Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ACS Synth Biol ; 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39353593

RESUMO

Artificial cells with reconstructed cellular functions could serve as practical protocell models for studying the early cellular life on the Earth. Investigating the viability of protocell models in extreme environments where life may have arisen is important for advancing origin-of-life research. Here, we tested the survivability of lipid membrane vesicles in deep-sea environments. The vesicles were submerged in the deep-sea floor with a human-occupied vehicle. Although most of the vesicles were broken, some vesicles maintained a spherical shape after the dives. When a cell-free protein synthesis system was encapsulated inside, a few vesicles remained even after a 1,390 m depth dive. Interestingly, such artificial cells could subsequently synthesize protein in a nutrient-rich buffer solution. Together with on shore experiments showing artificial cells synthesized protein under high pressure, our results suggest artificial cells may be able to express genes in deep-sea environments where thermal energy is available from hydrothermal vents.

2.
Adv Funct Mater ; 34(28)2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-39386164

RESUMO

Blood scarcity is one of the main causes of healthcare disruptions worldwide, with blood shortages occurring at an alarming rate. Over the last decades, blood substitutes has aimed at reinforcing the supply of blood, with several products (e.g., hemoglobin-based oxygen carriers, perfluorocarbons) achieving a limited degree of success. Regardless, there is still no widespread solution to this problem due to persistent challenges in product safety and scalability. In this Review, we describe different advances in the field of blood substitution, particularly in the development of artificial red blood cells, otherwise known as engineered erythrocytes. We categorize the different strategies into natural, synthetic, or hybrid approaches, and discuss their potential in terms of safety and scalability. We identify synthetic engineered erythrocytes as the most powerful approach, and describe erythrocytes from a materials engineering perspective. We review their biological structure and function, as well as explore different methods of assembling a material-based cell. Specifically, we discuss how to recreate size, shape, and deformability through particle fabrication, and how to recreate the functional machinery through synthetic biology and nanotechnology. We conclude by describing the versatile nature of synthetic erythrocytes in medicine and pharmaceuticals and propose specific directions for the field of erythrocyte engineering.

3.
Adv Biol (Weinh) ; : e2400353, 2024 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-39334525

RESUMO

The regulation of protein uptake and secretion by cells is paramount for intercellular signaling and complex multicellular behavior. Mimicking protein-mediated communication in artificial cells holds great promise to elucidate the underlying working principles, but remains challenging without the stimulus-responsive regulatory machinery of living cells. Therefore, systems to precisely control when and where protein release occurs should be incorporated in artificial cells. Here, a light-activatable TEV protease (LaTEV) is presented that enables spatiotemporal control over protein release from a coacervate-based artificial cell platform. Due to the presence of Ni2+-nitrilotriacetic acid moieties within the coacervates, His-tagged proteins are effectively sequestered into the coacervates. LaTEV is first photocaged, effectively blocking its activity. Upon activation by irradiation with 365 nm light, LaTEV cleaves the His-tags from sequestered cargo proteins, resulting in their release. The successful blocking and activation of LaTEV provides control over protein release rate and triggerable protein release from specific coacervates via selective irradiation. Furthermore, light-activated directional transfer of proteins between two artificial cell populations is demonstrated. Overall, this system opens up avenues to engineer light-responsive protein-mediated communication in artificial cell context, which can advance the probing of intercellular signaling and the development of protein delivery platforms.

4.
Angew Chem Int Ed Engl ; : e202414970, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39348462

RESUMO

Lipid nanoparticles have important applications as biomedical delivery platforms and broader engineering biology applications in artificial cell technologies. These emerging technologies often require changes in the shape and topology of biological or biomimetic membranes. Here we show that topologically-active lyotropic liquid crystal nanoparticles (LCNPs) can trigger such transformations in the membranes of giant unilamellar vesicles (GUVs). Monoolein (MO) LCNPs, cubosomes with an internal nanostructure of space group Im3m incorporate into 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) GUVs creating excess membrane area with stored curvature stress. Using time-resolved fluorescence confocal and lattice light sheet microscopy, we observe and characterise various life-like dynamic events in these GUVs, including growth, division, tubulation, membrane budding and fusion. Our results shed new light on the interactions of LCNPs with bilayer lipid membranes, providing insights relevant to how these nanoparticles might interact with cellular membranes during drug delivery and highlighting their potential as minimal triggers of topological transitions in artificial cells.

