Next Article in Journal
Molecular Characterization of Dehydrin in Azraq Saltbush among Related Atriplex Species
Previous Article in Journal
Computational Screening of Approved Drugs for Inhibition of the Antibiotic Resistance Gene mecA in Methicillin-Resistant Staphylococcus aureus (MRSA) Strains
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biomolecular Liquid–Liquid Phase Separation for Biotechnology

Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Hyogo, Japan
*
Author to whom correspondence should be addressed.
BioTech 2023, 12(2), 26; https://doi.org/10.3390/biotech12020026
Submission received: 9 March 2023 / Revised: 28 March 2023 / Accepted: 30 March 2023 / Published: 1 April 2023

Abstract

:
The liquid–liquid phase separation (LLPS) of biomolecules induces condensed assemblies called liquid droplets or membrane-less organelles. In contrast to organelles with lipid membrane barriers, the liquid droplets induced by LLPS do not have distinct barriers (lipid bilayer). Biomolecular LLPS in cells has attracted considerable attention in broad research fields from cellular biology to soft matter physics. The physical and chemical properties of LLPS exert a variety of functions in living cells: activating and deactivating biomolecules involving enzymes; controlling the localization, condensation, and concentration of biomolecules; the filtration and purification of biomolecules; and sensing environmental factors for fast, adaptive, and reversible responses. The versatility of LLPS plays an essential role in various biological processes, such as controlling the central dogma and the onset mechanism of pathological diseases. Moreover, biomolecular LLPS could be critical for developing new biotechnologies such as the condensation, purification, and activation of a series of biomolecules. In this review article, we introduce some fundamental aspects and recent progress of biomolecular LLPS in living cells and test tubes. Then, we discuss applications of biomolecular LLPS toward biotechnologies.
Key Contribution: This review introduces insights into the underlying principles governing the phase behavior of biomolecules and provides comprehensive lists of biomolecular liquid–liquid phase separation which has been garnering attention in broad research fields. This review also highlights its potential applications in biotechnology, including drug delivery, biomolecular purification, biocatalysts, drug protection, and microgel formation.

1. Introduction

A living cell is made up of different kinds of organelles. Some of these are covered by membranes and some of these are membrane-less [1,2]. Almost one century ago, British scientist J. Haldane (1929) and Russian biochemist A. Oparin (1965) independently proposed the first genesis of life theory. Both theories, incidentally, describe the original concept of prebiotic compartmentalization. According to Oparin, the earliest polymers that resembled proteins and carbohydrates would form into colloidal particles, and these particles would subsequently evolve a rudimentary metabolism resembling fermentation. He postulated that after other nutrients became rare, the colloidal particles would start to develop photosynthesis and absorb the available resources through fermentation. These particles, known as “coacervates”, are organic-rich droplets produced by LLPS [3]. Despite such great proposals for the origin of the prebiotic compartmentalization with the coacervates, the membranelles organelles in living cells have very recently been discovered compared with the membrane organelles [4]. Examples of some membrane-less organelles include the stress granule, Nucleolus, P-body, U-body, Gem granule, Cajal body, and Histone locus body (Figure 1) [5]. These are formed through the LLPS of biomolecules such as proteins and nucleic acids [6].
LLPS can be useful in controlling (activation and suppression) many important cellular reactions because it can significantly increase the local concentration of the participating molecule in the cellular reaction [7,8]. For example, LLPS is important for a variety of biological processes, including ribosomal biogenesis [9], transcription [10], cell signaling [11], stress response [12], cytoskeletal regulation [13], cell polarization [14], cytoplasmic branching [15], nucleolus formation [16], miRISC assembly [17], innate immune signaling [18], stress granule assembly [19], and autophagy [20]. Furthermore, LLPS has a significant role in various diseases such as cancer [21], Alzheimer’s disease [22], Parkinson’s disease [22], Huntington’s disease, and amyotrophic lateral sclerosis [23,24,25]. Importantly, recent studies have demonstrated that biomolecular LLPS can also be imposed in various applied fields of biotechnology, synthetic biology, food technology, environmental engineering, etc. In this review, we discuss the recent trends in biomolecular LLPS research and its application.

2. Principles of Biomolecular LLPS

A multi-component system, such as cellular cytoplasm, can exist as a homogenous and well-mixed mixture or a soup of separate phases, depending on the interactions of the constituent molecules, including the solvent that is water in the case of living cells [3]. There are three types of LLPS that can commonly be distinguished (Figure 2a) [26]. In a simple LLPS, a single molecular attractive interaction proceeds that generate coactivates depending on molecular environmental factors, such as temperature, pH, and salt concentration. Many proteins with disordered regions have been found to undergo simple LLPS, driven by a combination of electrostatic interaction and other weak interactions such as hydrogen bonding, π–π stacking, cation–π, and dipole–dipole interactions (Figure 2b) [26]. In associative LLPS, two soluble molecules end up in the same phase, due to attractive interactions between them. This condensed phase is called a droplet; it is enriched in both solutes, but still contains a significant amount of solvent. A classic example of associative LLPS is that of two oppositely charged polymers, such as alginate (a negatively charged polysaccharide) and gelatin (a positively charged protein) (Figure 3a) [26]. In segregative LLPS, two soluble molecules (e.g., peptides, polymers, and nucleotides) do not mix despite a favorable mixing entropy due to repulsive interactions between them. As a result, they end up in two separate phases, each enriched in one of the solutes. A classic example of segregative LLPS is that of poly(ethylene glycol) and dextran (Figure 3b) [26].
Proteins are the primary cause of biomolecular LLPS. Notably, several multi-domain protein systems display LLPS behavior [27,28,29]. As shown in Figure 3c, the so-called stickers-and-spacers approach can be used to conceptually examine multi-domain proteins and intrinsically disordered proteins (IDPs) [29,30]. IDPs have less defined three-dimensional structures in physiological conditions [31]. It was discovered that significant amounts of intrinsically disordered regions (IDRs) can be found in many LLPS systems discovered in living cells [32]. The stickers-and-spacers model divides the target protein into two regions: molecular fragments responsible for chain–chain interactions (stickers), and the remainder of the molecule, which is not involved in the interaction (spacer). Although spacers are suggested to modify chain characteristics, they have a smaller impact on chain–chain interactions than stickers do. Multi-domain proteins can easily undergo LLPS because interacting domains serve as stickers and disordered linkers serve as spacers. Recently, scientists have discovered that divalent ions have some important role in the formation of biomolecular droplets [33]. Zinc ions, which greatly increase the tendency for tau to undergo LLPS by reducing the critical concentration of protein, were found to influence the LLPS of tau protein [34]. Both directly and through interactions with other proteins, divalent cations can influence phase transitions. Recent studies have shown that the EF-hand domain protein (EFhd2) directly affects tau’s liquid phase behavior to form solid-like structures in vitro. This modification is caused by calcium ions [35].

3. Biomolecular Droplets and Their Functions

The number of biomolecular droplets produced via LLPS is rapidly growing, and their biological functions have been identified [36,37]. In this section, we present biomolecular droplets which occur naturally via LLPS, as well as artificial biomolecules which undergo LLPS to form droplets.

3.1. Droplets in Cytoplasm

Biomolecular droplets which are present in the cytoplasm have been identified. Stress granules are a typical example and are the most extensively studied in the cytoplasmic droplets. Stress granules are membrane-less organelles, ranging in size from 0.1 to 2 μm [38]. The essential components for stress granule formation are T-cell-restricted intracellular antigen-1 (TIA-1) and Ras-GTPase-activating protein SH3-domain-binding protein 1 (G3BP1) and RNAs. The primary function of stress granules is to promote cell survival by condensing translationally stalled mRNAs, ribosomal components, translation initiation factors, and RNA-binding proteins (RBPs). On the other hand, certain transcripts such as heat shock protein 70 are excluded from stress granules which are selectively translated under the stress conditions [39]. Therefore, stress granules can control protein expression (translation) through the inclusion and exclusion of certain mRNAs in response to unfavorable conditions for cells. Stress granules are formed under acute stress conditions such as hypoxia, oxidative stress, osmotic stress, and temperature change [40]. The timescale of the disassembly of stress granules varies depending on the stress factors. For example, cold-shock-induced stress granules disassemble within minutes after returning to normal temperature [41]. On the other hand, recovery after arsenate stress, H2O2 treatment, osmotic stress, or heat shock occurs between 60 and 120 min [42]. In addition to the recovery time varies, this range of time is much shorter than the gene expression response. Therefore, LLPS including the assembly of stress granules is critical for promptly controlling cellular functions to protect cells from death under adverse conditions. Moreover, stress granules under stress conditions alter nuclear events, providing a linkage between the nuclear and the cytoplasmic processes [41]. Stress granules also respond to diseases such as viral infections and cancer [43]. Stress granules are further recognized as potential precursors of pathological aggregates in neurodegenerative diseases [44]. The position, function, chemical composition, and detection technique of cytoplasmic droplets are briefly listed in Table 1 [45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63].

3.2. Droplets in Nucleus

The interior of a cell nucleus is a complex environment: a crowded mixture of biomolecules, including very long DNA strands in the form of chromatin with histone proteins, mRNAs that are newly transcribed, other RNAs for controlling gene expressions, and proteins for transcription and other processes. A wide variety of droplets are required to proceed biologically critical reactions under the complex environment.
One of the most well-known cellular droplets is the nucleolus, which, in the 1830s, was the first membrane-less component to be identified [5]. The number (usually 2–5 per cell) and size of the nucleoli depend on the cell type, cell cycle phase, and metabolic conditions. The nucleolus provides a site for the transcription of ribosomal RNA from ribosomal DNA and ribosome assembly for ribosome biogenesis. The nucleolus also serves other processes, such as maintaining cell homeostasis [64]. Recently, new roles of the nucleoli have attracted attention: as stress granules, the nucleoli act as sensors and regulators for cellular stresses such as RNA polymerase I inhibitors, prevalent cytotoxic agents, viral proteins, UV radiation, heat shock, and DNA damage, apoptosis, and senescence [65].
A nucleolus contains several functional modules, each constituting three sub-compartments or layers. From the inner to the periphery, the three layers are the fibrillar center, the dense fibrillar component, and the granular component, responsible for different steps of ribosomal biogenesis. The nucleolus is composed of hundreds of copies of ribosomal genes, newly synthesized ribosomal RNA (rRNA), ribosomal proteins, and ribonucleoproteins. Other droplets found in the nucleus are listed in Table 2 [8,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92].

3.3. Droplets in Membranes

Although they do not occur via LLPS, in cell membranes, biomolecular droplets are also produced, such as membrane clusters, as listed in Table 3 [13,28,93,94,95,96,97,98,99,100,101,102,103,104,105]. A membrane cluster is a lipid droplet consisting of triacylglycerols, phospholipids, sphingolipids, cholesterol, and proteins [106]. These clusters play important roles in various cellular processes, including signaling, and the transport of cell take and cell release material such as lipids, amino acids, ions water, and hormones, amines, and peptides [93]. The membrane cluster has a significant role not only in the uptake of lipids, but also in the distribution and storage of lipids. A representative example of a membrane cluster is the photosystem II (PSII) complex, which is involved in the light-dependent reactions of photosynthesis in plants, algae, and some bacteria [107]. PSII is a large and complex protein complex that contains over 20 different subunits, most of which are membrane-bound. It consists of a core antenna complex that captures light energy, a reaction center that uses this energy to split water into oxygen, and electron carriers that transfer the electrons to other components of the photosynthetic system [108]. The formation of PSII clusters is essential for their proper functioning. PSII clusters help to organize the various components and to create a favorable environment for the transport of electrons [109].

3.4. Enzymes and Transcription Factors Undergoing LLPS

Recent studies suggest that some enzymes show different activity inside droplets. For example, Saini et al. recently discovered that macromolecular crowding induces LLPS, which leads to an increase in the intrinsic catalytic efficiencies of horseradish peroxidase (HRP) and glucose oxidase (GOx) [110]. Transcription factors (TFs) and RNAs also induce the formation of transcriptional condensates via LLPS, which contain clusters of multiple enhancers (super-enhancers) [111]. This phenomenon is supported by the dynamic interaction of TFs with RNA polymerase II (Pol II) clusters [112]. To form transcriptional condensates, TFs bind to various cis-regulatory DNA elements (e.g., promoters and enhancers) and stimulate the transcription of active genes in proximity, facilitating the precise control of gene expression [113]. Other examples of enzymes and transcription factors which undergo LLPS are listed in Table 4 [110,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138].

3.5. Droplets Discovered in Various Biological Processes

The list of biomolecular condensates is increasing rapidly. For example, rubisco (pyrenoids) plays a crucial role in photosynthesis acceleration and in carbon fixation [139,140,141]. Another interesting droplet recently found is the Wnt droplet [142]. The Wnt droplet consists of proteins such as kinase that regulate β-catenin stability. Wnt droplets play a vital role in stem cell differentiation. These findings demonstrate that LLPS is pivotal and versatile not only in controlling the central dogma, but also in various biological processes. Therefore, it is considerable that LLPS is one of the fundamental characteristics of biomolecules. The location, name, component, biological role, and observation procedure of droplets discovered in various biological processes are listed in Table 5 [139,140,141,142,143,144,145,146].

3.6. Artificial Droplet System

Artificial and model droplet systems are gaining popularity. Artificial and model droplet systems have various uses because of their controllable size, concentration inside the droplet, and the component of the droplet. Researchers are focusing on the development of new artificial droplets as well as artificial systems. Artificial cells are simplified models of living cells for investigations of the molecular basis of life. Artificial cells are generally constructed using a water-in-oil (W/O) microdroplet. Water in an oil microdroplet is a micrometer-sized water droplet dispersed in an immiscible oil phase [147]. Another artificial droplet system that gains immense popularity is the droplet reactor system. Droplet reactor systems have considerable biochemical applications such as single-cell analysis, kinetic study, and controlled drug release [148]. Another interesting example of an artificial droplet system is DNA nanostructures. DNA nanostructures were employed by Sato and Takinoue to induce LLPS [149]. The DNA nanostructures localize at the oil–water interface when they are added to the oil–water system. Different two-dimensional phase separation patterns could be induced depending on the DNA sequences. Hydrogels were created as a result of the DNA nanostructures’ phase separation [149].
Recently, a novel class of short peptide derivatives that undergo LLPS has been created [150]. The peptide is made up of phenylalanine dipeptides joined by hydrophilic spacers (cystamine moiety). Disulfide bonds formed among spacers enable redox-chemistry-based dynamic regulation of the assembly. Additionally, researchers might functionalize the coacervates to act as a catalyst in the aldol and hydrazone production reaction [150]. Other examples of model droplet systems are given in Table 6 [151,152,153,154,155].

4. Biomolecular LLPS towards Biotechnology

Due to its special characteristics, such as the differential partitioning and compartmentalization of biomolecules, biodegradability, and biocompatibility of biomaterials, LLPS has been used in biotechnological and biomedical processes [156,157,158,159]. Some examples of biomolecular LLPS applications in biotechnology and the biomedical process are given in Table 7 and schematically shown in Figure 4. The chemistry of the three different LLPS systems, simple phase separation, segregation, and associative phase separation, varies from one to the other. In the case of simple phase separation, separation happens based on the density and other physical properties. For segregation phase separation, separation happens based on the size, shape, or charge of the components. In the case of associative phase separation, separation happens based on specific molecular interactions. The affinities for different chemical compounds to participate in phase-separated droplets are strongly influenced by the chemical component and the nature of both the LLPS system’s constituent parts and the chemicals. To account for this, each molecule and LLPS system pair has a “partition coefficient”, which is a parameter of how likely a molecule is to segregate into a droplet in comparison to the surrounding solution. The partition coefficient is more precisely defined as the ratio of the concentration of the molecule outside the droplet to the molecule inside the droplet in an equilibrium state. The partition coefficients of numerous compounds in phase-separated systems have been extensively studied [160,161,162,163,164,165,166]. Bioprocess researchers and engineers have made use of the differential partition coefficients of molecules in diverse LLPS systems as a method for selective biomolecule extraction and purification, which are processes frequently used in commercial biomolecule productions [167]. Recent discoveries demonstrated that biomacromolecules, such as nucleic acids, can be purified based on their phase separation behavior. It was discovered that an ATPS system (PEG-phosphate) in the purification of plasmid DNA from pure E. coli lysate had a yield of 80–85% of plasmid DNA [168]. Using a similar system, it was possible to separate plasmid DNA from RNA (from a mixture that solely contained plasmid DNA and RNA) with yields of 89% (plasmid DNA) and 70% (RNA), depending on pH [169].
It was demonstrated that LLPS is not only very useful for biomolecule separation and purification systems, but also for drug delivery systems. Small-molecule drugs, many of which are not easily soluble in aqueous solution, have been compartmentalized using phase-separated droplets [3,170,171]. An additional good reason to use LLPS for drug delivery is that it offers drug protection from biodegradations. Sodium-alginate beads combined with a coacervate system are used in the encapsulation of small-molecule drugs to protect the drug against low pH levels generated by stomach fluids, allowing the drug to be released further downstream in the digestive tract [162]. Such an application of LLPS could be used to prevent drugs from degrading before they reach their destination (target cells) and could improve the efficacy of the drug formulation without altering the chemical structure of the molecule [172]. Moreover, coacervates can provide encapsulated proteins with protection from temperature, pH, and even denaturants such as urea [173]. For example, Nojima et al. produced phase-separated protein condensate with a high concentration of over 600 distinct proteins while maintaining their natural structure using ionic surfactants [174,175]. Due to the protection ability of LLPS from biodegradation, drug delivery systems based on the LLPS of artificial polymers have been reported. Polyester microspheres are an example of such an artificial polymer system which can be charged with a variety of small-molecule medicines [176,177]. Other examples of artificial polymer systems which demonstrate LLPS are polylactate, polyglycolate, poly(lactate/glycolate) co-polymers, and other aliphatic polyesters [178,179,180]. These polymers are composed of simple aliphatic residues, and a straightforward hydrolysis mechanism allows them to break down into their monomer species (lactic acid and/or glycolic acid). The biocompatibility and biosafety of such polymers are suggested by the fact that lactic acid can be safely degraded by a number of natural processes, including neuron metabolism, glucogenesis in the liver, oxidation to pyruvate in muscle cells, or excretion through urine or breath as carbon dioxide [181,182]. Current research is looking at new ways to control the rate of degradation, such as the addition of other polymers [183], the development of novel polyester materials, such as foams [184], or the engineering of polyesters that may selectively degrade in response to specific external stimuli [185]. On the same note, currently, we have seen a substantial increase in therapeutic protein therapies with LLPS. One such demonstration even delivered functional myoglobin to human stem cells using amylose-based coacervates [186]. When more effective protein therapies are developed, the usage of phase-separated protein complexes may become even more prevalent [187]. Some recent research showed that large biomacromolecules, such as antibodies, can be delivered into the cytosol through coacervation using a peptide. T. Iwata et al. discovered that a mixture of Alexa488-IgG and FcB(L17E)3 formed liquid droplets; the hydrophobicity and basicity of the Alexa488-IgG and FcB(L17E)3 segment are important for liquid droplet formation [188]. These findings open a new horizon for small drugs and protein delivery inside cells. In recent years, scientists have utilizing the LLPS system for personalized genetic medicine treatments, such as DNA or RNA medicine, including miRNA and siRNA therapy [189,190,191,192,193]. Currently, mRNA vaccines against SARS-CoV-2 use lipid nanoparticles [194,195], and early LLPS systems have demonstrated the capacity to concentrate or shelter nucleic acids [119]. A more recent study has started to concentrate on the stabilization of encapsulated viruses (as vaccine vectors) in coacervates [196]. Future studies might be interestingly directed toward using LLPS systems as genetic carriers in RNA or DNA vaccines. Despite having all these applications, biomolecular LLPS also have other application in diverse field of biotechnology. LLPS plays an important role in material science. Recently, scientists have discovered that, by applying LLPS systems we can form new materials such as microgels [197]. Biomolecular LLPS systems have potential applications in environmental science. In a recent study, Zhou et al. efficiently removed organic pollutant from water using poly-lipoic ester base coactivates [198]. Another example of the application of LLPS systems in environmental science is the removal of phosphate from wastewater; environmental scientists have used microorganisms to extract phosphorus from wastewater by taking advantage of their proclivity to incorporate exogenous phosphates into internal phase-separated organelles [199]. In addition to these applications, biomolecular LLPS systems have applications in the food industry. One of the most common uses of LLPS is to create desirable textures in food products. LLPS causes the production of W/W emulsions containing biopolymers dispersed disproportionately between separated liquid phases [200]. Another use of LLPS systems in the food industry is the encapsulation of ingredients [201]. LLPS based on carbohydrates or proteins is often utilized to encapsulate active substances within the dispersion phase. In food items, encapsulation serves several functions, including hiding undesirable odors, stabilizing reactive substances, and managing the release of active compounds [202].
Table 7. Biomolecular LLPS systems and their application in biotechnology.
Table 7. Biomolecular LLPS systems and their application in biotechnology.
Biomolecular LLPS SystemApplication in BiotechnologyRef.
ATPS system (PEG-phosphate)Purification and extraction of biomolecules (DNA, RNA)[168,169]
Sodium-alginate beads combined coacervate systemDrug protection[162,173,174,175,176]
Polyester microspheres and artificial polymer systemDrug delivery[177,178,179,180,181,182,183,184,185,186]
Amylose-based coacervatesProtein delivery[187,188,189]
Lipid nanoparticlesDNA, RNA vaccines[190,191,192,193,194,195,196,197]
Poly-lipoic ester base coactivatesOrganic pollutant remover[199]
LLPS system from the gel–sol transition of protein (gelatin solution) in a macromolecular crowding agent (PEG solution).New material synthesis (protein microgel)[198]
Carbohydrates and proteins-based LLPS system Encapsulation of active substances (food industry)[201,202]

5. Conclusions and Perspectives

The importance of biomolecular LLPS and its roles in diverse biological processes is evident, as well as its involvement in a series of diseases. This review has provided an overview of biomolecular LLPS systems across various applied fields of biology, including biotechnology and synthetic biology. The unique properties of LLPS systems, such as their ability to concentrate biomolecules, organize biochemical reactions, and generate membrane-less organelles, make them ideal candidates for a wide range of applications, including drug delivery, enzyme immobilization, and biocatalysis. However, there are still many challenges to be addressed for practical applications of LLPS in biotechnology, such as controlling the size and stability of the resulting droplets, optimizing the conditions for LLPS, and scaling up production. More rational design and controlling procedures of LLPS are highly required. Another area of future research is the integration of LLPS systems into more complex biotechnological processes, such as bioreactors or biosensors, because the selective enrichment of molecules inside droplets could be useful as a pretreatment of reactors and sensors. Additionally, the development of high-throughput screening methods based on biomolecular LLPS could enable the discovery of new functional molecules involving small- to large-sized drugs targeting biomolecular droplets. Such drugs controlling the LLPS of biomolecules will be a new modality for broad spectrums of diseases. Overall, the potential of biomolecular LLPS for biotechnology is vast, and continued research in this field is likely to lead to significant advances in the coming years.

