Or Gozani’s research while affiliated with Stanford University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (183)


SMYD5 is largely dispensable in mice and there is no evidence for SMYD5 having histone methylation activity in vitro or in cells
a, SMYD5 expression levels in gastric cancer negatively correlate with GAC patient progression free survival (n = 498 patients). P-values determined by log-rank test. b, Summary of full embryonic Smyd5 knockout mice phenotype data from the IMPC (International Mouse Phenotyping Consortium). Smyd5 deletion has no impact on viability and fertility and mice have only minor phenotypes as shown in the table. c, Top, main domain structure of SMYD5. SMYD5, like the four other SMYD family members (SMYDs 1–4), contains a catalytic methyltransferase SET domain that is disrupted due to insertion of a MYND domain. When the protein folds, the two parts of the SET domain form a continuous functional domain as shown. Bottom, alignment of the putative catalytic region of SMYD5 with the catalytic regions of SMYD2 and SMYD3. Grey box, homology, or similarity. Red box, catalytic tyrosine in SMYD2 and SMYD3 that was used to generate a catalytic mutant for SMYD5. d, SMYD5 depletion has no effect on the levels of H3K36me3 and H4K20me3 in three different human cell lines. Western blots with the indicated antibodies of WCEs from control or SMYD5-depleted A549, KKLS, and U2OS cells. Total H3 and tubulin are shown as loading controls. e, SMYD5 does not methylate free histones, octamers, or nucleosomes in vitro. In vitro methylation assays with the indicated enzymes and substrates. H3: free histone H3; H4: free histone H4; Oct: recombinant octamers (2 each of H2A, H2B, H3 and H4); rNuc: recombinant nucleosome. SETD2 and SUV420H1 are included as positive controls for methylation of H3K36 and H4K20, respectively. Note that SUV420H1’s preferred substrate is H4K20me1 nucleosome and as indicated, H4K20me1 nucleosomes were used in the SUV420H1 methylation assays. Top panel, ³H-SAM was used as methyl donor and the methylation was visualized by autoradiography. Bottom panel, Coomassie stain of proteins present in the in vitro reactions. f, SMYD5 localizes to the cytoplasm. Western blots with the indicated antibodies of biochemically separated 293 T WCEs into nuclear and cytoplasmic fractions. Tubulin and H3 are shown as controls for the integrity of the fractionation with Tubulin exclusively present in cytoplasmic fractions and H3 exclusively present in nuclear fractions.
Purification of SMYD5 catalytic activity in cell lysates to identify UBA52 as candidate substrate
a, Western blots of WCEs from control or SMYD5-depleted KKLS and U2OS cells used in b as substrates. b, SMYD5 methylates a ~ 10 kDa protein in KKLS and U2OS cell lysates. In vitro methylation assays as in Fig. 1d with the indicated enzymes and WCEs in a as substrates. c, The candidate SMYD5 substrate migrates faster than histone H4. In vitro methylation reactions as in b with the indicated KMT enzymes or GST alone as a negative control using SMYD5-depleted A549 WCEs as substrates as in Fig. 1d. Red arrowhead, SMYD5-dependent methylated protein. Blue arrowhead, SETD8-dependent histone H4 methylated at K20. d, Substantial enrichment of SMYD5-dependent methylated protein in extracts at 4th fractionation step (see schematic Fig. 1e). In vitro methylation assays as in Fig. 1d with the indicated enzymes using as substrate SMYD5-depleted A549 WCE, Input (lysate after 3 fractionation steps: (1) cytoplasmic isolation, (2) ammonium sulfate salt precipitation, and (3) hydrophobic interaction chromatography), and the indicated fractions (1–13) of the input material separated by size-exclusion chromatography. The Coomassie staining demonstrates the enrichment of the activity relative to total proteins present in the starting material. The region around the ~10 kDa band signal in Fraction 13, which showed the highest degree of purification, was analyzed by mass spectrometry (See Supplementary Tables 1–4). e, 2D gel electrophoresis separation of Fraction 13 in d further purifies the SMYD5-dependent methylated ~10kD protein. Red asterisk, SMYD5-dependent methyl band. The region around the red asterisk was analyzed by mass spectrometry to obtain UBA52 as the top hit. Left: Silver stain of 2D separation of Fraction 13 proteins; Middle: ³H-SAM was used as methyl donor and the methylation was visualized by autoradiography. Right: merge of the two panels.