5.
Proc Natl Acad Sci U S A ; 121(36): e2404790121, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39186653

RESUMO

Eukaryotic cells are characterized by multiple chemically distinct compartments, one of the most notable being the nucleus. Within these compartments, there is a continuous exchange of information, chemicals, and signaling molecules, essential for coordinating and regulating cellular activities. One of the main goals of bottom-up synthetic biology is to enhance the complexity of synthetic cells by establishing functional compartmentalization. There is a need to mimic autonomous signaling between compartments, which in living cells, is often regulated at the genetic level within the nucleus. This advancement is key to unlocking the potential of synthetic cells as cell models and as microdevices in biotechnology. However, a technological bottleneck exists preventing the creation of synthetic cells with a defined nucleus-like compartment capable of genetically programmed intercompartment signaling events. Here, we present an approach for creating synthetic cells with distinct nucleus-like compartments that can encapsulate different biochemical mixtures in discrete compartments. Our system enables in situ protein expression of membrane proteins, enabling autonomous chemical communication between nuclear and cytoplasmic compartments, leading to downstream activation of enzymatic pathways within the cell.


Assuntos
Células Artificiais , Núcleo Celular , Biologia Sintética , Biologia Sintética/métodos , Núcleo Celular/metabolismo , Núcleo Celular/genética , Células Artificiais/metabolismo , Transdução de Sinais , Citoplasma/metabolismo , Comunicação Celular
6.
ACS Appl Mater Interfaces ; 16(32): 43016-43025, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39088740

RESUMO

Motile droplets using Marangoni convection are attracting attention for their potential as cell-mimicking small robots. However, the motion of droplets relative to the internal and external environments that generate Marangoni convection has not been quantitatively described. In this study, we used an aqueous two-phase system [poly(ethylene glycol) (PEG) and dextran] in an elongated chamber to generate motile dextran droplets in a constant PEG concentration gradient. We demonstrated that dextran droplets move by Marangoni convection, resulting from the PEG concentration gradient and the active transport of PEG and dextran into and out of the motile dextran droplet. Furthermore, by spontaneously incorporating long DNA into the dextran droplets, we achieved cell-like motility changes controlled by coexisting environment-sensing molecules. The DNA changes its position within the droplet and motile speed in response to external conditions. In the presence of Mg2+, the coil-globule transition of DNA inside the droplet accelerates the motile speed due to the decrease in the droplet's dynamic viscosity. Globule DNA condenses at the rear part of the droplet along the convection, while coil DNA moves away from the droplet's central axis, separating the dipole convections. These results provide a blueprint for designing autonomous small robots using phase-separated droplets, which change the mobility and molecular distribution within the droplet in reaction with the environment. It will also open unexplored areas of self-assembly mechanisms through phase separation under convections, such as intracellular phase separation.


Assuntos
DNA , Dextranos , Polietilenoglicóis , Dextranos/química , Polietilenoglicóis/química , DNA/química , Viscosidade , Soluções
7.
ACS Synth Biol ; 13(3): 888-900, 2024 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-38359048

RESUMO

Methanol has gained substantial attention as a substrate for biomanufacturing due to plentiful stocks and nonreliance on agriculture, and it can be sourced renewably. However, due to inevitable complexities in cell metabolism, microbial methanol conversion requires further improvement before industrial applicability. Here, we present a novel, parallel strategy using artificial cells to provide a simplified and well-defined environment for methanol utilization as artificial methylotrophic cells. We compartmentalized a methanol-utilizing enzyme cascade, including NAD-dependent methanol dehydrogenase (Mdh) and pyruvate-dependent aldolase (KHB aldolase), in cell-sized lipid vesicles using the inverted emulsion method. The reduction of cofactor NAD+ to NADH was used to quantify the conversion of methanol within individual artificial methylotrophic cells via flow cytometry. Compartmentalization of the reaction cascade in liposomes led to a 4-fold higher NADH production compared with bulk enzyme experiments, and the incorporation of KHB aldolase facilitated another 2-fold increase above the Mdh-only reaction. This methanol-utilizing platform can serve as an alternative route to speed up methanol biological conversion, eventually shifting sugar-based bioproduction toward a sustainable methanol bioeconomy.