Author Contributions

S.S., M.T., K.K. and D.M. conceptualized the manuscript’s content; S.S. and M.T. were involved in data collection and preparation of the first draft; K.K. and D.M. performed the editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by JSPS (KAKENHI Grant No. 21H02062, 20K21259, and 17H06351), and 22J21167 (Grant-in-Aid for JSPS Fellows); A Research Grant of the Asahi Glass Foundation, Japan; and the Hirao Taro Foundation of Konan Gakuen for Academic Research, Japan.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Harmon, T.S.; Holehouse, A.S.; Rosen, M.K.; Pappu, R.V. Intrinsically disordered linkers determine the interplay between phase separation and gelation in multivalent proteins. eLife 2017, 6, e30294. [Google Scholar] [CrossRef]
  2. Kim, C.G.; Hwang, D.E.; Kumar, R.; Chung, M.; Eom, Y.G.; Kim, H.; Koo, D.H.; Choi, J.M. Recent trends in studies of biomolecular phase separation. BMB Rep. 2022, 55, 363–369. [Google Scholar] [CrossRef] [PubMed]
  3. Jia, T.Z.; Wang, P.H.; Niwa, T.; Mamajanov, I. Connecting primitive phase separation to biotechnology, synthetic biology, and engineering. J. Biosci. 2021, 46, 79. [Google Scholar] [CrossRef] [PubMed]
  4. Shin, Y.; Brangwynne, C.P. Liquid phase condensation in cell physiology and disease. Science 2017, 357, eaaf4382. [Google Scholar] [CrossRef] [Green Version]
  5. Gomes, E.; Shorter, J. The molecular language of membraneless organelles. J. Biol. Chem. 2019, 294, 7115–7127. [Google Scholar] [CrossRef] [Green Version]
  6. Ryu, J.K.; Hwang, D.E.; Choi, J.M. Current Understanding of Molecular Phase Separation in Chromosomes. Int. J. Mol. Sci. 2021, 22, 10736. [Google Scholar] [CrossRef] [PubMed]
  7. Banani, S.F.; Rice, A.M.; Peeples, W.B.; Lin, Y.; Jain, S.; Parker, R.; Rosen, M.K. Compositional Control of Phase-Separated Cellular Bodies. Cell 2016, 166, 651–663. [Google Scholar] [CrossRef] [Green Version]
  8. Feric, M.; Vaidya, N.; Harmon, T.S.; Mitrea, D.M.; Zhu, L.; Richardson, T.M.; Kriwacki, R.W.; Pappu, R.V.; Brangwynne, C.P. Coexisting Liquid Phases Underlie Nucleolar Subcompartments. Cell 2016, 165, 1686–1697. [Google Scholar] [CrossRef] [Green Version]
  9. Peng, P.H.; Hsu, K.W.; Wu, K.J. Liquid-liquid phase separation (LLPS) in cellular physiology and tumor biology. Am. J. Cancer Res. 2021, 11, 3766–3776. [Google Scholar]
  10. Cho, W.K.; Spille, J.H.; Hecht, M.; Lee, C.; Li, C.; Grube, V.; Cisse, I.I. Mediator and RNA polymerase II clusters associate in transcription-dependent condensates. Science 2018, 361, 412–415. [Google Scholar] [CrossRef] [Green Version]
  11. Su, Q.; Mehta, S.; Zhang, J. Liquid-liquid phase separation: Orchestrating cell signaling through time and space. Mol. Cell 2021, 81, 4137–4146. [Google Scholar] [CrossRef]
  12. Kroschwald, S.; Alberti, S. Gel or Die: Phase Separation as a Survival Strategy. Cell 2017, 168, 947–948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Banjade, S.; Rosen, M.K. Phase transitions of multivalent proteins can promote clustering of membrane receptors. eLife 2014, 3, e04123. [Google Scholar] [CrossRef] [PubMed]
  14. Riback, J.A.; Katanski, C.D.; Kear-Scott, J.L.; Pilipenko, E.V.; Rojek, A.E.; Sosnick, T.R.; Drummond, D.A. Stress-Triggered Phase Separation Is an Adaptive, Evolutionarily Tuned Response. Cell 2017, 168, 1028–1040.e1019. [Google Scholar] [CrossRef] [Green Version]
  15. Molliex, A.; Temirov, J.; Lee, J.; Coughlin, M.; Kanagaraj, A.P.; Kim, H.J.; Mittag, T.; Taylor, J.P. Phase separation by low complexity domains promotes stress granule assembly and drives pathological fibrillization. Cell 2015, 163, 123–133. [Google Scholar] [CrossRef] [Green Version]
  16. Nicolas, E.; Parisot, P.; Pinto-Monteiro, C.; de Walque, R.; De Vleeschouwer, C.; Lafontaine, D.L. Involvement of human ribosomal proteins in nucleolar structure and p53-dependent nucleolar stress. Nat. Commun. 2016, 7, 11390. [Google Scholar] [CrossRef] [Green Version]
  17. Sheu-Gruttadauria, J.; MacRae, I.J. Phase Transitions in the Assembly and Function of Human miRISC. Cell 2018, 173, 946–957.e916. [Google Scholar] [CrossRef]
  18. Xiao, Q.; McAtee, C.K.; Su, X. Phase separation in immune signalling. Nat. Rev. Immunol. 2022, 22, 188–199. [Google Scholar] [CrossRef] [PubMed]
  19. Wippich, F.; Bodenmiller, B.; Trajkovska, M.G.; Wanka, S.; Aebersold, R.; Pelkmans, L. Dual specificity kinase DYRK3 couples stress granule condensation/dissolution to mTORC1 signaling. Cell 2013, 152, 791–805. [Google Scholar] [CrossRef] [Green Version]
  20. Fujioka, Y.; Alam, J.M.; Noshiro, D.; Mouri, K.; Ando, T.; Okada, Y.; May, A.I.; Knorr, R.L.; Suzuki, K.; Ohsumi, Y.; et al. Phase separation organizes the site of autophagosome formation. Nature 2020, 578, 301–305. [Google Scholar] [CrossRef]
  21. Tong, X.; Tang, R.; Xu, J.; Wang, W.; Zhao, Y.; Yu, X.; Shi, S. Liquid-liquid phase separation in tumor biology. Signal. Transduct. Target. Ther. 2022, 7, 221. [Google Scholar] [CrossRef] [PubMed]
  22. Sawner, A.S.; Ray, S.; Yadav, P.; Mukherjee, S.; Panigrahi, R.; Poudyal, M.; Patel, K.; Ghosh, D.; Kummerant, E.; Kumar, A.; et al. Modulating α-Synuclein Liquid-Liquid Phase Separation. Biochemistry 2021, 60, 3676–3696. [Google Scholar] [CrossRef] [PubMed]
  23. Boyko, S.; Surewicz, W.K. Tau liquid-liquid phase separation in neurodegenerative diseases. Trends Cell Biol. 2022, 32, 611–623. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, B.; Zhang, L.; Dai, T.; Qin, Z.; Lu, H.; Zhang, L.; Zhou, F. Liquid-liquid phase separation in human health and diseases. Signal. Transduct. Target. Ther. 2021, 6, 290. [Google Scholar] [CrossRef]
  25. Alberti, S.; Dormann, D. Liquid-Liquid Phase Separation in Disease. Annu. Rev. Genet. 2019, 53, 171–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Abbas, M.; Lipiński, W.P.; Wang, J.; Spruijt, E. Peptide-based coacervates as biomimetic protocells. Chem. Soc. Rev. 2021, 50, 3690–3705. [Google Scholar] [CrossRef]
  27. Espinosa, J.R.; Joseph, J.A.; Sanchez-Burgos, I.; Garaizar, A.; Frenkel, D.; Collepardo-Guevara, R. Liquid network connectivity regulates the stability and composition of biomolecular condensates with many components. Proc. Natl. Acad. Sci. USA 2020, 117, 13238–13247. [Google Scholar] [CrossRef]
  28. Li, P.; Banjade, S.; Cheng, H.C.; Kim, S.; Chen, B.; Guo, L.; Llaguno, M.; Hollingsworth, J.V.; King, D.S.; Banani, S.F.; et al. Phase transitions in the assembly of multivalent signalling proteins. Nature 2012, 483, 336–340. [Google Scholar] [CrossRef] [Green Version]
  29. Freeman Rosenzweig, E.S.; Xu, B.; Kuhn Cuellar, L.; Martinez-Sanchez, A.; Schaffer, M.; Strauss, M.; Cartwright, H.N.; Ronceray, P.; Plitzko, J.M.; Förster, F.; et al. The Eukaryotic CO(2)-Concentrating Organelle Is Liquid-like and Exhibits Dynamic Reorganization. Cell 2017, 171, 148–162.e119. [Google Scholar] [CrossRef] [Green Version]
  30. Roden, C.; Gladfelter, A.S. RNA contributions to the form and function of biomolecular condensates. Nat. Rev. Mol. Cell Biol. 2021, 22, 183–195. [Google Scholar] [CrossRef]
  31. Choi, J.M.; Holehouse, A.S.; Pappu, R.V. Physical Principles Underlying the Complex Biology of Intracellular Phase Transitions. Annu. Rev. Biophys. 2020, 49, 107–133. [Google Scholar] [CrossRef] [Green Version]
  32. Hong, K.; Song, D.; Jung, Y. Behavior control of membrane-less protein liquid condensates with metal ion-induced phase separation. Nat. Commun. 2020, 11, 5554. [Google Scholar] [CrossRef]
  33. Onuchic, P.L.; Milin, A.N.; Alshareedah, I.; Deniz, A.A.; Banerjee, P.R. Divalent cations can control a switch-like behavior in heterotypic and homotypic RNA coacervates. Sci. Rep. 2019, 9, 12161. [Google Scholar] [CrossRef] [Green Version]
  34. Singh, V.; Xu, L.; Boyko, S.; Surewicz, K.; Surewicz, W.K. Zinc promotes liquid-liquid phase separation of tau protein. J. Biol. Chem. 2020, 295, 5850–5856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Vega, I.E.; Umstead, A.; Kanaan, N.M. EFhd2 Affects Tau Liquid-Liquid Phase Separation. Front. Neurosci. 2019, 13, 845. [Google Scholar] [CrossRef] [Green Version]
  36. Oldfield, C.J.; Dunker, A.K. Intrinsically disordered proteins and intrinsically disordered protein regions. Annu. Rev. Biochem. 2014, 83, 553–584. [Google Scholar] [CrossRef]
  37. Choi, J.M.; Hyman, A.A.; Pappu, R.V. Generalized models for bond percolation transitions of associative polymers. Phys. Rev. E 2020, 102, 042403. [Google Scholar] [CrossRef]
  38. Banani, S.F.; Lee, H.O.; Hyman, A.A.; Rosen, M.K. Biomolecular condensates: Organizers of cellular biochemistry. Nat. Rev. Mol. Cell Biol. 2017, 18, 285–298. [Google Scholar] [CrossRef] [PubMed]
  39. Moser, J.J.; Fritzler, M.J. Cytoplasmic ribonucleoprotein (RNP) bodies and their relationship to GW/P bodies. Int. J. Biochem. Cell Biol. 2010, 42, 828–843. [Google Scholar] [CrossRef]
  40. Crisp, M.; Liu, Q.; Roux, K.; Rattner, J.B.; Shanahan, C.; Burke, B.; Stahl, P.D.; Hodzic, D. Coupling of the nucleus and cytoplasm: Role of the LINC complex. J. Cell Biol. 2006, 172, 41–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Hofmann, S.; Kedersha, N.; Anderson, P.; Ivanov, P. Molecular mechanisms of stress granule assembly and disassembly. Biochim. Biophys. Acta Mol. Cell Res. 2021, 1868, 118876. [Google Scholar] [CrossRef] [PubMed]
  42. Hofmann, S.; Cherkasova, V.; Bankhead, P.; Bukau, B.; Stoecklin, G. Translation suppression promotes stress granule formation and cell survival in response to cold shock. Mol. Biol. Cell 2012, 23, 3786–3800. [Google Scholar] [CrossRef] [PubMed]
  43. Campos-Melo, D.; Hawley, Z.C.E.; Droppelmann, C.A.; Strong, M.J. The Integral Role of RNA in Stress Granule Formation and Function. Front. Cell Dev. Biol. 2021, 9, 621779. [Google Scholar] [CrossRef] [PubMed]
  44. Wolozin, B.; Ivanov, P. Stress granules and neurodegeneration. Nat. Rev. Neurosci. 2019, 20, 649–666. [Google Scholar] [CrossRef] [PubMed]
  45. Decker, C.J.; Parker, R. P-bodies and stress granules: Possible roles in the control of translation and mRNA degradation. Cold Spring Harb. Perspect. Biol. 2012, 4, a012286. [Google Scholar] [CrossRef] [Green Version]
  46. Van Treeck, B.; Parker, R. Principles of Stress Granules Revealed by Imaging Approaches. Cold Spring Harb. Perspect. Biol. 2019, 11, a033068. [Google Scholar] [CrossRef] [Green Version]
  47. Woodruff, J.B.; Ferreira Gomes, B.; Widlund, P.O.; Mahamid, J.; Honigmann, A.; Hyman, A.A. The Centrosome Is a Selective Condensate that Nucleates Microtubules by Concentrating Tubulin. Cell 2017, 169, 1066–1077.e1010. [Google Scholar] [CrossRef] [Green Version]
  48. Frikstad, K.M.; Schink, K.O.; Gilani, S.; Pedersen, L.B.; Patzke, S. 3D-Structured Illumination Microscopy of Centrosomes in Human Cell Lines. Bio Protoc. 2022, 12, e4360. [Google Scholar] [CrossRef]
  49. Gonçalves, A.B.; Hasselbalch, S.K.; Joensen, B.B.; Patzke, S.; Martens, P.; Ohlsen, S.K.; Quinodoz, M.; Nikopoulos, K.; Suleiman, R.; Damsø Jeppesen, M.P.; et al. CEP78 functions downstream of CEP350 to control biogenesis of primary cilia by negatively regulating CP110 levels. eLife 2021, 10, e63731. [Google Scholar] [CrossRef]
  50. Liu, J.L.; Gall, J.G. U bodies are cytoplasmic structures that contain uridine-rich small nuclear ribonucleoproteins and associate with P bodies. Proc. Natl. Acad. Sci. USA 2007, 104, 11655–11659. [Google Scholar] [CrossRef] [Green Version]
  51. O’Flynn, B.G.; Mittag, T. The role of liquid-liquid phase separation in regulating enzyme activity. Curr. Opin. Cell Biol. 2021, 69, 70–79. [Google Scholar] [CrossRef] [PubMed]
  52. Liu, J.; Xu, Y.; Stoleru, D.; Salic, A. Imaging protein synthesis in cells and tissues with an alkyne analog of puromycin. Proc. Natl. Acad. Sci. USA 2012, 109, 413–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Jin, M.; Fuller, G.G.; Han, T.; Yao, Y.; Alessi, A.F.; Freeberg, M.A.; Roach, N.P.; Moresco, J.J.; Karnovsky, A.; Baba, M.; et al. Glycolytic Enzymes Coalesce in G Bodies under Hypoxic Stress. Cell Rep. 2017, 20, 895–908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Luo, Y.; Na, Z.; Slavoff, S.A. P-Bodies: Composition, Properties, and Functions. Biochemistry 2018, 57, 2424–2431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Elbaum-Garfinkle, S.; Kim, Y.; Szczepaniak, K.; Chen, C.C.; Eckmann, C.R.; Myong, S.; Brangwynne, C.P. The disordered P granule protein LAF-1 drives phase separation into droplets with tunable viscosity and dynamics. Proc. Natl. Acad. Sci. USA 2015, 112, 7189–7194. [Google Scholar] [CrossRef] [Green Version]
  56. Boke, E.; Ruer, M.; Wühr, M.; Coughlin, M.; Lemaitre, R.; Gygi, S.P.; Alberti, S.; Drechsel, D.; Hyman, A.A.; Mitchison, T.J. Amyloid-like Self-Assembly of a Cellular Compartment. Cell 2016, 166, 637–650. [Google Scholar] [CrossRef] [Green Version]
  57. Jamieson-Lucy, A.; Mullins, M.C. The vertebrate Balbiani body, germ plasm, and oocyte polarity. Curr. Top. Dev. Biol. 2019, 135, 1–34. [Google Scholar] [CrossRef]
  58. Lee, K.L.; Marlow, F.L. Visualizing the Balbiani Body in Zebrafish Oocytes. Methods Mol. Biol. 2019, 1920, 277–293. [Google Scholar] [CrossRef]
  59. Voronina, E.; Seydoux, G.; Sassone-Corsi, P.; Nagamori, I. RNA granules in germ cells. Cold Spring Harb. Perspect. Biol. 2011, 3, a002774. [Google Scholar] [CrossRef] [Green Version]
  60. Brangwynne, C.P.; Eckmann, C.R.; Courson, D.S.; Rybarska, A.; Hoege, C.; Gharakhani, J.; Jülicher, F.; Hyman, A.A. Germline P granules are liquid droplets that localize by controlled dissolution/condensation. Science 2009, 324, 1729–1732. [Google Scholar] [CrossRef]
  61. Saha, S.; Weber, C.A.; Nousch, M.; Adame-Arana, O.; Hoege, C.; Hein, M.Y.; Osborne-Nishimura, E.; Mahamid, J.; Jahnel, M.; Jawerth, L.; et al. Polar Positioning of Phase-Separated Liquid Compartments in Cells Regulated by an mRNA Competition Mechanism. Cell 2016, 166, 1572–1584.e1516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Kiebler, M.A.; Bassell, G.J. Neuronal RNA granules: Movers and makers. Neuron 2006, 51, 685–690. [Google Scholar] [CrossRef] [Green Version]
  63. Bauer, K.E.; Bargenda, N.; Schieweck, R.; Illig, C.; Segura, I.; Harner, M.; Kiebler, M.A. RNA supply drives physiological granule assembly in neurons. Nat. Commun. 2022, 13, 2781. [Google Scholar] [CrossRef] [PubMed]
  64. Pederson, T. The nucleolus. Cold Spring Harb. Perspect. Biol. 2011, 3, a000638. [Google Scholar] [CrossRef]
  65. Yang, K.; Yang, J.; Yi, J. Nucleolar Stress: Hallmarks, sensing mechanism and diseases. Cell Stress 2018, 2, 125–140. [Google Scholar] [CrossRef] [PubMed]
  66. Boisvert, F.M.; van Koningsbruggen, S.; Navascués, J.; Lamond, A.I. The multifunctional nucleolus. Nat. Rev. Mol. Cell Biol. 2007, 8, 574–585. [Google Scholar] [CrossRef]
  67. Brangwynne, C.P.; Mitchison, T.J.; Hyman, A.A. Active liquid-like behavior of nucleoli determines their size and shape in Xenopus laevis oocytes. Proc. Natl. Acad. Sci. USA 2011, 108, 4334–4339. [Google Scholar] [CrossRef] [Green Version]
  68. Yoneda, M.; Nakagawa, T.; Hattori, N.; Ito, T. The nucleolus from a liquid droplet perspective. J. Biochem. 2021, 170, 153–162. [Google Scholar] [CrossRef]
  69. Nizami, Z.; Deryusheva, S.; Gall, J.G. The Cajal body and histone locus body. Cold Spring Harb. Perspect. Biol. 2010, 2, a000653. [Google Scholar] [CrossRef] [Green Version]
  70. Duronio, R.J.; Marzluff, W.F. Coordinating cell cycle-regulated histone gene expression through assembly and function of the Histone Locus Body. RNA Biol. 2017, 14, 726–738. [Google Scholar] [CrossRef] [Green Version]
  71. Larson, A.G.; Elnatan, D.; Keenen, M.M.; Trnka, M.J.; Johnston, J.B.; Burlingame, A.L.; Agard, D.A.; Redding, S.; Narlikar, G.J. Liquid droplet formation by HP1α suggests a role for phase separation in heterochromatin. Nature 2017, 547, 236–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Chang, C.W.; Shen, Y.C.; Yan, S.J. HP1a-mediated heterochromatin formation inhibits high dietary sugar-induced tumor progression. Cell Death Dis. 2021, 12, 1130. [Google Scholar] [CrossRef]
  73. Liu, Y.; Zhang, D. HP1a/KDM4A is involved in the autoregulatory loop of the oncogene gene c-Jun. Epigenetics 2015, 10, 453–459. [Google Scholar] [CrossRef] [Green Version]
  74. Tatarakis, A.; Behrouzi, R.; Moazed, D. Evolving Models of Heterochromatin: From Foci to Liquid Droplets. Mol. Cell 2017, 67, 725–727. [Google Scholar] [CrossRef] [Green Version]
  75. Strom, A.R.; Biggs, R.J.; Banigan, E.J.; Wang, X.; Chiu, K.; Herman, C.; Collado, J.; Yue, F.; Ritland Politz, J.C.; Tait, L.J.; et al. HP1α is a chromatin crosslinker that controls nuclear and mitotic chromosome mechanics. eLife 2021, 10, e63972. [Google Scholar] [CrossRef] [PubMed]
  76. Erdel, F.; Rademacher, A.; Vlijm, R.; Tünnermann, J.; Frank, L.; Weinmann, R.; Schweigert, E.; Yserentant, K.; Hummert, J.; Bauer, C.; et al. Mouse Heterochromatin Adopts Digital Compaction States without Showing Hallmarks of HP1-Driven Liquid-Liquid Phase Separation. Mol. Cell. 2020, 78, 236–249.e237. [Google Scholar] [CrossRef]
  77. Capitanio, J.S.; Montpetit, B.; Wozniak, R.W. Human Nup98 regulates the localization and activity of DExH/D-box helicase DHX9. eLife 2017, 6, e18825. [Google Scholar] [CrossRef] [PubMed]
  78. Wong, R.W. New Activities of the Nuclear Pore Complexes. Cells 2021, 10, 2123. [Google Scholar] [CrossRef]
  79. Beck, M.; Hurt, E. The nuclear pore complex: Understanding its function through structural insight. Nat. Rev. Mol. Cell Biol. 2017, 18, 73–89. [Google Scholar] [CrossRef] [PubMed]
  80. Kumanski, S.; Viart, B.T.; Kossida, S.; Moriel-Carretero, M. Lipid Droplets Are a Physiological Nucleoporin Reservoir. Cells 2021, 10, 472. [Google Scholar] [CrossRef]
  81. Spector, D.L.; Lamond, A.I. Nuclear speckles. Cold Spring Harb. Perspect. Biol. 2011, 3, a000646. [Google Scholar] [CrossRef] [Green Version]
  82. Kim, J.; Han, K.Y.; Khanna, N.; Ha, T.; Belmont, A.S. Nuclear speckle fusion via long-range directional motion regulates speckle morphology after transcriptional inhibition. J. Cell Sci. 2019, 132, jcs226563. [Google Scholar] [CrossRef] [Green Version]
  83. Rothkamm, K.; Barnard, S.; Moquet, J.; Ellender, M.; Rana, Z.; Burdak-Rothkamm, S. DNA damage foci: Meaning and significance. Environ. Mol. Mutagen. 2015, 56, 491–504. [Google Scholar] [CrossRef]
  84. Oshidari, R.; Huang, R.; Medghalchi, M.; Tse, E.Y.W.; Ashgriz, N.; Lee, H.O.; Wyatt, H.; Mekhail, K. DNA repair by Rad52 liquid droplets. Nat. Commun. 2020, 11, 695. [Google Scholar] [CrossRef] [Green Version]
  85. Morimoto, M.; Boerkoel, C.F. The role of nuclear bodies in gene expression and disease. Biology 2013, 2, 976–1033. [Google Scholar] [CrossRef] [Green Version]
  86. Cauchi, R.J. Gem formation upon constitutive Gemin3 overexpression in Drosophila. Cell Biol. Int. 2011, 35, 1233–1238. [Google Scholar] [CrossRef] [PubMed]
  87. Liu, Q.; Dreyfuss, G. A novel nuclear structure containing the survival of motor neurons protein. Embo J. 1996, 15, 3555–3565. [Google Scholar] [CrossRef] [PubMed]
  88. Pirrotta, V.; Li, H.B. A view of nuclear Polycomb bodies. Curr. Opin. Genet. Dev. 2012, 22, 101–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Smigová, J.; Juda, P.; Cmarko, D.; Raška, I. Fine structure of the “PcG body” in human U-2 OS cells established by correlative light-electron microscopy. Nucleus 2011, 2, 219–228. [Google Scholar] [CrossRef] [Green Version]
  90. Fox, A.H.; Lamond, A.I. Paraspeckles. Cold Spring Harb. Perspect. Biol. 2010, 2, a000687. [Google Scholar] [CrossRef] [Green Version]
