footprints (21-nt RFPs), elevated densities were observed for multiple codon classes when followed by GNN. This striking translation slowdown at adjacent 5′-NNN GNN codon pairs is likely mediated, in part, by the ribosome's CAR surface, which acts as an extension of the A-site tRNA anticodon during ribosome translocation and interacts through hydrogen bonding and pi stacking with the GNN codon. The functional consequences of 5′-NNN GNN codon adjacency are expected to influence the evolution of protein coding sequences. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. The intersection of sleep and synaptic translation in synaptic plasticity deficits in neurodevelopmental disorders.
- Author
-
Coulson, Rochelle L., Mourrain, Philippe, and Wang, Gordon X.
- Subjects
- *
NEUROPLASTICITY , *GENETIC translation , *NEURAL development , *SLEEP , *CONDITIONED response - Abstract
Individuals with neurodevelopmental disorders experience persistent sleep deficits, and there is increasing evidence that sleep dysregulation is an underlying cause, rather than merely an effect, of the synaptic and behavioral defects observed in these disorders. At the molecular level, dysregulation of the synaptic proteome is a common feature of neurodevelopmental disorders, though the mechanism connecting these molecular and behavioral phenotypes is an ongoing area of investigation. A role for eIF2α in shifting the local proteome in response to changes in the conditions at the synapse has emerged. Here, we discuss recent progress in characterizing the intersection of local synaptic translation and sleep and propose a reciprocal mechanism of dysregulation in the development of synaptic plasticity defects in neurodevelopmental disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. The Odds of Protein Translation Control Under Stress.
- Author
-
Chen, Qin M.
- Subjects
- *
TRANSFER RNA , *GENETIC translation , *INITIATION factors (Biochemistry) , *STRESS granules , *PROTEINS , *PROTEIN kinases - Abstract
Physical or chemical stress is commonly known to inhibit protein translation at the cellular level. Since the process of protein translation requires catalysis by a multi-component machinery containing eukaryotic initiation factors (eIFs) and ribosomes in a sequence of reactions, how the process fails to proceed and whether certain genes can escape such blockade have provoked research efforts. Lines of evidence have demonstrated that phosphorylation of eIF4E or dephosphorylation of 4E-binding proteins (4E-BPs) prevents the formation of the eukaryotic translation initiation factor 4F (eIF4F) complex, whereas phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) due to activation of heme-regulated inhibitor (HRI), general control nonderepressible 2 (GCN2), protein kinase RNA-like endoplasmic reticulum kinase (PERK), or protein kinase R (PKR) by a diverse array of stressors prevents eIF2-GTP-tRNAiMet ternary complex assembly. These signal the abandonment of translation initiation via 5′-7-methylguanine (m7G) cap recognition by eIF4E. Stress can promote cleavage of tRNAs, impediment of rRNA processing, changes in the epitranscriptomic landscape, ribosome stalling or collision, activation of ribosomal surveillance systems, and assembly of the stress granules. Although these events contribute to the general inhibition of protein translation, a few proteins can bypass such negativity and become translated selectively. Such selective protein translation is primarily m7G cap independent through the integrated stress response or Internal Ribosomal Entry Site (IRES). The newly synthesized proteins often influence cell fate, facilitate cell survival, and build endogenous defense. Insights into the general inhibition of protein translation and selective translation of specific proteins will advance our understanding of the etiology or progression of human diseases involving cellular stress from viral infection or inflammation to myocardial infarction, stroke, or neurodegenerative disease. Antioxid. Redox Signal. 40, 943–947. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. Corrigendum: Cytosolic protein translation regulates cell asymmetry and function in early TCR activation of human CD8+ T lymphocytes
- Author
-
Álvaro Gómez-Morón, Ilya Tsukalov, Camila Scagnetti, Clara Pertusa, Marta Lozano-Prieto, Pedro Martínez-Fleta, Silvia Requena, Pilar Martín, Aranzazu Alfranca, Enrique Martin-Gayo, and Noa B Martin-Cofreces
- Subjects
protein translation ,cell asymmetry ,cytotoxic CD8+ T lymphocytes ,cytoskeleton ,metabolism ,mitochondria ,Immunologic diseases. Allergy ,RC581-607 - Published
- 2024
- Full Text
- View/download PDF
25. Esrra regulates Rplp1-mediated translation of lysosome proteins suppressed in metabolic dysfunction-associated steatohepatitis and reversed by alternate day fasting
- Author
-
Madhulika Tripathi, Karine Gauthier, Reddemma Sandireddy, Jin Zhou, Priyanka Guptta, Suganya Sakthivel, Wei Wen Teo, Yadanar Than Naing, Kabilesh Arul, Keziah Tikno, Sung-Hee Park, Yajun Wu, Lijin Wang, Boon-Huat Bay, Lei Sun, Vincent Giguere, Pierce K.H. Chow, Sujoy Ghosh, Donald P. McDonnell, Paul M. Yen, and Brijesh K. Singh
- Subjects
Metabolic dysfunction-associated steatohepatitis (MASH) ,Lysosome ,Estrogen related receptor alpha (ERRα/Esrra) ,Ribosome ,Protein translation ,Internal medicine ,RC31-1245 - Abstract
Objective: Currently, little is known about the mechanism(s) regulating global and specific protein translation during metabolic dysfunction-associated steatohepatitis (MASH; previously known as non-alcoholic steatohepatitis, NASH). Methods: Unbiased label-free quantitative proteome, puromycin-labelling and polysome profiling were used to understand protein translation activity in vitro and in vivo. Results: We observed a global decrease in protein translation during lipotoxicity in human primary hepatocytes, mouse hepatic AML12 cells, and livers from a dietary mouse model of MASH. Interestingly, proteomic analysis showed that Rplp1, which regulates ribosome and translation pathways, was one of the most downregulated proteins. Moreover, decreased Esrra expression and binding to the Rplp1 promoter, diminished Rplp1 gene expression during lipotoxicity. This, in turn, reduced global protein translation and Esrra/Rplp1-dependent translation of lysosome (Lamp2, Ctsd) and autophagy (sqstm1, Map1lc3b) proteins. Of note, Esrra did not increase its binding to these gene promoters or their gene transcription, confirming its regulation of their translation during lipotoxicity. Notably, hepatic Esrra-Rplp1-dependent translation of lysosomal and autophagy proteins also was impaired in MASH patients and liver-specific Esrra knockout mice. Remarkably, alternate day fasting induced Esrra-Rplp1-dependent expression of lysosomal proteins, restored autophagy, and reduced lipotoxicity, inflammation, and fibrosis in hepatic cell culture and in vivo models of MASH. Conclusions: Esrra regulation of Rplp1-mediated translation of lysosome/autolysosome proteins was downregulated during MASH. Alternate day fasting activated this novel pathway and improved MASH, suggesting that Esrra and Rplp1 may serve as therapeutic targets for MASH. Our findings also provided the first example of a nuclear hormone receptor, Esrra, to not only regulate transcription but also protein translation, via induction of Rplp1.
