John W. R. Schwabe’s research while affiliated with Cambridge University Hospitals NHS Foundation Trust 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 (154)


Circular Engineered Sortase for Interrogating Histone H3 in Chromatin
  • Article

November 2024

·

6 Reads

Journal of the American Chemical Society

·

Kwangwoon Lee

·

Zhipeng A. Wang

·

[...]

·

Philip A. Cole

Reversible modification of the histone H3 N-terminal tail is critical in regulating the chromatin structure, gene expression, and cell states, while its dysregulation contributes to disease pathogenesis. Understanding the crosstalk between H3 tail modifications in nucleosomes constitutes a central challenge in epigenetics. Here, we describe an engineered sortase transpeptidase, cW11, that displays highly favorable properties for introducing scarless H3 tails onto nucleosomes. This approach significantly accelerates the production of both symmetrically and asymmetrically modified nucleosomes. We demonstrate the utility of asymmetrically modified nucleosomes produced in this way in dissecting the impact of multiple modifications on eraser enzyme processing and molecular recognition by a reader protein. Moreover, we show that cW11 sortase is very effective at cutting and tagging histone H3 tails from endogenous histones, facilitating multiplex “cut-and-paste” middle-down proteomics with tandem mass tags. This cut-and-paste proteomics approach permits the quantitative analysis of histone H3 modification crosstalk after treatment with different histone deacetylase inhibitors. We propose that these chemoenzymatic tail isolation and modification strategies made possible with cW11 sortase will broadly power epigenetic discovery and therapeutic development.


Fig. 2 (A) CI-994 was incorporated as the HDAC ligand in our previously reported PROTACs, 1 is a HDAC1 and HDAC2 selective inhibitor reported by Ibrahim et al. 16 and 2 is a direct analogue of 1 without the para-fluorine phenyl ring (B) Comparison of HDAC2 active site with the zinc bound to a benzamide inhibitor on the left (PDB: 3MAX) and the HDAC3 active site with acetate bound to the zinc on the right (PDB: 4A69), the para-fluorine phenyl ring in 1 has been proposed to occupy the larger foot pocket present in HDAC1/2 (C) A library of potential cereblon-recruiting PROTACs utilising 1 and 2 as the HDAC ligand in attempt to improve the physiochemical properties compared to previous PROTACs utilising CI-994.
Cereblon-recruiting proteolysis targeting chimeras (PROTACs) can determine the selective degradation of HDAC1 over HDAC3
  • Article
  • Full-text available

November 2024

·

39 Reads

Chemical Communications

Cereblon-recruiting PROTACs can degrade HDAC1 with selectivity over HDAC3.