  91. Hennig, S.; Kong, G.; Mannen, T.; Sadowska, A.; Kobelke, S.; Blythe, A.; Knott, G.J.; Iyer, K.S.; Ho, D.; Newcombe, E.A.; et al. Prion-like domains in RNA binding proteins are essential for building subnuclear paraspeckles. J. Cell Biol. 2015, 210, 529–539. [Google Scholar] [CrossRef] [Green Version]
  92. Ohnishi, Y.; Huber, W.; Tsumura, A.; Kang, M.; Xenopoulos, P.; Kurimoto, K.; Oleś, A.K.; Araúzo-Bravo, M.J.; Saitou, M.; Hadjantonakis, A.K.; et al. Cell-to-cell expression variability followed by signal reinforcement progressively segregates early mouse lineages. Nat. Cell Biol. 2014, 16, 27–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Jarc, E.; Petan, T. Lipid Droplets and the Management of Cellular Stress. Yale J. Biol. Med. 2019, 92, 435–452. [Google Scholar] [PubMed]
  94. Sheng, M.; Kim, E. The postsynaptic organization of synapses. Cold Spring Harb. Perspect. Biol. 2011, 3, a005678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Zeng, M.; Shang, Y.; Araki, Y.; Guo, T.; Huganir, R.L.; Zhang, M. Phase Transition in Postsynaptic Densities Underlies Formation of Synaptic Complexes and Synaptic Plasticity. Cell 2016, 166, 1163–1175.e1112. [Google Scholar] [CrossRef] [Green Version]
  96. Case, L.B.; Waterman, C.M. Integration of actin dynamics and cell adhesion by a three-dimensional, mechanosensitive molecular clutch. Nat. Cell Biol. 2015, 17, 955–963. [Google Scholar] [CrossRef] [PubMed]
  97. Case, L.B.; De Pasquale, M.; Henry, L.; Rosen, M.K. Synergistic phase separation of two pathways promotes integrin clustering and nascent adhesion formation. eLife 2022, 11, e72588. [Google Scholar] [CrossRef]
  98. Jones, N.; Blasutig, I.M.; Eremina, V.; Ruston, J.M.; Bladt, F.; Li, H.; Huang, H.; Larose, L.; Li, S.S.; Takano, T.; et al. Nck adaptor proteins link nephrin to the actin cytoskeleton of kidney podocytes. Nature 2006, 440, 818–823. [Google Scholar] [CrossRef]
  99. Martin, C.E.; New, L.A.; Phippen, N.J.; Keyvani Chahi, A.; Mitro, A.E.; Takano, T.; Pawson, T.; Blasutig, I.M.; Jones, N. Multivalent nephrin-Nck interactions define a threshold for clustering and tyrosine-dependent nephrin endocytosis. J. Cell Sci. 2020, 133, jcs236877. [Google Scholar] [CrossRef]
  100. Dustin, M.L.; Groves, J.T. Receptor signaling clusters in the immune synapse. Annu. Rev. Biophys. 2012, 41, 543–556. [Google Scholar] [CrossRef] [Green Version]
  101. Su, X.; Ditlev, J.A.; Hui, E.; Xing, W.; Banjade, S.; Okrut, J.; King, D.S.; Taunton, J.; Rosen, M.K.; Vale, R.D. Phase separation of signaling molecules promotes T cell receptor signal transduction. Science 2016, 352, 595–599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Calle, Y.; Burns, S.; Thrasher, A.J.; Jones, G.E. The leukocyte podosome. Eur. J. Cell Biol. 2006, 85, 151–157. [Google Scholar] [CrossRef] [PubMed]
  103. Vincent, C.; Siddiqui, T.A.; Schlichter, L.C. Podosomes in migrating microglia: Components and matrix degradation. J. Neuroinflamm. 2012, 9, 190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Goode, B.L.; Eskin, J.A.; Wendland, B. Actin and endocytosis in budding yeast. Genetics 2015, 199, 315–358. [Google Scholar] [CrossRef] [Green Version]
  105. Moseley, J.B.; Goode, B.L. The yeast actin cytoskeleton: From cellular function to biochemical mechanism. Microbiol. Mol. Biol. Rev. 2006, 70, 605–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Olzmann, J.A.; Carvalho, P. Dynamics and functions of lipid droplets. Nat. Rev. Mol. Cell Biol. 2019, 20, 137–155. [Google Scholar] [CrossRef] [PubMed]
  107. Johnson, V.M.; Pakrasi, H.B. Advances in the Understanding of the Lifecycle of Photosystem II. Microorganisms 2022, 10, 836. [Google Scholar] [CrossRef] [PubMed]
  108. Müh, F.; Zouni, A. Structural basis of light-harvesting in the photosystem II core complex. Protein Sci. 2020, 29, 1090–1119. [Google Scholar] [CrossRef] [Green Version]
  109. Järvi, S.; Suorsa, M.; Aro, E.M. Photosystem II repair in plant chloroplasts--Regulation, assisting proteins and shared components with photosystem II biogenesis. Biochim. Biophys. Acta 2015, 1847, 900–909. [Google Scholar] [CrossRef] [Green Version]
  110. Saini, B.; Mukherjee, T.K. Biomolecular Condensates Regulate Enzymatic Activity under a Crowded Milieu: Synchronization of Liquid–Liquid Phase Separation and Enzymatic Transformation. J. Phys. Chem. B 2023, 127, 180–193. [Google Scholar] [CrossRef]
  111. Tang, S.C.; Vijayakumar, U.; Zhang, Y.; Fullwood, M.J. Super-Enhancers, Phase-Separated Condensates, and 3D Genome Organization in Cancer. Cancers 2022, 14, 2866. [Google Scholar] [CrossRef]
  112. Richter, W.F.; Nayak, S.; Iwasa, J.; Taatjes, D.J. The Mediator complex as a master regulator of transcription by RNA polymerase II. Nat. Rev. Mol. Cell Biol. 2022, 23, 732–749. [Google Scholar] [CrossRef] [PubMed]
  113. Sharp, P.A.; Chakraborty, A.K.; Henninger, J.E.; Young, R.A. RNA in formation and regulation of transcriptional condensates. RNA 2022, 28, 52–57. [Google Scholar] [CrossRef]
  114. Bauer, J.A.; Zámocká, M.; Majtán, J.; Bauerová-Hlinková, V. Glucose Oxidase, an Enzyme “Ferrari”: Its Structure, Function, Production and Properties in the Light of Various Industrial and Biotechnological Applications. Biomolecules 2022, 12, 472. [Google Scholar] [CrossRef]
  115. Koga, S.; Williams, D.S.; Perriman, A.W.; Mann, S. Peptide-nucleotide microdroplets as a step towards a membrane-free protocell model. Nat. Chem. 2011, 3, 720–724. [Google Scholar] [CrossRef] [PubMed]
  116. Lindhoud, S.; Norde, W.; Cohen Stuart, M.A. Effects of Polyelectrolyte Complex Micelles and Their Components on the Enzymatic Activity of Lipase. Langmuir 2010, 26, 9802–9808. [Google Scholar] [CrossRef]
  117. Wang, L.; Lin, Y.; Zhou, Y.; Xie, H.; Song, J.; Li, M.; Huang, Y.; Huang, X.; Mann, S. Autonomic Behaviors in Lipase-Active Oil Droplets. Angew. Chem. Int. Ed. Engl. 2019, 58, 1067–1071. [Google Scholar] [CrossRef] [Green Version]
  118. Strulson, C.A.; Molden, R.C.; Keating, C.D.; Bevilacqua, P.C. RNA catalysis through compartmentalization. Nat. Chem. 2012, 4, 941–946. [Google Scholar] [CrossRef] [PubMed]
  119. Drobot, B.; Iglesias-Artola, J.M.; Le Vay, K.; Mayr, V.; Kar, M.; Kreysing, M.; Mutschler, H.; Tang, T.D. Compartmentalised RNA catalysis in membrane-free coacervate protocells. Nat. Commun. 2018, 9, 3643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Fuller, G.G.; Han, T.; Freeberg, M.A.; Moresco, J.J.; Ghanbari Niaki, A.; Roach, N.P.; Yates, J.R., III; Myong, S.; Kim, J.K. RNA promotes phase separation of glycolysis enzymes into yeast G bodies in hypoxia. eLife 2020, 9, e48480. [Google Scholar] [CrossRef]
  121. Zhu, J.; Zhou, Q.; Xia, Y.; Lin, L.; Li, J.; Peng, M.; Zhang, R.; Zhang, M. GIT/PIX Condensates Are Modular and Ideal for Distinct Compartmentalized Cell Signaling. Mol. Cell 2020, 79, 782–796.e786. [Google Scholar] [CrossRef] [PubMed]
  122. Watanabe, K.; Ohtsuki, T. Inhibition of HSF1 and SAFB Granule Formation Enhances Apoptosis Induced by Heat Stress. Int. J. Mol. Sci. 2021, 22, 4982. [Google Scholar] [CrossRef] [PubMed]
  123. Gaglia, G.; Rashid, R.; Yapp, C.; Joshi, G.N.; Li, C.G.; Lindquist, S.L.; Sarosiek, K.A.; Whitesell, L.; Sorger, P.K.; Santagata, S. HSF1 phase transition mediates stress adaptation and cell fate decisions. Nat. Cell Biol. 2020, 22, 151–158. [Google Scholar] [CrossRef]
  124. Rawat, P.; Boehning, M.; Hummel, B.; Aprile-Garcia, F.; Pandit, A.S.; Eisenhardt, N.; Khavaran, A.; Niskanen, E.; Vos, S.M.; Palvimo, J.J.; et al. Stress-induced nuclear condensation of NELF drives transcriptional downregulation. Mol. Cell 2021, 81, 1013–1026.e1011. [Google Scholar] [CrossRef]
  125. Boija, A.; Klein, I.A.; Sabari, B.R.; Dall’Agnese, A.; Coffey, E.L.; Zamudio, A.V.; Li, C.H.; Shrinivas, K.; Manteiga, J.C.; Hannett, N.M.; et al. Transcription Factors Activate Genes through the Phase-Separation Capacity of Their Activation Domains. Cell 2018, 175, 1842–1855.e1816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Safari, M.S.; Wang, Z.; Tailor, K.; Kolomeisky, A.B.; Conrad, J.C.; Vekilov, P.G. Anomalous Dense Liquid Condensates Host the Nucleation of Tumor Suppressor p53 Fibrils. iScience 2019, 12, 342–355. [Google Scholar] [CrossRef] [Green Version]
  127. Park, S.K.; Park, S.; Pentek, C.; Liebman, S.W. Tumor suppressor protein p53 expressed in yeast can remain diffuse, form a prion, or form unstable liquid-like droplets. iScience 2021, 24, 102000. [Google Scholar] [CrossRef]
  128. Kamagata, K.; Kanbayashi, S.; Honda, M.; Itoh, Y.; Takahashi, H.; Kameda, T.; Nagatsugi, F.; Takahashi, S. Liquid-like droplet formation by tumor suppressor p53 induced by multivalent electrostatic interactions between two disordered domains. Sci. Rep. 2020, 10, 580. [Google Scholar] [CrossRef] [Green Version]
  129. Lemos, C.; Schulze, L.; Weiske, J.; Meyer, H.; Braeuer, N.; Barak, N.; Eberspächer, U.; Werbeck, N.; Stresemann, C.; Lange, M.; et al. Identification of Small Molecules that Modulate Mutant p53 Condensation. iScience 2020, 23, 101517. [Google Scholar] [CrossRef]
  130. Petronilho, E.C.; Pedrote, M.M.; Marques, M.A.; Passos, Y.M.; Mota, M.F.; Jakobus, B.; Sousa, G.d.S.d.; Pereira da Costa, F.; Felix, A.L.; Ferretti, G.D.S.; et al. Phase separation of p53 precedes aggregation and is affected by oncogenic mutations and ligands. Chem. Sci. 2021, 12, 7334–7349. [Google Scholar] [CrossRef]
  131. Park, J.E.; Zhang, L.; Bang, J.K.; Andresson, T.; DiMaio, F.; Lee, K.S. Phase separation of Polo-like kinase 4 by autoactivation and clustering drives centriole biogenesis. Nat. Commun. 2019, 10, 4959. [Google Scholar] [CrossRef] [Green Version]
  132. Vitiello, E.; Moreau, P.; Nunes, V.; Mettouchi, A.; Maiato, H.; Ferreira, J.G.; Wang, I.; Balland, M. Acto-myosin force organization modulates centriole separation and PLK4 recruitment to ensure centriole fidelity. Nat. Commun. 2019, 10, 52. [Google Scholar] [CrossRef]