SMYD5 specifically methylates rpL40 and has no activity on many other putative substrates in vitro
a, The sequence of human rpL40 protein surrounding lysine 22. The site of methylation at K22 is indicated. Due to the many charged residues and the specific residues surrounding K22, irrespective of proteases, it is challenging to obtain suitable peptides for MS-based identification. b, SMYD5 methylates recombinant un-cleaved UBA52 protein in vitro. c, SMYD5 methylates recombinant rpL40 but not ubiquitin alone. d, Besides UBA52, SMYD5 did not in vitro methylate several other candidate proteins identified as potential candidates by MS/MS analysis of fraction 13 described in Extended Data Fig. 2d. e, SMYD5 does not in vitro methylate HIV-1 Tat protein. rpL40 is shown as a positive control.
Generation of a specific rpL40K22me3 antibody and evidence that SMYD5 is the principal enzyme that physiologically generates rpL40K22me3
a, rpL40K22me3 antibody recognizes cognate methylated peptide sequence but not unmethylated peptide. Dot blot analysis with the indicated rpL40 peptides spanning amino acids (16–26) at the indicated concentrations probed with the rpL40K22me3 antibody (top) or stained with Ponceau S (bottom) to control for loading. b, rpL40K22me3 antibody recognizes cognate methylated peptide sequence but does not recognize several other histone and non-histone peptides harboring trimethylation. Dot bot analysis as in a. c, Western blot with the rpL40K22me3 antibody of in vitro methylation reactions with the indicated enzyme and substrates. d-e, Western blots with the indicated antibodies of WCEs from d, LMSU and e, SH-10-TC cell lines expressing CRISPR-Cas9 and two independent sgRNAs targeting SMYD5 or a control sgRNA. Tubulin was used as loading control. f, Western blots of SMYD5-depleted LMSU cells complemented with CRISPR-resistant SMYD5WT, SMYD5Y351A catalytic mutant, or control plasmid. g, Western blots with indicated antibodies of sucrose-cushion purified ribosome-depleted cytoplasmic lysates and ribosomes-enriched fractions from KKLS cells.
Alphafold analysis of SMYD5-rpL40 interaction and structural analysis of rpL40 in human ribosomes
a-b, UBA52 + SMYD5 predicted structures (AlphaFold v2.3; see Methods) a, UBA52/rpL40 depicted by blue cartoon and SMYD5 by red molecular surface, as indicated. K22 of rpL40 the SMYD5 catalytic tunnel-forming residue (Y351) are shown. K22 of rpL40 directly inserts into SMYD5’s catalytic tunnel. b, UBA52 + SMYD5 predicted structures coloured by electrostatic charge. Colouring key depicts predict Coulombic electrostatic potential, with red corresponding to negative charge and blue corresponding to positive charge. rpL40 substrate lysine K22 and SMYD5 catalytic Y351 are shown. c-e, Ribosome-bound rpL40 undergoes rearrangements through different stages of elongation factor association. c, Top, ribbon representation of the overall complex of human 80 S ribosomes bound to eEF1A (PDB: 6ZMO), bottom, human 80 S ribosomes bound to eEF2 (PDB: 6D9J). rpL40, eEF1A and eEF2 are shown in surface representations and differentially coloured as indicated. d-e, close-up view of potential intermolecular interaction between rpL40K22 and adjacent 28 S rRNA. Distances are provided of hydrogen-bonding interactions depicted as dashed lines. d, rpL40K22 (shown in blue) from apo human 80 S ribosome structure (PDB: 4UG0) that is not bound to initiation/elongation factors and has close interactions with the 28 S rRNA at bases C4412 and C4413. rpL40K22 (shown in red) from 80 S ribosome bound to eEF1a (PDB: 6ZMO) leads to a shift in K22 of 5.3 Å toward an ionic interaction with G1945 of the 28 S rRNA. e, as in d but replacing eEF1A with 80 S ribosome bound to eEF2 (PDB: 6D9J), rpL40K22 shifts by 6.6 Å in favor of more distant interactions with G4411.

