Shiying Huang’s research while affiliated with Cornell 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 (6)


A pool of PLEKHA5 translocates to the PM during mitosis by binding to PI(4,5)P2
(a) Live-cell confocal microscopy of HeLa cells transiently expressing GFP-tagged PLEKHA5 constructs in interphase and mitosis. Imaging experiment was repeated three times independently with similar results. (b) Representative series of still images from a time-lapse movie in asynchronous, stable H2B-mCherry-expressing (magenta) HeLa cells transfected with PLEKHA5WW-H-BP-PH-GFP (green). Numbers indicate time before/after nuclear envelope breakdown (defined as t = 0 min). Imaging experiment was repeated three times independently with similar results. (c) Schematic representation of proximity biotinylation strategy using Lyn10-TurboID-V5 to biotinylate PM-proximal proteins. (d) Lyn10-TurboID showed expected PM-specific localization and biotinylation activity as demonstrated by staining for V5 (green) and biotin (streptavidin-Alexa Fluor 568, magenta). Imaging experiment was repeated three times independently with similar results. (e) Western blot analysis confirmed that Lyn10-TurboID biotinylates a protein marker of the PM but not markers of other subcellular localizations. The experiment was repeated two times independently with similar results. (f) Clustal Omega alignment of the primary amino acid sequences of the PH domain of PLEKHA4 and PLEKHA5. Highlighted in red are two arginine residues previously established to be required for binding PI(4,5)P2 in PLEKHA4. (g) Confocal microscopy of HeLa cells transfected with the R190A and R244 A mutants of PLEKHA5WW-H-BP-PH-GFP and PLEKHA5H-BP-PH-GFP. Imaging experiment was repeated two times independently with similar results. Unprocessed blots are available in source data.
Source data
Design and engineering of MARS and MARS-nGFP
(a) Clustal Omega alignment of primary amino acid sequences around the ‘4A’ region of PLEKHA4 and PLEKHA5. (b) Confocal microscopy of the K157A and R158A mutants of PLEKHA5H-BP-PH-GFP. Imaging experiment was repeated two times independently with similar results. (c, d) Representative series of still images from time-lapse movies in asynchronous HeLa cells stably expressing H2B-tagBFP and transfected with either MARS (c) or 2xMARS-nGFP (d). MARS constructs are colored magenta and H2B-tagBFP is colored green. Numbers indicate time before/after nuclear envelope breakdown (defined as t = 0 min). Each imaging experiment was repeated two times independently with similar results.
Characterization of stoichiometry of 2xMARS-nGFP and effects of MARS systems on cell proliferation rates
(a) Four stable cell lines were generated via lentiviral transduction that express similar levels of 2xMARS-nGFP and varying levels of GFP, which was under control of either a PGK or CMV promoter (the three CMV-GFP-expressing lines were isolated by FACS, sorting for different levels of GFP expression). Shown are representative Western blots showing GFP (asterisk denotes a longer CMV-GFP product potentially arising from an upstream alternative start codon) and 2xMARS-nGFP (mCherry blot) expression levels in these cell lines alongside blots of purified GFP and mCherry protein standards. (b) Quantification of GFP to 2xMARS-nGFP molar ratios in HeLa cells stably expressing 2xMARS-nGFP and various levels of GFP from (A) (n = 3 biological replicates, one-way ANOVA with Tukey post hoc multiple comparisons test). (c, d) Imaging analysis of GFP recruitment to the PM during mitosis at different stoichiometries of GFP to 2xMARS-nGFP determined by quantitative image analysis. (c) Quantification of the PM to cytosolic fluorescence ratios of GFP as a readout for measuring GFP recruitment levels by 2xMARS-nGFP. For PGK-GFP: n = 8 cells for low GFP expression, n = 4 cells for medium expression, and n = 7 cells for high expression. For CMV-GFP cell lines, n = 10 cells each for low, medium, and high GFP expression level. Cells analyzed came from two independent experiments (one-way ANOVA with Tukey post hoc multiple comparisons test). Note: CMV-GFP cells were sorted by FACS to generate three separate cell lines with low, medium, and high GFP expression, and images were acquired for each cell line. For PGK-GFP cells, the low, medium, and high cells were all from a single cell line and are categorized based on GFP fluorescence intensity during imaging analysis by ImageJ post-acquisition. (d) Representative micrographs for HeLa cells stably expressing 2xMARS-nGFP and varying levels of GFP analyzed in (c). Brightness of the GFP channel was adjusted post-acquisition for better visualization of the weaker fluorescence signals for PGK-GFP and CMV-GFP low cells. (e) Quantification of mean fluorescence intensity in the cytosol during interphase as a means to determine GFP expression levels in PGK-GFP low, medium, high cells and CMV-GFP low cells (n = 40 cells from two independent experiments). Compared to CMV-GFP low cells, PGK-GFP low cells exhibited an average of 7.6x lower expression, PGK-GFP medium cells exhibited ~2.2x lower expression, and PGK-GFP-high cells exhibited ~1.1x higher expression. Converting to GFP:2xMARS-nGFP molar ratio in these cells, the estimated ratios are 1.8 (low), 6.2 (medium), and 15.1 (high). (f, g) Ectopic expression of MARS and 2xMARS-nGFP did not negatively impact rates of cell division and proliferation. (f) Representative growth curves for wild-type (WT) HeLa cells and those stably expressing MARS and 2xMARS-nGFP (n = 2 technical replicates for each curve). Raw data (faded-color curves with error bar) acquired by IncuCyte Live-Cell Analysis System were fitted using GraphPad Prism (Exponential growth curve model). Fitted curves are shown as solid lines. (g) Doubling time of the three cell lines calculated from the fitted growth curves (n = 3 biological replicates, one-way ANOVA with Tukey post hoc multiple comparisons test). Source numerical data and unprocessed blots are available in Source data.
Source data
Mutational analysis reveals no contributions of phosphorylation of residues other than S161 to the cell cycle-dependent localization of PLEKHA5
Live-cell confocal microscopy of phosphodeficient (Ser/Thr to Ala and Tyr to Phe) and phosphomimetic (Ser/Thr to Asp and Tyr to Glu) mutants of GFP-tagged PLEKHA5 full-length protein (a) or WW-H-BP-PH fragment (b). Imaging experiment was repeated two times independently with similar results.
S161 phosphorylation status influences the PM localization of EGFP-PLEKHA5FL and PLEKHA5WW-H-BP-PH-EGFP
(a, b) Live-cell confocal microscopy of the subcellular localizations of the WT, S161A, and S161D variants of full-length PLEKHA5FL (a) and the WW-H-BP-PH motifs (b). (c, d) Quantification of PM localization for GFP-PLEKHA5FL (c) and PLEKHA5WW-H-BP-PH-GFP (d) using colocalization analysis. Pearson coefficients were computed from images acquired in HeLa cells transiently expressing either of the PLEKHA5 constructs with a transfectable PM marker, Lyn10-LOV-mCherry. For GFP-PLEKHA5FL in interphase: n = 37 cells for WT, n = 35 cells for S161A, and n = 30 cells for S161D; in mitosis: n = 12 cells for WT and S161D and n = 11 cells for S161A. For PLEKHA5WW-H-BP-PH-GFP in interphase, n = 26 cells for WT, n = 29 cells for S161A, and n = 25 cells for S161D; in mitosis: n = 13 cells for WT and S161D and n = 16 cells for S161A. Cells analyzed came from three independent experiments (one-way ANOVA with Tukey post hoc multiple comparisons test). (e, f) Quantification of PM association for transfected GFP-PLEKHA5FL (e) and PLEKHA5WW-H-BP-PH-GFP (f) using PM-targeted proximity biotinylation. Representative western blots are shown at top with quantification of GFP signals in the streptavidin pulldowns at bottom (n = 4 biological replicates, one-way ANOVA with Tukey post hoc multiple comparisons test). Source numerical data and unprocessed blots are available in Source data.
Source data

+9

A phosphorylation-controlled switch confers cell cycle-dependent protein relocalization
  • Article
  • Publisher preview available

August 2024

·

101 Reads

·

2 Citations

Nature Cell Biology

·

Shiying Huang

·

Mateusz M. Wagner

·

[...]

·

Tools for acute manipulation of protein localization enable elucidation of spatiotemporally defined functions, but their reliance on exogenous triggers can interfere with cell physiology. This limitation is particularly apparent for studying mitosis, whose highly choreographed events are sensitive to perturbations. Here we exploit the serendipitous discovery of a phosphorylation-controlled, cell cycle-dependent localization change of the adaptor protein PLEKHA5 to develop a system for mitosis-specific protein recruitment to the plasma membrane that requires no exogenous stimulus. Mitosis-enabled anchor-away/recruiter system comprises an engineered, 15 kDa module derived from PLEKHA5 capable of recruiting functional protein cargoes to the plasma membrane during mitosis, either through direct fusion or via GFP–GFP nanobody interaction. Applications of the mitosis-enabled anchor-away/recruiter system include both knock sideways to rapidly extract proteins from their native localizations during mitosis and conditional recruitment of lipid-metabolizing enzymes for mitosis-selective editing of plasma membrane lipid content, without the need for exogenous triggers or perturbative synchronization methods.

View access options

Figure 2. Design and engineering of Mitosis-enabled Anchor-away/Recruiter System (MARS), based on the H-BP-PH motifs of PLEKHA5. (A) Domain map of the PLEKHA5 H-BP-PH motifs
Figure 4. S161 phosphorylation and PLEKHA5 PM localization are modulated by protein kinase C (PKC) activity. (A) Top 10 kinase candidates for phosphorylating S161. PKC isoforms are bolded. (B) S161 phosphorylation increased upon stimulation of PKC activity using 100 nM PMA or 200 nM Bryostatin 1, and the increase was attenuated with pre-treatment of PKC inhibitors (PKCi, 100 nM Gö 6983 and 100 nM Sotrastaurin/AEB071). Representative western blots (left) and quantification (right) from Phos-tag and regular SDS-PAGE analysis probing PLEKHA5 H-BP-PH -GFP are shown (n=3 biological replicates, one-way ANOVA with Sidak post hoc multiple comparisons test). (C-D) PM localization of PLEKHA5 H-BP-PH was decreased upon activation of PKC by 100 nM PMA (C) or 200 nM Bryostatin 1 (D). Representative confocal micrographs before and after agonist treatment for each condition are shown at left, with quantification of the ratios of PM to cytosolic fluorescence before and after treatment are shown at right (n=12 cells from three independent experiments, one-way ANOVA with Sidak post hoc multiple comparisons test).
Figure 5. MARS recruits polo-like kinase 1 (PLK1) and phospholipase D (PLD) to the PM specifically during mitosis. (A) Cartoon illustration of the working principle of MARS for either
FIGURES
A phosphorylation-controlled switch confers cell cycle-dependent protein relocalization

June 2024

·

81 Reads

Tools for acute manipulation of protein localization enable elucidation of spatiotemporally defined functions, but their reliance on exogenous triggers can interfere with cell physiology. This limitation is particularly apparent for studying mitosis, whose highly choreographed events are sensitive to perturbations. Here we exploit the serendipitous discovery of a phosphorylation-controlled, cell cycle-dependent localization change of the adaptor protein PLEKHA5 to develop a system for mitosis-specific protein recruitment to the plasma membrane that requires no exogenous stimulus. Mitosis-enabled Anchor-away/Recruiter System (MARS) comprises an engineered, 15-kDa module derived from PLEKHA5 capable of recruiting functional protein cargoes to the plasma membrane during mitosis, either through direct fusion or via GFP-GFP nanobody interaction. Applications of MARS include both knock sideways to rapidly extract proteins from their native localizations during mitosis and conditional recruitment of lipid-metabolizing enzymes for mitosis-selective editing of plasma membrane lipid content, without the need for exogenous triggers or perturbative synchronization methods.


Mapping the global interactome of the ARF family reveals spatial organization in cellular signaling pathways

April 2024

·

68 Reads

·

4 Citations

Journal of Cell Science

The ADP-ribosylation factors (ARFs) and ARF-like (ARLs) GTPases serve as essential molecular switches governing a wide array of cellular processes. In this study, we utilized proximity-dependent biotin identification (BioID) to comprehensively map the interactome of 28 out of 29 ARF and ARL proteins in two cellular models. Through this approach, we identified ∼3000 high-confidence proximal interactors, enabling us to assign subcellular localizations to the family members. Notably, we uncovered previously undefined localizations for ARL4D and ARL10. Clustering analyses further exposed the distinctiveness of the interactors identified with these two GTPases. We also reveal that the expression of the understudied member ARL14 is confined to the stomach and intestines. We identified phospholipase D1 (PLD1) and the ESCPE-1 complex, more precisely SNX1, as proximity interactors. Functional assays demonstrated that ARL14 can activate PLD1 in cellulo and is involved in cargo trafficking via the ESCPE-1 complex. Overall, the BioID data generated in this study provide a valuable resource for dissecting the complexities of ARF and ARL spatial organization and signaling.


ARF-family global interactome mapping uncovers spatial organization of cellular signaling pathways

March 2023

·

157 Reads

·

1 Citation

The ADP-ribosylation factors (ARFs) and ARF-like (ARLs) GTPases serve as essential molecular switches governing a wide array of cellular processes. In this study, we utilized proximity-dependent biotin identification (BioID) to comprehensively map the interactome of 28 out of 29 ARF and ARL proteins in two cellular models. Through this approach, we identified ∼3000 high-confidence proximal interactors, enabling us to assign subcellular localizations to the family members. Notably, we uncovered previously undefined localizations for ARL4D and ARL10. Clustering analyses further exposed the distinctiveness of the interactors identified with these two GTPases. We also reveal that the expression of the understudied member ARL14 is confined to the stomach and intestines. We identified phospholipase D1 (PLD1) and the ESCPE-1 complex, more precisely SNX1, as proximity interactors. Functional assays demonstrated that ARL14 can activate PLD1 in cellulo and is involved in cargo trafficking via the ESCPE-1 complex. Overall, the BioID data generated in this study provide a valuable resource for dissecting the complexities of ARF and ARL spatial organization and signaling. SUMMARY STATEMENT Generation of the ARF family interactome allowed the attribution of potential localizations and functions to previously understudied members. We found that ARL14 activates PLD1 and contributes to ESCPE-1-mediated trafficking.


Adding a Chemical Biology Twist to CRISPR Screening

October 2022

·

49 Reads

·

1 Citation

Israel Journal of Chemistry

In less than a decade, CRISPR screening has revolutionized forward genetics and cell and molecular biology. Advances in screening technologies, including sgRNA libraries, Cas9‐expressing cell lines, and streamlined sequencing pipelines, have democratized pooled CRISPR screens at genome‐wide scale. Initially, many such screens were survival‐based, identifying essential genes in physiological or perturbed processes. With the application of new chemical biology tools to CRISPR screening, the phenotypic space is no longer limited to live/dead selection or screening for levels of conventional fluorescent protein reporters. Further, the resolution has been increased from cell populations to single cells or even the subcellular level. We highlight advances in pooled CRISPR screening, powered by chemical biology, that have expanded phenotypic space, resolution, scope, and scalability as well as strengthened the CRISPR/Cas enzyme toolkit to enable biological hypothesis generation and discovery.


Click chemistry–enabled CRISPR screening reveals GSK3 as a regulator of PLD signaling

November 2021

·

25 Reads

·

15 Citations

Proceedings of the National Academy of Sciences

Significance The application of CRISPR-Cas9 to genome-wide screening has revolutionized our ability to discover new functions for genes and regulators of physiological processes. However, a major constraint placed on such screens is the choice of phenotypic selection step, typically limited to live/dead or conventional fluorescent reporter/labeling. Here, we develop a CRISPR interference–based screening platform harnessing click chemistry tagging to reveal regulators of a specific signaling pathway. Using this tool, we discovered a regulatory circuit involving glycogen synthase kinase 3 and protein kinase C in the control of phospholipase D signaling. More broadly, this work illustrates how emergent chemical biology approaches can expand the power of genome-wide CRISPR screening to elucidate mechanisms regulating specific enzyme-driven signaling pathways.

Citations (3)


... Recent proximity labeling assays across the ARF/ARL family further validated CNNM2-4 as ARL15 interacting partners in HEK and HeLa cell lines, highlighting the specificity of this interaction. Notably, no other ARF/ARL proteins labeled the endogenous CNNMs in this investigation, suggesting the exclusive role of ARL15 in regulating these proteins [222]. The crystal structure (8F6D) reveals that PRL and ARL15 binding sites on the CNNM proteins are adjacent and partially overlap ( Figure 2), suggesting that these proteins compete for binding. ...

Reference:

The PACT Network: PRL, ARL, CNNM, and TRPM Proteins in Magnesium Transport and Disease
Mapping the global interactome of the ARF family reveals spatial organization in cellular signaling pathways
  • Citing Article
  • April 2024

Journal of Cell Science

... Collectively, this poses a problem for extrapolating potentially co-acting GEF-GAP-ARF-Effector modules from classical biochemical data. Indeed, two recent proteomic studies examining the repertoire of what GTP-loaded ARFs can bind to indicate a highly, but not completely, overlapping effector set [43,44]. If, stated crudely, most things can bind most things in this ARF-centred network, how is specificity in ARF GTPase signalling generated? ...

ARF-family global interactome mapping uncovers spatial organization of cellular signaling pathways

... In contrast, the control experiment with H372BocK (protein with BCN in 372 is replaced by a Boc group) did not show any signals in the Cy-5 channel, supporting labeling specificity of this novel method. In short, this photo-tetrazole-BCN click reaction is faster than many other bioorthogonal reactions with BCN, allowing for live cell labeling of proteins within 15 s.Besides the methods discussed in the section, there are many other applications of direct conjugation using click chemistry and new methods are being added to the list5,8,49,[111][112][113][114][115] . For instance, there is a novel cysteine-specific modification of proteins or bacteriophage using isonitrile-chloroxime ligation chemistry with a rate constant of 306 L/mol·s116 . ...

Click chemistry–enabled CRISPR screening reveals GSK3 as a regulator of PLD signaling
  • Citing Article
  • November 2021

Proceedings of the National Academy of Sciences