Download

Cereblon-Based Compounds for Induction of HbF

November 2024

·

8 Reads

Blood

Fetal hemoglobin (HbF) induction is a well-known strategy for the treatment of hemoglobinopathies such as Sickle Cell Disease (SCD) and Thalassemia, worldwide life-threatening conditions. Increased levels of HbF are able to prevent HbS polymerization and mitigate disease manifestations, resulting in lower morbidity and mortality in SCD. Nowadays there are only four drugs approved by the FDA for SCD treatment: Hydroxyurea, voxelotor, L-glutamine and crizanlizumab, while the search for new effective and safe treatments are scarce. Controlling the expression of the gamma-globin gene by inhibiting or degrading epigenetic targets has been shown to be promising for HbF induction, and histone deacetylases (HDACs 1 and 2) stand out as potential targets. Targeted protein degradation (TPD) was explored in this work with the synthesis of new cereblon-based molecules able to anchor to cereblon and HDACs 1 and 2 simultaneously, leading to their dose dependent decrease via polyubiquitination and proteasome degradation. Six new cereblon derivatives containing a HDAC inhibitor subunit were designed, synthesized and evaluated in HCT-116 cells for HDAC 1, 2 and 3 degradation via quantitative western blot at concentrations ranging from 0.01 to 10 µM. The promising results showed that three of these compounds (ARP-26, ARP-37 and ARP-49) were able to degrade HDAC-1 more selectively, when compared to HDAC-2 and HDAC-3, achieving DC50 values of 2.5 µM. A derivative with no ability to bind to cereblon was also synthesized resulting in no degradation of HDACs 1-3, suggesting that these compounds act via cereblon binding. ARP-49 was then selected for further studies in HUDEP-2 cell culture at concentrations of 500 and 750nM, and incubation time of 72 and 96h. After the treatment, cells were stained with HbF-antibody and the percentage of HbF positive cells was assessed by flow cytometry. The quantification of HBG1/2 mRNA was obtained by qPCR. HUDEP-2 cells exhibit a basal level of HbF positive cells lower than 1%, then the results obtained from our compounds were normalized based on the control. At 72h, the concentrations of 500nM and 750nM were able to significantly increase the HBG1/2 mRNA levels by 26.9-fold and 35.5-fold, respectively (CTRL = 0.027 ± 0.006 arbitrary units (a.u.) vs 500nM = 0.73 ±0.25 a.u. and 750nM = 0.96 ± 0.11 a.u., p<0.0001, n=4). At 96h, both concentrations were also able to significantly induce the expression of the HBG1/2 genes by 34-fold and 44.5-fold, respectively (CTRL = 0.034 ± 0.012 a.u. vs 500nM = 1.12 ± 0.35 a.u. and 750nM = 1.52 ± 0.37 a.u., p<0.0001, n=4). HbF-positive cells at 72h presented a significantly increase of 7.5-fold and 9-fold at 500 and 750nM, respectively (CTRL = 0.58 ± 0.15 % vs 500nM = 4.43 ± 0.88 % and 750nM = 5.29 ± 1.52 %, p<0.0001, n=4), while at 96h the increase was 8.37-fold and 10.6-fold, respectively (CTRL = 0.65 ± 0.15 % vs 500nM = 5.44 ± 0.79 % and 750nM = 6.89 ± 1.05 %, p<0.0001, n=4). HUDEP-2 cells were also treated with HU at 100 µM, reaching its highest level of HBG1/2 expression at 72h (10-fold), when compared to control, and an increase in the HbF-positive cells population by 8.5-fold and 8.9-fold at 72h and 96h, respectively (CTRL 72h = 0.66 ± 0.23% vs HU 72h = 5.65 ± 0.59%, CTRL 96h = 0.64 ± 0.15% vs HU 96h = 5.73 ± 0.52%; p<0.0001, n=2). Compounds obtained in this study present a degradation selectivity towards HDAC-1 with no previous description in the literature. Furthermore, the ability to significantly induce HBG1/2 expression in HUDEP-2 cells was superior to the one seen for the standard SCD treatment, HU, with levels of HBG1/2 expression 2.6-fold higher (500nM, 72h), at a concentration 200-fold lower. These findings support that ARP-49 shows promising potential for further pre-clinical studies for increasing the production of HbF.


Damaging mutations in liver X receptor-α are hepatotoxic and implicate cholesterol sensing in liver health

September 2024

·

125 Reads

·

1 Citation

Nature Metabolism

Liver X receptor-α (LXRα) regulates cellular cholesterol abundance and potently activates hepatic lipogenesis. Here we show that at least 1 in 450 people in the UK Biobank carry functionally impaired mutations in LXRα, which is associated with biochemical evidence of hepatic dysfunction. On a western diet, male and female mice homozygous for a dominant negative mutation in LXRα have elevated liver cholesterol, diffuse cholesterol crystal accumulation and develop severe hepatitis and fibrosis, despite reduced liver triglyceride and no steatosis. This phenotype does not occur on low-cholesterol diets and can be prevented by hepatocyte-specific overexpression of LXRα. LXRα knockout mice exhibit a milder phenotype with regional variation in cholesterol crystal deposition and inflammation inversely correlating with steatosis. In summary, LXRα is necessary for the maintenance of hepatocyte health, likely due to regulation of cellular cholesterol content. The inverse association between steatosis and both inflammation and cholesterol crystallization may represent a protective action of hepatic lipogenesis in the context of excess hepatic cholesterol.