+10

SMYD5 methylation of rpL40 links ribosomal output to gastric cancer
  • Article
  • Publisher preview available

July 2024

·

188 Reads

·

3 Citations

Nature

Juhyung Park

·

Jibo Wu

·

·

[...]

·

Dysregulated transcription due to disruption in histone lysine methylation dynamics is an established contributor to tumorigenesis1,2. However, whether analogous pathologic epigenetic mechanisms act directly on the ribosome to advance oncogenesis is unclear. Here we find that trimethylation of the core ribosomal protein L40 (rpL40) at lysine 22 (rpL40K22me3) by the lysine methyltransferase SMYD5 regulates mRNA translation output to promote malignant progression of gastric adenocarcinoma (GAC) with lethal peritoneal ascites. A biochemical–proteomics strategy identifies the monoubiquitin fusion protein partner rpL40 (ref. ³) as the principal physiological substrate of SMYD5 across diverse samples. Inhibiting the SMYD5–rpL40K22me3 axis in GAC cell lines reprogrammes protein synthesis to attenuate oncogenic gene expression signatures. SMYD5 and rpL40K22me3 are upregulated in samples from patients with GAC and negatively correlate with clinical outcomes. SMYD5 ablation in vivo in familial and sporadic mouse models of malignant GAC blocks metastatic disease, including peritoneal carcinomatosis. Suppressing SMYD5 methylation of rpL40 inhibits human cancer cell and patient-derived GAC xenograft growth and renders them hypersensitive to inhibitors of PI3K and mTOR. Finally, combining SMYD5 depletion with PI3K–mTOR inhibition and chimeric antigen receptor T cell administration cures an otherwise lethal in vivo mouse model of aggressive GAC-derived peritoneal carcinomatosis. Together, our work uncovers a ribosome-based epigenetic mechanism that facilitates the evolution of malignant GAC and proposes SMYD5 targeting as part of a potential combination therapy to treat this cancer.