- Published
- 2024
- Full Text
- View/download PDF
26. FTH1 overexpression using a dCasRx translation enhancement system protects the kidney from calcium oxalate crystal-induced injury
- Author
-
Ziqi He, Caitao Dong, Tianbao Song, Jiawei Zhou, Tao Xu, Ruyuan He, and Sheng Li
- Subjects
CRISPR–Cas system ,dCasRx ,eIF4GI ,Gene regulation ,Protein translation ,Nephrolithiasis ,Cytology ,QH573-671 - Abstract
Abstract The engineered clustered regularly interspaced short palindromic repeats (CRISPR)–CRISPR-associated protein (Cas) system is currently widely applied in genetic editing and transcriptional regulation. The catalytically inactivated CasRx (dCasRx) has the ability to selectively focus on the mRNA coding region without disrupting transcription and translation, opening up new avenues for research on RNA modification and protein translation control. This research utilized dCasRx to create a translation-enhancement system for mammals called dCasRx-eIF4GI, which combined eukaryotic translation initiation factor 4G (eIF4GI) to boost translation levels of the target gene by recruiting ribosomes, without affecting mRNA levels, ultimately increasing translation levels of different endogenous proteins. Due to the small size of dCasRx, the dCasRx-eIF4GI translation enhancement system was integrated into a single viral vector, thus optimizing the delivery and transfection efficiency in subsequent applications. Previous studies reported that ferroptosis, mediated by calcium oxalate (CaOx) crystals, significantly promotes stone formation. In order to further validate its developmental potential, it was applied to a kidney stone model in vitro and in vivo. The manipulation of the ferroptosis regulatory gene FTH1 through single-guide RNA (sgRNA) resulted in a notable increase in FTH1 protein levels without affecting its mRNA levels. This ultimately prevented intracellular ferroptosis and protected against cell damage and renal impairment caused by CaOx crystals. Taken together, this study preliminarily validated the effectiveness and application prospects of the dCasRx-eIF4GI translation enhancement system in mammalian cell-based disease models, providing novel insights and a universal tool platform for protein translation research and future therapeutic approaches for nephrolithiasis.
- Published
- 2024
- Full Text
- View/download PDF
27. Sub-network transcriptome dataset for diseases associated with exposure to bisphenol F and bisphenol S in human SH-SY5Y neuroblastoma cells
- Author
-
Guzman, Andrea, Sanchez, Christina L., Ivantsova, Emma, Watkins, Jacqueline, Sutton, Sara, Souders, Christopher L., II, and Martyniuk, Christopher J.
- Published
- 2025
- Full Text
- View/download PDF
28. Ribosomal profiling of human endogenous retroviruses in healthy tissues
- Author
-
Dopkins, Nicholas, Singh, Bhavya, Michael, Stephanie, Zhang, Panpan, Marston, Jez L., Fei, Tongyi, Singh, Manvendra, Feschotte, Cedric, Collins, Nicholas, Bendall, Matthew L., and Nixon, Douglas F.
- Published
- 2024
- Full Text
- View/download PDF
29. FTH1 overexpression using a dCasRx translation enhancement system protects the kidney from calcium oxalate crystal-induced injury.
- Author
-
He, Ziqi, Dong, Caitao, Song, Tianbao, Zhou, Jiawei, Xu, Tao, He, Ruyuan, and Li, Sheng
- Abstract
The engineered clustered regularly interspaced short palindromic repeats (CRISPR)–CRISPR-associated protein (Cas) system is currently widely applied in genetic editing and transcriptional regulation. The catalytically inactivated CasRx (dCasRx) has the ability to selectively focus on the mRNA coding region without disrupting transcription and translation, opening up new avenues for research on RNA modification and protein translation control. This research utilized dCasRx to create a translation-enhancement system for mammals called dCasRx-eIF4GI, which combined eukaryotic translation initiation factor 4G (eIF4GI) to boost translation levels of the target gene by recruiting ribosomes, without affecting mRNA levels, ultimately increasing translation levels of different endogenous proteins. Due to the small size of dCasRx, the dCasRx-eIF4GI translation enhancement system was integrated into a single viral vector, thus optimizing the delivery and transfection efficiency in subsequent applications. Previous studies reported that ferroptosis, mediated by calcium oxalate (CaOx) crystals, significantly promotes stone formation. In order to further validate its developmental potential, it was applied to a kidney stone model in vitro and in vivo. The manipulation of the ferroptosis regulatory gene FTH1 through single-guide RNA (sgRNA) resulted in a notable increase in FTH1 protein levels without affecting its mRNA levels. This ultimately prevented intracellular ferroptosis and protected against cell damage and renal impairment caused by CaOx crystals. Taken together, this study preliminarily validated the effectiveness and application prospects of the dCasRx-eIF4GI translation enhancement system in mammalian cell-based disease models, providing novel insights and a universal tool platform for protein translation research and future therapeutic approaches for nephrolithiasis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. Viral RNA capping: Mechanisms and antiviral therapy.
- Author
-
Chen, Saini, Jiang, Zhimin, Li, Qiuchen, Pan, Wenliang, Chen, Yu, and Liu, Jinhua
- Subjects
RNA ,GENETIC translation ,VIRAL genes ,ANIMAL diseases ,VIRUS diseases - Abstract
RNA capping is an essential trigger for protein translation in eukaryotic cells. Many viruses have evolved various strategies for initiating the translation of viral genes and generating progeny virions in infected cells via synthesizing cap structure or stealing the RNA cap from nascent host messenger ribonucleotide acid (mRNA). In addition to protein translation, a new understanding of the role of the RNA cap in antiviral innate immunity has advanced the field of mRNA synthesis in vitro and therapeutic applications. Recent studies on these viral RNA capping systems have revealed startlingly diverse ways and molecular machinery. A comprehensive understanding of how viruses accomplish the RNA capping in infected cells is pivotal for designing effective broad‐spectrum antiviral therapies. Here we systematically review the contemporary insights into the RNA‐capping mechanisms employed by viruses causing human and animal infectious diseases, while also highlighting its impact on host antiviral innate immune response. The therapeutic applications of targeting RNA capping against viral infections and the development of RNA‐capping inhibitors are also summarized. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Tbx4 and Tbx5 gene expression associated with appendage development and its relationship with the absence of the pelvic fin in Pampus argenteus.
- Author
-
Zhang, Shun, Zhu, Xiaojing, Hu, Lingzhu, Liao, Kai, Xu, Shanliang, Wang, Danli, and Guo, Chunyang
- Subjects
- *
GENE expression , *STROMATEIDAE , *MARINE fishes , *NUCLEOTIDE sequence , *POLYMERASE chain reaction - Abstract
The Tbx family is first known through the study of their functions in the body and limbs, and its members Tbx4 and Tbx5 genes are important factors in determining the characteristics of the appendages. Pampus argenteus is one of the important economical marine fishes widely distributed in offshore areas. Therefore, it is necessary to study the role of Tbx family genes in the deletion of pelvic fin in P. argenteus. In this study, we cloned Tbx4 and Tbx5 cDNA sequence of P. argenteus (GenBank: MH709128 and MH712458). The Western blot and real time PCR were used to detect the expressions of Tbx4 and Tbx5 in different developmental stages and tissues of P. argenteus. In addition, whole-mount in-situ hybridization was used to study the localization of Tbx4 and Tbx5 genes in different developmental stages of P. argenteus. Results show that the translation of Tbx4 mRNA was inhibited during the critical period of pelvic fin development. Among different tissues, Tbx4 protein levels were the lowest in the abdominal epithelium, and even lower than that in the pectoral fin, suggesting that the protein expression was also inhibited in the abdominal epithelium of adult P. argenteus. Therefore, the results indicated that upstream genes regulation led to the key stage-specific and low expression of Tbx4 during pelvic fin development and in the abdominal epithelium. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Maize DDK1 encoding an Importin‐4 β protein is essential for seed development and grain filling by mediating nuclear exporting of eIF1A.
- Author
-
Huang, Xing, Huang, Yongcai, Qin, Li, Xiao, Qiao, Wang, Qiong, Wang, Jiechen, Wang, Wenqin, Lu, Xiaoduo, and Wu, Yongrui
- Subjects
- *
SEED development , *NUCLEAR membranes , *IMMOBILIZED proteins , *EUKARYOTIC cells , *PROTEIN synthesis , *CORN - Abstract
Summary: Nuclear‐cytoplasmic trafficking is crucial for protein synthesis in eukaryotic cells due to the spatial separation of transcription and translation by the nuclear envelope. However, the mechanism underlying this process remains largely unknown in plants.In this study, we isolated a maize (Zea mays) mutant designated developmentally delayed kernel 1 (ddk1), which exhibits delayed seed development and slower filling. Ddk1 encodes a plant‐specific protein known as Importin‐4 β, and its mutation results in reduced 80S monosomes and suppressed protein synthesis.Through our investigations, we found that DDK1 interacts with eIF1A proteins in vivo. However, in vitro experiments revealed that this interaction exhibits low affinity in the absence of RanGTP. Additionally, while the eIF1A protein primarily localizes to the cytoplasm in the wild‐type, it remains significantly retained within the nuclei of ddk1 mutants. These observations suggest that DDK1 functions as an exportin and collaborates with RanGTP to facilitate the nuclear export of eIF1A, consequently regulating endosperm development at the translational level.Importantly, both DDK1 and eIF1A are conserved among various plant species, implying the preservation of this regulatory module across diverse plants. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. A model organism pipeline provides insight into the clinical heterogeneity of TARS1 loss-of-function variants
- Author
-
Rebecca Meyer-Schuman, Allison R. Cale, Jennifer A. Pierluissi, Kira E. Jonatzke, Young N. Park, Guy M. Lenk, Stephanie N. Oprescu, Marina A. Grachtchouk, Andrzej A. Dlugosz, Asim A. Beg, Miriam H. Meisler, and Anthony Antonellis
- Subjects
aminoacyl-tRNA synthetases ,threonyl-tRNA synthetase ,Mendelian disease ,recessive disease ,protein translation ,threonine ,Genetics ,QH426-470 - Abstract
Summary: Aminoacyl-tRNA synthetases (ARSs) are ubiquitously expressed, essential enzymes that complete the first step of protein translation: ligation of amino acids to cognate tRNAs. Genes encoding ARSs have been implicated in myriad dominant and recessive phenotypes, the latter often affecting multiple tissues but with frequent involvement of the central and peripheral nervous systems, liver, and lungs. Threonyl-tRNA synthetase (TARS1) encodes the enzyme that ligates threonine to tRNATHR in the cytoplasm. To date, TARS1 variants have been implicated in a recessive brittle hair phenotype. To better understand TARS1-related recessive phenotypes, we engineered three TARS1 missense variants at conserved residues and studied these variants in Saccharomyces cerevisiae and Caenorhabditis elegans models. This revealed two loss-of-function variants, including one hypomorphic allele (R433H). We next used R433H to study the effects of partial loss of TARS1 function in a compound heterozygous mouse model (R432H/null). This model presents with phenotypes reminiscent of patients with TARS1 variants and with distinct lung and skin defects. This study expands the potential clinical heterogeneity of TARS1-related recessive disease, which should guide future clinical and genetic evaluations of patient populations.
- Published
- 2024
- Full Text
- View/download PDF
34. Cytosolic protein translation regulates cell asymmetry and function in early TCR activation of human CD8+ T lymphocytes
- Author
-
Álvaro Gómez-Morón, Ilya Tsukalov, Camila Scagnetti, Clara Pertusa, Marta Lozano-Prieto, Pedro Martínez-Fleta, Silvia Requena, Pilar Martín, Aranzazu Alfranca, Enrique Martin-Gayo, and Noa B Martin-Cofreces
- Subjects
protein translation ,cell asymmetry ,cytotoxic CD8+ T lymphocytes ,cytoskeleton ,metabolism ,mitochondria ,Immunologic diseases. Allergy ,RC581-607 - Abstract
IntroductionCD8+ cytotoxic T lymphocytes (CTLs) are highly effective in defending against viral infections and tumours. They are activated through the recognition of peptide–MHC-I complex by the T-cell receptor (TCR) and co-stimulation. This cognate interaction promotes the organisation of intimate cell–cell connections that involve cytoskeleton rearrangement to enable effector function and clearance of the target cell. This is key for the asymmetric transport and mobilisation of lytic granules to the cell–cell contact, promoting directed secretion of lytic mediators such as granzymes and perforin. Mitochondria play a role in regulating CTL function by controlling processes such as calcium flux, providing the necessary energy through oxidative phosphorylation, and its own protein translation on 55S ribosomes. However, the effect of acute inhibition of cytosolic translation in the rapid response after TCR has not been studied in mature CTLs.MethodsHere, we investigated the importance of cytosolic protein synthesis in human CTLs after early TCR activation and CD28 co-stimulation for the dynamic reorganisation of the cytoskeleton, mitochondria, and lytic granules through short-term chemical inhibition of 80S ribosomes by cycloheximide and 80S and 55S by puromycin.ResultsWe observed that eukaryotic ribosome function is required to allow proper asymmetric reorganisation of the tubulin cytoskeleton and mitochondria and mTOR pathway activation early upon TCR activation in human primary CTLs.DiscussionCytosolic protein translation is required to increase glucose metabolism and degranulation capacity upon TCR activation and thus to regulate the full effector function of human CTLs.
- Published
- 2024
- Full Text
- View/download PDF
35. Macrophage mitochondrial bioenergetics and tissue invasion are boosted by an Atossa‐Porthos axis in Drosophila
- Author
-
Emtenani, Shamsi, Martin, Elliot T, Gyoergy, Attila, Bicher, Julia, Genger, Jakob‐Wendelin, Köcher, Thomas, Akhmanova, Maria, Guarda, Mariana, Roblek, Marko, Bergthaler, Andreas, Hurd, Thomas R, Rangan, Prashanth, and Siekhaus, Daria E
- Subjects
Medical Biochemistry and Metabolomics ,Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Underpinning research ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Aetiology ,Generic health relevance ,Affordable and Clean Energy ,Animals ,Drosophila ,Energy Metabolism ,Macrophages ,Mitochondria ,RNA ,Messenger ,Saccharopine Dehydrogenases ,immune cell infiltration ,mitochondrial bioenergetics ,oxidative phosphorylation ,protein translation ,transcription factor ,Information and Computing Sciences ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Cellular metabolism must adapt to changing demands to enable homeostasis. During immune responses or cancer metastasis, cells leading migration into challenging environments require an energy boost, but what controls this capacity is unclear. Here, we study a previously uncharacterized nuclear protein, Atossa (encoded by CG9005), which supports macrophage invasion into the germband of Drosophila by controlling cellular metabolism. First, nuclear Atossa increases mRNA levels of Porthos, a DEAD-box protein, and of two metabolic enzymes, lysine-α-ketoglutarate reductase (LKR/SDH) and NADPH glyoxylate reductase (GR/HPR), thus enhancing mitochondrial bioenergetics. Then Porthos supports ribosome assembly and thereby raises the translational efficiency of a subset of mRNAs, including those affecting mitochondrial functions, the electron transport chain, and metabolism. Mitochondrial respiration measurements, metabolomics, and live imaging indicate that Atossa and Porthos power up OxPhos and energy production to promote the forging of a path into tissues by leading macrophages. Since many crucial physiological responses require increases in mitochondrial energy output, this previously undescribed genetic program may modulate a wide range of cellular behaviors.
- Published
- 2022
36. Polyamines: Their Role in Plant Development and Stress.
- Author
-
Blázquez, Miguel A.
- Abstract
This review focuses on the intricate relationship between plant polyamines and the genetic circuits and signaling pathways that regulate various developmental programs and the defense responses of plants when faced with biotic and abiotic aggressions. Particular emphasis is placed on genetic evidence supporting the involvement of polyamines in specific processes, such as the pivotal role of thermospermine in regulating xylem cell differentiation and the significant contribution of polyamine metabolism in enhancing plant resilience to drought. Based on the numerous studies describing effects of the manipulation of plant polyamine levels, two conceptually different mechanisms for polyamine activity are discussed: direct participation of polyamines in translational regulation and the indirect production of hydrogen peroxide as a defensive mechanism against pathogens. By describing the multifaceted functions of polyamines, this review underscores the profound significance of these compounds in enabling plants to adapt and thrive in challenging environments. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. Utilities of Isolated Nerve Terminals in Ex Vivo Analyses of Protein Translation in (Patho)physiological Brain States: Focus on Alzheimer's Disease.
- Author
-
Ibrahim, Mohammad Jasim, Baiju, Viswanath, Sen, Shivam, Chandran, Pranav Prathapa, Ashraf, Ghulam Md, Haque, Shafiul, and Ahmad, Faraz
- Abstract
Synapses are the cellular substrates of higher-order brain functions, and their dysfunction is an early and primary pathogenic mechanism across several neurological disorders. In particular, Alzheimer's disease (AD) is categorized by prodromal structural and functional synaptic deficits, prior to the advent of classical behavioral and pathological features. Recent research has shown that the development, maintenance, and plasticity of synapses depend on localized protein translation. Synaptosomes and synaptoneurosomes are biochemically isolated synaptic terminal preparations which have long been used to examine a variety of synaptic processes ex vivo in both healthy and pathological conditions. These ex vivo preparations preserve the mRNA species and the protein translational machinery. Hence, they are excellent in organello tools for the study of alterations in mRNA levels and protein translation in neuropathologies. Evaluation of synapse-specific basal and activity-driven de novo protein translation activity can be conveniently performed in synaptosomal/synaptoneurosomal preparations from both rodent and human brain tissue samples. This review gives a quick overview of the methods for isolating synaptosomes and synaptoneurosomes before discussing the studies that have utilized these preparations to study localized synapse-specific protein translation in (patho)physiological situations, with an emphasis on AD. While the review is not an exhaustive accumulation of all the studies evaluating synaptic protein translation using the synaptosomal model, the aim is to assemble the most relevant studies that have done so. The hope is to provide a suitable research platform to aid neuroscientists to utilize the synaptosomal/synaptoneurosomal models to evaluate the molecular mechanisms of synaptic dysfunction within the specific confines of mRNA localization and protein translation research. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Assembly and function of the amyloid‐like translational repressor Rim4 is coupled with nutrient conditions.
- Author
-
Ottoz, Diana SM, Tang, Lauren C, Dyatel, Annie E, Jovanovic, Marko, and Berchowitz, Luke E
- Subjects
- *
RNA-binding proteins , *MESSENGER RNA , *PROTEIN kinases , *CELL anatomy , *MEIOSIS , *RIBOSOMES , *AMYLOID beta-protein - Abstract
Amyloid‐like protein assemblies have been associated with toxic phenotypes because of their repetitive and stable structure. However, evidence that cells exploit these structures to control function and activity of some proteins in response to stimuli has questioned this paradigm. How amyloid‐like assembly can confer emergent functions and how cells couple assembly with environmental conditions remains unclear. Here, we study Rim4, an RNA‐binding protein that forms translation‐repressing assemblies during yeast meiosis. We demonstrate that in its assembled and repressive state, Rim4 binds RNA more efficiently than in its monomeric and idle state, revealing a causal connection between assembly and function. The Rim4‐binding site location within the transcript dictates whether the assemblies can repress translation, underscoring the importance of the architecture of this RNA‐protein structure for function. Rim4 assembly depends exclusively on its intrinsically disordered region and is prevented by the Ras/protein kinase A signaling pathway, which promotes growth and suppresses meiotic entry in yeast. Our results suggest a mechanism whereby cells couple a functional protein assembly with a stimulus to enforce a cell fate decision. Synopsis: The RNA‐binding protein Rim4, required for meiosis in yeast, forms amyloid‐like assemblies that repress translation of select messenger RNAs. Here, it is shown that PKA inactivation triggers the formation of Rim4 assemblies and that these structures repress translation by promoting efficient RNA binding and by preventing start codon access by ribosomes. Rim4 forms SDS‐resistant assemblies that repress translation of select messenger RNAs during meiosis.Rim4 assembly is necessary for function as it promotes efficient binding to target mRNAs.The architecture of the RNA‐Rim4 assembly structure dictates productive translational regulation.Active PKA prevents Rim4 amyloid‐like assembly. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