  133. Krainer, G.; Welsh, T.J.; Joseph, J.A.; Espinosa, J.R.; Wittmann, S.; de Csilléry, E.; Sridhar, A.; Toprakcioglu, Z.; Gudiškytė, G.; Czekalska, M.A.; et al. Reentrant liquid condensate phase of proteins is stabilized by hydrophobic and non-ionic interactions. Nat. Commun. 2021, 12, 1085. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, B.; Gan, W.; Han, X.; Liu, N.; Ma, T.; Li, D. The positive regulation loop between NRF1 and NONO-TFE3 fusion promotes phase separation and aggregation of NONO-TFE3 in NONO-TFE3 tRCC. Int. J. Biol. Macromol. 2021, 176, 437–447. [Google Scholar] [CrossRef]
  135. Chen, D.; Wang, Z.; Zhao, Y.G.; Zheng, H.; Zhao, H.; Liu, N.; Zhang, H. Inositol Polyphosphate Multikinase Inhibits Liquid-Liquid Phase Separation of TFEB to Negatively Regulate Autophagy Activity. Dev. Cell 2020, 55, 588–602.e587. [Google Scholar] [CrossRef]
  136. Liu, S.; Wang, T.; Shi, Y.; Bai, L.; Wang, S.; Guo, D.; Zhang, Y.; Qi, Y.; Chen, C.; Zhang, J.; et al. USP42 drives nuclear speckle mRNA splicing via directing dynamic phase separation to promote tumorigenesis. Cell Death Differ. 2021, 28, 2482–2498. [Google Scholar] [CrossRef] [PubMed]
  137. Yu, M.; Peng, Z.; Qin, M.; Liu, Y.; Wang, J.; Zhang, C.; Lin, J.; Dong, T.; Wang, L.; Li, S.; et al. Interferon-γ induces tumor resistance to anti-PD-1 immunotherapy by promoting YAP phase separation. Mol. Cell 2021, 81, 1216–1230.e1219. [Google Scholar] [CrossRef]
  138. Cai, D.; Feliciano, D.; Dong, P.; Flores, E.; Gruebele, M.; Porat-Shliom, N.; Sukenik, S.; Liu, Z.; Lippincott-Schwartz, J. Phase separation of YAP reorganizes genome topology for long-term YAP target gene expression. Nat. Cell Biol. 2019, 21, 1578–1589. [Google Scholar] [CrossRef] [PubMed]
  139. Barrett, J.; Girr, P.; Mackinder, L.C.M. Pyrenoids: CO(2)-fixing phase separated liquid organelles. Biochim. Biophys. Acta Mol. Cell Res. 2021, 1868, 118949. [Google Scholar] [CrossRef]
  140. Wunder, T.; Mueller-Cajar, O. Biomolecular condensates in photosynthesis and metabolism. Curr. Opin. Plant Biol. 2020, 58, 1–7. [Google Scholar] [CrossRef]
  141. Gao, Z.; Zhang, W.; Chang, R.; Zhang, S.; Yang, G.; Zhao, G. Liquid-Liquid Phase Separation: Unraveling the Enigma of Biomolecular Condensates in Microbial Cells. Front. Microbiol. 2021, 12, 751880. [Google Scholar] [CrossRef] [PubMed]
  142. Lach, R.S.; Qiu, C.; Kajbaf, E.Z.; Baxter, N.; Han, D.; Wang, A.; Lock, H.; Chirikian, O.; Pruitt, B.; Wilson, M.Z. Nucleation of the destruction complex on the centrosome accelerates degradation of β-catenin and regulates Wnt signal transmission. Proc. Natl. Acad. Sci. USA 2022, 119, e2204688119. [Google Scholar] [CrossRef] [PubMed]
  143. Cheng, Y.; Xie, W.; Pickering, B.F.; Chu, K.L.; Savino, A.M.; Yang, X.; Luo, H.; Nguyen, D.T.; Mo, S.; Barin, E.; et al. N(6)-Methyladenosine on mRNA facilitates a phase-separated nuclear body that suppresses myeloid leukemic differentiation. Cancer Cell 2021, 39, 958–972.e958. [Google Scholar] [CrossRef] [PubMed]
  144. Marschallinger, J.; Iram, T.; Zardeneta, M.; Lee, S.E.; Lehallier, B.; Haney, M.S.; Pluvinage, J.V.; Mathur, V.; Hahn, O.; Morgens, D.W.; et al. Lipid-droplet-accumulating microglia represent a dysfunctional and proinflammatory state in the aging brain. Nat. Neurosci. 2020, 23, 194–208. [Google Scholar] [CrossRef]
  145. Yue, F.; Oprescu, S.N.; Qiu, J.; Gu, L.; Zhang, L.; Chen, J.; Narayanan, N.; Deng, M.; Kuang, S. Lipid droplet dynamics regulate adult muscle stem cell fate. Cell Rep. 2022, 38, 110267. [Google Scholar] [CrossRef]
  146. Scholz, P.; Chapman, K.D.; Mullen, R.T.; Ischebeck, T. Finding new friends and revisiting old ones—How plant lipid droplets connect with other subcellular structures. New Phytol. 2022, 236, 833–838. [Google Scholar] [CrossRef]
  147. Sato, Y.; Takinoue, M. Creation of Artificial Cell-Like Structures Promoted by Microfluidics Technologies. Micromachines 2019, 10, 216. [Google Scholar] [CrossRef] [Green Version]
  148. Zhao, C.; Li, J.; Wang, S.; Xu, Z.; Wang, X.; Liu, X.; Wang, L.; Huang, X. Membranization of Coacervates into Artificial Phagocytes with Predation toward Bacteria. ACS Nano 2021, 15, 10048–10057. [Google Scholar] [CrossRef]
  149. Sato, Y.; Takinoue, M. Capsule-like DNA Hydrogels with Patterns Formed by Lateral Phase Separation of DNA Nanostructures. JACS Au 2022, 2, 159–168. [Google Scholar] [CrossRef]
  150. Abbas, M.; Lipiński, W.P.; Nakashima, K.K.; Huck, W.T.S.; Spruijt, E. A short peptide synthon for liquid–liquid phase separation. Nat. Chem. 2021, 13, 1046–1054. [Google Scholar] [CrossRef]
  151. Wang, Z.; Huang, P.; Jacobson, O.; Wang, Z.; Liu, Y.; Lin, L.; Lin, J.; Lu, N.; Zhang, H.; Tian, R.; et al. Biomineralization-Inspired Synthesis of Copper Sulfide-Ferritin Nanocages as Cancer Theranostics. ACS Nano 2016, 10, 3453–3460. [Google Scholar] [CrossRef] [Green Version]
  152. Nakatani, N.; Sakuta, H.; Hayashi, M.; Tanaka, S.; Takiguchi, K.; Tsumoto, K.; Yoshikawa, K. Specific Spatial Localization of Actin and DNA in a Water/Water Microdroplet: Self-Emergence of a Cell-Like Structure. Chembiochem 2018, 19, 1370–1374. [Google Scholar] [CrossRef] [Green Version]
  153. Bachler, S.; Haidas, D.; Ort, M.; Duncombe, T.A.; Dittrich, P.S. Microfluidic platform enables tailored translocation and reaction cascades in nanoliter droplet networks. Commun. Biol. 2020, 3, 769. [Google Scholar] [CrossRef] [PubMed]
  154. Wang, X.; Huang, Y.; Lin, Y.; Chen, H.; Li, J.; Zhao, C.; Liu, X.; Yang, L.; Huang, X. Life-Inspired Endogenous Dynamic Behavior of Lipid Droplet-like Microcompartments in Artificial Adipocyte-like Structures. CCS Chem. 2020, 3, 2782–2794. [Google Scholar] [CrossRef]
  155. Tsuruta, M.; Torii, T.; Kohata, K.; Kawauchi, K.; Tateishi-Karimata, H.; Sugimoto, N.; Miyoshi, D. Controlling liquid-liquid phase separation of G-quadruplex-forming RNAs in a sequence-specific manner. Chem. Commun. 2022, 58, 12931–12934. [Google Scholar] [CrossRef]
  156. Shin, H.; Park, Y.H.; Kim, Y.G.; Lee, J.Y.; Park, J. Aqueous two-phase system to isolate extracellular vesicles from urine for prostate cancer diagnosis. PLoS ONE 2018, 13, e0194818. [Google Scholar] [CrossRef] [PubMed]
  157. Mastiani, M.; Firoozi, N.; Petrozzi, N.; Seo, S.; Kim, M. Polymer-Salt Aqueous Two-Phase System (ATPS) Micro-Droplets for Cell Encapsulation. Sci. Rep. 2019, 9, 15561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Chao, Y.; Shum, H.C. Emerging aqueous two-phase systems: From fundamentals of interfaces to biomedical applications. Chem. Soc. Rev. 2020, 49, 114–142. [Google Scholar] [CrossRef] [PubMed]
  159. Ma, Q.; Song, Y.; Sun, W.; Cao, J.; Yuan, H.; Wang, X.; Sun, Y.; Shum, H.C. Cell-Inspired All-Aqueous Microfluidics: From Intracellular Liquid-Liquid Phase Separation toward Advanced Biomaterials. Adv. Sci. 2020, 7, 1903359. [Google Scholar] [CrossRef]
  160. Iqbal, M.; Tao, Y.; Xie, S.; Zhu, Y.; Chen, D.; Wang, X.; Huang, L.; Peng, D.; Sattar, A.; Shabbir, M.A.; et al. Aqueous two-phase system (ATPS): An overview and advances in its applications. Biol. Proced. Online 2016, 18, 18. [Google Scholar] [CrossRef] [Green Version]
  161. Diamond, A.D.; Hsu, J.T. Aqueous two-phase systems for biomolecule separation. Adv. Biochem. Eng. Biotechnol. 1992, 47, 89–135. [Google Scholar] [CrossRef]
  162. Albertsson, P.A. Partition of cell particles and macromolecules in polymer two-phase systems. Adv. Protein Chem. 1970, 24, 309–341. [Google Scholar] [CrossRef] [PubMed]
  163. Jia, T.Z.; Fraccia, T.P. Liquid Crystal Peptide/DNA Coacervates in the Context of Prebiotic Molecular Evolution. Crystals 2020, 10, 964. [Google Scholar] [CrossRef]
  164. Frankel, E.A.; Bevilacqua, P.C.; Keating, C.D. Polyamine/Nucleotide Coacervates Provide Strong Compartmentalization of Mg2+, Nucleotides, and RNA. Langmuir 2016, 32, 2041–2049. [Google Scholar] [CrossRef] [PubMed]
  165. Jia, T.Z.; Hentrich, C.; Szostak, J.W. Rapid RNA exchange in aqueous two-phase system and coacervate droplets. Orig. Life Evol. Biosph. 2014, 44, 1–12. [Google Scholar] [CrossRef] [Green Version]
  166. Keating, C.D. Aqueous phase separation as a possible route to compartmentalization of biological molecules. Acc. Chem. Res. 2012, 45, 2114–2124. [Google Scholar] [CrossRef] [PubMed]
  167. McQueen, L.; Lai, D. Ionic Liquid Aqueous Two-Phase Systems From a Pharmaceutical Perspective. Front. Chem. 2019, 7, 135. [Google Scholar] [CrossRef] [Green Version]
  168. Yau, Y.K.; Ooi, C.W.; Ng, E.-P.; Lan, J.C.-W.; Ling, T.C.; Show, P.L. Current applications of different type of aqueous two-phase systems. Bioresour. Bioprocess. 2015, 2, 49. [Google Scholar] [CrossRef] [Green Version]
  169. Frerix, A.; Müller, M.; Kula, M.R.; Hubbuch, J. Scalable recovery of plasmid DNA based on aqueous two-phase separation. Biotechnol. Appl. Biochem. 2005, 42, 57–66. [Google Scholar] [CrossRef]
  170. Frerix, A.; Schönewald, M.; Geilenkirchen, P.; Müller, M.; Kula, M.R.; Hubbuch, J. Exploitation of the coil-globule plasmid DNA transition induced by small changes in temperature, pH salt, and poly(ethylene glycol) compositions for directed partitioning in aqueous two-phase systems. Langmuir 2006, 22, 4282–4290. [Google Scholar] [CrossRef] [Green Version]
  171. Indulkar, A.S.; Gao, Y.; Raina, S.A.; Zhang, G.G.; Taylor, L.S. Exploiting the Phenomenon of Liquid-Liquid Phase Separation for Enhanced and Sustained Membrane Transport of a Poorly Water-Soluble Drug. Mol. Pharm. 2016, 13, 2059–2069. [Google Scholar] [CrossRef]
  172. Feng, C.; Song, R.; Sun, G.; Kong, M.; Bao, Z.; Li, Y.; Cheng, X.; Cha, D.; Park, H.; Chen, X. Immobilization of Coacervate Microcapsules in Multilayer Sodium Alginate Beads for Efficient Oral Anticancer Drug Delivery. Biomacromolecules 2014, 15, 985–996. [Google Scholar] [CrossRef]
  173. Zhao, M.; Zacharia, N.S. Protein encapsulation via polyelectrolyte complex coacervation: Protection against protein denaturation. J. Chem. Phys. 2018, 149, 163326. [Google Scholar] [CrossRef]
  174. Nojima, T.; Niwa, T.; Taguchi, H. Proteome Analysis of Phase-Separated Condensed Proteins with Ionic Surfactants Revealed Versatile Formation of Artificial Biomolecular Condensates. Biomacromolecules 2019, 20, 539–545. [Google Scholar] [CrossRef] [PubMed]
  175. Nojima, T.; Iyoda, T. Water-Rich Fluid Material Containing Orderly Condensed Proteins. Angew. Chem. Int. Ed. 2017, 56, 1308–1312. [Google Scholar] [CrossRef] [PubMed]
  176. Mohamed, F.; van der Walle, C.F. Engineering biodegradable polyester particles with specific drug targeting and drug release properties. J. Pharm. Sci. 2008, 97, 71–87. [Google Scholar] [CrossRef] [PubMed]
  177. Husmann, M.; Schenderlein, S.; Lück, M.; Lindner, H.; Kleinebudde, P. Polymer erosion in PLGA microparticles produced by phase separation method. Int. J. Pharm. 2002, 242, 277–280. [Google Scholar] [CrossRef]
  178. Karthick, V.; Panda, S.; Kumar, V.G.; Kumar, D.; Shrestha, L.K.; Ariga, K.; Vasanth, K.; Chinnathambi, S.; Dhas, T.S.; Suganya, K.S.U. Quercetin loaded PLGA microspheres induce apoptosis in breast cancer cells. Appl. Surf. Sci. 2019, 487, 211–217. [Google Scholar] [CrossRef]
  179. Washington, K.E.; Kularatne, R.N.; Karmegam, V.; Biewer, M.C.; Stefan, M.C. Recent advances in aliphatic polyesters for drug delivery applications. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2017, 9, e1446. [Google Scholar] [CrossRef]
  180. Abu Ghalia, M. Biodegradable poly(lactic acid)-based scaffolds: Synthesis and biomedical applications. J. Polym. Res. 2017, 24, 74. [Google Scholar] [CrossRef]
  181. Shive, M.S.; Anderson, J.M. Biodegradation and biocompatibility of PLA and PLGA microspheres. Adv. Drug Deliv. Rev. 1997, 28, 5–24. [Google Scholar] [CrossRef]
  182. Kucharczyk, P.; Pavelková, A.; Stloukal, P.; Sedlarík, V. Degradation behaviour of PLA-based polyesterurethanes under abiotic and biotic environments. Polym. Degrad. Stab. 2016, 129, 222–230. [Google Scholar] [CrossRef] [Green Version]
  183. Carvalho, J.R.G.; Conde, G.; Antonioli, M.L.; Dias, P.P.; Vasconcelos, R.O.; Taboga, S.R.; Canola, P.A.; Chinelatto, M.A.; Pereira, G.T.; Ferraz, G.C. Biocompatibility and biodegradation of poly(lactic acid) (PLA) and an immiscible PLA/poly(ε-caprolactone) (PCL) blend compatibilized by poly(ε-caprolactone-b-tetrahydrofuran) implanted in horses. Polym. J. 2020, 52, 629–643. [Google Scholar] [CrossRef]
  184. Mikos, A.G.; Thorsen, A.J.; Czerwonka, L.A.; Bao, Y.; Langer, R.; Winslow, D.N.; Vacanti, J.P. Preparation and characterization of poly(l-lactic acid) foams. Polymer 1994, 35, 1068–1077. [Google Scholar] [CrossRef]
  185. Urbánek, T.; Jäger, E.; Jäger, A.; Hrubý, M. Selectively Biodegradable Polyesters: Nature-Inspired Construction Materials for Future Biomedical Applications. Polymers 2019, 11, 1061. [Google Scholar] [CrossRef] [Green Version]
  186. Xiao, W.; Jakimowicz, M.D.; Zampetakis, I.; Neely, S.; Scarpa, F.; Davis, S.A.; Williams, D.S.; Perriman, A.W. Biopolymeric Coacervate Microvectors for the Delivery of Functional Proteins to Cells. Adv. Biosyst. 2020, 4, 2000101. [Google Scholar] [CrossRef]
  187. Lagassé, H.A.; Alexaki, A.; Simhadri, V.L.; Katagiri, N.H.; Jankowski, W.; Sauna, Z.E.; Kimchi-Sarfaty, C. Recent advances in (therapeutic protein) drug development. F1000Res 2017, 6, 113. [Google Scholar] [CrossRef] [Green Version]
  188. Iwata, T.; Hirose, H.; Sakamoto, K.; Hirai, Y.; Arafiles, J.V.V.; Akishiba, M.; Imanishi, M.; Futaki, S. Cover Picture: Liquid Droplet Formation and Facile Cytosolic Translocation of IgG in the Presence of Attenuated Cationic Amphiphilic Lytic Peptides (Angew. Chem. Int. Ed. 36/2021). Angew. Chem. Int. Ed. 2021, 60, 19493. [Google Scholar] [CrossRef]
  189. Manolio, T.A.; Bult, C.J.; Chisholm, R.L.; Deverka, P.A.; Ginsburg, G.S.; Goldrich, M.; Jarvik, G.P.; Mensah, G.A.; Ramos, E.M.; Relling, M.V.; et al. Genomic medicine year in review: 2021. Am. J. Hum. Genet. 2021, 108, 2210–2214. [Google Scholar] [CrossRef] [PubMed]
  190. Hu, B.; Zhong, L.; Weng, Y.; Peng, L.; Huang, Y.; Zhao, Y.; Liang, X.J. Therapeutic siRNA: State of the art. Signal Transduct. Target. Ther. 2020, 5, 101. [Google Scholar] [CrossRef]
  191. Horton, R.H.; Lucassen, A.M. Recent developments in genetic/genomic medicine. Clin. Sci. 2019, 133, 697–708. [Google Scholar] [CrossRef] [Green Version]
  192. Nair, S.R. Personalized medicine: Striding from genes to medicines. Perspect. Clin. Res. 2010, 1, 146–150. [Google Scholar] [CrossRef]
  193. Silveira, M.M.; Moreira, G.; Mendonça, M. DNA vaccines against COVID-19: Perspectives and challenges. Life Sci. 2021, 267, 118919. [Google Scholar] [CrossRef] [PubMed]
  194. Ura, T.; Yamashita, A.; Mizuki, N.; Okuda, K.; Shimada, M. New vaccine production platforms used in developing SARS-CoV-2 vaccine candidates. Vaccine 2021, 39, 197–201. [Google Scholar] [CrossRef]
  195. Walsh, E.E.; Frenck, R.W., Jr.; Falsey, A.R.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Neuzil, K.; Mulligan, M.J.; Bailey, R.; et al. Safety and Immunogenicity of Two RNA-Based Covid-19 Vaccine Candidates. N. Engl. J. Med. 2020, 383, 2439–2450. [Google Scholar] [CrossRef]
  196. Mi, X.; Blocher McTigue, W.C.; Joshi, P.U.; Bunker, M.K.; Heldt, C.L.; Perry, S.L. Thermostabilization of viruses via complex coacervation. Biomater. Sci. 2020, 8, 7082–7092. [Google Scholar] [CrossRef]
  197. Xu, Y.; Qi, R.; Zhu, H.; Li, B.; Shen, Y.; Krainer, G.; Klenerman, D.; Knowles, T.P.J. Liquid-Liquid Phase-Separated Systems from Reversible Gel-Sol Transition of Protein Microgels. Adv. Mater. 2021, 33, e2008670. [Google Scholar] [CrossRef]
  198. Zhang, Z.; Liu, Q.; Sun, Z.; Phillips, B.K.; Wang, Z.; Al-Hashimi, M.; Fang, L.; Olson, M.A. Poly-Lipoic Ester-Based Coacervates for the Efficient Removal of Organic Pollutants from Water and Increased Point-of-Use Versatility. Chem. Mater. 2019, 31, 4405–4417. [Google Scholar] [CrossRef]
  199. Bunce, J.T.; Ndam, E.; Ofiteru, I.D.; Moore, A.; Graham, D.W. A Review of Phosphorus Removal Technologies and Their Applicability to Small-Scale Domestic Wastewater Treatment Systems. Front. Environ. Sci. 2018, 6, 8. [Google Scholar] [CrossRef] [Green Version]
  200. Tolstoguzov, V. Texturising by phase separation. Biotechnol. Adv. 2006, 24, 626–628. [Google Scholar] [CrossRef] [PubMed]
  201. Wu, B.C.; Degner, B.; McClements, D.J. Soft matter strategies for controlling food texture: Formation of hydrogel particles by biopolymer complex coacervation. J. Phys. Condens. Matter 2014, 26, 464104. [Google Scholar] [CrossRef] [PubMed]
  202. Eghbal, N.; Choudhary, R. Complex coacervation: Encapsulation and controlled release of active agents in food systems. LWT 2018, 90, 254–264. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of eukaryotic cell and various biomolecular droplet observed inside the cytoplasm, nucleolus, and cell membranes. Certain droplets are unique to particular cell types. For example, balbiani bodies and germ granules are unique to germ cells, and RNA transport granules and synaptic densities are unique to neuronal cells.
Figure 1. Schematic representation of eukaryotic cell and various biomolecular droplet observed inside the cytoplasm, nucleolus, and cell membranes. Certain droplets are unique to particular cell types. For example, balbiani bodies and germ granules are unique to germ cells, and RNA transport granules and synaptic densities are unique to neuronal cells.
Biotech 12 00026 g001
Figure 2. (a) Schematic representation of three different types of LLPS and the formation droplet. (b) Schematic representation of different kinds of interaction involved for undergoing LLPS.
Figure 2. (a) Schematic representation of three different types of LLPS and the formation droplet. (b) Schematic representation of different kinds of interaction involved for undergoing LLPS.
Biotech 12 00026 g002
Figure 3. (a) Schematic representation of associative phase separation where a negatively charged alginate reacts with positively charged gelatine and produces droplets through associative phase separation. (b) Schematic representation of segregative phase separation induced by PEG (Poly-ethylene glycol) with DEX (Dextrin) to generate an artificial droplet. (c) Schematic representation of multivalent protein and intrinsically disordered protein as the sticker.
Figure 3. (a) Schematic representation of associative phase separation where a negatively charged alginate reacts with positively charged gelatine and produces droplets through associative phase separation. (b) Schematic representation of segregative phase separation induced by PEG (Poly-ethylene glycol) with DEX (Dextrin) to generate an artificial droplet. (c) Schematic representation of multivalent protein and intrinsically disordered protein as the sticker.
Biotech 12 00026 g003
Figure 4. Schematic representation of the application of biomolecular LLPS in various field of biotechnology.
Figure 4. Schematic representation of the application of biomolecular LLPS in various field of biotechnology.
Biotech 12 00026 g004
Table 1. Components, roles, and observation procedures of droplets in cytoplasm.
Table 1. Components, roles, and observation procedures of droplets in cytoplasm.
DropletMain ComponentRoleObservationRef.
Stress granule Proteins and RNAs Translational regulation mRNA storage CM (1), smFISH (2)[42,43]
CentrosomePericentriolar material Formation of mitotic spindles during mitosis CM (1)[44,45]
U body Uridine-rich small nuclear ribonucleoproteins Storage and assembly of snRNPsCM (1)[46,47]
G bodyLipid and proteinControlling the rate of glycolysisCM (1)[48,49]
P body Translationally repressed mRNAs and proteins related to mRNA decay mRNA decay and silencingCM (1)[50,51]
Balbiani body (germ cells)Endoplasmic reticulum/Golgi-like vesicles, mitochondria, and specific RNAs transporter.Storage and facilitating the organization of the oocyte into a polarized cellCM (1), EM (3)[52,53,54,55]
Germ granules (germ cells) Proteins and RNAs Storage of proteins and RNAs that are required for germ cell developmentEM (3)[56,57,58]
RNA transport granule (neuronal cell) mRNAs and proteins Storage and transport of mRNAsCM (1)[59,60]
(1) CM, confocal microscopy; (2) smFISH, single molecular fluorescence in situ hybridization; (3) EM, electron microscopy.
Table 2. Components, roles, and observation procedures of droplets in the nucleus.
Table 2. Components, roles, and observation procedures of droplets in the nucleus.
DropletMain ComponentRoleObservationRef.
Nucleolus Proteins and canonical nucleic acids, non-coding RNA Ribosome biogenesis FM 1[8,63,64]
Histone locus body NPAT 10, SLBP 11, the U7 spliceosomal snRNP-specific components, such as Sm proteins, LSm10 and LSm11, and the U7 spliceosomal snRNA, FLASH 12Histone mRNA biogenesis BFM 2[65,66]
Heterochromatin HP1 13, nucleosomal DNA Promote the formation of heterochromatin CM 3[67,68,69,70,71,72]
Nuclear pore central transport channel and nuclear pore complex Nups 14, FG 15Chromosomal translocations, change in protein expression levels.
Fuse with oncoproteins, nuclear import/export
HS-AFM 4, FM 1[73,74,75,76]
Nuclear specklesRNAs and proteinsmRNA splicingCM 3[77,78]
DNA damage foci Rad52 DNA repair proteins DNA damage repair Live-cell CM 5[79,80]
Gem SMN complex, ZPR1, GEMIN2–8 16. Storage aid histone, mRNA processingCM 3[81,82,83]
PcG body Transcriptional repression IM 6, EM 7[84,85]
ParaspeckleNONO 17, PSP1 17, PSP2 17, SFPQ 18, CFIm68 19, CFIm, hnRNPs 20 NEAT1 21RNA processingCM 3[86,87,88]
OPT domain The RNA polymerases and the general transcription factors Transcriptional regulation FM 1, EM 7[89,90,91]
Cajal body Coilin, CB-specific RNAs Assembly and/or modification of splicing machineryBFM 2[66,92,93]
Perinuclear compartment RNA-binding proteins and pol III RNA Associated with malignancy EM 7, IM 6[92]
Cleavage body snRNPs 22, p80-coilin protein, RNA polymerases, transcriptional factors, nucleolar constituents mRNA processing IL 8[94]
Nuclear bodies (NBs) Protein and non-protein components, heat shock transcription factors, HSF1 23 and HSF2 24, SAF-B 25, Sam68 26, SRSF1 27, SRSF7 27 and SRSF9 27. RNA Pol II. Regulation of genome functionIM 6, SRM 9[66,95]
PML body DAXX, SUMO 28Transcriptional regulation; apoptosis signaling; antiviral defense EM 7[82,96]
1 FM, fluorescence microscopy; 2 BFM: bright-field microscopy; 3 CM, confocal microscopy; 4 HS-AFM, high-speed atomic force microscopy; 5 live-cell CM; 6 IM, immunofluorescence microscopy; 7 EM, electron microscopy; 8 IL, immunofluorescence labelling; 9 SRM, super-resolution microscopy; 10 NPAT, factors required for processing histone pre-mRNAs, nuclear protein, ataxia–telangiectasia locus; 11 SLBP, stem-loop binding protein; 12 FLASH, FLICE-associated huge protein; 13 HP1, heterochromatin protein; 14 Nups, nucleoporins; 15 FG, phenylalanine–glycine; 16 GEMIN2–8, gem-associated proteins 2–8; 17 NONO, PSP1, PSP2, paraspeckle component proteins; 18 SFPQ, splicing factor proline/glutamine-rich; 19 CFIm68, mammalian cleavage factor I 68; 20 hnRNPs, Heterogeneous nuclear ribonucleoproteins; 21 NEAT1, long ncRNA nuclear-enriched abundant transcript 1; 22 snRNPs, small nuclear ribonucleoproteins; 23 HSF1, heat shock factor 1; 24 HSF2, heat shock factor 2; 25 SAF-B, scaffold attachment factor B; 26 Sam68: Src-associated mitosis 68 kDa protein; 27 SRSF1, SRSF7, SRSF9, SRSF family members; 28 SUMO, a potent repressor of transcription and modulator of apoptosis, ubiquitin-like protein.
Table 3. Components, roles, and observation procedures of droplets in membranes.
Table 3. Components, roles, and observation procedures of droplets in membranes.
DropletMain ComponentRoleObservationRef.
Membrane cluster Triacylglycerols, phospholipid, protein Lipid uptake, distribution, storage, and use in the cell.-[98]
Synaptic densities Actin’s cytoskeleton, kinases, phosphatases, and regulators, GTPases, subunits of AMPA and NMDA receptors, Catenin, N-CadherinNeurotransmissionAEM 1[99,100]
Focal adhesionsp130Cas (‘Cas’) and FAK 7Cell adhesion/migration SDCM 2[101,102]
Nephrin clusters Cytoplasmic adaptor protein Nck, the nephrin–Nck–N-WASp complex Glomerular filtration barrier SRSIM 5[13,28,103]
TCR clustersLAT 8Immune synapseTIRF 3[104,105,106]
Podosomes F-actin and its regulatory molecules, structural proteins Cell adhesion/migration PCM 4[107,108]
Actin patches Actin-associated proteins, upstream signaling molecules EndocytosisEM 6[109,110]
1 AEM, advanced electron microscopy; 2 SDCM, spinning disk confocal microscopy; 3 TIRF, total internal reflection fluorescence microscopy; 4 PCM, phase-contrast microscopy; 5 SRSIM, super-resolution structured illumination microscopy; 6 EM, electron microscopy; 7 FAK, focal adhesion kinase; 8 LAT, linker protein for activation of T cells.
Table 4. Components, roles, and observation procedures of droplet enzymes.
Table 4. Components, roles, and observation procedures of droplet enzymes.
EnzymeRoleObservation Ref.
Horseradish peroxidase (HRP) Catalyst (horseradish peroxidase (HRP) CM 1[115]
Glucose oxidase (GOx)Catalyst (oxidation of β-d-glucose to d-glucono-δ-lactone)CM 1[119]
HexokinaseCatalyst, catalyzing the phosphorylation of keto- and aldohexosesOM 2[120]
LipaseFat breakdown CM 1[121,122]
Hammerhead ribozymeCleavage and ligation of RNA molecule FRET 6, CD 4, CM 1[123,124]
Pfk2, Eno1, Eno2, Fba1 GlycolysisFM 3[125]
GIT1GTPase activatorFRAP 5, CM 1[126]
HSF1Transcription factor FM 3[127,128]
NELFETranscriptional regulationFM 3[129]
p53Transcription factorFM 3[130,131,132,133,134,135]
PLK4Serine/threonine-protein kinaseCM 1[136,137]
SOX-2Transcription factor FM 3[138]
TFE3 Transcription factor FM 3[139]
TFEBTranscription factorFRAP 5[140]
USP42Deubiquitinating enzyme FM 3[141]
YAPTranscription factor FM 3[142,143]
1 CM, confocal microscopy; 2 OM, optical microscopy; 3 FM, fluorescence microscopy; 4 CD, circular dichroism; 5 FRAP, fluorescence recovery after photobleaching; 6 FRET, fluorescence resonance energy transfer microscopy.
Table 5. Location, components, roles, and observation procedures of droplets discovered in various biological processes.
Table 5. Location, components, roles, and observation procedures of droplets discovered in various biological processes.
DropletLocationRoleMain ComponentObservationRef.
Pyrenoids (Rubisco), carboxysomesChloroplastPhotosynthesis, metabolism (Carbon fixation) Carboxysomal linker proteins CsoS2 and CcmM, Rubisco large subunitMicroscopy and sedimentation assay[144,145,146]
Wnt dropletCell cytoplasmStem cell differentiation, controlling Wnt pathwayScaffold proteins and kinases that regulate β-catenin stabilityCRISPR-engineered fluorescent tags, optogenetic tools[147]
YTHDC droplet (nuclear bodies)NucleusAML cell survival, differentiation state, leukemogenesisYTHDC1 protein, m6 A-containing RNAIF 1, SEM 2.[148]
LDAM 3HippocampusPromotion of pathogenesis, neuroinflammation LipidCARS 4[149]
Lipid dropletsCell cytoplasm (Stem cell)Skeletal muscle satellite cell fate determinationLipidTEM 5[150]
Plant lipid droplets: LD-Erm LD-Peroxisomes Plant cellUnknown Triacylglycerols (TAGs), sterol esters (SEs) FM 6, CM 7[151]
1 IF, immunofluorescent imaging; 2 SEM, scanning electron microscopy; 3 LDAM, hippocampus lipid droplet accumulating microglia; 4 CARS, coherent anti-Stokes Raman scattering microscopy; 5 TEM, transmission electron microscopy; 6 FM, fluorescence microscope; 7 CM, confocal microscopy.
Table 6. Components, application, and observation procedures of artificial droplets.
Table 6. Components, application, and observation procedures of artificial droplets.
DropletComponentObservationApplicationRef.
Adiposomes (artificial lipid droplets (ALDs))Phospholipids and neutral lipids such as TAG LM 1, EM 2Potential usage in drug delivery.[156]
Cell-sized aqueous/aqueous microdroplets (CAMDs) PEG 3 and DEX 4, actinFM 5Provide cell-like crowded microenvironments[157]
Microfluidic platform in a defined patternHexadecane/squalene with dissolved lipids Broad range of applications in the field of artificial cells, bioreactors, and pharmacological studies.[158]
Lipase-stabilized tributyrin microcompartment and amylose-polymer-stabilized 2-ethyl-1-hexanol microcompartmentAmy-PNIPAAm 6, BSA-PNIPAAm 7, LipaseOM 8Synthetic biology, bottom-up reaction[159]
G-quadruplex-forming oligonucleotides and R-rich oligopeptidesFMR1 RNA, C9orf72 RNA, peptide derived from FMRPCM 9Droplet redissolution in a sequence-specific manner[160]
1 LM, light microscopy; 2 EM, electron microscopy; 3 PEG, poly (ethylene glycol); 4 DEX, dextran; 5 FM, fluorescence microscopy; 6 Amy-PNIPAAm, Poly(N-isopropylacrylamide; 7 BSA-PNIPAAm, bovine serum albumin-Poly(N-isopropylacrylamide; 8 OM, optical microscopy; 9 CM, confocal microscopy.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shil, S.; Tsuruta, M.; Kawauchi, K.; Miyoshi, D. Biomolecular Liquid–Liquid Phase Separation for Biotechnology. BioTech 2023, 12, 26. https://doi.org/10.3390/biotech12020026

AMA Style

Shil S, Tsuruta M, Kawauchi K, Miyoshi D. Biomolecular Liquid–Liquid Phase Separation for Biotechnology. BioTech. 2023; 12(2):26. https://doi.org/10.3390/biotech12020026

Chicago/Turabian Style

Shil, Sumit, Mitsuki Tsuruta, Keiko Kawauchi, and Daisuke Miyoshi. 2023. "Biomolecular Liquid–Liquid Phase Separation for Biotechnology" BioTech 12, no. 2: 26. https://doi.org/10.3390/biotech12020026

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop