Sunday, September 11, 2022
HomeBiologyStress-dependent cell stiffening by tardigrade tolerance proteins that reversibly type a filamentous...

Stress-dependent cell stiffening by tardigrade tolerance proteins that reversibly type a filamentous community and gel


Summary

Tardigrades are in a position to tolerate nearly full dehydration by getting into a reversible ametabolic state referred to as anhydrobiosis and resume their animation upon rehydration. Dehydrated tardigrades are exceptionally steady and face up to numerous bodily extremes. Though trehalose and late embryogenesis considerable (LEA) proteins have been extensively studied as potent protectants in opposition to dehydration in different anhydrobiotic organisms, tardigrades produce excessive quantities of tardigrade-unique protecting proteins. Cytoplasmic-abundant heat-soluble (CAHS) proteins are uniquely invented within the lineage of eutardigrades, a serious class of the phylum Tardigrada and are important for his or her anhydrobiotic survival. Nonetheless, the exact mechanisms of their motion on this protecting position will not be absolutely understood. Within the current research, we first postulated the presence of tolerance proteins that type protecting condensates through section separation in a stress-dependent method and looked for tardigrade proteins that reversibly type condensates upon dehydration-like stress. Via a complete search utilizing a desolvating agent, trifluoroethanol (TFE), we recognized 336 proteins, collectively dubbed “TFE-Dependent ReversiblY condensing Proteins (T-DRYPs).” Unexpectedly, we rediscovered CAHS proteins as extremely enriched in T-DRYPs, 3 of which had been main elements of T-DRYPs. We revealed that these CAHS proteins reversibly polymerize into many cytoskeleton-like filaments relying on hyperosmotic stress in cultured cells and bear reversible gel-transition in vitro. Moreover, CAHS proteins elevated cell stiffness in a hyperosmotic stress-dependent method and counteract the cell shrinkage attributable to osmotic stress, and even improved the survival in opposition to hyperosmotic stress. The conserved putative helical C-terminal area is critical and enough for filament formation by CAHS proteins, and mutations disrupting the secondary construction of this area impaired each the filament formation and the gel transition. On the premise of those outcomes, we suggest that CAHS proteins are novel cytoskeleton-like proteins that type filamentous networks and bear gel-transition in a stress-dependent method to offer on-demand bodily stabilization of cell integrity in opposition to deformative forces throughout dehydration and will contribute to the distinctive bodily stability in a dehydrated state.

Introduction

Water is a necessary molecule for sustaining the metabolic exercise and mobile integrity of dwelling organisms. Some organisms, nevertheless, can tolerate nearly full dehydration by getting into a reversible ametabolic state referred to as anhydrobiosis [1]. Tardigrades, also referred to as water bears, are a distinguished instance of such desiccation-tolerant animals [2]. Below a drying setting, tardigrades regularly lose nearly all physique water and concurrently contract their our bodies to a shrunken spherical type referred to as a tun. Dehydrated tardigrades are exceptionally steady and may face up to numerous bodily excessive environments together with publicity to house [3,4]. Even after publicity to excessive stressors, tardigrades can reanimate inside a number of dozen minutes after rehydration.

A number of tolerance molecules in opposition to dehydration stress have been recognized in numerous organisms. One of the vital analyzed molecules is the nonreducing disaccharide, trehalose. A big quantity of trehalose accumulates throughout desiccation in a number of anhydrobiotic animals, resembling sleeping chironomids [5], brine shrimp [6], and a few nematodes [7], a few of which require trehalose synthesis for anhydrobiotic survival [8]. Trehalose is proposed to play its protecting roles via 2 modes of motion: water alternative, during which trehalose substitutes for water molecules, and vitrification, during which trehalose preserves cell elements in an amorphous strong (glassy) state [9]. In tardigrades, nevertheless, no or solely slightly quantity of trehalose accumulates, even in dehydrated states of the anhydrobiotic species [10], and a current research instructed that trehalose synthesis genes in tardigrades are acquired in solely restricted lineages through horizontal switch after the institution of the anhydrobiotic capability in ancestral eutardigrades [11], suggesting the presence of a trehalose-independent anhydrobiosis mechanism in tardigrades.

Late embryogenesis considerable (LEA) proteins are one other instance of tolerance molecules. LEA proteins are principally unstructured proteins initially recognized in desiccating plant seeds and later present in a number of anhydrobiotic animals [12]. LEA proteins have many proposed roles, together with stabilization of vitrified trehalose, molecular shielding of shopper biomolecules, and sequestration of ions [12]. LEA proteins can suppress dehydration-dependent denaturation of enzymes and have robust synergistic protecting results with trehalose [13]. The LEA proteins of brine shrimp had been not too long ago reported to bear section separation to type droplet condensates upon dehydration and to extend the desiccation tolerance of insect cells [14].

Via a seek for LEA-like heat-soluble proteins that stay soluble even after boiling in tardigrades, we beforehand recognized cytoplasmic-abundant heat-soluble (CAHS) proteins from one of many hardest tardigrade species, Ramazzottius varieornatus [15]. CAHS proteins exhibited nearly no similarity with non-tardigrade proteins, and later genome and transcriptome analyses instructed that CAHS proteins are current solely in eutardigrades, one of many main courses of the phylum Tardigrada [11,1620]. Regardless of the absence of sequence similarity between CAHS proteins and LEA proteins, they share comparable biochemical properties, e.g., high-hydrophilicity supporting heat-solubility and structural transition from the disordered state in hydration to a helix beneath desolvating or dehydrated circumstances [12,15]. Like LEA proteins, CAHS proteins can defend enzymes from dehydration stress [18] and R. varieornatus produces a exceptional quantity of CAHS proteins moderately than trehalose and LEA proteins. Knockdown of a number of CAHS genes that impaired the anhydrobiotic survival revealed that CAHS proteins are concerned within the desiccation tolerance of eutardigrades [18]. Though CAHS proteins had been proposed to behave as a vitrifying agent based mostly on a shift in differential scanning calorimetry (DSC), this speculation was not too long ago counter-argued as such a shift might be defined by the evaporation of residual water [21], and the molecular mechanism stays to be elucidated.

Dehydration stress results in the cell shrinkage, inflicting extreme deformative mechanical stress affecting the integrity of cell constructions. To counteract the deformative forces, cytoskeletons like intermediate filaments (IFs) are typically principal gamers in extraordinary animal cells [22,23]. Apparently, canonical cytoplasmic IFs are lacking in Panarthropoda together with tardigrades and arthropods. Tardigrades have a tardigrade-unique IF protein referred to as cytotardin, which isn’t homologous to any cytoplasmic IFs in different animals and moderately derives from the nuclear filament protein lamin [24]. Cytotardin doesn’t localize to the nucleus as a result of it lacks a nuclear localization sign and as an alternative varieties belt-like filaments beneath the plasma membrane encircling epithelial cells, suggesting its contribution to the mechanical strengthening of epithelial tissues. In tardigrades, no IFs are identified to type scaffold-like filamentous networks within the cytosol, which is assumed to successfully counteract the deformative forces in lots of different animal cells [25,26].

On this research, we postulated the presence of tolerance proteins that type protecting condensates in a stress-dependent method and looked for such proteins in tardigrade lysate utilizing a desolvating agent, trifluoroethanol (TFE). Amongst greater than 300 recognized proteins that we collectively dubbed “TFE-dependent reversibly condensing proteins (T-DRYPs),” we unexpectedly rediscovered CAHS proteins as extremely enriched and main elements of T-DRYPs. Additional analyses revealed that in response to emphasize, CAHS reversibly varieties many cytoskeleton-like filaments in cultured cells and likewise reveals reversible gelation in vitro. CAHS proteins enhance the mechanical energy of cultured cells and enhance their resistance to dehydration-like stress. We additionally examined the structural foundation required for filament formation by deletion and level mutation analyses. By finding out the generated filament-impaired mutants, we confirmed that the filament-forming capability is the premise for the gel transition of CAHS proteins. On the premise of those outcomes, we suggest a brand new tolerance mannequin during which CAHS proteins act as a form of cytoskeleton that reversibly varieties intracellular filamentous networks in response to dehydration and induces gel transition that will increase mechanical energy of cells and contributes to the desiccation tolerance of tardigrades.

Outcomes

Trifluoroethanol-dependent reversibly condensing proteins (T-DRYPs) are recognized from Ramazzottius varieornatus

We designed the experimental scheme proven in Fig 1A to establish tardigrade proteins that type condensates in response to dehydration-like stress in a reversible method. We started with the lysate of the desiccation-tolerant tardigrade species R. varieornatus, as a result of this species constitutively expresses the tolerance proteins and its genome sequence is on the market [16]. First, we added a desolvating agent, TFE to a soluble fraction of R. varieornatus lysate to induce condensation in a dehydration-like state. TFE is a cosolvent that impacts the protein conformation by displacing water molecules from the floor of polypeptides [27] and/or destabilizing an aqueous solvation of polypeptide spine [28], which not directly promotes intramolecular hydrogen bonding and stabilizes the secondary constructions of proteins. TFE can be identified to advertise alpha-helix formation in a number of desiccation-tolerance proteins, resembling LEA and CAHS proteins as dehydration do [15,29,30]. The TFE-condensed proteins had been collected as precipitates and resolubilized with TFE-free PBS to imitate rehydration (resolvation). Therapy with increased focus of TFE elevated the variety of proteins detected within the resolubilized fraction (Figs 1B and S1). As remedy with 20% and 30% TFE had comparable results, we thought of 20% TFE to be an satisfactory stress situation for this screening (S1 Fig). When handled with TFE at 20% or increased, many proteins, particularly these with a excessive molecular weight, had been detected within the irreversibly precipitated fraction, indicating that solely the chosen proteins had been retrieved within the resolubilized fraction.

thumbnail

Fig 1. Isolation and characterization of T-DRYPs.