View access options



Fig. 1 (See legend on next page.)
Fig. 3 BCAR3 methylation promotes cell migration and invasiveness. a, b Representative single-cell migration tracks monitored by time-lapse live microscopy (a) and quantification of single-cell migration velocity (b) of MDA-MB-231 cells with indicated engineering. Boxes: 25th to 75th percentile, whiskers: 10 to 90 percentile, center line: median. P-values were calculated by ANOVA with Tukey's testing for multiple comparisons. c, d Representative images of three-dimensional collagen I matrix invasion (c) and spreading quantification (d) of basement membrane enclosed MDA-MB-231 with indicated engineering. P-values were calculated by ANOVA with Tukey's testing for multiple comparisons. Scale bars, 50 μm. e 3D image reconstruction of spheroid edge depicting engineered MDA-MB-231 cells protrusions, related to (c). Scale bars represent 10 µm. In all box plots, the center line indicates the median, the box marks the 75th and 25th percentiles and the whiskers indicate 10 to 90 percentile values.
Fig. 4 FMNLs are BCAR3 methyl-specific interactors. a Identification of K334 methylation-specific BCAR3 binding partners using SILAC-based quantitative proteomics screen. A scatter plot of the log 2 transformed protein-normalized methyl-sensitive peptide SILAC ratios. The x-axis shows the Log 2 ratio of proteins binding to BCAR3 K334me1 versus BCAR3 K334me0 (Forward). The y-axis shows the Log 2 ratio of a label-swap replicate experiment (Reverse). Specific interactors of methylated BCAR3 reside in the upper right quadrant. b, c Immunoblot analysis with the indicated antibodies of protein pulldowns using unmethylated (me0) or K334 monomethylated (me1) BCAR3 peptides from MDA-MB-231 cell extracts (b) or recombinant GST-FMNLs proteins (c). d Co-immunoprecipitation of endogenous BCAR3 after enrichment of endogenous FMNL2/3 in MDA-MB-231 cells upon genetic or pharmacologic SMYD2 repression. Tubulin is shown as a loading control. e, f Representative images (e) and signal quantification (f) of PLA monitoring BCAR3-FMNL3 interaction in situ, coupling HA-BCAR3 and Flag-FMNL3 antibodies in MDA-MB-231 cells with indicated engineering. Dotted red lines represent cells periphery and dotted white square represents the enlarged area depicted in Supplementary Fig. S4c. P-values were calculated by BrownForsythe and Welch ANOVA tests with Dunnett's T3 testing for multiple comparisons. Scale bars, 10 μm. In all box plots, the center line indicates the median, the box marks the 75th and 25th percentiles and the whiskers indicate 10 to 90 percentile values.
Fig. 5 FMNLs contains a novel methyl-binding domain. a Immunoblot analysis with the indicated antibodies of protein pulldowns using unmethylated (me0) or K334 monomethylated (me1) BCAR3 peptides from extracts of 293 T cells expressing full-length, GBD-FH3 or FH1-FH2-DAD domains of FMNL3. b Predicted structure model of FMNL3 GBD-FH3 domain interaction with BCAR3 K334me1 peptide, based on the available structure of FMNL2 GBD-FH3/CDC42 GppNHp (PDBe code: 4YC7). The putative methylated lysine-binding hydrophobic pocket of FMNL3 containing two aromatic residues (W124 and Y237) surrounded by aliphatic residues (L117, I122, V125, A233) is shown. c Sequence alignment of FMNL1, 2 and 3 showing a strict conservation of all aromatic and aliphatic residues identified in the structural model of FMNL3 hydrophobic pocket responsible for BCAR3me1 recognition. d Immunoblot analysis with the indicated antibodies of peptide pulldowns using unmethylated (me0) or K334 monomethylated (me1) BCAR3 peptides and recombinant GST-FMNL3 proteins either WT or harboring W124A or Y237A point mutations.
Fig. 6 BCAR3 methylation recruits FMNLs to lamellipodia and regulates cytoskeleton protrusions. a, b Representative immunofluorescence images (a) and signal quantification (b) of F-Actin (phalloidin) and Flag-FMNL3 in MDA-MB-231 cells with indicated engineering. Magnifications of the leading edge of lamellipodia protrusions are provided. P-values were calculated by Brown-Forsythe and Welch ANOVA tests with Dunnett's T3 testing for multiple comparisons. Scale bars, 5 μm. c, d Representative images of lamellipodia visualized by Z-projection of F-Actin staining (c) and lamellipodia F-Actin density quantification (d) in MDA-MB-231 cells with indicated engineering. Dotted red lines represent lamellipodia area. P-values were calculated by Brown-Forsythe and Welch ANOVA tests with Dunnett's T3 testing for multiple comparisons. Scale bars, 5 μm. e, f Representative visualization (kymograph, distance vs time) of lamellipodia protrusion rate (e) and quantification of lamellipodia protrusion velocity (f) in MDA-MB-231 cells with indicated modifications. P-values were calculated by Kruskal-Wallis test with Dunn's testing for multiple comparisons. In all box plots, the center line indicates the median, the box marks the 75th and 25th percentiles and the whiskers indicate 10 to 90 percentile values.
Cytoskeleton remodeling induced by SMYD2 methyltransferase drives breast cancer metastasis

January 2024

·

90 Reads

·

4 Citations

Cell Discovery

Malignant forms of breast cancer refractory to existing therapies remain a major unmet health issue, primarily due to metastatic spread. A better understanding of the mechanisms at play will provide better insights for alternative treatments to prevent breast cancer cell dispersion. Here, we identify the lysine methyltransferase SMYD2 as a clinically actionable master regulator of breast cancer metastasis. While SMYD2 is overexpressed in aggressive breast cancers, we notice that it is not required for primary tumor growth. However, mammary-epithelium specific SMYD2 ablation increases mouse overall survival by blocking the primary tumor cell ability to metastasize. Mechanistically, we identify BCAR3 as a genuine physiological substrate of SMYD2 in breast cancer cells. BCAR3 monomethylated at lysine K334 (K334me1) is recognized by a novel methyl-binding domain present in FMNLs proteins. These actin cytoskeleton regulators are recruited at the cell edges by the SMYD2 methylation signaling and modulate lamellipodia properties. Breast cancer cells with impaired BCAR3 methylation lose migration and invasiveness capacity in vitro and are ineffective in promoting metastases in vivo. Remarkably, SMYD2 pharmacologic inhibition efficiently impairs the metastatic spread of breast cancer cells, PDX and aggressive mammary tumors from genetically engineered mice. This study provides a rationale for innovative therapeutic prevention of malignant breast cancer metastatic progression by targeting the SMYD2-BCAR3-FMNL axis.