A circular engineered sortase for interrogating histone H3 in chromatin

September 2024

·

8 Reads

Reversible modification of the histone H3 N-terminal tail is critical in regulating chromatin structure, gene expression, and cell states, while its dysregulation contributes to disease pathogenesis. Understanding the crosstalk between H3 tail modifications in nucleosomes constitutes a central challenge in epigenetics. Here we describe an engineered sortase transpeptidase, cW11, that displays highly favorable properties for introducing scarless H3 tails onto nucleosomes. This approach significantly accelerates the production of both symmetrically and asymmetrically modified nucleosomes. We demonstrate the utility of asymmetrically modified nucleosomes produced in this way in dissecting the impact of multiple modifications on eraser enzyme processing and molecular recognition by a reader protein. Moreover, we show that cW11 sortase is very effective at cutting and tagging histone H3 tails from endogenous histones, facilitating multiplex “cut-and-paste” middle down proteomics with tandem mass tags. This cut-and-paste proteomics approach permits the quantitative analysis of histone H3 modification crosstalk after treatment with different histone deacetylase inhibitors. We propose that these chemoenzymatic tail isolation and modification strategies made possible with cW11 sortase will broadly power epigenetics discovery and therapeutic development.


Figure 1. Identification of loss of function and dominant negative mutations in the ligand binding domain of LXRa carried by participants in population biobanks. We characterised 65 coding variants in the ligand binding domain of LXRa in two separate assays. A: Stacked bar chart illustrating LXRa mutant activity. HEK293 cells were transfected with an LXR-luciferase reporter construct and wildtype or mutant LXRa. 4 hours after transfection, cells were treated with indicated concentration of the LXRa agonist T0901317 and luminescence was measured as an index of LXRa activity 20 hours later. The dashed coloured lines indicate average activity of wildtype LXRa at the corresponding concentration of T0901317. Statistical analysis of mutant vs WT activity was assessed by two-way ANOVA with Geisser-Greenhouse correction and multiple comparison controlling for FDR at each ligand concentration by two-stage step-up method of Benjamini, Krieger and Yekutieli. FDR-corrected P-values are presented in Supplementary Table 1. N=3-6 experiments were performed on diZerent day per variant in the same cell line. Height of bars and error bars represent mean +/-standard deviation. B: Forest plot of mutant LXRa activity in a co-expression assay. Wildtype and mutant LXRa and a LXR-luciferase reporter construct were transfected into HEK293 cells. 24 hours later luminescence was measured as an index of LXRa activity. Assuming normal distribution, significant increases and decreases according to a two-tailed one-sample t-test are depicted in green and red, respectively. N=4-8 independent experiments were performed on diZerent days in the same cell line. Dots and error bars represent mean +/-95% confidence intervals. EV= empty vector, WT= wildtype, DM = an artificial double mutant which is strongly dominant negative.
Damaging mutations in LXRα uncouple lipogenesis from hepatotoxicity and implicate hepatic cholesterol sensing in human liver health

April 2024

·

123 Reads

·

1 Citation

The nuclear receptor Liver X Receptor-α (LXR α) activates lipogenic gene expression in hepatocytes. Its inhibition has therefore been proposed as a strategy to treat metabolic dysfunction-associated steatotic liver disease (MASLD). In order to understand the impact of reducing LXRα activity on human health we first examined the association between the carriage of rare loss of function mutations in NR1H3 (encoding LXRα) and metabolic and hepatic phenotypes. We identified 63 rare predicted damaging variants in the ligand binding domain of LXRα in 454,787 participants in UK Biobank. On functional characterisation, 42 of these were found to be severely impaired. Consistent with loss of the lipogenic actions of LXRα, carriers of damaging mutations in LXRα had reduced serum triglycerides (Beta=-0.13 s.d. [plusmn]0.03, P=2.7x10 ⁻⁵ , N(carriers)=971). Surprisingly, these carriers also have elevated concentrations of serum liver enzymes (e.g. ALT: Beta=0.17s.d.±0.03, P=1.1x10 ⁻⁸ , N(carriers)=972) with a 35% increased risk of clinically significant elevations in ALT (OR=1.32, 95%CI:1.15-1.53, P=1.2x10 ⁻⁴ , N(carriers)=972), suggestive of hepatotoxicity. We generated a knock-in mouse carrying one of the most severely damaging mutations (Nr1h3 p.W441R) which we demonstrated to have dominant negative properties. Homozygous knock-in mice rapidly developed severe hepatitis and fibrotic liver injury following exposure to western diet despite markedly reduced steatosis, liver triglycerides and lipogenic gene expression. This phenotype was completely rescued by viral over-expression of wildtype LXRα specifically in hepatocytes, indicating a cell-autonomous effect of the mutant on hepatocyte health. While homozygous LXRα knockout mice showed some evidence of hepatocyte injury under similar dietary conditions, the phenotype of the LXRα W441R/W441R mouse was much more severe, suggesting that dominant negative mutations that actively co-repress target genes can result in pathological impacts significantly more severe than those seen with simple absence of the receptor. In summary, our results show that loss of function mutations in LXRα occur in at least 1/450 people and are associated with evidence of liver dysfunction. These findings implicate LXRα in the maintenance of human liver health, identify a new murine model of rapidly progressive fibrotic liver disease and caution against LXR antagonism as a therapeutic strategy for MASLD.


Aster-dependent nonvesicular transport facilitates dietary cholesterol uptake

November 2023

·

104 Reads

·

23 Citations

Science

Intestinal absorption is an important contributor to systemic cholesterol homeostasis. Niemann-Pick C1 Like 1 (NPC1L1) assists in the initial step of dietary cholesterol uptake, but how cholesterol moves downstream of NPC1L1 is unknown. We show that Aster-B and Aster-C are critical for nonvesicular cholesterol movement in enterocytes. Loss of NPC1L1 diminishes accessible plasma membrane (PM) cholesterol and abolishes Aster recruitment to the intestinal brush border. Enterocytes lacking Asters accumulate PM cholesterol and show endoplasmic reticulum cholesterol depletion. Aster-deficient mice have impaired cholesterol absorption and are protected against diet-induced hypercholesterolemia. Finally, the Aster pathway can be targeted with a small-molecule inhibitor to manipulate cholesterol uptake. These findings identify the Aster pathway as a physiologically important and pharmacologically tractable node in dietary lipid absorption.



Figure 4. The amino-terminal region of MIER1(aa:1-177) binds a full histone octamer. (A) Alphafold2 model of the interaction of MIER1(aa:17-75) (lime green/cyan) with an H2A:H2B dimer (grey cartoon). (B) Alphafold2 model of the the interaction of MIER1(aa:17-75) (lime green/cyan) binding to H2A:H2B (electrostatic surface) and the model rotated 260 • clockwise about y-axis. (C) The FLAG tagged amino-terminus of wild type MIER1(aa:1-177) (red symbols), Mutation 1 (S71A, L72A) (green symbols), Mutation 2 (F22A, M28A, L29A) (purple symbols) purified with histone H2A:H2B dimer, the Mutation 3 (I66A, L69A, L70A) (orange symbols) does not bind to histones (sequences shown in Supplement Figure S3). (D) GST pull down experiments confirm the ability of the amino-terminus of MIER1(aa:1-177) to bind a full histone octamer. (E) GST pull down experiments show that the amino-terminus of MIER1(aa:1-177) does not bind to a DNA-wrapped nucleosome. (F, G) Comparison of the Alphafold2 model for the binding of the amino-terminal region of MIER1(aa:17-75) to a histone octamer with a DNA-bound nucleosome.
A potential histone-chaperone activity for the MIER1 histone deacetylase complex