Assuntos
Células Artificiais , Metanol , Metanol/metabolismo , NAD/metabolismo , Frutose-Bifosfato Aldolase , Aldeído Liases/metabolismo
8.
Synth Biol (Oxf) ; 9(1): ysae004, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38327596

RESUMO

Synthetic cells are a novel class of cell-like bioreactors, offering the potential for unique advancements in synthetic biology and biomedicine. To realize the potential of those technologies, synthetic cell-based drugs need to go through the drug approval pipeline. Here, we discussed several regulatory challenges, both unique to synthetic cells, as well as challenges typical for any new biomedical technology. Overcoming those difficulties could bring transformative therapies to the market and will create a path to the development and approval of cutting-edge synthetic biology therapies. Graphical Abstract.

9.
Cell Syst ; 15(1): 49-62.e4, 2024 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-38237551

RESUMO

Synthetic minimal cells are a class of bioreactors that have some, but not all, functions of live cells. Here, we report a critical step toward the development of a bottom-up minimal cell: cellular export of functional protein and RNA products. We used cell-penetrating peptide tags to translocate payloads across a synthetic cell vesicle membrane. We demonstrated efficient transport of active enzymes and transport of nucleic acid payloads by RNA-binding proteins. We investigated influence of a concentration gradient alongside other factors on the efficiency of the translocation, and we show a method to increase product accumulation in one location. We demonstrate the use of this technology to engineer molecular communication between different populations of synthetic cells, to exchange protein and nucleic acid signals. The synthetic minimal cell production and export of proteins or nucleic acids allows experimental designs that approach the complexity and relevancy of natural biological systems. A record of this paper's transparent peer review process is included in the supplemental information.


Assuntos
Células Artificiais , Peptídeos Penetradores de Células , Ácidos Nucleicos , Ácidos Nucleicos/metabolismo , Células Artificiais/metabolismo , Proteínas , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/metabolismo
10.
Adv Healthc Mater ; 13(12): e2303699, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38277695

RESUMO

Artificial cells are engineered units with cell-like functions for different purposes including acting as supportive elements for mammalian cells. Artificial cells with minimal liver-like function are made of alginate and equipped with metalloporphyrins that mimic the enzyme activity of a member of the cytochrome P450 family namely CYP1A2. The artificial cells are employed to enhance the dealkylation activity within 3D bioprinted structures composed of HepG2 cells and these artificial cells. This enhancement is monitored through the conversion of resorufin ethyl ether to resorufin. HepG2 cell aggregates are 3D bioprinted using an alginate/gelatin methacryloyl ink, resulting in the successful proliferation of the HepG2 cells. The composite ink made of an alginate/gelatin liquid phase with an increasing amount of artificial cells is characterized. The CYP1A2-like activity of artificial cells is preserved over at least 35 days, where 6 nM resorufin is produced in 8 h. Composite inks made of artificial cells and HepG2 cell aggregates in a liquid phase are used for 3D bioprinting. The HepG2 cells proliferate over 35 days, and the structure has boosted CYP1A2 activity. The integration of artificial cells and their living counterparts into larger 3D semi-synthetic tissues is a step towards exploring bottom-up synthetic biology in tissue engineering.


Assuntos
Bioimpressão , Citocromo P-450 CYP1A2 , Impressão Tridimensional , Humanos , Células Hep G2 , Bioimpressão/métodos , Citocromo P-450 CYP1A2/metabolismo , Alginatos/química , Gelatina/química , Engenharia Tecidual/métodos , Proliferação de Células/efeitos dos fármacos , Metaloporfirinas/química , Metaloporfirinas/farmacologia
11.
Adv Sci (Weinh) ; 11(6): e2308030, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38054641

RESUMO

Cells are small, closed spaces filled with various types of macromolecules. Although it is shown that the characteristics of biochemical reactions in vitro are quite different from those in living cells, the role of the co-existence of various macromolecules in cell-size space remains still elusive. Here, using a constructive approach, it is demonstrated that the co-existence of various macromolecules themselves has the ability to tune protein localization for spatiotemporal regulation and a biochemical reaction system in a cell-size space. Both experimental and theoretical analyses reveal that enhancement of interfacial effects by a large surface-area-to-volume ratio facilitates membrane localization of molecules in the cell-size space, and the interfacial effects are alleviated by competitive binding to lipid membranes among multiple proteins even if their membrane affinities are weak. These results indicate that competition for membrane binding among various macromolecules in the cell-size space plays a role in regulating the spatiotemporal molecular organization and biochemical reaction networks. These findings shed light on the importance of surrounding molecules for biochemical reactions using purified elements in small spaces.


Assuntos
Proteínas , Proteínas/química , Transporte Proteico
12.
Adv Sci (Weinh) ; 11(8): e2305837, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37984885

RESUMO

The exponential growth of research on artificial cells and organelles underscores their potential as tools to advance the understanding of fundamental biological processes. The bottom-up construction from a variety of building blocks at the micro- and nanoscale, in combination with biomolecules is key to developing artificial cells. In this review, artificial cells are focused upon based on compartments where polymers are the main constituent of the assembly. Polymers are of particular interest due to their incredible chemical variety and the advantage of tuning the properties and functionality of their assemblies. First, the architectures of micro- and nanoscale polymer assemblies are introduced and then their usage as building blocks is elaborated upon. Different membrane-bound and membrane-less compartments and supramolecular structures and how they combine into advanced synthetic cells are presented. Then, the functional aspects are explored, addressing how artificial organelles in giant compartments mimic cellular processes. Finally, how artificial cells communicate with their surrounding and each other such as to adapt to an ever-changing environment and achieve collective behavior as a steppingstone toward artificial tissues, is taken a look at. Engineering artificial cells with highly controllable and programmable features open new avenues for the development of sophisticated multifunctional systems.


Assuntos
Células Artificiais , Polímeros/química , Organelas
13.
ACS Nano ; 17(23): 23772-23783, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38038709

RESUMO

There is currently no plausible path for the emergence of a self-replicating protocell, because prevalent formulations of model protocells are built with fatty acid vesicles that cannot withstand the concentrations of Mg2+ needed for the function and replication of nucleic acids. Although prebiotic chelates increase the survivability of fatty acid vesicles, the resulting model protocells are incapable of growth and division. Here, we show that protocells made of mixtures of cyclophospholipids and fatty acids can grow and divide in the presence of Mg2+-citrate. Importantly, these protocells retain encapsulated nucleic acids during growth and division, can acquire nucleotides from their surroundings, and are compatible with the nonenzymatic extension of an RNA oligonucleotide, chemistry needed for the replication of a primitive genome. Our work shows that prebiotically plausible mixtures of lipids form protocells that are active under the conditions necessary for the emergence of Darwinian evolution.


Assuntos
Células Artificiais , Ácidos Nucleicos , RNA , Ácidos Graxos , Citratos
14.
Mater Today Bio ; 23: 100877, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38075249

RESUMO

Artificial cells have received much attention in recent years as cell mimics with typical biological functions that can be adapted for therapeutic and diagnostic applications, as well as having an unlimited supply. Although remarkable progress has been made to construct complex multifunctional artificial cells, there are still significant differences between artificial cells and natural cells. It is therefore important to understand the techniques and challenges for the fabrication of artificial cells and their applications for further technological advancement. The key concepts of top-down and bottom-up methods for preparing artificial cells are summarized, and the advantages and disadvantages of the bottom-up methods are compared and critically discussed in this review. Potential applications of artificial cells as drug carriers (microcapsules), as signaling regulators for coordinating cellular communication and as bioreactors for biomolecule fabrication, are further discussed. The challenges and future trends for the development of artificial cells simulating the real activities of natural cells are finally described.

15.
Cancers (Basel) ; 15(24)2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38136352

RESUMO

Artificial cells have been extensively used in many fields, such as nanomedicine, biotherapy, blood substitutes, drug delivery, enzyme/gene therapy, cancer therapy, and the COVID-19 vaccine. The unique properties of superparamagnetic Fe3O4 nanoparticles have contributed to increased interest in using superparamagnetic artificial cells (PLGA-Fe3O4 micro/nanocapsules) for targeted therapy. In this review, the preparation methods of Fe3O4 NPs and superparamagnetic artificial cell PLGA-drug-Fe3O4 micro/nanocapsules are discussed. This review also focuses on the recent progress of superparamagnetic PLGA-drug-Fe3O4 micro/nanocapsules as targeted therapeutics. We shall concentrate on the use of superparamagnetic artificial cells in the form of PLGA-drug-Fe3O4 nanocapsules for magnetic hyperthermia/photothermal therapy and cancer therapies, including lung breast cancer and glioblastoma.

