Karla-Luise Herpoldt’s research while affiliated with University of Washington 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 (7)


Controlled encapsulation of macromolecular cargoes by in vitro assembly of I53‐50‐V5 nanoparticles. a) Schematic of the computationally designed two‐component icosahedral protein nanoparticle I53‐50. Each nanoparticle consists of 12 copies of a pentameric building block (orange) and 20 copies of a trimeric building block (gray). These assemble spontaneously in E. coli cells when co‐expressed, or upon controlled mixing in vitro after independent expression and purification. b) SEC of I53‐50 variants produced by co‐expression in E. coli (top) or in vitro assembly (bottom). Absorbance was measured at 260 and 280 nm to detect the presence of packaged nucleic acid contaminants. Each chromatogram was normalized such that the peak absorbance at 280 nm equaled 1 for ease of comparison. Full chromatograms are provided in Figure S1d (Supporting Information). c) Representative negatively stained electron micrograph and 2D class averages of in vitro‐assembled I53‐50‐V5 particles. d) DLS measurements of the nanoparticles from panel (b) assembled by co‐expression in E. coli or by mixing in vitro assembly. Hydrodynamic radii (Rh) are indicated. e) Schematic of electrostatic encapsulation of negatively charged cargoes via in vitro assembly. f–h) Biophysical characterization of I53‐50‐V5 assembled in the presence of two different sizes of nucleic acid cargoes, 400 nt (top) and 2500 nt (bottom), by (f) SEC, (g) DLS, and (h) nsEM (left, field view; right, selected 2D class averages).
Protection of encapsulated ssRNA from nuclease challenge. a) Native agarose gel electrophoresis of encapsulation reactions containing increasing amounts of 400 nt ssRNA. P:N, protein:nucleic acid ratio (moles nanoparticle:ssRNA). b) RT‐qPCR quantitation of ssRNA recovery after Benzonase treatment of encapsulation reactions in panel (a). c) RT‐qPCR quantitation of ssRNA recovery after Benzonase treatment of 800 and 1600 nt ssRNA encapsulation reactions at 25 and 37 °C. The 25 °C data also appear in panel (d). d) RT‐qPCR quantitation of ssRNA recovery after Benzonase treatment of 800 and 1600 nt ssRNA encapsulation reactions at 25 °C and various CHAPS concentrations. e) Decay of encapsulated ssRNA of various lengths as function of the duration of Benzonase challenge. RT‐qPCR was used to quantify full‐length ssRNA present after incubation with Benzonase for 1 min, 20 min, 1 h, 3 h, 9 h, 24 h, 3 days, 9 days, and 27 days. In all panels, error bars indicate standard deviations from triplicate measurements.
Encapsulation of non‐biological polymeric nanoparticles inside a designed protein nanoparticle. a) Cartoon representation of the structure of a radiant star polymer nanoparticle. b) Native agarose gel electrophoresis of increasing sizes of RSN present upon encapsulation. c) SEC trace of RSN alone (top) and RSN encapsulated within I53‐50‐V5 (bottom). Small polymers, once encapsulated, eluted at the same elution volume as empty I53‐50‐V5 (dotted vertical line). d) DLS size and polydispersity measurements of RSN alone or encapsulated within I53‐50‐V5. e) Representative negatively stained electron micrograph and class averages of DP25 encapsulated within I53‐50‐V5 particles. f) Negatively stained electron micrograph of a DP200 encapsulation reaction. The data shown are from representative experiments that were performed at least twice.
Encapsulated polymeric adjuvants improve immune responses in vitro and in vivo. a) Schematic of resiquimod prodrug monomer that is polymerized into RSNs via RAFT. b) SEC trace of pResi alone (top) and pResi encapsulated within I53‐50‐V5 (bottom). Once encapsulated, the polymer nanoparticle elutes earlier from the column, at the same elution volume as empty I53‐50‐V5, indicating it is now trapped within the protein nanoparticle. c) Kinetics of release of free resiquimod from pResi alone in human serum or buffer at pH 5 and encapsulated pResi in human pooled serum (HPS). d) In vitro cytokine response from human PBMCs (each of three donors represented by a distinct symbol) incubated for 24 h with free resiquimod or encapsulated pResi. e) Serum IgG responses against I53‐50‐V5 after three immunizations of BALB/c mice (n = 5), measured as EC50 titer. f) Anti‐I53‐50‐V5 antibody titers measured over 24 weeks for groups receiving encapsulated pResi (green), free resiquimod (gray), or no adjuvant (purple). g) In vivo systemic cytokine responses measured 1 h post‐immunization. Only free resiquimod at a dose commonly used in the literature induced TNF‐α and IL‐6 levels above background. The data shown are from representative experiments that were performed at least twice. Error bars in panel (f) indicate standard deviation. Statistical significance was determined by an independent t‐test with Bonferroni correction; ****, p ≤ 1 × 10⁻⁴; ***, p ≤ 1 × 10⁻³; **, p ≤ 1 × 10⁻²; *, p ≤ 5 × 10⁻².
Macromolecular Cargo Encapsulation via In Vitro Assembly of Two‐Component Protein Nanoparticles
  • Article
  • Full-text available

February 2024

·

32 Reads

·

3 Citations

Karla‐Luise Herpoldt

·

Ciana L. López

·

Isaac Sappington

·

[...]

·

Neil P. King

Self‐assembling protein nanoparticles are a promising class of materials for targeted drug delivery. Here, the use of a computationally designed, two‐component, icosahedral protein nanoparticle is reported to encapsulate multiple macromolecular cargoes via simple and controlled self‐assembly in vitro. Single‐stranded RNA molecules between 200 and 2500 nucleotides in length are encapsulated and protected from enzymatic degradation for up to a month with length‐dependent decay rates. Immunogenicity studies of nanoparticles packaging synthetic polymers carrying a small‐molecule TLR7/8 agonist show that co‐delivery of antigen and adjuvant results in a more than 20‐fold increase in humoral immune responses while minimizing systemic cytokine secretion associated with free adjuvant. Coupled with the precise control over nanoparticle structure offered by computational design, robust and versatile encapsulation via in vitro assembly opens the door to a new generation of cargo‐loaded protein nanoparticles that can combine the therapeutic effects of multiple drug classes.

Download


Figure 2. JUPITER's first phase of research focused on studying individual previously designed successful nanoparticles. Analysis of I3-01 31 through the pSUFER protocol. A-C, pSUFER overview: A, The interface between two nanoparticle subunits of a relaxed design model, here with side chains highlighted in stick representation, is selected using ResidueSelectors; B, each interface residue (here, K23) is computationally mutated to all possible amino acids; and C, the free energy difference compared to the wild-type (ddG) is calculated. D-F, Data visualization by the students. D, A representative barplot showing the energy difference for each possible mutation at a given sequence position (K32), highlighting a favorable mutation (K23I, green) and an unfavorable mutation (K23Y, pink). E, PyMOL visualization of pSUFER scores for the whole interface by
Figure 4. JUPITER students further refined their hypotheses and developed new tests. A, Students updated their hypotheses to analyze residues neighboring but not directly participating in the previously analyzed interfaces. The students found that residues neighboring the interface had the most favorable mutations halfway between the center of the mass of the interface and the furthest residue from the interface. Each circle on the graph represents a single position of a single nanoparticle. B, The Rosetta score function can be refined by adding distance-and burial-dependent score terms to reward favorable electrostatic interactions (distance) or penalize buried polar residues (burial) based on their depth from the protein surface. By applying two different variants of the score function, the students observed that the addition of burial-and distance-dependent score terms shifted the degree to which different residues lead to favorable mutations across all nanoparticles.
Increasing computational protein design literacy through cohort-based learning for undergraduate students

May 2022

·

70 Reads

Undergraduate research experiences can improve student success in graduate education and STEM careers. During the COVID-19 pandemic, undergraduate researchers at our institution and many others lost their work-study research positions due to interruption of in-person research activities. This imposed a financial burden on the students and eliminated an important learning opportunity. To address these challenges, we created a paid, fully-remote, cohort-based research curriculum in computational protein design. Our curriculum used existing protein design methods as a platform to first educate and train undergraduate students and then to test research hypotheses. In the first phase, students learned computational methods to assess the stability of designed protein assemblies. In the second phase, students used a larger dataset to identify factors that could improve the accuracy of current protein design algorithms. This cohort-based program created valuable new research opportunities for undergraduates at our institute and enhanced the undergraduates’ feeling of connection with the lab. Students learned transferable and useful skills such as literature review, programming basics, data analysis, hypothesis testing, and scientific communication. Our program provides a model of structured computational research training opportunities for undergraduate researchers in any field for organizations looking to expand educational access. Graphical Abstract


Fig. 2 Direct and Indirect antigens derived from P. gingivalis in induction of T-cell functions. In the left panel mFA is shown to induce a Th2 response whereas FimA elicits Th1 activity, it is not known if these effects are direct or indirect but FimA can signal through TLR2 to upregulate NO and CD11b/CD18 integrin expression. FimA also acts as a ligand to the CD11b/CD18 integrin. In the right panel, the products that purify out from the LPS-extraction method are shown; antagonist and agonist LPS (pink and purple) and the lipoprotein from gene product PG1828 (orange). Together, this 'LPS' can elicit CD4 T-cell, and Th17 expansion as well as IL-17 from γδTc subsets. Although the signaling pathways for T-cell induction are not yet identified, LPS typically signals through TLR4 but can also be presented (lipid A) by DCs via CD1b or CD1c.
Mucosal pathogens that require Th17-mediated protection.
Is the oral microbiome a source to enhance mucosal immunity against infectious diseases?

December 2021

·

98 Reads

·

27 Citations

npj Vaccines

Mucosal tissues act as a barrier throughout the oral, nasopharyngeal, lung, and intestinal systems, offering first-line protection against potential pathogens. Conventionally, vaccines are applied parenterally to induce serotype-dependent humoral response but fail to drive adequate mucosal immune protection for viral infections such as influenza, HIV, and coronaviruses. Oral mucosa, however, provides a vast immune repertoire against specific microbial pathogens and yet is shaped by an ever-present microbiome community that has co-evolved with the host over thousands of years. Adjuvants targeting mucosal T-cells abundant in oral tissues can promote soluble-IgA (sIgA)-specific protection to confer increased vaccine efficacy. Th17 cells, for example, are at the center of cell-mediated immunity and evidence demonstrates that protection against heterologous pathogen serotypes is achieved with components from the oral microbiome. At the point of entry where pathogens are first encountered, typically the oral or nasal cavity, the mucosal surfaces are layered with bacterial cohabitants that continually shape the host immune profile. Constituents of the oral microbiome including their lipids, outer membrane vesicles, and specific proteins, have been found to modulate the Th17 response in the oral mucosa, playing important roles in vaccine and adjuvant designs. Currently, there are no approved adjuvants for the induction of Th17 protection, and it is critical that this research is included in the preparedness for the current and future pandemics. Here, we discuss the potential of oral commensals, and molecules derived thereof, to induce Th17 activity and provide safer and more predictable options in adjuvant engineering to prevent emerging infectious diseases.


DMS-guided structure-based design of repacked (“Rpk”) SARS-CoV-2 RBDs. (A) Molecular surface representation of the SARS-CoV-2 S trimer ectodomain (PDB 6VYB), with a close-up view of the RBD (PDB 6VXX) which highlights both the location of the linoleic acid-binding pocket and the receptor-binding motif (RBM). Each protomer is colored distinctly, and N-linked glycans are rendered dark green. (B) The linoleic acid-binding pocket within the RBD, which was targeted for stabilizing mutations. The left panel shows the apo structure (PDB 6VXX) and the right panel shows conformational changes with linoleic acid (black) bound (PDB 6ZB5). (C) Mutations that increased RBD expression, identified by DMS of the RBD using yeast display (57) were used to guide Rosetta-based design of stabilized RBDs. Structural models of stabilized RBDs were generated from PDB 6VXX for Rpk4 and Rpk9, and PDB 6YZ5 for Rpk11. All experimentally tested stabilizing mutations are shown in Supplementary Table 1 . (D) Cropped reducing and non-reducing SDS-PAGE of supernatants from HEK293F cells after small-scale expression of stabilized RBD designs genetically fused to the I53-50A trimer. “Negative” refers to a negative control plasmid that does not encode a secreted protein. Uncropped gels are shown in Supplementary Figure 1 .
Expression, thermal stability, and structural order of stabilized RBDs is improved while remaining antigenically intact. (A) SEC purification of wild-type and stabilized RBDs after expression from equal volumes of HEK293F cultures followed by IMAC purification and concentration. Monomeric RBDs (left) were purified using a Superdex 75 Increase 10/300 GL while fusions to the I53-50A trimer (right) were purified using a Superdex 200 Increase 10/300 GL. Cropped gels show equivalently diluted SEC load samples. Uncropped gels are shown in Supplementary Figure 2 . (B) Thermal denaturation of wild-type and stabilized RBD monomers (left) and fusions to the I53-50A trimer (right), monitored by nanoDSF using intrinsic tryptophan fluorescence. Top panels show the barycentric mean (BCM) of each fluorescence emission spectrum as a function of temperature, while lower panels show smoothed first derivatives used to calculate melting temperatures. (C) HDX-MS of wild-type and stabilized RBDs fused to I53-50A trimers. The structural model from PDB 6W41 is shown with differences in deuterium uptake at the 1 minute timepoint highlighted (top). Both Rpk4-I53-50A and Rpk9-I53-50A showed similar increases in exchange protection in similar regions. The red box highlights the peptide segment from residues 392–399, with exchange for this peptide shown at 3 sec, 15 sec, 1 min, 30 min, and 20 h timepoints (bottom). Each point is an average of two measurements. Standard deviations are shown unless smaller than the points plotted. A complete set of plots for all peptide segments is shown in Supplementary Figure 3 . (D) Fluorescence of SYPRO Orange when mixed with equal concentrations of wild-type and stabilized RBD monomers. (E) Binding kinetics of immobilized CV30 and CR3022 monoclonal antibodies to monomeric wild-type and stabilized RBDs as assessed by BLI. Experimental data from five concentrations of RBDs in two-fold dilution series (colored traces) were fitted (black lines) with binding equations describing a 1:1 interaction. Structural models (left) were generated by structural alignment of the SARS-CoV-2 bound to CV30 Fab (PDB 6XE1) and CR3022 Fab (PDB 6W41).
Stabilized RBDs presented on assembled I53-50 nanoparticles enhance solution stability compared to the wild-type RBD. (A) Schematic of assembly of I53-50 nanoparticle immunogens displaying RBD antigens. (B) nsEM of RBD-I53-50, Rpk4-I53-50, and Rpk9-I53-50 (scale bar, 200 nm). (C–E) show summarized quality control results for RBD-I53-50, Rpk4-I53-50, and Rpk9-I53-50 before and after a single freeze/thaw cycle in four different buffers. Complete data available in Supplementary Information 3 . (C) The ratio of absorbance at 320 to 280 nm in UV-Vis spectra, an indicator of the presence of soluble aggregates. (D) DLS measurements, which monitor both proper nanoparticle assembly and formation of aggregates. (E) Fractional reactivity of I53-50 nanoparticle immunogens against immobilized hACE2-Fc receptor (top) and CR3022 (bottom). The pre-freeze and post-freeze data were separately normalized to the respective CHAPS-containing samples for each nanoparticle.
Potent immunogenicity of the parental RBD-I53-50 nanoparticle immunogen is maintained with addition of Rpk mutations. (A) Female BALB/c mice (six per group) were immunized at weeks 0 and 3. Each group received equimolar amounts of RBD antigen adjuvanted with AddaVax, which in total antigen equates to 5 μg per dose for HexaPro-foldon and 0.9 μg per dose for all other immunogens. Serum collection was performed at weeks 2 and 5. The RBD-I53-50 immunogen was prepared in two different buffer conditions, with one group including CHAPS as an excipient to bridge to previous studies. (B) Binding titers against HexaPro-foldon at weeks 2 and 5, as assessed by AUC from ELISA measurements of serial dilutions of serum. Each circle represents the AUC measurement from an individual mouse and horizontal lines show the geometric mean of each group. One mouse with a near-zero AUC at week 2 for group four was not plotted but still included in the geometric mean calculation. Midpoint titers are shown in Supplementary Figure 5A . (C) Autologous (D614G) pseudovirus neutralization using a lentivirus backbone. Each circle represents the neutralizing antibody titer at 50% inhibition (IC50) for an individual mouse and horizontal lines show the geometric mean of each group. Pseudovirus neutralization titers using an MLV backbone are shown in Supplementary Figure 5B . Statistical analysis was performed using one-sided nonparametric Kruskal–Wallis test with Dunn’s multiple comparisons. *p < 0.05; **p < 0.01; ***p < 0.001.
Shelf-life stability of RBD-based nanoparticle immunogens is improved by Rpk mutations. (A) Summary of DLS measurements over four weeks. Hydrodynamic diameter remained consistent for all nanoparticles except wild-type RBD-I53-50 at 35-40°C, which showed signs of aggregation after 28 days of storage. (B) Binding against immobilized hACE2-Fc receptor (dashed lines) and CR3022 mAb (solid lines) by BLI, normalized to -80°C sample for each time point. Antigenic integrity remained consistent for the stabilized nanoparticle immunogens, while the binding signal of wild-type RBD-I53-50 incubated at 35-40°C decreased by 60% (hACE2-Fc) and 30% (CR3022). (C) Summary of SDS-PAGE and nsEM over four weeks. No degradation was observed by SDS-PAGE. Partial aggregation was only observed by nsEM on day 28 for the wild-type nanoparticle stored at 35-40°C. Electron micrographs for day 28 after storage at 35-40°C are shown, with red boxes indicating instances of aggregates (scale bar, 200 nm). All samples were formulated in TBS, 5% glycerol, 100 mM L-arginine. All raw data provided in Supplementary Figure 4 .
Stabilization of the SARS-CoV-2 Spike Receptor-Binding Domain Using Deep Mutational Scanning and Structure-Based Design

June 2021

·

147 Reads

·

39 Citations

The unprecedented global demand for SARS-CoV-2 vaccines has demonstrated the need for highly effective vaccine candidates that are thermostable and amenable to large-scale manufacturing. Nanoparticle immunogens presenting the receptor-binding domain (RBD) of the SARS-CoV-2 Spike protein (S) in repetitive arrays are being advanced as second-generation vaccine candidates, as they feature robust manufacturing characteristics and have shown promising immunogenicity in preclinical models. Here, we used previously reported deep mutational scanning (DMS) data to guide the design of stabilized variants of the RBD. The selected mutations fill a cavity in the RBD that has been identified as a linoleic acid binding pocket. Screening of several designs led to the selection of two lead candidates that expressed at higher yields than the wild-type RBD. These stabilized RBDs possess enhanced thermal stability and resistance to aggregation, particularly when incorporated into an icosahedral nanoparticle immunogen that maintained its integrity and antigenicity for 28 days at 35-40°C, while corresponding immunogens displaying the wild-type RBD experienced aggregation and loss of antigenicity. The stabilized immunogens preserved the potent immunogenicity of the original nanoparticle immunogen, which is currently being evaluated in a Phase I/II clinical trial. Our findings may improve the scalability and stability of RBD-based coronavirus vaccines in any format and more generally highlight the utility of comprehensive DMS data in guiding vaccine design.


Figure 3 | Stabilized RBDs presented on assembled I53-50 nanoparticles enhance solution stability compared to the wild-type RBD. (A) Schematic of assembly of I53-50 nanoparticle immunogens displaying RBD antigens. (B) nsEM of RBD-I53-50, Rpk4-I53-50, and Rpk9-I53-50 (scale bar, 200 nm). (C-E) show summarized quality control results for RBD-I53-50, Rpk4-I53-50, and Rpk9-I53-50 before and after a single freeze/thaw cycle in four different buffers. Complete data available in Supplementary Information 3. (C) The ratio of absorbance at 320 to 280 nm in UV-Vis spectra, an indicator of the presence of soluble aggregates. (D) DLS measurements, which monitor both proper nanoparticle assembly and formation of aggregates. (E) Fractional reactivity of I53-50 nanoparticle immunogens against immobilized hACE2-Fc receptor (top) and CR3022 (bottom). The pre-freeze and post-freeze data were separately normalized to the respective CHAPS-containing samples for each nanoparticle.
Figure 4 Potent immunogenicity of the parental RBD-I53-50 nanoparticle immunogen is maintained with addition of Rpk mutations. (A) Female BALB/c mice (six per group) were immunized at weeks 0 and 3. Each group received equimolar amounts of RBD antigen adjuvanted with AddaVax, which in total antigen equates to 5 μg per dose for HexaPro-foldon and 0.9 μg per dose for all other immunogens. Serum collection was performed at weeks 2 and 5. The RBD-I53-50 immunogen was prepared in two different buffer conditions, with one group including CHAPS as an excipient to bridge to previous studies. (B) Binding titers against HexaPro-foldon at weeks 2 and 5, as assessed by AUC from ELISA measurements of serial dilutions of serum. Each circle represents the AUC measurement from an individual mouse and horizontal lines show the geometric mean of each group. One mouse with a near-zero AUC at week 2 for group four was not plotted but still included in the geometric mean calculation. Midpoint titers are shown in Supplementary Figure 5A. (C) Autologous (D614G) pseudovirus neutralization using a lentivirus backbone. Each circle represents the neutralizing antibody titer at 50% inhibition (IC 50 ) for an individual mouse and horizontal lines show the geometric mean of each group. Pseudovirus neutralization titers using an MLV backbone are shown in Supplementary Figure 5B. Statistical analysis was performed using one-sided nonparametric Kruskal-Wallis test with Dunn's multiple comparisons. *, p < 0.05; **, p < 0.01; ***, p < 0.001.
Figure 5 | Shelf-life stability of RBD-based nanoparticle immunogens is improved by Rpk mutations. (A) Summary of DLS measurements over four weeks. Hydrodynamic diameter remained consistent for all nanoparticles except wild-type RBD-I53-50 at 35-40°C, which showed signs of aggregation after 28 days of storage. (B) Binding against immobilized hACE2-Fc receptor (dashed lines) and CR3022 mAb (solid lines) by BLI, normalized to -80°C sample for each time point. Antigenic integrity remained consistent for the stabilized nanoparticle immunogens, while the binding signal of wild-type RBD-I53-50 incubated at 35-40°C decreased by 60% (hACE2-Fc) and 30% (CR3022). (C) Summary of SDS-PAGE and nsEM over four weeks. No degradation was observed by SDS-PAGE. Partial aggregation was only observed by nsEM on day 28 for the wild-type nanoparticle stored at 35-40°C. Electron micrographs for day 28 after storage at 35-40°C are shown, with red boxes indicating instances of aggregates (scale bar, 200 nm). All samples were formulated in TBS, 5% glycerol, 100 mM L-arginine. All raw data provided in Supplementary Figure 4.
Stabilization of the SARS-CoV-2 Spike receptor-binding domain using deep mutational scanning and structure-based design

May 2021

·

150 Reads

The unprecedented global demand for SARS-CoV-2 vaccines has demonstrated the need for highly effective vaccine candidates that are thermostable and amenable to large-scale manufacturing. Nanoparticle immunogens presenting the receptor-binding domain (RBD) of the SARS-CoV-2 Spike protein (S) in repetitive arrays are being advanced as second-generation vaccine candidates, as they feature robust manufacturing characteristics and have shown promising immunogenicity in preclinical models. Here, we used previously reported deep mutational scanning (DMS) data to guide the design of stabilized variants of the RBD. The selected mutations fill a cavity in the RBD that has been identified as a linoleic acid binding pocket. Screening of several designs led to the selection of two lead candidates that expressed at higher yields than the wild-type RBD. These stabilized RBDs possess enhanced thermal stability and resistance to aggregation, particularly when incorporated into an icosahedral nanoparticle immunogen that maintained its integrity and antigenicity for 28 days at 35-40°C, while corresponding immunogens displaying the wild-type RBD experienced aggregation and loss of antigenicity. The stabilized immunogens preserved the potent immunogenicity of the original nanoparticle immunogen, which is currently being evaluated in a Phase I/II clinical trial. Our findings may improve the scalability and stability of RBD-based coronavirus vaccines in any format and more generally highlight the utility of comprehensive DMS data in guiding vaccine design.


Aldehyde Oxidase Contributes to All- Trans -Retinoic Acid Biosynthesis in Human Liver

December 2020

·

26 Reads

·

20 Citations

Drug Metabolism and Disposition: the Biological Fate of Chemicals

All-trans-retinoic acid (atRA) is a critical endogenous signaling molecule. atRA is predominantly synthesized from retinaldehyde by aldehyde dehydrogenase 1A1 (ALDH1A1), but aldehyde oxidase (AOX) may also contribute to atRA biosynthesis. The goal of this study was to test the hypothesis that AOX contributes significantly to atRA formation in human liver. Human recombinant AOX formed atRA from retinaldehyde (Km ∼1.5 ± 0.4 µM; kcat ∼3.6 ± 2.0 minute-1). In human liver S9 fractions (HLS9), atRA formation was observed in the absence of NAD+, suggesting AOX contribution to atRA formation. In the presence of NAD+, Eadie-Hofstee plots of atRA formation in HLS9 indicated that two enzymes contributed to atRA formation. The two enzymes were identified as AOX and ALDH1A1 based on inhibition of atRA formation by AOX inhibitor hydralazine (20%-50% inhibition) and ALDH1A1 inhibitor WIN18,446 (50%-80%inhibition). The expression of AOX in HLS9 was 9.4-24 pmol mg-1 S9 protein, whereas ALDH1A1 expression was 156-285 pmol mg-1 S9 protein measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS) quantification of signature peptides. The formation velocity of atRA in the presence of NAD+ correlated significantly with the expression of ALDH1A1 and AOX protein. Taken together, the data show that both AOX and ALDH1A1 contribute to atRA biosynthesis in the human liver, with ALDH1A1 being the high-affinity, low-capacity enzyme and AOX being the low-affinity, high-capacity enzyme. The results suggest that in the case of ALDH1A dysfunction or excess vitamin A, AOX may play an important role in regulating hepatic vitamin A homeostasis and that inhibition of AOX may alter atRA biosynthesis and signaling. SIGNIFICANCE STATEMENT: This study provides direct evidence to show that human AOX converts retinaldehyde to atRA and contributes to hepatic atRA biosynthesis. The finding that AOX may be responsible for 20%-50% of overall hepatic atRA formation suggests that alterations in AOX activity via drug-drug interactions, genetic polymorphisms, or disease states may impact hepatic atRA concentrations and signaling and alter vitamin A homeostasis.

Citations (5)


... The prospective applications of computerdesigned self-assembling protein nanoparticles are extensive, spanning fields such as biomedicine, drug delivery, and nanomaterials [94,95]. For example, they can be utilized as drug carriers to facilitate the precise delivery of drugs to target tissues or cells, thereby enhancing therapeutic efficacy while reducing the incidence of adverse effects [96]. Additionally, these nanoparticles can also be employed in the construction of biosensors, tissue engineering scaffolds, and novel nanoelectronic devices [97]. ...

Reference:

Nanocarriers for intracellular delivery of proteins in biomedical applications: strategies and recent advances
Macromolecular Cargo Encapsulation via In Vitro Assembly of Two‐Component Protein Nanoparticles

... [54] Despite the numerous and diverse applications now in reach, several challenges remain in the field. While structure prediction algorithms can be used as a validation step prior to experimental testing (Figure 5a and 5b), typical experimental success rates for the design of polyhedral protein assemblies, where success is defined as the production of proteins that selfassemble into the designed architecture, lay around 10 %, [136] with even lower success rates observed for fully AI-based approaches. [67,68] One way to improve individual pipelines is to combine different methods and subsequent design rounds, which can diversify and optimise generated backbones and sequences towards user defined parameters (e. g. Figure 5c). ...

Increasing Computational Protein Design Literacy through Cohort-Based Learning for Undergraduate Students
  • Citing Article
  • August 2022

Journal of Chemical Education

... Moreover, certain small molecules like formaldehyde [25], linoleic acid [26][27][28], and an ultrapotent synthetic nanobody Nb6 [29] can stabilize the SARS-CoV-2 S trimer in its down conformation by creating specific cross-links between RBD protomers and S2 subunit residues. Developing methods to trap the SARS-CoV-2 S trimer in its down state could assist in treating . ...

Stabilization of the SARS-CoV-2 Spike Receptor-Binding Domain Using Deep Mutational Scanning and Structure-Based Design

... Sundry studies have shown that the microbiome is essential for the establishment of the mucosa immune system, therefore, inappropiate manipulations in the mouth can cause microbiological desequillibriums, making the baby more susceptible to infections. Seeing the dearth of scientific content about the influence of the cleaning mouth of teethless babies on the oral microbiome, this mini-review aims to describe the main findings about this influence, enabling an evidence-based in pediatric dentistry (8)(9)(10) . ...

Is the oral microbiome a source to enhance mucosal immunity against infectious diseases?

npj Vaccines

... In vitro studies by Ozaki et al. demonstrated that fetal bovine serum inhibited keratinization of mouse epithelial cells, but the addition of a retinoic acid receptor inhibitor reversed this effect [34]. Additionally, Miyazono et al. con rmed that ATRA inhibits keratinization of epithelial cells in mice, pigs, and humans [35].In the vitamin A metabolic pathway, retinol is oxidized by alcohol dehydrogenase to retinal, which is further oxidized by aldehyde dehydrogenase and aldehyde oxidase to retinoic acid [36,37], and then metabolized by cytochrome P450 enzymes (particularly CYP26) [38,39]. In our study, genes related to the retinoic acid metabolic pathway were signi cantly enriched in alveolar mucosa (ALV). ...

Aldehyde Oxidase Contributes to All- Trans -Retinoic Acid Biosynthesis in Human Liver
  • Citing Article
  • December 2020

Drug Metabolism and Disposition: the Biological Fate of Chemicals