(A) Experimental scheme of T-DRYP isolation from tardigrade lysate. (B) SDS-PAGE picture of the resolubilized fractions with 0%, 10%, or 20% TFE remedy. (C) Comparability of the unstructured rating distributions between all tardigrade proteins and T-DRYPs. (D) Enrichment evaluation of the PANTHER protein class in T-DRYPs. Ribosomal proteins and cytoskeletal proteins had been considerably enriched. The numbers of the corresponding proteins detected in T-DRYPs and all tardigrade proteomes are proven on the appropriate, respectively. (E) Enrichment evaluation of stress-related proteins in T-DRYPs. CAHS proteins had been considerably enriched in T-DRYPs. (F) Venn diagram of T-DRYPs categorised by up- or down-regulation upon desiccation in orthologs of two different tardigrade species. (G) Comparability of unstructured rating distributions among the many in another way regulated protein teams in T-DRYPs. “Up-regulated” and “down-regulated” point out up-regulated or down-regulated proteins in each species, respectively. Proteins up-regulated upon desiccation exhibited increased unstructured scores. Pink and a pair of black horizontal bars in violin plot point out the fiftieth, twenty fifth, and seventy fifth percentiles, respectively. Statistical analyses had been carried out with the Wilcoxon rank sum check in (C) and the Metal–Dwass check in (G). The underlying numerical information can be found in S4 Knowledge (C, D, and G) in S2 Knowledge (E) and in S1 Knowledge (F). CAHS, cytoplasmic-abundant heat-soluble; T-DRYP, TFE-dependent reversibly condensing protein; TFE, trifluoroethanol.


https://doi.org/10.1371/journal.pbio.3001780.g001

We recognized 336 proteins within the resolubilized fraction (20% TFE) by nanoflow liquid chromatography-tandem mass spectrometry (nanoLC-MS/MS) and collectively termed these proteins “TFE-Dependent ReversiblY condensing Proteins (T-DRYPs)” (S1 Knowledge). As a result of reversible condensation is a attribute property anticipated for unstructured proteins, we calculated the unstructured rating of every protein in T-DRYPs by IUPred2A and in contrast the rating distribution with these of all tardigrade proteins. As anticipated, unstructured proteins had been considerably enriched in T-DRYPs (p < 2.2e-16, Wilcoxon rank sum check; Fig 1C). We assigned Drosophila melanogaster orthologs for tardigrade proteins and carried out enrichment evaluation of PANTHER Protein class or Gene Ontology time period in T-DRYPs. The outcomes revealed that ribosomal proteins and actin-related cytoskeletal proteins had been properly concentrated in T-DRYPs (Figs 1D and S2). Amongst T-DRYPs, nevertheless, 105 (31%) proteins had no obvious fly orthologs and T-DRYPs include many tardigrade-unique proteins (21%) together with identified tolerance proteins like CAHS proteins (S1 Knowledge). Subsequently, we expanded the enrichment analyses to the beforehand annotated tardigrade tolerance protein households that include greater than 5 members [16] and revealed the numerous enrichment of CAHS, LEA, HSP20, HSP70, and peroxiredoxin households in T-DRYPs (p < 0.01, chi-square check; Fig 1E and S2 Knowledge), suggesting that our new screening scheme concentrates desiccation-tolerance associated proteins to the resolubilized fraction. To judge this chance additional, we categorised T-DRYPs into 3 teams: stress-up-regulated teams, stress-down-regulated teams, and the others. R. varieornatus is without doubt one of the hardest tardigrade species that constitutively expresses stress-related genes [16]. Thus, we utilized gene expression information of two intently associated tardigrades, Hypsibius exemplaris and Paramacrobiotus metropolitanus (previously Paramacrobiotus sp. TYO), each of which exhibit robust up-regulation of tolerance gene expression upon desiccation [11,17,31]. Of 336 T-DRYPs, 315 proteins had orthologs in each species and 72 genes had been up-regulated throughout dehydration (Fig 1F and S1 Knowledge). Statistical evaluation indicated that the up-regulated proteins had been considerably enriched in T-DRYPs in contrast with the tardigrade proteome (p = 9.53e-29, chi-square check; S2 Knowledge). As well as, the up-regulated proteins additionally exhibited a a lot increased unstructured rating (Fig 1G), suggesting that tolerance-related unstructured proteins had been properly concentrated within the resolubilized fraction in our scheme. As a result of CAHS proteins had been extremely enriched within the T-DRYPs (Fig 1E), and likewise 3 main bands within the resolubilized fraction had been individually recognized as CAHS12, CAHS3, and CAHS8 (Figs 1B and S3), we centered on these 3 CAHS proteins for additional analyses.

CAHS3, CAHS8, and CAHS12 reversibly assemble into filaments or granules in animal cells relying on hyperosmotic stress

To visualise the stress-dependent condensation, 3 CAHS proteins, resembling CAHS3, CAHS8, and CAHS12 proteins had been individually expressed as a GFP-fused protein in human cultured HEp-2 cells and the distribution adjustments of those fusion proteins had been examined upon publicity to a hyperosmotic stress, which induces water efflux like dehydration stress [32]. In an unstressed situation, CAHS3-GFP broadly distributed within the cytosol, whereas CAHS8-GFP and CAHS12-GFP distributed broadly in each the cytosol and the nucleus with CAHS12-GFP displaying a slight choice for the nucleus (Fig 2A). When uncovered to hyperosmotic medium supplemented with 0.4 M trehalose, CAHS3-GFP condensed and shaped a filamentous community within the cytosol (Fig 2A and 2B). Comparable filament formation was noticed when CAHS3 alone was expressed with out GFP (S4 Fig), suggesting that filament formation is an intrinsic characteristic of CAHS3 protein moderately than artifact of fusion with GFP. CAHS12-GFP additionally shaped filaments within the cytosol and extra prominently within the nucleus in a majority of cells, although granule-like condensates had been additionally noticed within the nucleus of roughly 34% of the cells (Figs 2B and S5). CAHS8-GFP predominantly shaped granule-like condensates particularly within the nucleus, however filaments had been additionally noticed within the cytosol in a small inhabitants (roughly 3%) of the cells. Comparable distribution adjustments had been noticed even when GFP was fused to the other website in CAHS proteins (S6 Fig), whereas GFP alone didn’t exhibit such drastic adjustments. When hyperosmotic stress was eliminated by changing with isosmotic medium, all CAHS condensates, each filaments and granules, quickly dispersed (Fig 2A and 2B). Hyperosmotic stress by different supplemented osmolytes, resembling 0.2 M NaCl or 0.4 M sorbitol, which have an equal osmolarity to 0.4 M trehalose, induces comparable filament or granule formation, suggesting that the hyperosmotic stress itself is the motive force of condensation moderately than particular results of every osmolyte (S7 Fig). Comparable reversible condensations of CAHS proteins had been additionally noticed when expressed in Drosophila cultured S2 cells (S8 Fig and S1 Film), indicating that the stress-dependent filament/granule condensations are intrinsic options of CAHS proteins generally noticed in animal cells of taxonomically distant species.

thumbnail

Fig 2. Reversible formation of filaments or granules by CAHS3, CAHS8, and CAHS12 proteins in response to a hyperosmotic stress.

(A) Distribution adjustments in AcGFP1-tagged CAHS3, CAHS8, or CAHS12 proteins in HEp-2 cells throughout the transient hyperosmotic remedy with HBSS containing 0.4 M trehalose. Blue signifies Hoechst33342 staining of nuclei. (B) The proportion of distribution patterns (filaments, granules, or dispersed) of every CAHS protein in human cells. (C and D) FRAP analyses of CAHS3-GFP in human cells in dispersed state beneath an isosmotic situation (C, n = 7) and in a filament-formed state beneath a hyperosmotic situation (D, n = 6). (E) Confocal photographs of AcGFP1-tagged CAHS3 proteins and fluorescently labeled cytoskeletal proteins in HEp-2 cells beneath a hyperosmotic situation. White arrowheads point out slight co-localization of CAHS3 proteins and actin filaments. (F and G) Time-lapse photographs of filament formation or deformation of CAHS3-GFP in human cells (see additionally S2 and S3 Motion pictures). CAHS3-GFP first condensed into granules (155 s) after which elongated into filaments (355 s) as indicated by white arrowheads (F). CAHS3-GFP filaments concurrently collapsed and dispersed (398 s) (G). Time because the medium alternate to hyperosmotic (F) or isosmotic (G) resolution is proven in every picture. (H) Fluorescent photographs of HEp-2 cells co-expressing pairs of CAHS3, CAHS8, and CAHS12 proteins with a distinct fluorescent-tag beneath a hyperosmotic situation. CAHS3 co-localized with neither CAHS8 nor CAHS12. In distinction, CAHS8 properly co-localized with CAHS12 filaments. White arrowheads point out consultant co-localization. Scale bar, 10 μm in (A, E, and H), 2 μm in (F and G). The underlying numerical information can be found in S4 Knowledge (B–D). CAHS, cytoplasmic-abundant heat-soluble; FRAP, fluorescence restoration after photobleaching; HBSS, Hanks’ Balanced Salt Resolution.


https://doi.org/10.1371/journal.pbio.3001780.g002

Granule-like condensates of CAHS8 resemble droplet constructions shaped by intrinsically disordered proteins through liquid–liquid section separation. To check this chance, we examined the impact of 1,6-hexanediol, a disruption reagent of liquid-like condensates. After remedy with 5% 1,6-hexanediol for 30 min, the well-known droplet-forming protein FUS successfully dispersed, whereas a number of CAHS8 granules within the nucleus additionally dispersed however a lot much less successfully than FUS protein granules (S9 Fig), suggesting that CAHS8 granules had been partly liquid like. In distinction, the filament constructions of CAHS3 or CAHS12 weren’t affected by the hexanediol remedy, suggesting that CAHS3 and CAHS12 filaments had been in a static solid-like state. To additional assess the staticity of CAHS filaments, we carried out fluorescence restoration after photobleaching (FRAP) evaluation on CAHS3-GFP each earlier than and after publicity to hyperosmotic stress. In unstressed cultured cells, CAHS3-GFP was broadly distributed within the cytosol and the bleached fluorescence was quickly recovered (Fig 2C), indicating their excessive mobility nature. In distinction, beneath hyperosmotic stress, CAHS3-GFP filaments exhibited nearly no fluorescence restoration after bleaching (Fig 2D), suggesting that CAHS3 proteins shaped static filaments in response to a hyperosmotic stress. The filamentous networks shaped by CAHS proteins resembled cytoskeletal construction. To look at attainable cooperation between CAHS filament formation and different cytoskeletal constructions or organelles, we carried out co-localization analyses and noticed no co-localization between filament-forming CAHS proteins and any examined intracellular constructions aside from slight co-localization with actin filaments (Figs 2E and S10A–S10C). GFP alone additionally exhibited slight co-localization with actin filaments and actin polymerization inhibitor didn’t disrupt filament formation of CAHS3 and CAHS12 proteins (S10D, S10E, and S11 Figs), suggesting that CAHS filament formation can be impartial from actin filaments. These outcomes instructed that CAHS molecules freely disperse in an unstressed situation, however upon the publicity to hyperosmotic stress, CAHS molecules are firmly built-in into a further cytoskeleton-like filaments.

To elucidate the method of filament formation and deformation in additional element, we captured time-lapse photographs of cells expressing CAHS3-GFP whereas altering the stress circumstances by high-speed super-resolution microscopy. Roughly 2.5 min after the medium was modified to a hyperosmotic situation by a perfusion system, CAHS3-GFP started to condense concurrently at many websites within the cells and quickly shaped fibril constructions. The fibrils then additional prolonged in a number of dozen seconds (Fig 2F and S2 Film). When the hyperosmotic stress was eliminated by altering to an isosmotic medium, CAHS3 filaments concurrently started to loosen and regularly dispersed in roughly 6 min (Fig 2G and S3 Film). The preliminary condensation of CAHS3 and the granule formation of CAHS8 probably occurred through section separation, which incessantly results in co-condensation of a number of proteins, particularly these containing comparable motifs [33]. CAHS proteins share a number of conserved motifs and will thus cooperatively type the identical condensates. To look at this, we co-expressed pairs of the three CAHS proteins labeled with totally different fluorescent proteins in human cells. Below hyperosmotic stress, CAHS3 filaments didn’t co-localize with CAHS8 granules or CAHS12 filaments (Fig 2H). In distinction, CAHS8 largely co-localized with CAHS12 filaments all through the cell, suggesting that the granule-forming CAHS8 cooperatively varieties the filament construction with different CAHS proteins resembling CAHS12.

Secondary construction within the conserved C-terminal area is liable for CAHS filament formation

To disclose the structural foundation of CAHS filament formation, we first carried out de novo motif search and located 10 conserved motifs by evaluating 40 CAHS proteins of three tardigrade species, R. varieornatus, H. exemplaris, and P. metropolitanus (S12 and S13 Figs and S3 Knowledge). Specifically, we discovered that 2 C-terminal motifs (CR1 and CR2) are extremely conserved in all CAHS relations besides 1 CAHS protein of H. exemplaris (S12 Fig). To find out the area liable for filament formation, we generated a collection of truncated mutant proteins of CAHS3 or CAHS12 both N-terminally or C-terminally, and examined their filament formation in human cultured cells beneath a hyperosmotic stress (Figs 3A, 3B, and S14). In CAHS3, N-terminal deletion to motif 3 or C-terminal deletion to CR2 drastically impaired filament formation and as an alternative granule formation was incessantly noticed within the cytosol (Figs 3B, S15A and S15B). Accordingly, we designed a truncated mutant consisting of the minimal required area from motif 3 to CR2 (motif 3-motif H1-CR1-CR2) and revealed that this area is enough for the filament formation by CAHS3 protein (Figs 3B and S15C). Equally, in CAHS12 protein, the area consisting of CR1, CR2, and the two previous motifs (motif H2-motif H3-CR1-CR2) was proven to be essential and enough for the filament formation (S14 Fig). These outcomes indicated that 2 extremely conserved motifs (CR1 and CR2) and a pair of previous motifs (65 to 85 residues) play a necessary position within the filament formation of each CAHS3 and CAHS12 proteins.

In these recognized areas liable for the filament formation, intensive helix and coiled-coil constructions had been predicted by the secondary construction prediction software, JPred4 and COILS (Figs 3C and S16). The coiled-coil construction is the important thing structural foundation for the polymerization of IFs [34]. To check the contribution of those predicted secondary constructions in filament formation, we generated 2 mutants for every CAHS3 and CAHS12 by substituting leucine with proline, that are predicted to disrupt the helical and coiled-coil constructions of CR1 or CR2, respectively (Figs 3C and S17) [35]. As anticipated, all coiled-coil disruption mutants exhibited considerably impaired filament formation and as an alternative shaped granules (Figs 3C and S17S19). The double mutation (CAHS3-L207P-L236P) additional suppressed filaments formation and even lowered granule formation (Figs 3C and S20). These outcomes instructed that the secondary constructions of each CR1 and CR2 are an necessary foundation for the filament formation of CAHS3 and CAHS12 proteins.

thumbnail

Fig 3. Secondary construction within the conserved C-terminal area is liable for the CAHS filament formation and gelation.

(A) Schematic diagrams of CAHS3 proteins. “CR1” and “CR2” point out putative helical motifs extremely conserved amongst nearly all CAHS relations. “H1” point out putative helical conserved motifs; “1,” “2,” and “3” point out different conserved motifs. (B) Schematic diagrams and the corresponding distribution patterns of the truncated mutants of CAHS3. Quantified cell proportions of the distribution patterns beneath a hyperosmotic situation are proven as a stacked bar graph. Confocal photographs are proven for the consultant distribution sample of the corresponding CAHS mutants. Blue signifies Hoechst33342 staining of nuclei. (C) Results of a helix-disrupting mutation by substituting leucine with proline on filament formation of CAHS3. Schematic construction and the coiled-coil rating predicted by COILS are proven for each wild-type and proline substitution mutants. Asterisks point out the websites of proline substitutions. Substitution with proline considerably decreased the coiled-coil rating within the corresponding area. Confocal photographs present consultant distribution patterns of the corresponding CAHS proteins. Enlarged picture is proven as superimposition in every panel. Blue signifies Hoechst33342 staining of nuclei. Quantified cell proportions of every distribution sample are proven as stacked bar plots on the appropriate. (D) In vitro time-lapse confocal photographs of fibril formation of CAHS3-GFP proteins (1.24 mg/mL) after including TFE (closing 20%). GFP is a non-filament forming management. (E) TFE-dependent reversible gel-formation of CAHS3 proteins. By including TFE (closing 20%), CAHS3 protein options (4.0 mg/mL) turned turbid and transited right into a gel-like state. The gels spontaneously liquefied inside a number of minutes (proven in white letters) after publicity to air. (F) Filament-impaired CAHS3-L207P mutant protein options did not transit right into a gel-like state beneath 20% TFE. (G) Minimal filament-forming CAHS3 truncated protein (CAHS3-min) resolution reversibly solidified beneath 20% TFE like full-length CAHS3 protein. Scale bar, 10 μm in (B and C), 2.5 μm in superimposition in (C), 20 μm in (D), 2 mm in (E–G). The underlying numerical information can be found in S4 Knowledge (B and C). CAHS, cytoplasmic-abundant heat-soluble; TFE, trifluoroethanol.


https://doi.org/10.1371/journal.pbio.3001780.g003

In vitro reversible gel transition of CAHS proteins relying on desolvating agent and salt

To look at whether or not CAHS proteins alone are enough to type filaments, we carried out in vitro experiments utilizing purified CAHS3-GFP proteins. Below an unstressed situation, the uniform distribution of CAHS3-GFP proteins was noticed beneath a confocal microscope (Fig 3D). When the desolvating agent TFE was added to induce a dehydration-like conformational change as in our preliminary screening, CAHS3-GFP instantly condensed and shaped mesh-like fibril networks after 1 min. This outcome indicated that CAHS3 proteins alone can sense the adjustments within the situation and type filaments with out the help of different proteins.

When TFE was added to the answer containing a better focus of purified CAHS3 protein (closing 4 mg/mL; S21 Fig), the protein resolution instantly turned turbid, and the answer was solidified right into a gel-like state (Fig 3E). When the CAHS3 gel within the tube was unfold onto parafilm, the CAHS3 gel spontaneously liquefied inside roughly 10 min. We speculated that volatilization of TFE relieved the desolvating stress, thereby making the CAHS3 gel resoluble. Constantly, washing with TFE-free PBS additionally redissolved the gelated CAHS3 (S22A Fig). Whereas the management protein BSA was not solidified in the identical situation (S22B Fig), CAHS8 and CAHS12 exhibited the same TFE-dependent reversible gel-transition like CAHS3, however the gel of CAHS8 was a lot smaller than these of different CAHS proteins (S22C and S22D Fig), suggesting variations within the propensity for gelation amongst CAHS proteins. We additionally examined whether or not different stressors that might emerge throughout dehydration induce CAHS gelation and revealed that an elevated focus of salt (2 M NaCl) additionally induced the gel transition of CAHS3 proteins, whereas a molecular crowding agent (20% polyethylene glycol) precipitated turbidity, however no gelation (S23 Fig). The salt-induced gel persevered even after publicity to air on parafilm, presumably as a result of salt can not evaporate (S23A Fig). The granule-forming CAHS8 solely shaped a really small gel in vitro, implying a attainable relationship between the filament-forming capability in cells and the gel-forming capability in vitro. This notion was supported by the truth that the filament-impaired CAHS3-L207P mutant protein did not type the gel in vitro (Fig 3F). In distinction, minimal CAHS3 protein possessing the filament-forming capability (CAHS3-min) efficiently shaped the gel in vitro upon the addition of TFE and this transition was reversible as in full-length CAHS3 (Fig 3G), suggesting that the filament-forming capability underlies the gel transition of CAHS proteins in vitro.

CAHS confers the mechanical resistance in opposition to deformation forces on cell-like microdroplets and bug cells

To disclose what the gelation of CAHS proteins gives, we evaluated the results of CAHS gelation on the mechanical properties of cells utilizing cell-like microdroplets coated with a lipid layer. The elasticity of the microdroplets was examined by measuring the elongation size in a micropipette whereas aspirating with a sure stress. Microdroplets containing uniformly distributed CAHS3-GFP exhibited steady elongation exceeding 50 μm beneath very small stress (<<0.5 kPa), indicating that they weren’t elastic and in a liquid section (Fig 4A–4C). Then again, the addition of salt induced the filament formation of CAHS3-GFP and the corresponding microdroplets exhibited important elasticity (Younger’s modulus roughly 2.0 kPa in common), indicating that the CAHS3-GFP droplets gelated after which bodily hardened. Microdroplets containing GFP alone weren’t elastic whatever the addition of salt (Fig 4B and 4C).

thumbnail

Fig 4. CAHS confers the mechanical resistance in opposition to deformation forces on cell-like microdroplets and bug cells.

(A) Consultant fluorescent photographs of a microdroplet containing CAHS3-GFP within the absence or presence of extra NaCl. Scale bar, 5 μm. (B) Consultant response curves of the elongation size of microdroplets containing CAHS3-GFP or GFP alone beneath a really small stress (<<0.5 kPa). Steady elongation exceeding 50 μm signifies not elastic and in a liquid section. (C) Comparability of the elasticity (Younger’s modulus) amongst droplets containing CAHS3-GFP or GFP with or with out NaCl addition. Knowledge are introduced as common ± SE. (D) The consequences of CAHS3-expression on the cortical elasticity of Drosophila S2 cells beneath hyperosmosis. CAHS3-stably expressing cells exhibited increased elasticity in comparison with the management cells transfected with empty vector beneath a hyperosmotic situation supplemented with 0.4 M trehalose for 3 h. Grey and pink dots point out the values of every measurement. Black dots and bars point out averages and customary errors, respectively. (E) Comparability of cell quantity adjustments by hyperosmotic stress between CAHS3-expressing cells and management cells. The relative cell quantity was calculated by dividing the quantity beneath hyperosmotic stress by the averaged cell quantity beneath isosmotic circumstances. Middle bar and edges point out fiftieth, twenty fifth, and seventy fifth percentiles, respectively, and whiskers correspond to the 1.5 interquartile vary. (F and G) Comparability of cell viability between CAHS3-expressing cells and management cells beneath an isosmotic situation (F) and a hyperosmotic situation for 48 h (G). PI was used to find out lifeless cells. Survival charges had been examined in 6 wells for every situation by counting >500 cells/properly. Statistical analyses had been carried out with the Wilcoxon rank sum check in (D) and (E), and Pupil t check in (F) and (G); n.s. means not important within the statistical exams. The underlying numerical information can be found in S4 Knowledge (B–G). CAHS, cytoplasmic-abundant heat-soluble; PI, propidium iodide.


https://doi.org/10.1371/journal.pbio.3001780.g004

To additional decide whether or not CAHS proteins additionally stiffen animal cells, we established a Drosophila S2 cell line stably expressing CAHS3. S2 cells lack canonical cytoplasmic IFs as tardigrade cells do [24] and thus it will be appropriate to measure the impact of CAHS filamentation. Measuring the cell stiffness by atomic pressure microscope (AFM) revealed that beneath an unstressed situation, the CAHS3-expressing cells exhibited no important distinction within the elasticity with that of the management cells transfected with empty vector. Below a hyperosmotic situation, management cells exhibited increased elasticity than that in an unstressed situation, however the CAHS3-expressing cells exhibited considerably additional increased elasticity than that of the management cells beneath the identical situation (p < 0.05; Fig 4D), which is per the outcomes utilizing microdroplets. Hyperosmotic stress reduces the cell quantity via osmotic pressures [32]. As CAHS3-expressing cells exhibited increased elasticity beneath a hyperosmotic situation, they considerably counteract the osmotic stress and would possibly resist the cell shrinkage. To look at this chance, we measured the cell quantity adjustments by publicity to hyperosmotic stress. As proven in Fig 4E, CAHS3-expressing cells retained the cell quantity considerably higher than the management cells (p < 0.001; Fig 4E). These outcomes counsel that beneath a filament-forming situation, CAHS3 proteins stiffen cells and defend them from deformation stress attributable to water-efflux stress. Moreover, we additionally examined the impact of CAHS3 on cell viability after publicity to hyperosmotic stress. Cell viability was evaluated by the exclusion of propidium iodide (PI), which is an indicator of cell integrity. Below an unstressed situation, CAHS3 expression didn’t have an effect on the cell viability, however after 48 h remedy with hyperosmotic stress, CAHS3-expressing cells exhibited the elevated cell viability (Fig 4F and 4G). The cell stiffening by CAHS proteins could contribute to the stabilization of cell construction and the survival of cells throughout the dehydration-like course of.

Dialogue

Our research gives proof that CAHS proteins reversibly condense in a stress-dependent method and type a cytoskeleton-like filamentous community in animal cells or bear gel-transition in vitro (Figs 2A, 3E, S22C and S22D), and we additional demonstrated that the CAHS proteins enhance the mechanical energy of cell-like microdroplets and enhance the resistance in opposition to deformation stress of insect cells (Fig 4). Within the earlier research, CAHS proteins had been instructed to behave as a vitrifying agent like trehalose throughout dehydration based mostly on the shift in DSC [18], however this speculation was not too long ago counter-argued with information demonstrating that the shift in DSC may be defined by water retention of CAHS proteins [21]. As a result of hydrogel typically has excessive water retention properties, our commentary of gel transition by CAHS proteins helps the water retention within the counterargument moderately than vitrification. In vitro gel transition was noticed when utilizing a comparatively excessive focus (roughly 4 mg/mL) of CAHS protein resolution (Figs 3E, S22C and S22D), and the filament-impaired CAHS mutants failed in transition to gel (Fig 3F), suggesting {that a} dense filament formation is the structural foundation for the gel transition of CAHS proteins. To verify the protein focus utilized in gel transition in vitro is physiologically related, we estimated the quantity of endogenous CAHS3 proteins in R. varieornatus by immunoblotting evaluation, indicating that the quantity of CAHS3 proteins is about 3.8 ng per particular person (S24 Fig; see Supplies and strategies). The moist weight of a single particular person of R. varieornatus was reported to be 1.84 μg [36], which roughly corresponds 1.84 nL, and thus our tough estimate of the focus of endogenous CAHS3 protein is 2 mg/mL. Contemplating that CAHS3 proteins are current solely within the cytosol and never within the nucleus or extracellular house, the physiological focus of CAHS3 proteins can be a lot increased than our estimate, and we assumed it’s in the same vary of the focus used within the gel transition experiments in vitro. Contemplating the cell quantity discount throughout dehydration that results in a major enhance in each the protein focus and ion energy that could be one of many gel-inducing components as proven in S23A Fig, the intracellularly considerable CAHS proteins may bear gel transition in tardigrade cells and supply mechanical stabilization of cell integrity throughout dehydration (Fig 4C). This gel transition may partly account for the distinctive stability of dehydrated tardigrades. CAHS3-expression additionally stiffened insect cells, elevated the mechanical resistance, and improved hyperosmotic tolerance (Fig 4D–4G). The stress-dependent enhance of cell elasticity and the suppression of cell shrinkage beneath hyperosmosis are in a very good settlement with the truth that CAHS3 proteins shaped cytoskeleton-like filamentous community in animal cells in a stress-dependent method and help its performance, however it isn’t ascertained whether or not this enhancement of tolerance absolutely relies upon upon the filament formation by CAHS3. CAHS3 would possibly contribute to the tolerance in another means, and our mannequin doesn’t additionally exclude different attainable contributions or performance of CAHS proteins. The sol-gel transition and filament formation of CAHS proteins had been extremely reversible and stress dependent, and FRAP analyses revealed that CAHS proteins had been motionless solely when filaments shaped beneath a stress situation. Subsequently, we suppose that CAHS proteins are freely dispersed in a hydrated situation to attenuate interference with different organic processes, whereas in a dehydrated situation, CAHS proteins type an intracellular filamentous community and elastic hydrogel to offer mechanical stabilization of cell integrity.

Though CAHS proteins exhibit no sequence similarity with every other cytoskeletal proteins, they shaped cytoskeleton-like filamentous networks independently from the opposite cytoskeleton beneath a hyperosmotic stress (Figs 2E, S10 and S11) and CAHS-expressing cells exhibited increased resistance in opposition to the deformative mechanical forces beneath the hyperosmotic stress (Fig 4D and 4E). Therefore, CAHS proteins could also be a novel cytoskeletal protein household with stress-dependence and gel-forming capability. Though no identified motifs are discovered within the major sequence of CAHS proteins, the C-terminal area together with the extremely conserved CR1 and CR2 motifs was important and enough for the filament formation (Figs 3B and S14). This area was principally predicted as helical and to type a coiled-coil construction (Figs 3C and S17). This prediction was additionally supported by the earlier round dichroism (CD) spectroscopy of CAHS1 protein of R. varieornatus, one other member of the CAHS household [15]. In the course of the assessment of this manuscript, 2 associated papers had been printed [37,38], which reported that 2 different CAHS proteins, i.e., CAHS1 of R. varieornatus and CAHS8 of H. exemplaris, shaped fibrous construction and gel in a concentration-dependent method in vitro, and the enriched helix construction within the C-terminal areas in both CAHS proteins had been demonstrated by elaborate NMR analyses and/or CD spectroscopy beneath the situation forming filaments or gels. These current structural analyses are in a very good settlement with our structural predictions (S16 Fig), though the impact of disturbance of such helix construction on filament/gel formation had not been examined. Our commentary of the extreme impairments within the filament/gel formation by proline substitutions in both the CR1 or CR2 area (Fig 3C and 3F) point out that the secondary construction of CR1 and CR2 performs necessary roles in CAHS filament/gel formation. Some intrinsically disordered proteins are reported to type a gel-like granule condensate through promiscuous binding via multivalent interplay websites [39], however in CAHS3 and CAHS12, single amino acid substitution is sufficient to disrupt each filament formation and gel transition, suggesting that the mechanism of filament/gel formation of CAHS proteins is probably going not attributable to multivalent interactions, however moderately to polymerization based mostly on the secondary construction. The prediction of 3D constructions by AlphaFold2 [40,41] instructed that CAHS3-min proteins type a helix within the CR1+CR2 area with excessive confidence (pLDDT = 70 to 90) and a pair of CAHS3-min proteins type an antiparallel dimer with the juxtaposition of every helical area the place the cost and hydrophobicity distribution is per the stabilization of two helix interactions (S25 Fig). This antiparallel alignment is just like the lamin tetramer formation [34], suggesting that the method of filament formation of CAHS proteins could also be considerably just like IFs.

In distinction to filament-forming CAHS3 and CAHS12, CAHS8 alone shaped granule-like condensates in each human and bug cells beneath a hyperosmotic situation (Figs 2A and S8). Lately, CAHS1 protein from R. varieornatus was additionally reported to type granules in response to hyperosmotic stress in human cultured cells [37], and these stress-dependent granule condensation by CAHS8 and CAHS1 resembled the stress-granule formation in mammalian cells that happens via section separation to create protecting membrane-less compartments in opposition to stress [42,43]. A current research revealed that one other desiccation tolerance protein, AfrLEA6, which is a bunch 6 LEA protein of Artemia franciscana, additionally undergoes section separation to type granules in insect cells [14] and protects enzyme exercise from desiccation stress in vitro [44]. Like stress granules and AfrLEA6 granules, CAHS8 granules exhibited sure sensitivity in opposition to 1,6-hexanediol remedy (S9 Fig). CAHS8 and CAHS1 would possibly exert comparable protecting features through granule condensation beneath stress circumstances. Alternatively, in cells co-expressing CAHS8 and CAHS12, as proven in Fig 2H, CAHS8 contributes to filament formation with CAHS12 in tardigrades.

Intriguingly, the desiccation-induced (up-regulated) tardigrade proteins had been considerably enriched in T-DRYPs (S2 Knowledge), implying that our TFE-based isolation scheme may selectively seize the dehydration-responsive proteins that reversibly condense in response to desiccation stress. Two well-known desiccation-tolerance protein households, LEA and CAHS proteins, had been additionally captured as extremely enriched in T-DRYPs (Fig 1E). These 2 protein households are largely unstructured in hydration and mutually unrelated within the major sequence, however each develop into helix-rich construction upon dehydration [12,37]. TFE is also referred to as a stabilizer of helical construction [45], for which a number of stabilization mechanisms have been proposed, e.g., the destabilization of the interplay between polypeptides and water molecules promotes native intramolecular hydrogen bonding in polypeptides and consequently stabilize the helical construction [27,28,46]. Our T-DRYPs isolation methodology may seize CAHS and LEA proteins via TFE-induced helix formation as occurred in a dehydrating situation. We additionally can not exclude the likelihood that some proteins in T-DRYPs might be remoted via the helix-inducing property moderately than the desolvating property of TFE. Within the T-DRYPs, stress-related unstructured proteins had been enriched (Fig 1G), in addition to translational proteins and cytoskeleton-related proteins (Fig 1D). These proteins could be integrated into stress-dependent condensates like stress granules to be protected against stress. Alternatively, a few of them like cytoskeletal proteins could be co-precipitated via entangling with CAHS filaments. Though CAHS proteins are conserved solely in eutardigrades, proteins with comparable properties could be current in different desiccation-tolerant organisms and should contribute to emphasize resistance. It’s noteworthy that the associated animal teams resembling heterotardigrades or arthropods additionally lack the canonical cytoplasmic IFs however wonderful anhydrobiotic capability is noticed in some chosen species resembling Echiniscus testudo (a heterotardigrade), Polypedilum vanderplanki (a sleeping chironomid), and Artemia (a brine shrimp) [2,5,6]. These animals would possibly possess one other class of stress-dependent filament-forming proteins. Lately, a brand new heat-soluble protein household termed EtAHS was recognized in E. testudo [20]. This protein household or different new ones are probably good candidates. Our isolation scheme of T-DRYPs could present a helpful methodology to establish unstructured proteins that bear reversible condensation to filaments or granules in a stress-dependent method from numerous organisms. CAHS proteins had been initially recognized by looking for heat-soluble proteins to establish anhydrobiotic protectants in tardigrades [15]. Later, many heat-soluble proteins had been recognized from people and flies, dubbed Hero proteins [47], that exhibit no sequence similarity with CAHS proteins however present stabilization of different proteins as CAHS and LEA proteins do. Equally, future T-DRYPome evaluation could result in the identification of protecting phase-separating proteins even in non-anhydrobiotic organisms.

Within the current research, we established a brand new methodology to establish proteins which might be reversibly condensed in response to desolvating agent and located 336 such proteins from desiccation-tolerant tardigrades. The main elements, CAHS3 and CAHS12, had been proven to type cytoskeleton-like filaments and elastic hydrogel in a stress-dependent method. Moreover, we demonstrated that CAHS3 can confer mechanical resistance in opposition to deformation stress on insect cells and enhanced their tolerance to dehydration-like stress. We suggest that these CAHS proteins could operate as novel stress-dependent and gel-forming cytoskeletal proteins that present mechanical energy to stabilize mobile integrity throughout stress. Our information instructed a novel desiccation tolerance mechanism based mostly on filament/gel formation. The isolation scheme established on this research opens the best way to figuring out such novel stress-dependent cytoskeletal proteins from numerous organisms.

Supplies and strategies

Identification of trifluoroethanol-dependent reversibly condensing proteins

Previous to protein extraction, tardigrades had been starved for 1 day to get rid of digestive meals. Roughly 400 R. varieornatus had been collected and extensively washed with sterilized Milli-Q water to take away contaminants. Tardigrades had been rinsed with lysis buffer, phosphate-buffered saline (PBS; 137 mM NaCl, 2.7 mM KCl, 10 mM Na2HPO4, 1.76 mM KH2PO4 (pH 7.4)) containing full protease inhibitors (Roche) and transferred to a 1.7 mL tube. Tardigrades had been homogenized in 20 μL lysis buffer utilizing a plastic pestle on ice. The pestle was rinsed with a further 20 μL of lysis buffer collected in the identical tube. After centrifugation at 16,000 × g for 20 min at 4°C, the supernatant was recovered as a soluble protein extract. To imitate dehydration stress, the desolvating agent, TFE, was added (closing focus 10%, 20%, or 30%), and the combination was incubated on ice for 1 h to permit full induction of condensation. After centrifugation at 16,000 × g for 20 min, the supernatant was eliminated as a TFE-soluble fraction and the remaining precipitate was washed twice by lysis buffer containing TFE on the similar focus. The washed precipitate was resuspended in lysis buffer with out TFE and incubated at room temperature for 30 min to facilitate resolubilization. After centrifugation at 16,000 × g at 4°C, the supernatant was recovered as a resolubilized fraction. The fractions had been analyzed by SDS-PAGE and proteins had been visualized utilizing a Silver Stain MS Equipment (Fujifilm). Three chosen bands had been excised and individually subjected to mass spectrometry. Complete identification of T-DRYPs was achieved by shot-gun proteomics of the resolubilized fraction. Briefly, proteins in gel slices or within the fraction had been digested with trypsin and fragmented peptides had been analyzed by nanoLC-MS/MS. Proteins had been recognized utilizing MASCOT software program (Matrix Science). The mass spectrometry proteomics information have been deposited to the ProteomeXchange Consortium through the jPOST repository with the dataset identifier PXD030241 and may be retrieved at http://proteomecentral.proteomexchange.org/cgi/GetDataset?ID=PXD030241.

Enrichment evaluation

To make the most of well-annotated info within the mannequin organism D. melanogaster, we assigned a D. melanogaster ortholog for every R. verieornatus protein by a reciprocal BLAST search. We assigned 231 fly orthologs for 336 T-DRYPs and seven,361 fly orthologs for all 19,521 R. varieornatus proteins. Utilizing the assigned fly orthologs, we carried out enrichment analyses with PANTHER Overrepresentation Check [53] (PANTHER Protein Class model 16.0, Fisher’s check; http://pantherdb.org/) and Metascape [54] (GO Mobile Elements; https://metascape.org/). The listing of fly orthologs for all R. varieornatus proteins was used as a reference within the enrichment analyses.

Amongst tardigrade stress-related proteins described beforehand [16], 7 protein households containing greater than 5 members had been chosen for the enrichment evaluation (S2 Knowledge). Enrichment of every household in T-DRYPs was statistically examined by Fisher’s actual check utilizing R. Enrichment of up-regulated genes was equally examined besides utilizing a chi-square check.

Differential gene expression evaluation

Transcriptome information at a hydrated state and a dehydrated state had been retrieved from the general public database (DRR144971-DRR144973 and DRR144978-DRR144980 for Paramacrobiotus metropolitanus; SRR5218239-SRR5218241 and SRR5218242-SRR5218244 for Hypsibius exemplaris, respectively). The genome sequence of P. metropolitanus was retrieved from the general public database beneath accession numbers BHEN01000001-BHEN01000684 [11]. The genome sequence of H. exemplaris v3.0 was retrieved from http://www.tardigrades.org. RNA-seq reads had been mapped to the genome sequence utilizing HISAT2 v.2.1.0 [55]. Learn counts for every gene area had been quantified by featureCounts in SubRead package deal v.1.6.3 [56] and statistically in contrast by R package deal DESeq2 [57]. The genes with FDR < 0.01 had been thought of as differentially expressed genes. Orthologous gene relationships had been decided by reciprocal BLAST searches amongst 3 tardigrade species.

Plasmids

CAHS3, CAHS8, and CAHS12 coding sequences had been amplified from the corresponding EST clones of R. varieornatus [16] and inserted into pAcGFP1-N1 or pAcGFP1-C1 (Clontech) with (GGGGS)3 linker utilizing In-Fusion HD Cloning Equipment (Takara). Plasmids to specific CAHS deletion mutants (CAHS3Δctail, CAHS3ΔCR2-C, CAHS3ΔN-M2, CAHS3ΔN-M3, CAHS3-min, CAHS12Δctail, CAHS12ΔCR2-C, CAHS12ΔN-M1, CAHS12ΔN-H2, and CAHS12-min) or leucine-to-proline substitution mutants (CAHS3-L207P, CAHS3-L236P, CAHS3-L207P-L236P, CAHS12-L204P, and CAHS12-L241P) had been generated by inverse PCR and ligation or PCR-based website directed mutagenesis. The CAHS3/8/12-mScarlet-I expression vector was generated from CAHS3/8/12-GFP expression vector by changing AcGFP1 coding sequences with mScarlet-I sequence fragments [58] synthesized artificially (IDT). Expression constructs for numerous cytoskeleton or organelle marker proteins had been obtained from Addgene (S1 Desk). For bacterial expression of His6-tagged CAHS proteins, CAHS3, CAHS8, or CAHS12 coding sequences had been amplified and inserted into pEThT vectors [15], and CAHS3-GFP was equally inserted right into a pCold-I vector (Takara). For expression in Drosophila cells, codon-optimized CAHS3, CAHS8, CAHS12, and AcGFP1 DNA fragments had been synthesized (Gene Common) and inserted into pAc5.1/V5-His A vector (Invitrogen). The FUS-Venus plasmid was a sort reward from Dr. Tetsuro Hirose.

Stay cell imaging beneath hyperosmosis

We used Hep-2 cells for live-imaging of fluorescently labeled proteins as a result of Hep-2 cell had been properly sticky even beneath a stress situation and enabled exact inspections. Hep-2 cells had been transiently transfected with an expression vector of fluorescently labeled proteins utilizing Lipofectamine LTX & Plus Reagent (Invitrogen) for 48 h earlier than stress publicity. Previous to microscopy, the medium was changed with Hanks’ Balanced Salt Resolution (HBSS) with out the dications and phenol pink. For publicity to hyperosmotic stress, the buffer was changed with HBSS containing 0.4 M trehalose. The cells had been stained with Hoechst 33342 (5 μg/mL, Lonza) to visualise nuclear DNA. Fluorescent alerts had been noticed utilizing a confocal microscope LSM710 (Carl Zeiss). The variety of cells for every CAHS distribution sample, resembling dispersed, granules, or filaments, had been counted by 2 impartial investigators and averaged counts had been used. For time-lapse imaging in 3D house, we used the LSM-980 with Airyscan to carry out super-resolution imaging. From the z-stack photographs, we generated orthogonal projections utilizing ZEN 2.6 software program. In time-lapse imaging experiments, a perfusion system KSX-Type1 (Tokai Hit) was used to switch the buffer. To visualise actin filaments by chemical staining, Hep-2 cells had been handled with silicon-rhodamine dye probing actin (SiR-actin, Spirochrome) in HBSS containing the drug efflux inhibitor verapamil (10 μM, Tokyo Chemical Business) for two h. For actin polymerization inhibition experiments, cells had been handled with cytochalasin B (5 μM, Nacalai Tesque) for 60 min. Cells had been then noticed by a confocal microscope LSM-710 (Carl Zeiss).

Sensitivity to 1,6-hexanediol remedy

Hep-2 cells had been transfected with expression vectors of CAHS3/8/12-AcGFP1 or FUS-Venus. After 48 h, cells had been uncovered on minimal important medium supplemented with 0.4 M trehalose and 10% FBS for 1 h to induce the formation of granules or filaments. FUS protein was used as a management as it’s identified to be integrated into liquid droplets beneath hyperosmosis [59]. After the addition of a liquid droplet disruptor, 1,6-hexanediol (closing 5%), fluorescent photographs had been captured at 0 and 30 min later by a confocal microscope LSM710 (Carl Zeiss). The fluorescence depth was measured by Fiji and normalized to the preliminary fluorescence depth of the granules or filaments.

Protein preparation

Recombinant proteins had been expressed as N-terminally His6-tagged proteins in Escherichia coli BL21(DE3) strains. CAHS3, CAHS8, and CAHS12 proteins had been expressed utilizing pET system (Novagen) basically as described beforehand [15]. CAHS3-GFP and AcGFP1 had been expressed utilizing a chilly shock expression system (Takara) basically as described beforehand [60]. Bacterial pellets had been lysed in PBS containing full EDTA-free protease inhibitors (Roche) by sonication. For CAHS3, CAHS8, and CAHS12, the supernatant was heated at 99°C for 15 min to retrieve heat-soluble CAHS proteins in a soluble fraction as described beforehand [15]. From the soluble fraction, His6-tagged proteins had been purified with Ni-NTA His-Bind Superflow (Novagen) and dialyzed in opposition to PBS utilizing a Pur-A-Lyzer Midi Dialysis Equipment (Merck).

Measurement of the elasticity of droplets by micropipette aspiration

The elasticity of the cell-like microdroplets was evaluated by a micropipette aspiration system as reported beforehand [61]. The floor elasticity (Younger’s modulus), E, is derived from the linear relationship between the elongation size into the micropipette, ΔL, and the aspiration stress, ΔP: E = (3ΔPRpΦ/2π)/ΔL, whereby Rp and Φ are the micropipette inside radius and wall operate, which is derived from the form of the micropipette. We used a micropipette with an Rp smaller than × 0.4 of the microdroplet radius R. The worth of Φ is 2.0. A rise in ΔL to above 50 μm beneath a really small ΔP (<<0.5 kPa) signifies that the microdroplet is in liquid section. Within the case of the elastic gel section, a linear relationship between ΔL and ΔP was confirmed for the small deformation inside ΔL < 5 μm and ΔP < 3 kPa. Below these circumstances, we derived the values of E. The temperature was roughly 25°C.

Institution of stably transfected cell line of Drosophila S2 cells

The expression vector Ac5-STABLE2-neo was obtained from Addgene (#32426) [62], after which the coding sequence of FLAG-mCherry was changed with the codon-optimized CAHS3 coding sequence (Gene Common) to specific CAHS3-T2A-EGFP-T2A-neoR beneath the management of Ac5 promoter. The empty vector was constructed by deleting FLAG-mCherry from Ac5-STABLE2-neo, which was designed to specific T2A-EGFP-T2A-neoR pushed by the identical Ac5 promoter. Drosophila S2 cells had been transfected utilizing a cationic liposome reagent Hilymax (Dojindo) with the expression assemble or the empty vector above. We established stably transfected cells by culturing for six weeks beneath the drug choice with G418 disulfate (2,000 μg/mL, Nacalai Tesque).

Measurement of cell quantity

The quantity of every cell was measured utilizing serial photographs of optical sections in keeping with the earlier publication [63]. Three-dimensional imaging was carried out for GFP fluorescence within the stably transfected cells at 1.05 μm z-axis intervals utilizing a 63 × /1.2 oil-immersion lens on a confocal microscope LSM710 (Carl Zeiss). Cross-sectional space of the cell was calculated from every sectioned picture utilizing Fiji software program, and cell quantity was estimated as a sum of them.