Cytoskeleton remodeling induced by SMYD2 methyltransferase drives breast cancer metastasis

September 2023

·

38 Reads

·

1 Citation

Malignant forms of breast cancer refractory to existing therapies remain a major unmet health issue, primarily due to metastatic spread. A better understanding of the mechanisms at play will provide better insights for alternative treatments to prevent breast cancer cells dispersion. Here, we identify the lysine methyltransferase SMYD2 as a clinically actionable master regulator of breast cancer metastasis. While SMYD2 is overexpressed in aggressive breast cancers, we notice that it is not required for primary tumor growth. However, mammary-epithelium specific SMYD2 ablation increases mouse overall survival by blocking the primary tumor cells ability to metastasize. Mechanistically, we identify BCAR3 as a genuine physiological substrate of SMYD2 in breast cancer cells. BCAR3 monomethylated at lysine K334 (K334me1) is recognized by a novel methyl-binding domain present in FMNLs proteins. These actin cytoskeleton regulators are recruited at the cell edges by the SMYD2 methylation signaling and modulates lamellipodia properties. Breast cancer cells with impaired BCAR3 methylation loose migration and invasiveness capacity in vitro and are ineffective in promoting metastases in vivo . Remarkably, SMYD2 pharmacologic inhibition efficiently impairs the metastatic spread of breast cancer cells, PDX and aggressive mammary tumors from genetically engineered mice. This study provides a rationale for innovative therapeutic prevention of malignant breast cancer metastatic progression by targeting the SMYD2-BCAR3-FMNL axis.



Targeting KDM2A Enhances T-cell Infiltration in NSD1-Deficient Head and Neck Squamous Cell Carcinoma

June 2023

·

36 Reads

·

4 Citations

Cancer Research

In head and neck squamous cell carcinoma (HNSCC), a significant proportion of tumors have inactivating mutations in the histone methyltransferase NSD1. In these tumors, NSD1 inactivation is a driver of T-cell exclusion from the tumor microenvironment (TME). A better understanding of the NSD1-mediated mechanism regulating infiltration of T cells into the TME could help identify approaches to overcome immunosuppression. Here, we demonstrated that NSD1 inactivation results in lower levels of H3K36 dimethylation and higher levels of H3K27 trimethylation, the latter being a known repressive histone mark enriched on the promoters of key T-cell chemokines CXCL9 and CXCL10. HNSCC with NSD1 mutations had lower levels of these chemokines and lacked responses to PD-1 immune checkpoint blockade. Inhibition of KDM2A, the primary lysine demethylase that is selective for H3K36, reversed the altered histone marks induced by NSD1 loss and restored T-cell infiltration into the TME. Importantly, KDM2A suppression decreased growth of NSD1-deficient tumors in immunocompetent, but not in immunodeficient, mice. Together, these data indicate that KDM2A is an immunotherapeutic target for overcoming immune exclusion in HNSCC. Significance The altered epigenetic landscape of NSD1-deficient tumors confers sensitivity to inhibition of the histone-modifying enzyme KDM2A as an immunotherapeutic strategy to stimulate T-cell infiltration and suppress tumor growth.


The FAM86 domain of FAM86A confers substrate specificity to promote EEF2-Lys525 methylation

May 2023

·

17 Reads

·

2 Citations

Journal of Biological Chemistry

FAM86A is a class I lysine methyltransferase (KMT) that generates trimethylation on the eukaryotic translation elongation factor 2 (EEF2) at Lys525. Publicly available data from The Cancer Dependency Map project indicate high dependence of hundreds of human cancer cell lines on FAM86A expression. This classifies FAM86A among numerous other KMTs as potential targets for future anticancer therapies. However, selective inhibition of KMTs by small molecules can be challenging due to high conservation within the S-Adenosyl methionine (SAM) co-factor binding domain amongst KMT subfamilies. Therefore, understanding the unique interactions within each KMT-substrate pair can facilitate developing highly specific inhibitors. The FAM86A gene encodes an N-terminal FAM86 domain of unknown function in addition to its C-terminal methyltransferase domain. Here, we used a combination of X-ray crystallography, the AlphaFold algorithms, and experimental biochemistry to identify an essential role of the FAM86 domain in mediating EEF2 methylation by FAM86A. To facilitate our studies, we also generated a selective EEF2K525 methyl antibody. Overall, this is the first report of a biological function for the FAM86 structural domain in any species, and an example of a non-catalytic domain participating in protein lysine methylation. The interaction between the FAM86 domain and EEF2 provides a new strategy for developing a specific FAM86A small molecule inhibitor and our results provide an example in which modeling a protein-protein interaction with AlphaFold expedites experimental biology.


Antibody toolkit to investigate eEF1A methylation dynamics in mRNA translation elongation

April 2023

·

38 Reads

·

4 Citations

Journal of Biological Chemistry

Protein synthesis is a fundamental step in gene expression, with modulation of mRNA translation at the elongation step emerging as an important regulatory node in shaping cellular proteomes. In this context, five distinct lysine methylation events on eEF1A (eukaryotic elongation factor 1A), a fundamental non-ribosomal elongation factor, are proposed to influence mRNA translation elongation dynamics. However, a lack of affinity tools has hindered progress in fully understanding how eEF1A lysine methylation impacts protein synthesis. Here we develop and characterize a suite of selective antibodies to investigate eEF1A methylation and provide evidence that methylation levels decline in aged tissue. Determination of the methyl state and stoichiometry on eEF1A in various cell lines by mass spectrometry shows modest cell-to-cell variability. We also find by Western blot analysis that knockdown of individual eEF1A-specific lysine methyltransferases (KMTs) leads to depletion of the cognate lysine methylation event and indicates active crosstalk between different sites. Further, we find that the antibodies are specific in immunohistochemistry (IHC) applications. Finally, application of the antibody toolkit suggests that several eEF1A methylation events decrease in aged muscle tissue. Together our study provides a roadmap for leveraging methyl state and sequence selective antibody reagents to accelerate discovery of eEF1A methylation-related functions and suggests a role for eEF1A methylation, via protein synthesis regulation, in aging biology.


Citations (75)


... Our results confirm two recent studies, which were published while this work was nearing completion 43,44 . Both groups also found that SMYD5 methylates RPL40 K22 but does not methylate histone substrates 43,44 . ...

Reference:

SMYD5 is a ribosomal methyltransferase which trimethylates RPL40 lysine 22 through recognition of a KXY motif
SMYD5 methylation of rpL40 links ribosomal output to gastric cancer

Nature

... A total of 39 hub genes were identified using eight different algorithms, encompassing 21 ribosomal proteins (involved in the translation process) and 18 additional proteins. These hub genes are predominantly involved in transcriptional regulation (Snrpd3), translation (21 ribosomal proteins, Eef2, and Eef1a1), protein folding and refolding (Hsp90ab1, Hspa1a, Tcp1, Cct5), and coagulation (C4bpa, Fgg, Serpine1) [9][10][11][12] during the critical phase of placental development. (Snrpd3), translation (21 ribosomal proteins, Eef2, and Eef1a1), protein folding and refolding (Hsp90ab1, Hspa1a, Tcp1, Cct5), and coagulation (C4bpa, Fgg, Serpine1) [9][10][11][12] during the critical phase of placental development. ...

FAM86A methylation of eEF2 links mRNA translation elongation to tumorigenesis
  • Citing Article
  • March 2024

Molecular Cell

... Specifically, through the monomethylation of lysine K334 in the adapter protein NSP family member (BCAR3), SMYD2 induces the recognition of the methyl-binding domain in formin-like (FMNL) proteins, thereby regulating the actin cytoskeleton. Through this action, it has been confirmed lamellipodia formation, an important feature of cell movement, is regulated by SMYD2 methylation 44 . Furthermore, SMYD2 regulates lung cancer metastasis by directly controlling SMAD family member 3 (SMAD3), which regulates the progression of lung cancer. ...

Cytoskeleton remodeling induced by SMYD2 methyltransferase drives breast cancer metastasis

Cell Discovery

... Interestingly, we found that gene KMT2A is typically wrapped into high-order TAD (Fig. 5c), whose copy gains and break aparts are associated with CTCF depletion and reduced binding [32]. KMT2A amplifications and translocations are prevalent in infant, adult, and therapy-induced leukemia. ...

Epigenetic balance ensures mechanistic control of MLL amplification and rearrangement
  • Citing Article
  • October 2023

Cell

... [6,7] The overall response rates to immune checkpoint inhibitors are generally limited and remain below 20% in LSCC. [8,9] Multiple interventions aimed at enhancing synthetic and endogenous immunemediated tumor cell rejection are currently being extensively investigated, [10] but these approaches face significant challenges that necessitate a comprehensive understanding of the intricate interactions between tumor cells and heterogeneous infiltrating immune cells within the tumor microenvironment (TME). ...

Targeting KDM2A Enhances T-cell Infiltration in NSD1-Deficient Head and Neck Squamous Cell Carcinoma
  • Citing Article
  • June 2023

Cancer Research

... In fact, only a handful of structures of 7βS methyltransferases bound to whole-protein substrates have been determined to-date: bacterial PrmA with ribosomal protein L11 (14), human DOT1L with a nucleosome substrate (15), and recently, human VCP-KMT with VCP/p97 J o u r n a l P r e -p r o o f (16,17). Additionally, a model of human eEF2-KMT/FAM86A bound to eEF2 has recently been determined using AlphaFold (18). All these structures show that contacts distal to the methyltransferase active site are critical for binding and methylation. ...

The FAM86 domain of FAM86A confers substrate specificity to promote EEF2-Lys525 methylation
  • Citing Article
  • May 2023

Journal of Biological Chemistry

... In this regard, there is growing appreciation that context-dependent dysregulation of mRNA translation influences tumorigenesis 1,2,44,45 . As several other ribosomal and elongation factors are methylated 46,47 , it will be interesting to understand how crosstalk between these methylation events, and with other key ribosomal modifications 48 , regulate the translatome in human health and disease. ...

Antibody toolkit to investigate eEF1A methylation dynamics in mRNA translation elongation

Journal of Biological Chemistry

... nucleosomes, which are enriched in promotor and enhancer regions to influence the transcription 12,18 . Furthermore, the acetylation of H2A.Z by the KAT5 subunit is tightly linked to transcription activation 19 . EP400 and KAT5 were also reported to function together to alter nucleosome stability during DNA double-strand break repair 11,20 . ...

Tip60-mediated H2A.Z acetylation promotes neuronal fate specification and bivalent gene activation
  • Citing Article
  • November 2022

Molecular Cell

... Notably, N84T is near the active site of the 2A protease and is known to confer resistance to the drug telaprevir but reduces the viral replication efficiency [23]. AA substitution T57A (2A) is in close proximity to G58, which has been identified as a critical contact residue involved in the interaction with the host cell protein SETD3 [24]. ...

Structure-function analysis of enterovirus protease 2A in complex with its essential host factor SETD3

... In contrast, NSD2 truncating and missense variants have been causally associated with Wolf-Hirschhorn syndrome, a genetic disorder characterized by intellectual and developmental delay [16][17][18]. Translocation-mediated overexpression and hyperactive variants of NSD2 and NSD3 have been identified in several types of cancer, notably in multiple myeloma, acute lymphoblastic leukemia, and lung squamous cell carcinoma, with elevated H3K36me2 being implicated in tumour progression and survival outcomes [19][20][21][22][23]. As an H3K36 methyltransferase, ASH1L remains relatively understudied. ...

NSD2 dimethylation at H3K36 promotes lung adenocarcinoma pathogenesis
  • Citing Article
  • September 2021

Molecular Cell