39. Repurposing an Antioxidant to Kill Mycobacterium tuberculosis by Targeting the 50S Subunit of the Ribosome.
- Author
-
Dong, Wenqi, Wang, Gaoyan, Bai, Yajuan, Li, Yuxin, Zhao, Liying, Lu, Wenjia, Wang, Chenchen, Zhang, Zhaoran, Lu, Hao, Wang, Xiangru, Chen, Huanchun, and Tan, Chen
- Subjects
- *
MYCOBACTERIUM tuberculosis , *RIBOSOMES , *DRUG repositioning , *NUCLEOTIDE sequencing , *PROTEIN synthesis , *SURVIVAL rate - Abstract
Tuberculosis and drug-resistant TB remain serious threats to global public health. It is urgent to develop novel anti-TB drugs in order to control it. In addition to redesigning and developing new anti-TB drugs, drug repurposing is also an innovative way to develop antibacterial drugs. Based on this method, we discovered SKQ-1 in the FDA-approved drug library and evaluated its anti-TB activity. In vitro, we demonstrated that SKQ-1 engaged in bactericidal activity against drug-sensitive and -resistant Mtb and confirmed the synergistic effects of SKQ1 with RIF and INH. Moreover, SKQ-1 showed a significant Mtb-killing effect in macrophages. In vivo, both the SKQ-1 treatment alone and the treatment in combination with RIF were able to significantly reduce the bacterial load and improve the survival rate of G. mellonella infected with Mtb. We performed whole-genome sequencing on screened SKQ-1-resistant strains and found that the SNP sites were concentrated in the 50S ribosomal subunit of Mtb. Furthermore, we proved that SKQ-1 can inhibit protein translation. In summary, from the perspective of drug repurposing, we discovered and determined the anti-tuberculosis effect of SKQ-1, revealed its synergistic effects with RIF and INH, and demonstrated its mechanism of action through targeting ribosomes and disrupting protein synthesis, thus making it a potential treatment option for DR-TB. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. Glycogen synthase kinase 3 signaling in neural regeneration in vivo.
- Author
-
Zhang, Jing, Yang, Shu-Guang, and Zhou, Feng-Quan
- Abstract
Glycogen synthase kinase 3 (GSK3) signaling plays important and broad roles in regulating neural development in vitro and in vivo. Here, we reviewed recent findings of GSK3-regulated axon regeneration in vivo in both the peripheral and central nervous systems and discussed a few controversial findings in the field. Overall, current evidence indicates that GSK3β signaling serves as an important downstream mediator of the PI3K–AKT pathway to regulate axon regeneration in parallel with the mTORC1 pathway. Specifically, the mTORC1 pathway supports axon regeneration mainly through its role in regulating cap-dependent protein translation, whereas GSK3β signaling might be involved in regulating N
6 -methyladenosine mRNA methylation-mediated, cap-independent protein translation. In addition, GSK3 signaling also plays a key role in reshaping the neuronal transcriptomic landscape during neural regeneration. Finally, we proposed some research directions to further elucidate the molecular mechanisms underlying the regulatory function of GSK3 signaling and discover novel GSK3 signaling-related therapeutic targets. Together, we hope to provide an updated and insightful overview of how GSK3 signaling regulates neural regeneration in vivo. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
41. Methods for In Vivo Characterization of Proteostasis in the Mouse Retina
- Author
-
Wang, Yixiao, Lobanova, Ekaterina S., Crusio, Wim E., Series Editor, Dong, Haidong, Series Editor, Radeke, Heinfried H., Series Editor, Rezaei, Nima, Series Editor, Steinlein, Ortrud, Series Editor, Xiao, Junjie, Series Editor, Ash, John D., editor, Pierce, Eric, editor, Anderson, Robert E., editor, Bowes Rickman, Catherine, editor, Hollyfield, Joe G., editor, and Grimm, Christian, editor
- Published
- 2023
- Full Text
- View/download PDF
42. mTORC1 promotes cell growth via m6A-dependent mRNA degradation
- Author
-
Cho, Sungyun, Lee, Gina, Pickering, Brian F, Jang, Cholsoon, Park, Jin H, He, Long, Mathur, Lavina, Kim, Seung-Soo, Jung, Sunhee, Tang, Hong-Wen, Monette, Sebastien, Rabinowitz, Joshua D, Perrimon, Norbert, Jaffrey, Samie R, and Blenis, John
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Cancer ,Genetics ,2.1 Biological and endogenous factors ,Aetiology ,Adenosine ,Animals ,Base Sequence ,Cell Cycle Proteins ,Cell Line ,Tumor ,Cell Proliferation ,Eukaryotic Initiation Factors ,HEK293 Cells ,Humans ,Male ,Mechanistic Target of Rapamycin Complex 1 ,Mice ,Models ,Biological ,Protein Biosynthesis ,Proto-Oncogene Proteins c-myc ,RNA Splicing Factors ,RNA Stability ,RNA ,Messenger ,Ribosomal Protein S6 Kinases ,Signal Transduction ,MXD2 ,Protein translation ,S6K1 ,WTAP ,YTHDF readers ,cMyc ,eIF4A ,m(6)A mRNA modification ,mRNA stability ,mTORC1 ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Dysregulated mTORC1 signaling alters a wide range of cellular processes, contributing to metabolic disorders and cancer. Defining the molecular details of downstream effectors is thus critical for uncovering selective therapeutic targets. We report that mTORC1 and its downstream kinase S6K enhance eIF4A/4B-mediated translation of Wilms' tumor 1-associated protein (WTAP), an adaptor for the N6-methyladenosine (m6A) RNA methyltransferase complex. This regulation is mediated by 5' UTR of WTAP mRNA that is targeted by eIF4A/4B. Single-nucleotide-resolution m6A mapping revealed that MAX dimerization protein 2 (MXD2) mRNA contains m6A, and increased m6A modification enhances its degradation. WTAP induces cMyc-MAX association by suppressing MXD2 expression, which promotes cMyc transcriptional activity and proliferation of mTORC1-activated cancer cells. These results elucidate a mechanism whereby mTORC1 stimulates oncogenic signaling via m6A RNA modification and illuminates the WTAP-MXD2-cMyc axis as a potential therapeutic target for mTORC1-driven cancers.
- Published
- 2021
43. Queuosine‐tRNA promotes sex‐dependent learning and memory formation by maintaining codon‐biased translation elongation speed.
- Author
-
Cirzi, Cansu, Dyckow, Julia, Legrand, Carine, Schott, Johanna, Guo, Wei, Perez Hernandez, Daniel, Hisaoka, Miharu, Parlato, Rosanna, Pitzer, Claudia, van der Hoeven, Franciscus, Dittmar, Gunnar, Helm, Mark, Stoecklin, Georg, Schirmer, Lucas, Lyko, Frank, and Tuorto, Francesca
- Subjects
- *
RIBOSOMES , *TRANSFER RNA , *COGNITIVE ability , *MEMORY disorders , *MULTIENZYME complexes , *PROTEIN synthesis , *GUT microbiome - Abstract
Queuosine (Q) is a modified nucleoside at the wobble position of specific tRNAs. In mammals, queuosinylation is facilitated by queuine uptake from the gut microbiota and is introduced into tRNA by the QTRT1‐QTRT2 enzyme complex. By establishing a Qtrt1 knockout mouse model, we discovered that the loss of Q‐tRNA leads to learning and memory deficits. Ribo‐Seq analysis in the hippocampus of Qtrt1‐deficient mice revealed not only stalling of ribosomes on Q‐decoded codons, but also a global imbalance in translation elongation speed between codons that engage in weak and strong interactions with their cognate anticodons. While Q‐dependent molecular and behavioral phenotypes were identified in both sexes, female mice were affected more severely than males. Proteomics analysis confirmed deregulation of synaptogenesis and neuronal morphology. Together, our findings provide a link between tRNA modification and brain functions and reveal an unexpected role of protein synthesis in sex‐dependent cognitive performance. Synopsis: Queuosine (Q) is a modified nucleoside introduced in specific tRNAs by a complex comprising queuine tRNA ribosyltransferase 1 (QTRT1). Loss of Q‐tRNA modification by the knockout of Qtrt1 gene in mice (Q1) leads to an imbalance in codon‐biased protein translation speed, resulting in alterations of hippocampal architecture and sex‐dependent reduction in learning and memory formation.Q‐tRNA levels vary considerably between murine tissues, with the brain showing a high queuosinylation level.Loss of Q results in learning and memory deficits, with females being more affected than male mice.Q1 mice exhibit altered neuronal cytoarchitecture in the hippocampus.Loss of Q leads to ribosome stalling on Q‐decoded codons and alteration of overall protein translation homeostasis.Translational stress and increased eIF2α phosphorylation impair neuronal cognitive functions in Q1 female mice. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
44. Ribosomal Biogenesis and Heterogeneity in Development, Disease, and Aging.
- Author
-
Islam, Rowshan Ara and Rallis, Charalampos
- Subjects
GENETIC regulation ,PHENOMENOLOGICAL biology ,ORGANELLE formation ,HETEROGENEITY ,CYTOLOGY - Abstract
Although reported in the literature, ribosome heterogeneity is a phenomenon whose extent and implications in cell and organismal biology is not fully appreciated. This has been the case due to the lack of the appropriate techniques and approaches. Heterogeneity can arise from alternative use and differential content of protein and RNA constituents, as well as from post-transcriptional and post-translational modifications. In the few examples we have, it is apparent that ribosomal heterogeneity offers an additional level and potential for gene expression regulation and might be a way towards tuning metabolism, stress, and growth programs to external and internal stimuli and needs. Here, we introduce ribosome biogenesis and discuss ribosomal heterogeneity in various reported occasions. We conclude that a systematic approach in multiple organisms will be needed to delineate this biological phenomenon and its contributions to growth, aging, and disease. Finally, we discuss ribosome mutations and their roles in disease. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
45. 4E-BP1 Protects Neurons from Misfolded Protein Stress and Parkinson's Disease Toxicity by Inducing the Mitochondrial Unfolded Protein Response
- Author
-
Dastidar, Somasish Ghosh, Pham, Michael T, Mitchell, Matthew B, Yeom, Steven G, Jordan, Sarah, Chang, Angela, Sopher, Bryce L, and La Spada, Albert R
- Subjects
Neurodegenerative ,Parkinson's Disease ,Aging ,Brain Disorders ,Neurosciences ,Neurological ,Adaptor Proteins ,Signal Transducing ,Animals ,Animals ,Newborn ,Brefeldin A ,Cell Cycle Proteins ,Female ,Male ,Mice ,Mice ,Transgenic ,Mitochondria ,Neurons ,Parkinson Disease ,Secondary ,Primary Cell Culture ,Protein Biosynthesis ,Protein Synthesis Inhibitors ,Protein Unfolding ,Proteostasis Deficiencies ,Rotenone ,Uncoupling Agents ,alpha-Synuclein ,4E-BP1 ,alpha-synuclein ,mitochondrial unfolded protein response ,neuroprotection ,Parkinson's disease ,protein translation ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Neurology & Neurosurgery - Abstract
Decline of protein quality control in neurons contributes to age-related neurodegenerative disorders caused by misfolded proteins. 4E-BP1 is a key node in the regulation of protein synthesis, as activated 4E-BP1 represses global protein translation. Overexpression of 4E-BP1 mediates the benefits of dietary restriction and can counter metabolic stress, and 4E-BP1 disinhibition on mTORC1 repression may be neuroprotective; however, whether 4E-BP1 overexpression is neuroprotective in mammalian neurons is yet to be fully explored. To address this question, we generated 4E-BP1-overexpressing transgenic mice and confirmed marked reductions in protein translation in 4E-BP1-overexpressing primary neurons. After documenting that 4E-BP1-overexpressing neurons are resistant to proteotoxic stress elicited by brefeldin A treatment, we exposed primary neurons to three different Parkinson's disease (PD)-linked toxins (rotenone, maneb, or paraquat) and documented significant protection in neurons from newborn male and female 4E-BP1-OE transgenic mice. We observed 4E-BP1-dependent upregulation of genes encoding proteins that comprise the mitochondrial unfolded protein response, and noted 4E-BP1 overexpression required activation of the mitochondrial unfolded protein response for neuroprotection against rotenone toxicity. We also tested whether 4E-BP1 could prevent α-synuclein neurotoxicity by treating 4E-BP1-overexpressing primary neurons with α-synuclein preformed fibrils, and we observed marked reductions in α-synuclein aggregation and neurotoxicity, thus validating that 4E-BP1 is a powerful suppressor of PD-linked pathogenic insults. Our results indicate that increasing 4E-BP1 expression or enhancing 4E-BP1 activation can robustly induce the mitochondrial unfolded protein response and thus could be an appealing strategy for treating a variety of neurodegenerative diseases, including especially PD.SIGNIFICANCE STATEMENT In neurodegenerative disease, misfolded proteins accumulate and overwhelm normal systems of homeostasis and quality control. One mechanism for improving protein quality control is to reduce protein translation. Here we investigated whether neuronal overexpression of 4E-BP1, a key repressor of protein translation, can protect against misfolded protein stress and toxicities linked to Parkinson's disease, and found that 4E-BP1 overexpression prevented cell death in neurons treated with brefeldin A, rotenone, maneb, paraquat, or preformed fibrils of α-synuclein. When we sought the basis for 4E-BP1 neuroprotection, we discovered that 4E-BP1 activation promoted the mitochondrial unfolded protein response. Our findings highlight 4E-BP1 as a therapeutic target in neurodegenerative disease and underscore the importance of the mitochondrial unfolded protein response in neuroprotection against various insults.
- Published
- 2020
46. Elucidating the Novel Role for Core Binding Factor beta in Osteosarcoma Protein Translation
- Author
-
Oldberg, Nick Alexander
- Subjects
Pharmacology ,Oncology ,Cellular biology ,CBFB ,Osteosarcoma ,Protein Translation ,RUNX2 ,Target Validation - Abstract
Osteosarcoma (OS) is the most common primary bone malignancy in humans and canines, and in humans primarily affects younger patients 10-14 years of age. While considerable efforts have been put forth in new therapeutic approaches to this disease, the treatment and prognosis for OS has changed very little since the 1980s. Targeted therapeutics have made considerable progress in other cancer types, leveraging characteristics of cancer cells which differentiate them from that of normal healthy cells. In comparison to other cancers, OS is highly heterogeneic, and no single unifying driver mutation has yet been found. Development of a therapy which could overcome the high degree of heterogeneity amongst OS tumors could go a long way in improving the lives of OS patients. Targeting protein translation has been proposed as one mechanism by which to overcome tumor heterogeneity, and this dissertation focuses on studying a noncanonical role of core binding factor beta (CBFβ) as a regulator of protein translation, and elucidating whether this could represent a potential therapeutic target for OS. Utilizing a wide array of in vitro assays, we have been able to demonstrate that loss of CBFβ reduces protein expression of RUNX2 in a post-transcriptional manner, and this decrease in RUNX2 protein level is not fully explained by alterations in RUNX2 stability brought about by loss of its binding partner CBFβ. Additionally, we demonstrate that loss of CBFβ also causes a decrease in global protein translation, and confirmed an interaction between CBFβ and hnRNPK which has thus far only been observed in breast cancer cells. Importantly, this interaction with hnRNPK is said to be the mechanism by which CBFβ influences protein translation, and our results corroborate those observed in breast cancer cells and suggest CBFβ may also perform this role in OS. Reports of the interactions between CBFβ and hnRNPK or RUNX2 allude to mutual exclusivity in interaction, and with the transcriptional role of CBFβ accomplished via binding to RUNX proteins, and the translational role of CBFβ accomplished via binding to hnRNPK, it is entirely possible these two roles are antagonistic in some fashion. To investigate the relevance of certain CBFβ residues in terms of this translational role of CBFβ, and avoid confounding variables from the transcriptional role of CBFβ, we utilized point mutations to interrupt CBFβ-RUNX2 interaction. Using various in vitro assays, we validated key residues of CBFβ which are involved in its interaction with RUNX2, re-introduced this mutant form into CBFβ knockout cells, and measured alterations to RUNX2 interaction and nuclear shuttling. We confirmed that our mutant displays reduced binding to RUNX2, and drastically reduced nuclear shuttling. Lastly, we expanded our studies from RUNX2 to the entire genome and proteome. Encouraging data thus far had suggested CBFβ may play a role in protein translation, and necessary next steps were to assess which proteins CBFβ may be interacting with in performance of this role, and elucidate which proteins may be under the translational purview of CBFβ. Using immunoprecipitation mass spectrometry we identified numerous specific interactors of CBFβ, with high enrichment in pathway analysis terms associated with protein translation. Additionally, using two different methods we generated a list of proteins which may be under the translational purview of CBFβ, and found strong enrichment of numerous cancer-associated terms among this list. These studies establish that CBFβ participates in protein translation in OS, with many genes under its purview associated strongly with cancer in general, and OS specifically. This provides justification for future studies delving deeper into this novel role of CBFβ, and opens up another mechanism by which protein translation could be targeted therapeutically in OS.
- Published
- 2024
47. Extracellular arginine availability modulates eIF2α O-GlcNAcylation and heme oxygenase 1 translation for cellular homeostasis
- Author
-
Yu-Wen Hung, Ching Ouyang, Xiaoli Ping, Yue Qi, Yi-Chang Wang, Hsing-Jien Kung, and David K. Ann
- Subjects
Arginine ,O-GlcNAcylation ,Eukaryotic initiation factor 2α ,Protein translation ,Heme oxygenase 1 ,Antioxidant defense ,Medicine - Abstract
Abstract Background Nutrient limitations often lead to metabolic stress during cancer initiation and progression. To combat this stress, the enzyme heme oxygenase 1 (HMOX1, commonly known as HO-1) is thought to play a key role as an antioxidant. However, there is a discrepancy between the level of HO-1 mRNA and its protein, particularly in cells under stress. O-linked β-N-acetylglucosamine (O-GlcNAc) modification of proteins (O-GlcNAcylation) is a recently discovered cellular signaling mechanism that rivals phosphorylation in many proteins, including eukaryote translation initiation factors (eIFs). The mechanism by which eIF2α O-GlcNAcylation regulates translation of HO-1 during extracellular arginine shortage (ArgS) remains unclear. Methods We used mass spectrometry to study the relationship between O-GlcNAcylation and Arg availability in breast cancer BT-549 cells. We validated eIF2α O-GlcNAcylation through site-specific mutagenesis and azido sugar N-azidoacetylglucosamine-tetraacylated labeling. We then evaluated the effect of eIF2α O-GlcNAcylation on cell recovery, migration, accumulation of reactive oxygen species (ROS), and metabolic labeling during protein synthesis under different Arg conditions. Results Our research identified eIF2α, eIF2β, and eIF2γ, as key O-GlcNAcylation targets in the absence of Arg. We found that O-GlcNAcylation of eIF2α plays a crucial role in regulating antioxidant defense by suppressing the translation of the enzyme HO-1 during Arg limitation. Our study showed that O-GlcNAcylation of eIF2α at specific sites suppresses HO-1 translation despite high levels of HMOX1 transcription. We also found that eliminating eIF2α O-GlcNAcylation through site-specific mutagenesis improves cell recovery, migration, and reduces ROS accumulation by restoring HO-1 translation. However, the level of the metabolic stress effector ATF4 is not affected by eIF2α O-GlcNAcylation under these conditions. Conclusions Overall, this study provides new insights into how ArgS fine-tunes the control of translation initiation and antioxidant defense through eIF2α O-GlcNAcylation, which has potential biological and clinical implications.
- Published
- 2023
- Full Text
- View/download PDF
48. The activation of mTOR signalling modulates DNA methylation by enhancing DNMT1 translation in hepatocellular carcinoma
- Author
-
Mengke Chen, Yi Fang, Meinong Liang, Ning Zhang, Xinyue Zhang, Lixia Xu, Xuxin Ren, Qingfeng Zhang, Yufeng Zhou, Sui Peng, Jun Yu, Judeng Zeng, and Xiaoxing Li
- Subjects
Hepatocellular carcinoma ,Protein translation ,DNA methylation ,DNMT1 ,Pyrimidine rich translational element ,Medicine - Abstract
Abstract Background Both dysregulation of mechanistic target of rapamycin (mTOR) signalling and DNA methylation patterns have been shown to be closely associated with tumor progression and serve as promising targets for hepatocellular carcinoma (HCC) therapy. Although their respective roles in HCC have been extensively revealed, the existence of molecular interactions between them remains largely unknown. Methods The association of DNA methylation and mTOR signalling in HCC tissues and cell lines was assessed. A Kaplan‒Meier analysis was applied to estimate the overall survival (OS) and recurrence-free survival (RFS) of HCC patients. The modulation of DNMT1 by mTOR in HCC cell lines was determined. The effect of the drug combination in cell lines and mouse models was examined. Results The results showed that the DNA methylation level was positively associated with the activation of mTOR signalling in HCC tissues and cell lines. Moreover, HCC patients with higher DNA methylation levels and enhanced activation of mTOR signalling exhibited the worst prognosis. Then, we screened methylation-related enzymes and found that the activation of mTOR signalling increased DNMT1 expression and activity. In addition, mTOR enhanced the translational efficiency of DNMT1 in a 4E-BP1-dependent manner, which is based on the pyrimidine rich translational element (PRTE)-containing 5′UTR of DNMT1. Moreover, we demonstrated that the combined inhibition of mTOR and DNMT synergistically inhibited HCC growth in vitro and in vivo. Conclusions In addition to some already identified pro-cancer downstream molecules, the activation of mTOR signalling was found to promote DNA methylation by increasing the translation of DNMT1. Furthermore, combined targeting of mTOR and DNMT1 has been demonstrated to have a more effective tumor suppressive function in HCC.
- Published
- 2023
- Full Text
- View/download PDF
49. PKR-mediated stress response enhances dengue and Zika virus replication
- Author
-
Taissa Ricciardi-Jorge, Edroaldo Lummertz da Rocha, Edgar Gonzalez-Kozlova, Gabriela Flavia Rodrigues-Luiz, Brian J. Ferguson, Trevor Sweeney, Nerea Irigoyen, and Daniel Santos Mansur
- Subjects
dengue virus ,Zika virus ,innate immunity ,protein translation ,PKR ,Microbiology ,QR1-502 - Abstract
ABSTRACT The mechanisms by which flaviviruses use non-canonical translation to support their replication in host cells are largely unknown. Here, we investigated how the integrated stress response (ISR), which promotes translational arrest by eIF2ɑ phosphorylation (p-eIF2ɑ), regulates flavivirus replication. During dengue virus (DENV) and Zika virus (ZIKV) infection, eIF2ɑ phosphorylation peaked at 24 hours post-infection and was dependent on protein kinase RNA-activated (PKR) but not type I interferon. The ISR is activated downstream of p-eIF2α during infection with either virus, but translation arrest only occurred following DENV4 infection. Despite this difference, both DENV4 and ZIKV replications were impaired in cells lacking PKR, independent of type I interferon/NF-kB signaling or cell viability. By using a ZIKV 5′-untranslated region (UTR) reporter system as a model, we found that this region of the genome is sufficient to promote an enhancement of viral mRNA translation in the presence of an active ISR. Together, we provide evidence that flaviviruses escape ISR translational arrest and co-opt this response to increase viral replication. IMPORTANCE One of the fundamental features that make viruses intracellular parasites is the necessity to use cellular translational machinery. Hence, this is a crucial checkpoint for controlling infections. Here, we show that dengue and Zika viruses, responsible for nearly 400 million infections every year worldwide, explore such control for optimal replication. Using immunocompetent cells, we demonstrate that arrest of protein translations happens after sensing of dsRNA and that the information required to avoid this blocking is contained in viral 5′-UTR. Our work, therefore, suggests that the non-canonical translation described for these viruses is engaged when the intracellular stress response is activated.
- Published
- 2023
- Full Text
- View/download PDF
50. The delayed bloodstream clearance of Plasmodium falciparum parasites after M5717 treatment is attributable to the inability to modify their red blood cell hosts Molly.
- Author
-
Schneider, Molly Parkyn, Looker, Oliver, Rebelo, Maria, Khoury, David S., Dixon, Matthew W. A., Oeuvray, Claude, Crabb, Brendan S., McCarthy, James, and Gilson, Paul R.
- Subjects
PLASMODIUM falciparum ,PARASITES ,PLASMODIUM ,PARASITE life cycles ,PROTEIN synthesis ,RIBOSOMES ,DRUG development ,ERYTHROCYTES - Abstract
M5717 is a promising antimalarial drug under development that acts against multiple stages of the life cycle of Plasmodium parasites by inhibiting the translation elongation factor 2 (PfeEF2), thereby preventing protein synthesis. The parasite clearance profile after drug treatment in preclinical studies in mice, and clinical trials in humans showed a notable delayed clearance phenotype whereby parasite infected red blood cells (iRBCs) persisted in the bloodstream for a significant period before eventual clearance. In a normal P. falciparum infection iRBCs sequester in the deep circulation by cytoadherence, allowing them to avoid surveillance and clearance in the spleen. We found that M5717 blocks parasite modification of their host red blood cells (RBCs) by preventing synthesis of new exported proteins, rather than by directly blocking the export of these proteins into the RBC compartment. Using in vitro models, we demonstrated that M5717 treated ring/trophozoite stage iRBCs became less rigid, and cytoadhered less well compared to untreated iRBCs. This indicates that in vivo persistence of M5717 treated iRBCs in the bloodstream is likely due to reduced cytoadherence and splenic clearance. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.