17.
Adv Sci (Weinh) ; 10(34): e2302461, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37807811

RESUMO

In the quest to produce artificial cells, one key challenge that remains to be solved is the recreation of a complex cellular membrane. Among the existing models, giant unilamellar vesicles (GUVs) are particularly interesting due to their intrinsic compartmentalisation ability and their resemblance in size and shape to eukaryotic cells. Many techniques have been developed to produce GUVs all having inherent advantages and disadvantages. Here, the authors show that fluorinated silica nanoparticles (FNPs) used to form Pickering emulsions in a fluorinated oil can destabilise lipid nanosystems to template the formation of GUVs. This technique enables GUV production across a broad spectrum of buffer conditions, while preventing the leakage of the encapsulated components into the oil phase. Furthermore, a simple centrifugation process is sufficient for the release of the emulsion-trapped GUVs, bypassing the need to use emulsion-destabilising chemicals. With fluorescent FNPs and transmission electron microscopy, the authors confirm that FNPs are efficiently removed, producing contaminant-free GUVs. Further experiments assessing the lateral diffusion of lipids and unilamellarity of the GUVs demonstrate that they are comparable to GUVs produced via electroformation. Finally, the ability of incorporating transmembrane proteins is demonstrated, highlighting the potential of this method for the production of GUVs for artificial cell applications.


Assuntos
Células Artificiais , Lipossomas Unilamelares , Emulsões , Membrana Celular , Proteínas de Membrana
18.
Int J Mol Sci ; 24(18)2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37762444

RESUMO

The recent and important advances in bottom-up synthetic biology (SB), in particular in the field of the so-called "synthetic cells" (SCs) (or "artificial cells", or "protocells"), lead us to consider the role of wetware technologies in the "Sciences of Artificial", where they constitute the third pillar, alongside the more well-known pillars hardware (robotics) and software (Artificial Intelligence, AI). In this article, it will be highlighted how wetware approaches can help to model life and cognition from a unique perspective, complementary to robotics and AI. It is suggested that, through SB, it is possible to explore novel forms of bio-inspired technologies and systems, in particular chemical AI. Furthermore, attention is paid to the concept of semantic information and its quantification, following the strategy recently introduced by Kolchinsky and Wolpert. Semantic information, in turn, is linked to the processes of generation of "meaning", interpreted here through the lens of autonomy and cognition in artificial systems, emphasizing its role in chemical ones.


Assuntos
Células Artificiais , Vida Artificial , Inteligência Artificial , Biologia Sintética , Cognição
19.
Small Methods ; 7(12): e2300257, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37599260

RESUMO

Modern medical research develops interest in sophisticated artificial nano- and microdevices for future treatment of human diseases related to biological dysfunctions. This covers the design of protocells capable of mimicking the structure and functionality of eukaryotic cells. The authors use artificial organelles based on trypsin-loaded pH-sensitive polymeric vesicles to provide macrophage-like digestive functions under physiological conditions. Herein, an artificial cell is established where digestive artificial organelles (nanosize) are integrated into a protocell (microsize). With this method, mimicking crossing of different biological barriers, capture of model protein pathogens, and compartmentalized digestive function are possible. This allows the integration of different components (e.g., dextran as stabilizing block) and the diffusion of pathogens in simulated cytosolic environment under physiological conditions. An integrated characterization approach is carried out, with identifying electrospray ionization mass spectrometry as an excellent detection method for the degradation of a small peptide such as ß-amyloid. The degradation of model enzymes is measured by enzyme activity assays. This work is an important contribution to effective biomimicry with the design of cell-like functions having potential for therapeutic action.


Assuntos
Células Artificiais , Humanos , Células Artificiais/química , Células Artificiais/metabolismo , Biomimética/métodos , Proteínas/química , Macrófagos , Digestão
20.
Proc Natl Acad Sci U S A ; 120(35): e2307772120, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37603747

RESUMO

Artificial cells are biomimetic structures formed from molecular building blocks that replicate biological processes, behaviors, and architectures. Of these building blocks, hydrogels have emerged as ideal, yet underutilized candidates to provide a gel-like chassis in which to incorporate both biological and nonbiological componentry which enables the replication of cellular functionality. Here, we demonstrate a microfluidic strategy to assemble biocompatible cell-sized hydrogel-based artificial cells with a variety of different embedded functional subcompartments, which act as engineered synthetic organelles. The organelles enable the recreation of increasingly biomimetic behaviors, including stimulus-induced motility, content release through activation of membrane-associated proteins, and enzymatic communication with surrounding bioinspired compartments. In this way, we showcase a foundational strategy for the bottom-up construction of hydrogel-based artificial cell microsystems which replicate fundamental cellular behaviors, paving the way for the construction of next-generation biotechnological devices.


Assuntos
Células Artificiais , Biomimética , Hidrogéis , Comunicação , Organelas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA