Styxl2 regulates sarcomere integrity during zebrafish muscle development.

(A) A schematic of mouse Styxl2 protein. DSPc (red): dual-specificity phosphatase, catalytic domain. The numbers denote the positions of various amino acids in mouse Styxl2 including Ser (S)-225. (B) The expression of Styxl2 protein in different mouse tissues. 100 μg of soluble mouse tissue lysates were subjected to Western blot analysis. GAPDH: glyceraldehyde-3-phosphate dehydrogenase. SM: skeletal muscles. (C) The expression of Styxl2 protein in C2C12 myoblasts before and after differentiation in culture. MHC: myosin heavy chain. (D) Zebrafish zygotes were either mock-injected or injected with a Styxl2-specific morpholino (MO). Sarcomeres in muscles at 48 hpf were revealed by transmission electron microscopy (TEM). Representative images were shown. Scale bar: 500 nm. (E) Sarcomeres in fast and slow muscles of zebrafish embryos at 24 hpf were revealed by immunostaining using specific antibodies as indicated. Ctrl: control (non-treated); MO: Styxl2-morpholino-treated. The nuclei of slow and fast muscle fibers were counter-stained with an anti-Prox1 antibody and DAPI, respectively. Scale bar: 10 μm.

Candidate genes regulated by both Jak1 and Stat1.

Selected candidate genes targeted by both si-Jak1 and si-Stat1 were shown. The fold change was determined by the relative levels of a gene in cells treated with si-Jak1 or si-Stat1 over that in control cells. Genes with fold change > 1.5 for si-Jak1 and > 2 for si-Stat1 were listed in this table. Genes with positive fold change are up-regulated (positive value), while those with negative fold change are down-regulated. P Stage and D Stage denote the proliferation stage (GM 24h) and early differentiation stage (DM 12h), respectively.

Conditional deletion of mouse Styxl2 leads to defective sarcomeres in striated muscles.

(A) The expression of Styxl2 protein in skeletal muscles of wild-type (+/+), heterozygous (+/−) and Styxl2 knockout (KO) (−/−) (driven by EIIA-Cre) mice. Soluble tissue lysates from limbs of E14.5 mouse embryos were subjected to Western blot analysis. N.S.: non-specific. (B) E14.5 embryos of control (Ctrl) and EIIA-Cre; Styxl2f/f KO mice. *: edema was found in some Styxl2 KO foetuses. (C) The sarcomere structures of cardiomyocytes in the heart of E14.5 embryos from control and EIIA-Cre; Styxl2f/f KO mice were revealed by TEM. Scale bar: 1 μm. (D) The expression of Styxl2 protein in skeletal muscles of control (Ctrl) and Styxl2 CKO P1 mice. (E) The new-born pups (P1) from control (Ctrl) and Pax7Cre/+; Styxl2f/f KO mice (CKO). (F) Lung sections of control and Styxl2 CKO P1 mice. Scale bar: 50 μm. (G) The sarcomere structure of diaphragm muscles from control (Ctrl) and Styxl2 CKO P1 mice revealed by TEM. Scale bar: 1 μm.

Styxl2 is not required for sarcomere maintenance but involved in de novo sarcomere assembly in adult muscles.

(A) The schematic to assess the role of Styxl2 in sarcomere maintenance in adult muscles. TMX: tamoxifen. (B) Styxl2 protein levels in Ctrl and MF-iKO mouse gastrocnemius muscles used for force test. (C, D) The twitch and tetanic force generated by gastrocnemius muscles of three Ctrl and four MF-iKO mice were measured. In (A-D), Ctrl: Styxl2f/f mice; MF-iKO: Tg: HSA-MerCreMer; Styxl2f/f mice. (E) The schematic to assess the role of Styxl2 in de novo sarcomere assembly in adult muscles. CTX: cardiotoxin. (F) Styxl2 protein levels in regenerated gastrocnemius muscles used for force test. N.S.: non-specific. (G, H) Twitch and tetanic force by regenerated gastrocnemius muscles were measured from four pairs of control and SC-iKO mice. (I) TEM images of the muscle used for force generation test. Scale bar: 500 nm. (J) Quantification of sarcomere length in (I). TEM images from three pairs of mice were examined and the length of 3-5 sarcomeres from each mouse was measured. In (E-J), Ctrl: Pax7CreERT2/CreERT2 mice; SC-iKO: Pax7CreERT2/CreERT2; Styxl2f/f mice. n.s.: not significant. *, p-value <0.05.

Styxl2 binds to non-muscle myosin IIs.

(A) The schematic of the fusion protein Styxl2-BirA*-HA. (B) The work flow of sample preparation for BioID. (C) Enriched (with Enrichment score >1) biotinylated proteins in cells expressing Styxl2-BirA*-HA relative to that in control cells were identified by mass spectrometry followed by Gene Ontology (GO) analysis. Myofibril-related terms were shown. (D) C2C12 cells were harvested after 2 days in DM and the soluble whole cell lysates were subjected to immunoprecipitation (IP) using either an anti-MEF2A (negative control) or an anti-Styxl2 antibody. The endogenous Myh9 or Myh10 was detected by Western blot. (E) The schematic showing the domains of non-muscle myosin IIs (NM II). (F) HEK 293T cells were transfected with Flag-Styxl2 (black arrow) together with HA-NM II-head (red arrowheads) or HA-NM II-tail (black arrowheads). The soluble whole cell lysates were subjected to IP with an anti-Flag antibody and the co-immunoprecipitated proteins were detected by Western blot.

Styxl2 protein levels inversely correlates with that of non-muscle myosin IIs.

(A) Protein levels of Styxl2 and non-muscle myosin IIs in skeletal muscles of wild-type mice of different ages were determined by Western blot. (B) Protein levels of Styxl2 and non-muscle myosin IIs in skeletal muscles of control and Styxl2 CKO P1 mice were determined by Western blot. (C) Relative ratios of non-muscle myosin II protein levels in control and Styxl2 CKO P1 mice were quantified by ImageJ based on the data from four (for Myh9) or five (for Myh10) sets of Western blots. (D) Protein levels of Styxl2 and non-muscle myosin IIs in regenerating tibialis anterior muscles were determined by Western blot. The experiments were done for four times with similar results and a representative gel was shown. Ctrl: Pax7CreERT2/CreERT2 mice; SC-iKO: Pax7CreERT2/CreERT2; Styxl2f/f mice; dpi: days post-injury. (E) Zebrafish zygotes were separately injected with the following morpholinos (MO): control (Ctrl-MO), Styxl2-MO, and mixed morpholinos targeting both Styxl2 and Myh10 (dMO). At 48 hpf, zebrafishes were poked by a fine needle and their responses were recorded. Quantification of responses of zebrafish to needle touch was shown. In each group, at least 8 zebrafishes were tested and the percentage of zebrafishes responding to needle touch was calculated. Each dot represents one group of experiment. (F) Zebrafishes from each condition in (E) were subjected to immunostaining for α-actinin to visualize sarcomere structures. Representative images were shown. Scale bar: 10 μm. *, p-value < 0.05. ****, p-value < 0.0001.

The DSPc domain of Styxl2 is dispensable for degradation of non-muscle myosin IIs.

(A) DNA constructs encoding Myh9, Styxl2 or Mst1 (negative control) were co-expressed in C2C12 cells. Soluble whole cell extracts were subjected to Western blot analysis. OE: overexpression. (B) HEK 293T cells were transfected with different plasmids as indicated. 24 h after transfection, cells were harvested and soluble whole cell extracts were subjected to Western blot analysis. The arrow indicates the head domain of Myh9, and the arrowhead indicates Styxl2 missing the N-terminal 509 aa (Styxl2ΔN509). Mst1: negative control. N.S.: non-specific. (C) Zebrafish zygotes were injected with various morpholinos as indicated with or without the co-injected Styxl2 mRNAs. zStyxl2FL: the full-length fish Styxl2 mRNA; zStyxl2ΔN493: the truncated fish Styxl2 mRNA missing the 5’ region encoding the N-terminal 493 aa. Representative α-actinin immunofluorescent images were shown. The number in numerator at the bottom left corner of each image represents fish embryos showing normal α-actinin staining pattern, while that in denominator represents the total number of embryos examined. Scale bar: 10 μm.

The Styxl2-mediated degradation of non-muscle myosin IIs is autophagy dependent.

(A) Plasmids encoding Myh9, Styxl2 or Mst1 (negative control) were co-expressed in C2C12 cells together with various siRNAs as indicated. Soluble whole cell extracts were subjected to Western blot analysis. OE, overexpression. N.S.: non-specific. (B) HEK 293T cells were transfected with various constructs as indicated. Various inhibitors of the autophagy-lysosome pathway (50 μM LY294002, 100 μM Chloroquine, 100 nM BafA1, 20 mM NH4Cl) were added to the cell culture 6 h before harvest. OE, overexpression. (C) HEK 293T cells were co-transfected with the plasmids as indicated. 24 h later, the soluble whole cell lysates were subjected to IP with an anti-HA antibody and the immunoprecipitated proteins were further analysed by Western blot. (D) The schematic showing that Styxl2 promotes sarcomere assembly by binding to and targeting non-muscle myosin IIs for degradation, which facilitates their eventual replacement by muscle myosin II during sarcomere maturation.

Styxl2 was downstream target of Jak1-Stat1 pathway.

(A) Multiple sequence alignment of the region surrounding the active motif of the DSPc domain among mouse dual-specificity phosphatases and Styxl2. The conserved Cys in functional phosphatases was highlighted in yellow. (B, C) C2C12 cells were transfected with various siRNAs as indicated. After growing in GM for 24 hours (h), cells were harvested either right away (GM 24h) or after growing in DM for an additional 12 h (DM 12h). The total RNA and soluble whole cell lysates were subjected to RT-qPCR (B) or Western blot analysis (C), respectively. (D, E) C2C12 cells were treated with 10 ng/ml of LIF or OSM for various times. Cell extracts were subjected to Western blot analysis. (F) Mouse embryos at E9.5 and E10.5 were subjected to in situ hybridization using either sense (control) or antisense probe specific for mouse Styxl2. (G) C2C12 cells were transfected with constructs expressing Flag- or HA-tagged Styxl2 for 24 h. Cells were then fixed and subjected to immunostaining for Flag or HA. (H) Zebrafish zygotes were co-injected with a plasmid encoding GFP fused with the sequence targeted by Styxl2-MO at the start codon together with or without Styxl2-MO. The phase-contrast and fluorescent images were taken and the representative images were shown.

Both germline and conditional deletion of Styxl2 caused lethality.

(A) The schematic of the genomic locus of mouse Styxl2 and the targeting strategy. The probes used for Southern blot are indicated by black bars. (B, C) The schematic of the mating strategy to generate Styxl2 germ-line KO mice was shown (B). The number of live pups (B) and that of embryos (C) of different genotypes at different embryonic stages were counted and presented. (D) The schematic of the mating strategy to generate conditional Styxl2 KO mice and the number of live pups (in brackets) older than 1 day were shown. (E) Hindlimb muscle sections from control (Ctrl) and Styxl2 CKO P1 mice were subjected to electron microscopy analysis. Scale bar, 1 μm.

Identification of Styxl2-interacting partners by BioID.

(A) A representative gel of samples from BioID before mass spectrometry analysis. Whole cell lysates from control (Ctrl) and Doxycycline-treated (Dox) cells were subjected to Western blot using Streptavidin-HRP (top panel) or an anti-Styxl2 antibody (middle panel). (B) The biotinylated proteins identified by mass spectrometry were plotted with the log2Enrichment scores on the X-axis and the minus log10(p.value) on the Y-axis. The Enrichment score denotes the ratio of the peak intensity of reporter ions. Red dots indicate proteins with the Enrichment score > 1.2 and p value < 0.05. Blue dots indicate proteins with the Enrichment score < 0.83 and p value < 0.05. (C) Selected candidate proteins with more than 10 recovered peptides were shown.

Confluent C2C12 cells were induced to differentiate in DM and harvested at different time points. Soluble whole cell lysates were subjected to Western blot analysis.