April 2023

·

118 Reads

·

7 Citations

Nucleic Acids Research

Histone deacetylases 1 and 2 (HDAC1/2) serve as the catalytic subunit of six distinct families of nuclear complexes. These complexes repress gene transcription through removing acetyl groups from lysine residues in histone tails. In addition to the deacetylase subunit, these complexes typically contain transcription factor and/or chromatin binding activities. The MIER:HDAC complex has hitherto been poorly characterized. Here, we show that MIER1 unexpectedly co-purifies with an H2A:H2B histone dimer. We show that MIER1 is also able to bind a complete histone octamer. Intriguingly, we found that a larger MIER1:HDAC1:BAHD1:C1QBP complex additionally co-purifies with an intact nucleosome on which H3K27 is either di- or tri-methylated. Together this suggests that the MIER1 complex acts downstream of PRC2 to expand regions of repressed chromatin and could potentially deposit histone octamer onto nucleosome-depleted regions of DNA.


Figure 3. NMR analysis of K-Ras4B G12V :Rgl2 RBD complex formation in solution (A) Rgl2 RBD solution structure determined by NMR. The 2D 1 H-15 N-HSQC spectrum of the assigned residues of the Rgl2 RBD is shown (left). The Rgl2 RBD retains the ββαββαβ ubiquitin-fold structure (right). 20 Rgl2 NMR structures are superimposed. (B) Comparison between the Rgl2 RBD
Structural and biochemical insights into heterotetramer formation between human oncogenic K-Ras4B G12V and Rgl2, a RalA/B activator

October 2022

·

78 Reads

About a quarter of total human cancers carry mutations in Ras isoforms. Accumulating evidence suggests that small GTPases, RalA and RalB, and their activators, Ral guanine nucleotide exchange factors (RalGEFs), play an essential role in oncogenic Ras-induced signalling. We studied the interaction between human K-Ras4B and the Ras association (RA) domain of Rgl2 (Rgl2 RA ), one of the RA-containing RalGEFs. We show that the G12V oncogenic K-Ras4B mutation increases the affinity with Rgl2 RA . The crystal structure of the K-Ras4B G12V : Rgl2 RA complex shows a 2:2 heterotetramer where the Switch I and Switch II regions of each K-Ras G12V interact with both Rgl2 RA molecules. This structural arrangement is highly similar to the H-Ras E31K :RALGDS RA crystal structure and is distinct from the well-characterised Ras:Raf complex. Importantly, the G12V mutation was found at the dimer interface of K-Ras4B G12V with its partner. Our study reveals a potentially distinct mode of Ras:effector complex formation by RalGEFs, and offers a possible mechanistic explanation for how the oncogenic K-Ras4B G12V hyperactivates the RalA/B pathway. Summary Blurb The work reports that the K-RAS4B.G12V oncogenic mutation increases the affinity with Rgl2, an activator of RalA/B and provides a structural explanation by analysing the K-Ras4B(G12V):Rgl2 complex crystal structure where the G12V mutation resides at the interface of the complex.


Citations (61)


... In support of the mouse models, genome-wide association studies in humans have identified variants in the human LXRα gene (NR1H3) linked to lipid levels and insulin sensitivity [37][38][39] . Rare variants in LXRα are also associated with markers of liver damage 40 suggesting an important role for LXRα in maintaining normal liver function. ...

Reference:

A mutation in LXRα uncovers a role for cholesterol sensing in limiting metabolic dysfunction-associated steatohepatitis
Damaging mutations in liver X receptor-α are hepatotoxic and implicate cholesterol sensing in liver health

Nature Metabolism

... Specifically, harmful mutations in the LXRα ligandbinding domain can result in abnormal lipid metabolism and are associated with decreased triglyceride levels and increased high-density lipoproteins (HDL) cholesterol. Despite these changes in lipid levels, these mutations were also linked to increased serum liver enzymes, indicating possible hepatotoxicity [1]. LXR alpha plays a complex role in type 2 diabetes because it affects several metabolic pathways, including the metabolism of fatty acids and cholesterol. ...

Damaging mutations in LXRα uncouple lipogenesis from hepatotoxicity and implicate hepatic cholesterol sensing in human liver health

... Gramd1b and Gramd1c are highly expressed in differentiated enterocytes, while Gramd1a is prevalent in other intestinal cell types, including crypt stem cells and T cells (120). Intestinal deletion of Aster-B and -C results in lower uptake of diet-derived cholesterol into enterocytes and into the circulation, and atten-uates diet-induced hypercholesterolemia (120). ...

Aster-dependent nonvesicular transport facilitates dietary cholesterol uptake
  • Citing Article
  • November 2023

Science

... Interestingly, KDM1A (LSD1), RCOR1, RCOR3, SIN3A, NCOR1, and MIER1 showed an even more pronounced degradation. All mentioned proteins represent family members of multi-subunit complexes (CoREST, SMRT/NCoR, SIN3, and MIER) which contain HDAC1/2 or 3.42,43 The phenomenon of degraded proteins within multi-subunit complexes that contain the POI can be explained by E3 ligase recruitment and ternary complex formation. This process not only leads to the polyubiquitination of the POI but also of other members within the entire complex. ...

A potential histone-chaperone activity for the MIER1 histone deacetylase complex

Nucleic Acids Research

... The mammalian cell lines are the dominant platforms for producing recombinant protein drugs. These cell lines function similarly to human natural proteins due to their ability to perform post-translational modifications, such as glycosylation and disulfide bonding [5,6]. However, it should be noted that compared to microbial cells, the productivity of mammalian cell culture is much lower, and their cultivation is laborious, costly, and complex [7,8]. ...

Recombinant Protein Expression for Structural Biology in HEK 293F Suspension Cells: A Novel and Accessible Approach
  • Citing Article
  • October 2014

Journal of Visualized Experiments

... Finally, the N501Y mutation, present in most VOCs except for Delta, significantly enhances binding affinity by introducing π-π interactions with the ACE2 residue Y41, as well as additional CH-π and cation-π interactions with K353. These findings align with prior studies that reported similar enhancements in binding due to these mutations, including a dramatic increase in binding affinity with combined mutations Q498R and N501Y in vitro [41]. ...

In vitro evolution predicts emerging SARS-CoV-2 mutations with high affinity for ACE2 and cross-species binding

... Nearly complete degradation of both HDAC1 and HDAC2 was observed after treatment of 10 μM of 40 in HCT116 cells, while HDAC3 levels were also decreased, to a lesser extent [91]. Similarly, Sinatra et al. developed another series of HDAC PROTACs [92]. PROTAC 41 could induce degradation of HDAC1 and HDAC 3 at low micromolar concentrations. ...

Optimization of Class I Histone Deacetylase PROTACs Reveals that HDAC1/2 Degradation is Critical to Induce Apoptosis and Cell Arrest in Cancer Cells

Journal of Medicinal Chemistry

... In addition, they provide the basis for the rationale design and development of thyromimetics able to efficiently dissociate CoRs from mutant TRs, thereby alleviating receptor dysfunction in RTH. Recently, the crystal structure of a TRα mutant (P393GX LBD), prototypic of RTH-associated TRα mutants, in complex with T3 oriented the synthesis of different thyroid hormone analogues, and allowed the identification of ES08 as a lead compound (31). In silico modeling of ES08 binding to P393GX shows that the sulfonate ester extension at the 4′-hydroxyl of the outer thyronine ring in this compound protrudes out from the LBP and thus could disrupt the CoR binding surface. ...

Structure-Guided Approach to Relieving Transcriptional Repression in Resistance to Thyroid Hormone α

... Caspase-8 has important residues for its function: the catalytic Cys360 [89], Asp374, and Asp384. These aspartic acid residues are the cleavage sites between the p18 and p12 subunits of Caspase-8 cleavage at these sites and are crucial for the maturation and full activation of Caspase-8 [90]. ...

Cryo-EM structural analysis of FADD:Caspase-8 complexes defines the catalytic dimer architecture for co-ordinated control of cell fate