Estimation of the quantity of endogenous CAHS3 protein by immunoblotting

After intensive washing with purer water, roughly 100 R. varieornatus had been lysed utilizing pestle in 30 μL PBS containing full EDTA-free protease inhibitors (Roche) and centrifuged at 16,000 × g for 10 min. The soluble fractions of tardigrade lysate had been combined with 5 × SDS pattern buffer (62.5 mM Tris-HCl (pH6.8), 25% glycerol, 10% sodium dodecyl sulfate, and 0.01% bromophenol blue) and 2-mercapto-ethanol. After heated at 100°C for 3 min, the samples had been resolved by SDS-PAGE evaluation and electroblotted onto PVDF membrane (Millipore). The membrane was blocked with 1% regular goat serum (Abcam) for 1 h at room temperature and reacted with the affinity-purified CAHS3 antibody diluted by 1% regular goat serum for 1 h at room temperature. After washed with TBS-T 3 instances, the membrane was reacted with diluted peroxidase labeled anti-rabbit IgG antibody (KPL) for 1 h at room temperature. The membrane was washed with TBS-T 3 instances, after which antibody-antigen advanced was detected by ImageQuant LAS 500 (Cytiva) utilizing enhanced chemiluminescence system (GE Healthcare). The diluted collection of recombinant CAHS3 proteins (2.5, 5.0, 10.0, 20.0, 40.0 ng) had been analyzed concurrently on the identical blot as quantification requirements. Sign depth of every corresponding band was measured by Fiji software program and a linear regression was used to generate a normal curve between the sign depth and the quantity of protein as [Signal intensity] = [Amount of protein (ng)] × 446595.3952–696903.625; R2 = 0.9962. Utilizing the well-fitted customary curve, the quantity of endogenous CAHS3 protein was calculated to be roughly 3.81 ng per tardigrade.

Supporting info

S1 Fig. Silver-stained gel photographs of every fraction within the T-DRYP isolation course of.

Every fraction was analyzed by SDS-PAGE and visualized by silver-staining. The picture of the resolubilized fraction is partly introduced in Fig 1B. Because the focus of TFE elevated (0% to twenty%), proteins decreased within the TFE soluble fraction, and proteins elevated in each the irreversible precipitate and the resolubilized fraction. Therapy with 20% and 30% TFE had largely comparable results.

https://doi.org/10.1371/journal.pbio.3001780.s001

(TIF)

S5 Fig. Consultant photographs for every distribution sample (filament, granule, or dispersed) of CAHS3-GFP, CAHS8-GFP, CAHS12-GFP, and GFP alone in human cultured HEp-2 cells.

N/A signifies that the corresponding distribution sample will not be or not often present in a hyperosmotic situation. Scale bar, 10 μm.

https://doi.org/10.1371/journal.pbio.3001780.s005

(TIF)

S1 Knowledge. Checklist of recognized 336 T-DRYPs from Ramazzottius varieornatus.

Every column point out as follows: (A) Protein ID (Ramazzottius varieornatus). (B) Rating, Mascot rating. (C) Protection, the protection by the detected peptides in whole residues. (D) # Proteins, the variety of the matched proteins. (E) # Distinctive peptides, the variety of the uniquely matched peptides. (F) # Peptides, the variety of matched peptides. (G) # PSMs, the variety of peptide spectrum matches. (H) #AAs, the variety of amino acids. (I) MW [kDa], molecular weight. (J) pI, isoelectric level. (Ok) IUPred2A, the averaged unstructured scores by IUPred2A. (L) classification, putative taxonomic origins. (M) Human ortholog, orthologous genes of Homo sapiens. (N) Fly ortholog, orthologous genes of Drosophila melanogaster. (O) Hypsibius exemplaris ortholog (nHd3.0), orthologous genes of Hypsibius exemplaris. (S) Paramacrobiotus metropolitanus ortholog, orthologous genes of Paramacrobiotus metropolitanus (previously Paramacrobiotus sp. TYO). (P and T)log2FC, log2 fold adjustments upon desiccation in H. exemplaris or P. metropolitanus. (Q and U) FDR, false discovery fee in differentially expression analyses between the hydrated and dehydrated states. (R and V) desiccation-change, the classification based mostly on gene regulation upon desiccation. Clean signifies insignificant change (FDR > 0.01), UP signifies log2FC is constructive and DOWN signifies log2FC is damaging.

https://doi.org/10.1371/journal.pbio.3001780.s030

(XLSX)

References

  1. 1.
    Keilin D. The issue of anabiosis or latent life: historical past and present idea. Proc R Soc London Ser B, Biol Sci. 1959;150:149–191. pmid:13633975
  2. 2.
    Møbjerg N, Halberg KA, Jørgensen A, Persson D, Bjørn M, Ramløv H, et al. Survival in excessive environments–on the present information of diversifications in tardigrades. Acta Physiol (Oxf). 2011;202:409–420. pmid:21251237
  3. 3.
    Jönsson KI, Rabbow E, Schill RO, Harms-Ringdahl M, Rettberg P. Tardigrades survive publicity to house in low Earth orbit. Curr Biol. 2008;18:729–731. pmid:18786368
  4. 4.
    Persson D, Halberg KA, Jørgensen A, Ricci C, Møbjerg N, Kristensen RM. Excessive stress tolerance in tardigrades: Surviving house circumstances in low earth orbit. J Zoolog Syst Evol Res. 2011;49:90–97.
  5. 5.
    Sakurai M, Furuki T, Akao KI, Tanaka D, Nakahara Y, Kikawada T, et al. Vitrification is important for anhydrobiosis in an African chironomid, Polypedilum vanderplanki. Proc Natl Acad Sci U S A. 2008;105:5093–5098. pmid:18362351
  6. 6.
    Clegg JS. The origin of threhalose and its significance throughout the formation of encysted dormant embryos of Artmia salina. Comp Biochem Physiol. 1965;14:135–143.
  7. 7.
    Madin KAC, Crowe JH. Anhydrobiosis in nematodes: Carbohydrate and lipid metabolism throughout dehydration. Exp Zool. 1975;193:335–342.
  8. 8.
    Erkut C, Penkov S, Khesbak H, Vorkel D, Verbavatz JM, Fahmy Ok, et al. Trehalose renders the dauer larva of caenorhabditis elegans proof against excessive desiccation. Curr Biol. 2011;21:1331–1336. pmid:21782434
  9. 9.
    Jain NK, Roy I. Impact of trehalose on protein construction. Protein Sci. 2009;18:24–36. pmid:19177348
  10. 10.
    Hengherr S, Heyer AG, Köhler HR, Schill RO. Trehalose and anhydrobiosis in tardigrades–Proof for divergence in responses to dehydration. FEBS J. 2008;275:281–288. pmid:18070104
  11. 11.
    Hara Y, Shibahara R, Kondo Ok, Abe W, Kunieda T. Parallel evolution of trehalose manufacturing equipment in anhydrobiotic animals through recurrent gene loss and horizontal switch. Open Biol. 2021;11:200413. pmid:34255978
  12. 12.
    Hand SC, Menze MA, Toner M, Boswell L, Moore D. LEA proteins throughout water stress: Not only for vegetation anymore. Annu Rev Physiol. 2011;73:115–134. pmid:21034219
  13. 13.
    Goyal Ok, Walton LJ, Tunnacliffe A. LEA proteins forestall protein aggregation attributable to water stress. Biochem J. 2005;388:151–157. pmid:15631617
  14. 14.
    Belott C, Janis B, Menze MA. Liquid-liquid section separation promotes animal desiccation tolerance. Proc Natl Acad Sci U S A. 2020;117:27676–27684. pmid:33077592
  15. 15.
    Yamaguchi A, Tanaka S, Yamaguchi S, Kuwahara H, Takamura C, Imajoh-Ohmi S, et al. Two novel heat-soluble protein households abundantly expressed in an anhydrobiotic tardigrade. PLoS ONE. 2012;7:e44209. pmid:22937162
  16. 16.
    Hashimoto T, Horikawa DD, Saito Y, Kuwahara H, Kozuka-Hata H, Shin-I T, et al. Extremotolerant tardigrade genome and improved radiotolerance of human cultured cells by tardigrade-unique protein. Nat Commun. 2016;7:12808. pmid:27649274
  17. 17.
    Yoshida Y, Koutsovoulos G, Laetsch DR, Stevens L, Kumar S, Horikawa DD, et al. Comparative genomics of the tardigrades Hypsibius dujardini and Ramazzottius varieornatus. PLoS Biol. 2017;15:e2002266. pmid:28749982
  18. 18.
    Boothby TC, Tapia H, Brozena AH, Piszkiewicz S, Smith AE, Giovannini I, et al. Tardigrades use intrinsically disordered proteins to outlive desiccation. Mol Cell. 2017;65:975–984. pmid:28306513
  19. 19.
    Kamilari M, Jørgensen A, Schiøtt M, Møbjerg N. Comparative transcriptomics counsel distinctive molecular diversifications inside tardigrade lineages. BMC Genomics. 2019;20:607. pmid:31340759
  20. 20.
    Murai Y, Yagi-Utsumi M, Fujiwara M, Tanaka S, Tomita M, Kato Ok, et al. Multiomics research of a heterotardigrade, Echinisicus testudo, suggests convergent evolution of anhydrobiosis-related proteins in Tardigrada. BMC Genomics. 2021;22:813.
  21. 21.
    Arakawa Ok, Numata Ok. Reconsidering the “glass transition” speculation of intrinsically unstructured CAHS proteins in desiccation tolerance of tardigrades. Mol Cell. 2021;81:409–410. pmid:33545053
  22. 22.
    Fletcher DA, Mullins RD. Cell mechanics and the cytoskeleton. Nature. 2010;463:485–492. pmid:20110992
  23. 23.
    Pegoraro AF, Janmey P, Weitz DA. Mechanical properties of the cytoskeleton and cells. Chilly Spring Harb Perspect Biol. 2017;9:a022038. pmid:29092896
  24. 24.
    Hering L, Bouameur JE, Reichelt J, Magin TM, Mayer G. Novel origin of lamin-derived cytoplasmic intermediate filaments in tardigrades. Elife. 2016;5:e11117. pmid:26840051
  25. 25.
    Goldman RD, Khuon S, Chou YH, Opal P, Steinert PM. The operate of intermediate filaments in cell form and cytoskeletal integrity. J Cell Biol. 1996;134:971–983. pmid:8769421
  26. 26.
    Peter A, Stick R. Evolutionary facets in intermediate filament proteins. Curr Opin Cell Biol. 2015;32:48–55. pmid:25576801
  27. 27.
    Roccatano D, Colombo G, Fioroni M, Mark AE. Mechanism by which 2,2,2-trifluoroethanol/water mixtures stabilize secondary-structure formation in peptides: a molecular dynamics research. Proc Natl Acad Sci U S A. 2002;99:12179–12184. pmid:12196631
  28. 28.
    Kentsis A, Sosnick TR. Trifluoroethanol promotes helix formation by destabilizing spine publicity: desolvation moderately than native hydrogen bonding defines the kinetic pathway of dimeric coiled coil folding. Biochemistry. 1998;37:14613–14622. pmid:9772190
  29. 29.
    Tolleter D, Jaquinod M, Teyssier E, Payet N. Construction and performance of a mitochondrial late embryogenesis considerable protein are revealed by desiccation. 2007;19:1580–1589. pmid:17526751
  30. 30.
    Koubaa S, Bremer A, Hincha DK, Brini F. Structural properties and enzyme stabilization operate of the intrinsically disordered LEA_4 protein TdLEA3 from wheat. Sci Rep. 2019;9:3720. pmid:30842512
  31. 31.
    Sugiura Ok, Matsumoto M, Kunieda T. Description of a mannequin tardigrade Paramacrobiotus metropolitanus sp. Nov. (Eutardigrada) from Japan with a abstract of its life historical past, copy and genomics. Zootaxa. 2022;5134:92–112.
  32. 32.
    Guo M, Pegoraro AF, Mao A, Zhou EH, Arany PR, Han Y, et al. Cell quantity change via water efflux impacts cell stiffness and stem cell destiny. Proc Natl Acad Sci U S A. 2017;114:E8618–E8627. pmid:28973866
  33. 33.
    Kato M, Han TW, Xie S, Shi Ok, Du X, Wu LC, et al. Cell-free formation of RNA granules: Low complexity sequence domains type dynamic fibers inside hydrogels. Cell. 2012;149:753–767. pmid:22579281
  34. 34.
    Herrmann H, Aebi U. Intermediate filaments: construction and meeting. Chilly Spring Harb Lab Press. 2016;8:a018242. pmid:27803112
  35. 35.
    Chou PY, Fasman GD. Prediction of protein conformation. Biochemistry. 1974;13:222–245. pmid:4358940
  36. 36.
    Horikawa DD, Kunieda T, Abe W, Watanabe M, Nakahara Y, Yukuhiro F, et al. Institution of a rearing system of the extremotolerant tardigrade Ramazzottius varieornatus: A brand new mannequin animal for astrobiology. Astrobiology. 2008;8:549–556. pmid:18554084
  37. 37.
    Yagi-Utsumi M, Aoki Ok, Watanabe H, Tune C, Nishimura S, Satoh T, et al. Desiccation-induced fibrous condensation of CAHS protein from an anhydrobiotic tardigrade. Sci Rep. 2021;11:21328. pmid:34737320
  38. 38.
    Malki AA, Teulon J, Zarco AC, Chen W, Adamski W, Maurin D, et al. Intrinsically Disordered Tardigrade Proteins Self-Assemble into Fibrous Gels in Response to Environmental Stress. Angew Chem Int Ed Engl. 2022;61:e202109961. pmid:34750927
  39. 39.
    Harmon TS, Holehouse AS, Rosen MK, Pappu RV. Intrinsically disordered linkers decide the interaction between section separation and gelation in multivalent proteins. Elife. 2017;6:e30294. pmid:29091028
  40. 40.
    Jumper J, Evans R, Pritzel A, Inexperienced T, Figurnov M, Ronneberger O, et al. Extremely correct protein construction prediction with AlphaFold. Nature. 2021;596:583–589. pmid:34265844
  41. 41.
    Mirdita M, Schütze Ok, Moriwaki Y, Heo L, Ovchinnikov S, Steinegger M. ColabFold: making protein folding accessible to all. Nat Strategies. 2022. pmid:35637307
  42. 42.
    Protter DSW, Parker R. Ideas and properties of stress granules. Tendencies Cell Biol. 2016;26:668–679. pmid:27289443
  43. 43.
    Franzmann TM, Alberti S. Protein section separation as a stress survival technique. Chilly Spring Harb Perspect Med. 2019;9:a034058. pmid:30617047
  44. 44.
    LeBlanc BM, Hand SC. Goal enzymes are stabilized by AfrLEA6 and a acquire of α-helix coincides with safety by a bunch 3 LEA protein throughout incremental drying. Biochim Biophys Acta–Proteins. Proteomics. 2021;1869:140642. pmid:33647452
  45. 45.
    Shiraki Ok, Nishikawa Ok, Goto Y. Trifluoroethanol-induced stabilization of the alpha-helical construction of beta-lactoglobulin: implication for non-hierarchical protein folding. J Mol Biol. 1995;245:180–194. pmid:7799434
  46. 46.
    Vincenzi M, Mercurio FA, Leone M. About TFE: Outdated and new findings. Curr Protein Pept Sci. 2019;20:425–451. pmid:30767740
  47. 47.
    Tsuboyama Ok, Osaki T, Matsuura-Suzuki E, Kozuka-Hata H, Okada Y, Oyama M, et al. A widespread household of heat-resistant obscure (Hero) proteins defend in opposition to protein instability and aggregation. PLoS Biol. 2020;18:e3000632. pmid:32163402
  48. 48.
    Mészáros B, Erdös G, Dosztányi Z. IUPred2A: Context-dependent prediction of protein dysfunction as a operate of redox state and protein binding. Nucleic Acids Res. 2018;46:W329–37. pmid:29860432
  49. 49.
    Bailey T, Elkan C. Becoming a mix mannequin by expectation maximization to find motifs in biopolymers. Proc Int Conf Intell Syst Mol Biol. 1994;2:28–36. pmid:7584402
  50. 50.
    Drozdetskiy A, Cole C, Procter J, Barton GJ. JPred4: A protein secondary construction prediction server. Nucleic Acids Res. 2015;43:W389–W394. pmid:25883141
  51. 51.
    Lupas A, Van Dyke M, Inventory J. Predicting coiled coils from protein sequences. Science. 1991;252:1162–1164. pmid:2031185
  52. 52.
    Pettersen EF, Goddard TD, Huang CC, Meng EC, Sofa GS, Croll TI, et al. UCSF ChimeraX: Construction visualization for researchers, educators, and builders. Protein Sci. 2021;30:70–82. pmid:32881101
  53. 53.
    Mi H, Ebert D, Muruganujan A, Mills C, Albou LP, Mushayamaha T, et al. PANTHER model 16: A revised household classification, tree-based classification software, enhancer areas and intensive API. Nucleic Acids Res. 2021;49:D394–D403. pmid:33290554
  54. 54.
    Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape gives a biologist-oriented useful resource for the evaluation of systems-level datasets. Nat Commun. 2019;10:1523. pmid:30944313
  55. 55.
    Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol. 2019;37:907–915. pmid:31375807
  56. 56.
    Liao Y, Smyth GK, Shi W. FeatureCounts: An environment friendly normal function program for assigning sequence reads to genomic options. Bioinformatics. 2014;30:923–930. pmid:24227677
  57. 57.
    Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq information with DESeq2. Genome Biol. 2014;15:550. pmid:25516281
  58. 58.
    Bindels DS, Haarbosch L, Van Weeren L, Postma M, Wiese KE, Mastop M, et al. MScarlet: A shiny monomeric pink fluorescent protein for mobile imaging. Nat Strategies. 2016;14:53–56. pmid:27869816
  59. 59.
    Sama RRK, Ward CL, Kaushansky LJ, Lemay N, Ishigaki S, Urano F, et al. FUS/TLS assembles into stress granules and is a prosurvival issue throughout hyperosmolar stress. J Cell Physiol. 2013;228:2222–2231. pmid:23625794
  60. 60.
    Tanaka S, Tanaka J, Miwa Y, Horikawa DD, Katayama T, Arakawa Ok, et al. Novel mitochondria-targeted heat-soluble proteins recognized within the anhydrobiotic tardigrade enhance osmotic tolerance of human cells. PLoS ONE. 2015;10:e0118272. pmid:25675104
  61. 61.
    Sakai A, Murayama Y, Fujiwara Ok, Fujisawa T, Sasaki S, Kidoaki S, et al. Rising elasticity via adjustments within the secondary construction of gelatin by gelation in a microsized lipid house. ACS Cent Sci. 2018;4:477–483. pmid:29721530
  62. 62.
    González M, Martín-Ruíz I, Jiménez S, Pirone L, Barrio R, Sutherland JD. Technology of steady Drosophila cell traces utilizing multicistronic vectors. Sci Rep. 2011;1:75. pmid:22355594
  63. 63.
    Bullard TA, Borg TK, Value RL. The Expression and Position of Protein Kinase C in Neonatal Cardiac Myocyte Attachment, Cell Quantity, and Myofibril Formation Is Depending on the Composition of the Extracellular Matrix. Microsc Microanal. 2005;11:224–234. pmid:16060975
RELATED ARTICLES

LEAVE A REPLY

Please enter your comment!
Please enter your name here

Most Popular

Recent Comments