Skip to main content

A potential paradigm in CRISPR/Cas systems delivery: at the crossroad of microalgal gene editing and algal-mediated nanoparticles

Abstract

Microalgae as the photosynthetic organisms offer enormous promise in a variety of industries, such as the generation of high-value byproducts, biofuels, pharmaceuticals, environmental remediation, and others. With the rapid advancement of gene editing technology, CRISPR/Cas system has evolved into an effective tool that revolutionised the genetic engineering of microalgae due to its robustness, high target specificity, and programmability. However, due to the lack of robust delivery system, the efficacy of gene editing is significantly impaired, limiting its application in microalgae. Nanomaterials have become a potential delivery platform for CRISPR/Cas systems due to their advantages of precise targeting, high stability, safety, and improved immune system. Notably, algal-mediated nanoparticles (AMNPs), especially the microalgae-derived nanoparticles, are appealing as a sustainable delivery platform because of their biocompatibility and low toxicity in a homologous relationship. In addition, living microalgae demonstrated effective and regulated distribution into specified areas as the biohybrid microrobots. This review extensively summarised the uses of CRISPR/Cas systems in microalgae and the recent developments of nanoparticle-based CRISPR/Cas delivery systems. A systematic description of the properties and uses of AMNPs, microalgae-derived nanoparticles, and microalgae microrobots has also been discussed. Finally, this review highlights the challenges and future research directions for the development of gene-edited microalgae.

Graphical Abstract

Introduction

Following the urgent demand for energy and food, it is critical to discover an alternative source to alleviate these issues. Due to simple cultivation conditions, rapid growth rate and high photosynthetic efficiency, microalgae have emerged as the promising renewable energy resource. They also have a significant potential to construct a productive biorefinery which is the process of converting carbon dioxide into value-added compounds, for example proteins, vitamins, fatty acids, carotenoids, and nucleic acids. Microalgae are recognised as the third generation of biofuels with the potential for high-density cultivation system designs without compete with food or agricultural crops [1]. Additionally, microalgae are now capable of producing wide range of fuels, including biodiesel [2], hydrogen [3], syngas [4], etc. They may also serve as a raw material for functional food, natural dyes, and pharmaceutical drugs [5]. Studies on microalgae have primarily concentrated on developing integrative process and culture techniques, such as photobioreactor designs [6], harvesting approaches in downstream processes [7] and extraction techniques for high-value compounds [8]. However, this field still has some drawbacks, such as the low carbon fixation efficiency, low lipid accumulation rate and long cultivation period, which need to be overcome with more robust technology.

Many microalgal genomes have been sequenced to date, providing a definite genetic background and a compelling argument for genetic modification. Zinc finger nuclease (ZFN) [9], transcription activator-like effector-based nuclease (TALEN) [10] and the clustered regularly interspaced short palindromic repeats-associated system (CRISPR/Cas system) are examples of gene editing tools that offer an useful approach to deal with microalgal issues and achieve mass production. ZFN and TALEN are now severely constrained due to low editing efficiency, costly and laborious vector construction. The CRISPR/Cas system, in contrast, has become a reliable genome editing platform for gene correction, transcriptional regulation, disease modelling, and nucleic acids imaging. Its advantages include simplicity of target design, high editing efficiency, multiplex knock-in/out ability, low cost, and a quick cycle time [11]. Since then, CRISPR/Cas systems have been used as the treatment of infectious and metabolic diseases, creating sustainable techniques to produce chemicals and fuel as well as enhance the features of food crops. The disadvantages of CRISPR/Cas systems are off-target effects [12], variable efficiency, and inactive mutant [13, 14], despite the implementation of some strategies, for example rational design and modification of sgRNA, application of Cas variants, and improvement of the repair efficiency of HDR pathway.

An ideal delivery vector is essential for the efficient intracellular distribution of CRISPR components. Due to their superior biocompatibility in a homologous relationship, algal-mediated nanoparticles (AMNPs) have a tremendous potential to provide the CRISPR/Cas system for microalgal genetic editing. Furthermore, AMNPs provide a variety of properties, including anti-bacterial, anti-fungal, anti-cancer, anti-fouling, bioremediation, and biosensing activities [15]. This, green synthesis of AMNPs has drawn a lot of interest because it is safe, simple, sustainable, cost-effective, and eco-friendly. This review provides a thorough summary of the CRISPR/Cas system's delivery status using nanoparticles (NPs), with a focus on providing an in-depth description of AMNPs from the perspectives of their synthesis, features, classifications, and numerous applications. Some strategies, like as the use of a different Cas nuclease, better codon harmonisation, the addition of introns, and proper removal of the cell wall, have been suggested to tackle the limitations of CRISPR/Cas system in microalgae [16]. This review also examines the prospects of the AMNPs-delivered CRISPR/Cas system in microalgae, which offers a clear and crucial roadmap for advancing the use of gene editing in microalgae.

Research status of microalgae gene editing

Although ZFN and TALEN have been applied in diatoms Phaeodactylum tricornutum and Chlamydomonas reinhardtii [17, 18], both technologies have not been widely used due to the limitations of laborious design steps, low editing efficiency and high off-target events. In comparison to ZFN and TALEN, the CRISPR/Cas system is regarded as a more sophisticated editing tool with several benefits, including the ease of design, increased effectiveness, and capacity to introduce mutations in many genes simultaneously [19]. In the past ten years, the CRISPR/Cas system has evolved from a groundbreaking genome-editing tool used in bacteria to a significant gene-editing tool utilised in plants, animals, and humans [20]. As a result, CRISPR technologies have advanced innovation by changing numerous eukaryotic genomes [21]. In the past, the CRISPR/Cas editing technology has been used to modify a number of microalgae species, including Nannochloropsis oceanica [22], P. tricornutum [23], Thalassiosira pseudonana [24], Chlorella vulgaris [25], and other microalgae species [26,27,28,29,30,31,32,33,34,35,36,37,38,39,40]. The editing process, transformation process, editing effectiveness, and experimental outcomes have all been explained in depth, as seen in Table 1 [41].

Table 1 Application status of CRISPR/Cas system in microalgae

The problem of ineffective intracellular delivery with gene editing in microalgae persists despite varied success rates. An ideal transformation strategy and delivery vector are required to transport foreign DNA into cells. Among the transformation methods, such as electroporation [42], glass beads [43], particle bombardment [44], and Agrobacterium tumefaciens-mediated transformation [45], electroporation is the preferred method for microalgae transformation because it is rapid and highly effective in the generation of transformants (approximately 0.4 ~ 3 × 103 transformants per µg of exogenous DNA) [42, 46]. There are few delivery carriers in CRISPR systems, such as viral vectors [47], extracellular vehicles [48], cell-penetrating peptides [49], and etc. AMNPs have a greater potential to deliver CRISPR/Cas components in microalgae gene editing due to better biocompatibility in a homologous relationship [50] than other delivery carriers in CRISPR systems. Their detailed delivery in microalgae have been described systematically as follows.

NP-based delivery system for CRISPR/Cas components

Characteristics of different delivery systems

Given the existing challenges of the CRISPR/Cas system, including as off-target effects [51], poor delivery efficiency [52], and unintended adverse effects [53], various techniques have been proposed to overcome these concerns [14, 51, 54]. Choosing the best delivery system has become the most important step, with strict requirements to boost not just loading efficiency, but also correct delivery to the specified location [55].

So far, several delivery mechanisms have sprung up, resulting in an exponential expansion in the distribution of CRISPR/Cas components (Table 2). Delivery mechanisms consists of three types which are viral vectors, non-viral vectors, and physical and chemical approaches [48, 56,57,58,59,60]. Viral vectors including adenovirus, adeno-associated virus and lentivirus [61,62,63], and physical methods like electroporation [64], microinjection [65], hydrodynamic injection [66] and ultrasound [67], are relatively well-established strategies for delivering CRISPR/Cas systems [68]. Using these two techniques, the delivery efficiency of CRISPR/Cas components was enhanced to around 98% [60, 69,70,71], signifying a substantial achievement in the field of gene editing. Viral vectors have become the most evident method for delivering genes owing to excellent transfer efficiency and stable gene expression [72, 73].

Table 2 Summary of common delivery strategies for CRISPR/Cas9 systems

Despite ongoing improvements in viral vectors [74], their applications in the delivery of CRISPR/Cas system are still hindered by the issues with immunogenicity [75], mutagenesis [76], limited packaging capacity as well as scale-up manufacturing [77, 78]. Combinatorial distribution of multiple components is a critical challenge that may prevent widespread and flexible implementation in the future [79]. Despite its great effectiveness in the laboratory, physical delivery was less practicable for in vivo distribution due to its low scalability [80, 81]. They are more suitable for in vitro applications than clinical translations because they can be conducted at the cellular level rather than the organism level [82]. Further, the implementation of physical delivery in non-specialist laboratories and high-throughput applications is difficult because to the expensive equipment and requirement of high skilled manipulation. Improper and unoptimized physical techniques may cause inevitable cell damage or even cell death [83, 84]. Alternately, it is desirable for the CRISPR/Cas system to deliver the product by changing it directly rather than causing alterations in target cells [80]. In view of this, chemical carriers provide a novel perspective due to their low immunogenicity, adjustable cargo size, and mass-production feasibility [85]. More importantly, the ease of alteration of chemical carriers may answer future issues in terms of targeting, biosafety, loading capacity, and spatiotemporal controllability, which are desirable options for in vivo precise gene editing delivery [86, 87]. Among the many chemical vectors, nanomaterial is an efficient platform to deliver small molecule medications, genes, peptides, and diagnostic agents [88, 89]. CRISPR/Cas components could be supplied via designed NPs with advantages such as precise targeting [90], high stability [91], safety [68], enhanced immune escape [92], and combinatorial delivery of several components [60].

NPs delivery mechanism

The journey of NPs in vivo is difficult and accompanied with numerous obstacles, including disturbance of ‘protein corona’ [93], clearance by reticuloendothelial system, as well as the barriers of vasculature, extracellular matrix and endosome. In particular, the intravenous injection of NPs would face all of the difficulties of beginning from scratch, with the most challenging obstacle being the efficient access to organelles at the subcellular level [94]. The most important process for almost all successful NP-based carriers is endocytosis, which is mediated by target cells [95]. By controlling and mediating numerous signal pathways, NPs-based carriers were engulfed into endocytic vesicles through the invasion of plasma membrane [96,97,98].

Theoretically, these pathways are subdivided into five categories based on the specific lipid and transport protein types they contain, such as surface receptors, membrane lipids, and adaptor proteins. Phagocytosis, caveolin-dependent endocytosis, clathrin- and caveolin-independent endocytosis, and macropinocytosis are all types of endocytosis [99, 100] (Fig. 1a). The fate of NPs within the cells can be determined by many types of endocytosis, which depends on the factors including cell type, size, charge, and stiffness of NPs, as well as receptor interaction [101]. Among these mechanisms, the majority of receptor-bound NPs could be translocated into the cells by clathrin-mediated and caveolin-mediated endocytosis, while non-targeting NPs in small or large size will be taken up non-specifically by macropinocytosis or phagocytosis [102]. However, a certain lipid composition (mostly cholesterol) was required for clathrin- and caveolin-independent endocytosis, which was thought to be a direct entrance route [101]. However, the uptake mechanism of each item is distinct and has not yet been thoroughly investigated. When NPs become entrapped in an endosome during endocytosis, they are unable to perform their intended biological and therapeutic roles due to a lack of quick access to cytoplasm or cellular organelles. Endosomes were frequently acidified during ageing processes, with pH variations ranging from pH 6.5–6.8 to pH 5.2–6.0. Eventually, the acidic pH and enzymatic breakdown caused the cargoes to be destroyed [103]. As a result, the inability to exert biological effects is currently a key hindrance to nanomedicine efforts, necessitating the urgent development of endosome escape to increase the effectiveness of NPs delivery.

Fig. 1
figure 1

Delivery mechanism of NPs into cells. a The cellular uptake routes of NPs. b Pros and cons of CRISPR/Cas9 formats. c Different formats of CRISPR/Cas9 components into nucleus

When NPs are subjected to the harsh environment of lysosome, endosome escape is a critical step in retaining the integrity of their cargoes and exerting their effects. According to the data, the greatest distribution effectiveness of NPs made of polymers and lipids was just 1–2%, and that the remaining 98% of NPs were useless because they were trapped in endosomes [104] or recycled back to the extracellular space [105]. It is frequently regarded as the rate-limiting step in the intracellular delivery of NPs-based systems [106, 107]. Therefore, an endosome escape technique could be used to transfer CRISPR/Cas components effectively without degrading them. However, various contentious ideas have been offered as endosome escape routes, such as proton sponge effect [108], lipid fusion with endosomal membrane [109, 110], nanoparticle swelling [111], membrane destabilisation [112], and cationic lipids induced hexagonal HII conformation [113]. These mechanisms can be categorised into three types: membrane disruption [114], membrane fusion [115], and surface modification [116]. However, it is well known that all mechanisms with endosomal pH variation will be a strong indicator of endosomal escape, which is advantageous for the development of high-efficiency gene editing. Thus, investigating pH-responsive nanomaterials, such as NPs expansion [111, 117], polymer depolymerization [118, 119], and pH buffering materials [120, 121], could be a future study topic. In addition to endocytosis, NPs can be directly internalized into cytoplasm by passive membrane fusion or pore formation, which bypasses the endosome engulfment [110, 122]. However, the understanding of the trafficking of NPs is still evolving, thus additional efforts need to be done to confirm the precise mechanism. Developing novel methods and tools to dynamically observe membrane perturbation events and real-time cargo releases through visualisation of intracellular trafficking will be crucial for emerging biomedical applications to gain an in-depth understanding of cellular uptake process. Aside from the direct progressive approaches, much effort has been invested on the use of endocytosis inhibitors to promote and encourage advancement in the field.

Selection of CRISPR/Cas formats

The ideal CRISPR/Cas format is essential for meeting the requirements for various applications, including plasmid DNA (pDNA), mRNA, and Cas9/sgRNA ribonucleoproteins (RNPs), to achieve high-efficiency gene editing [123]. When viewed in the context of NPs formulations and clinical or research application, each format offers unique characteristics [124, 125] (Fig. 1b). Due to its outstanding stability, affordable, and ease of preparation, CRISPR/Cas pDNA has become the most popular format. Additionally, it is practical to perform polygenic editing simultaneously at several sites using different sgRNA designs. However, the drawbacks of pDNA limited the applicability of the editing system. These drawbacks included the relatively low expression, delayed responsiveness, induction of immunogenic responses, and difficulty of encapsulating large molecules. Even worse, because Cas9 proteins are expressed for a longer period of time, on-target effects would be compromised and there would be a higher chance of uncontrolled plasmid sequence integration [126,127,128,129]. The delivery of mRNA or RNP complex demonstrated a rapid onset of gene editing in comparison to the pDNA format, bypassing the restrictions of pDNA and achieving functional complementation. The mRNA format can prevent undesirable insertional mutagenesis and makes it simpler to generate transient expression in the cytoplasm, which lowers the off-target effects [130,131,132]. However, Cas9 mRNA distribution is difficult because of the low stability which results from its fragile single-stranded structure and is susceptible to in vivo RNAse degradation [133]. Thus, attempts should be taken to improve its stability using different vectors or chemical modifications.

RNP complexes are the simplest delivery format, and they have the least amount of intracellular processing that can evade the transcription and translation process (Fig. 1c). To accomplish this, they equip components with a rapid onset and short duration, which results in substantially fewer potential side effects and cellular toxicity as well as high gene editing efficiency [134, 135]. Nevertheless, as a specific obstacle of RNP, exogenous risk and big molecule size would cause immunogenicity and difficulty in efficient nuclear entry, limiting the broad uses of the CRISPR/Cas system [136]. Additionally, the preparation of Cas9 proteins is time-consuming, complicated, and costly [137]. The compatibility between the cargo and the carrier is also important to consider, along with the reasonable choice of cargoes. Designing NPs should not compromise other qualities, such as loaded-cargo protection, targeting, and effective transfection, in order to accommodate a specific CRISPR/Cas system format [81]. In conclusion, every delivery metod hin a variety of formats has disadvantages of its own. Depending on the compatibility of the cargo and the carrier, as well as the requirements for diverse purposes, selecting the best CRISPR/Cas format is therefore essential for effective editing.

NPs-based CRISPR/Cas delivery systems

The development of delivery carriers is currently trailing behind with regard to the distribution of CRISPR/Cas systems. Hence, there is an urgent need for sophisticated delivery systems with high efficiency, targeting, controllability, and safety characteristics. Synthetic nano-delivery systems, for instance lipid NPs (LNPs) [97], polymeric NPs (PNPs) [138], and gold NPs (AuNPs) [139], showed significant potential in enhancing the editing efficacy for CRISPR/Cas systems. These systems have the advantages of transient expression patterns, feasibility for mass-production at lower cost, all-in-one delivery, and low immunogenicity risk. These systems are also more flexible to deliver various cargoes for different purposes, encompassing RNPs, pDNA, mRNA, and donor DNA [140, 141]. In conclusion, nanotechnology holds the promise of overcoming the drawbacks of traditional delivery systems through cell-specific targeting, precise molecular transport to certain organelles, and other innovative strategies.

Lipid NPs

LNPs have been extensively investigated as the cutting-edge delivery platform for the drugs [142], CRISPR/Cas components [143], and vaccines [144]. They normally consist of four main components, i.e., key cationic or ionizable lipids complexed with negatively charged genetic material, phospholipids for particle structure, cholesterol for stability and membrane fusion, and PEGylated lipids to increase stability and circulation [145, 146]. The structure and formulation of LNPs protect the cargoes from enzymatic degradation and immunological responses, which subsequently facilitated their transportation into host cells for genome editing [147]. Ionizable lipids eventually replace permanently charged lipids due to their greater transfection efficiency and reduced cytotoxicity. Uncharged ionizable lipids are likely to adhere to the cell surface when the pH is neutral due to hydrophobic interactions or receptor-mediated endocytosis [148]. Ionizable lipids change to cationic form at lower pH to enable endosome escape and promote cargo release [113]. LNPs also have various advantages such as large cargo packaging capacity, good biocompatibility and bioavailability, low cell toxicity and immunogenicity, and mature industrial manufacturing technology. However, due to low drug loading and non-targeted biodistribution, it is challenging to achieve safe, efficient, and targeted administration of CRISPR/Cas components in vivo using LNPs [149]. The rational design or modulation of LNP formulations with the aim of obtaining optimum safety profile and efficient nucleic acid delivery would be a feasible technique to achieve the desired effects mentioned above [150, 151].

Developing new LNPs composition, such as NTLA-2001 LNPs [152] and 306-O12B-LNPs [153], can achieve targeting delivery of the CRISPR/Cas system with excellent editing efficiency and safety. In the case of treating transthyretin amyloidosis, the use of NTLA-2001 LNPs, an intravenous formulation made using a patented ionizable lipid, has demonstrated liver-targeted delivery. These LNPs display a consistent editing effect that is dosage dependent, safe, and long lasting, with maximum suppression seen after 12 months. The editing efficacy of transthyretin reached up to 93.7%, resulting in 87% decrease in protein concentration. In comparison to the gold standard LNPs (MC-3 LNPs, 14.6%), 306-O12B LNPs had a more effective liver-targeted delivery efficacy of around 38.5% [154], which offered the possibility of liver-specific administration in therapeutic applications. The high liver-targeted delivery efficiency could be attributed to the strong liver tropism caused by the active-targeting mechanism mediated by apolipoprotein E (ApoE) [113, 155]. Plasma ApoE attaches to the surface of LNPs and then interacts with the low-density lipoprotein receptor to achieve active endocytosis via ligand-receptor transport. Given this, modifying LNPs with appropriate ligands or targeting moieties may be an effective method for delivering drugs to specific organs [139, 141]. Similar to this, adding functional components to LNPs can reduce the risks associated with immunogenicity. TCL053-LNPs might preferentially transfer Cas9 mRNA and sgRNA into skeletal muscle due to the pH-dependent ionizable lipid TCL053. This approach could treat skeletal muscle disorders by giving repeated intramuscular injections because of its low immunogenicity. Nevertheless, each TCL053-LNPs injection could result in a long-lasting restoration of the dystrophin protein for at least a year [156].

Additionally, altering the proportion of LNPs formulation can achieve targeted and secure delivery for the CRISPR/Cas system. Multiple CRISPR cargoes could be selectively delivered to the targeted organs using a new technique called selective organ targeting (SORT), which involves changing the biodistribution of SORT LNPs [157]. The delivery to hepatocytes attained the highest specific transfection rate of 93% by including a supplementary component to the initial LNPs formulation. By constantly controlling the SORT molecule proportion, this system offered a reliable and designable platform for extrahepatic delivery, such as lung and spleen, which is also applicable to different NPs systems for targeted delivery. The proportion of various lipids in LNPs was then optimised using SORT and MC-3 LNP nanotechnology to create a lung-targeted LNPs-mediated CRISPR/Cas13d delivery system [158]. The data showed that, when compared to control groups, this system significantly decreased mRNA and protein levels of cathepsin L only in mouse lungs and reduced lung virus infectivity by two orders of magnitude, indicating an excellent strategy with exceptional efficacy, specificity, and safety. Since it is impossible to completely eliminate off-target events, efforts must be taken to address safety issues with the CRISPR/Cas system in order to build a perfect genome editing tool [60]. These efforts include sgRNA preoptimization, transient expression of the Cas9 protein, and targeted delivery to the intended tissue.

Polymeric NPs

Using multivalent charge interactions, PNPs condensed charged genome editing cargoes into nano-sized packages, shielding them from deactivation and promoting intracellular transit for genome editing [159, 160]. Different PNPs such as polyethylenimine (PEI), poly(lactic-co-glycolic-acid) (PLGA), poly(β-amino ester) (PBAE), poly(ethylene–glycol) (PEG), and amine-terminated polyamidoamine dendrimers have become very popular to deliver CRISPR/Cas components due to their excellent pharmacokinetic control, high cargo encapsulation, minimal immunogenicity, relatively flexible functionalization, and high bioavailability [78, 161]. More significantly, PNPs were able to accurately adjust the loading effectiveness and release kinetics to perform the editing for various objectives by varying their composition, stability, responsiveness, and surface charge characteristics [162]. Increasing the effectiveness of gene editing has increasingly become the significant route for the CRISPR/Cas system. PEI-based formulations, including branching PEI 25 kDa, are frequently utilised as the carriers for efficient delivery of CRISPR/Cas plasmids into the appropriate target cells when using PNP alone. In this technique, guide RNA and Cas9 are both expressed simultaneously, producing indel efficiencies (24.4%) that are comparable to Lipofectamine 2000 (27.9%) [163]. In contrast, PLGA-NPs offered higher 38.4% indel efficacy and 70% PLGA encapsulation efficacy, making them a more effective and secure delivery method for CRISPR components. Copolymer strategy by combination of multiple PNPs could be used as an effective method for CRISPR/Cas system. For instance, PEG-b-PLGA based copolymer with a cationic lipid-assisted NPs achieved direct modulating to immune cells such as neutrophils [164], macrophages [165], and DCs [166], which induced editing efficiency about 25.5–32.7% in vitro. Based on the PEG-b-PLGA combination, PEI-coated PEG-b-PLGA produced 80% selective genome editing in endothelial cells with a 40–50% efficiency [167]. It is evident that the distribution technique of copolymer had a more pronounced editing efficacy than employing PNPs alone. Notably, the helper components, such as cationic lipids or PEI coatings, are crucial to the delivery system that encourages cellular absorption and permits endosomal escape [168]. In-depth research into their interaction in copolymers may be a future direction for PNPs to explore since our understanding of high-efficiency copolymer delivery strategies is still evolving.

Aside from improving editing effectiveness, PNPs may easily implement their multifunctionality because they have effective cargo encapsulation, good modification potential, and interesting stimuli-responsiveness. Due of the exceptional ability of cargo encapsulation, co-delivery strategies that combine medicines and CRISPR components into a single polymer carrier have a beneficial effect in a variety of applications [169, 170]. For instance, the dendrimer-based lipid NPs might co-encapsulate and deliver CRISPR components, including Cas9 mRNA, sgRNA, and donor DNA, as an all-in-one nanocarrier [171]. The findings demonstrated that more than 91% of all cells were altered in vitro with 56% HDR efficiency. In addition, PNPs were frequently used to modify existing delivery vehicles in order to provide them exceptional biocompatibility, stimuli-responsiveness, loading capacity, and other properties. As an illustration, bifunctionalized aminoguanidine-PEGylated periodic mesoporous organosilica NPs successfully delivered RNPs with 40% effectiveness rate for gene editing. In this study, PEG coatings provided the carriers with protection against opsonization, aggregation, and phagocytosis as well as many biofunctions, including exceptional storage stability, permeability, and long-lasting blood circulation of nanocarriers [172, 173]. Numerous stimuli-responsive PNPs delivery systems have been created to extend therapeutic efficacy at a lower dose frequency. These systems can remotely initiate the release of bioactive molecules in response to internal and external stimulation, such as second near-infrared light (NIR II) and pH change [174, 175]. CRISPR/Cas components can be remotely released by NIR-light trigger depending on the specified semiconducting polymer brush. Controllable release of delivery system might be assisted by photothermal conversion, which increased their editing efficiency (around 35%) [176]. Additionally, pH-responsive PNPs provide a novel method of delivering Cas9 RNPs and donor DNA to selected organs. Local administration at various sites can simultaneously provide targeted distribution to desired organs such as intravenous, intratracheal, and intramuscular delivery to the liver, lung, and skeletal muscle in mice, respectively [177]. As a result, the administration method may offer a viable strategy for overcoming the barrier of limited biodistribution at intended areas. Designing delivery systems with a low amount of positive charge or shielding the positive charge reversibly would be a promising avenue to develop a secure and effective platform in the future because most cationic polymeric materials may cause high cytotoxicity.

Gold NPs

Inorganic gold nanocarriers have been utilised extensively to deliver imaging agents, nucleic acids, and proteins because of their distinctive physicochemical qualities that are favourable size, minimal toxicity, optical properties, superior biocompatibility, and photothermal action [178, 179]. Among these, the most unique quality that triggers cargo release to control the expression and activity of Cas9 proteins is the photothermal effect provided by AuNPs core [180,181,182]. Converted heat promoted an endonuclear change of heat-shock factor (HSF) from inactive monomer to active trimer through external NIR laser irradiation. The combination transfection of AuNPs and Cas9 plasmid under the influence of active HSF could achieve 90% GFP-positive cells, which was significantly greater than the result of lipofectamine 2000 [183]. Similarly, after hybridising protective DNA-modified gold nanorod (GNR) with sgRNA, heat damaged the hybrids and released sgRNA into cells under regulated NIR laser irradiation [184]. Furthermore, the unique photothermal activity of AuNPs makes them an appealing candidate for disease treatment, owing to their synergistic photothermal/gene therapy effects. By way of illustration, the mCas9-sGNR nanocarrier not only used to transport CRISPR components needed for gene therapy, but it also lowered the tolerance of cancer cells to heat through photothermal therapy, which led to superadditive synergistic anticancer effects [185]. The development of AuNPs-based CRISPR-Cas12a/Cas13a systems as visual biosensors for large-scale population screening and nucleic acid bioimaging, including smartphone-based diagnostics [186,187,188]. Compared to earlier detection technology, these devices offer quick, ultrasensitive, specific, and on-site biosensing methods. AuNPs have also been thoroughly studied for tumour visualisation, cancer detection, and bioimaging due to their remarkable photoluminescent characteristics, with the ultimate goal of obtaining precision targeted therapy [189].

Microalgae-based delivery systems

AMNPs features

Nanotechnology has grown exponentially as an interdisciplinary field due to its exceptional size and properties. NPs proved to be an excellent material than other counterparts and may serve as the main target in pharmacology, biosensors, and medicine [190, 191]. Synthetic metallic NPs, which exhibit a range of unprecedented physical, chemical, and optoelectronic capabilities, have become effective tools in various sectors over the past ten years. The synthetic procedure is the most important component in evaluating their applicability. Although various cutting-edge techniques were used to boost performance of NPs, such as reliability and practicality, the consensus view is that traditional synthesis of NPs will have a significant negative influence on human well-being because their synthesis was not eco-friendly. Several hazardous reagents, including reducing agents, organic solvents, and stabilisers, are introduced during the synthesis of NPs. These reagents led to significant toxicity issues as well as undesirable byproducts. These synthetic techniques nevertheless have several drawbacks, including high cost, poor efficacy, and requirement of skilled manipulation. The tendency to use sustainable, affordable, and environmental friendly NPs, such as silver, gold, copper, and other metals, would be further increased as a result of the expanding momentum of green chemistry [192]. With regard to biosynthetic mediation, there are the remarkable capabilities to consume, accumulate, and eventually remodel metal ions into NPs, which reduce the toxicity of metal ions and minimise adverse environmental effects through the resistance mechanism by organisms [193]. Biosynthesized NPs have previously been used in antimicrobials, pathogen detection or associated protein identification, and cargo delivery [194].

Bacteria, algae, fungi, and their extracts are emerging as attractive biocatalysts for NPs synthesis. Algae also contain large amounts of natural biocompounds such phytochemicals, carotenoids, vitamins, and pigments. All of which could act as reducing and stabilising substances to hyper-accumulate metal ions and transform them into more flexible forms at the nanoscale [195]. Algae gradually emerged as the best candidate for green synthetic NPs due to their capacity for bioreduction or biosorption. A new term, phyconanotechnology, which combines phycology with nanotechnology, is proposed to describe this phenomenon [196, 197].

Although there has been substantial development in AMNPs, there are still several obstacles existing, such as the strict criteria for homogeneous NP size and shape, reduced kinetics, and difficulties in large-scale manufacture. Managing the NPs development, stability, and aggregation is essential for obtaining uniform NPs and this involves optimisation of the variables, including pH, temperature, concentration, and others [198]. Notably, multidimensional characterizations of generated NPs are a necessary step to evaluate their uses in the interdisciplinary field, as they could provide specific criteria that make NP management easier. Moreover, generating genetic modified strains or screening high-producing algae strains are attractive strategies to synthesis NPs at large scale. Importantly, due to their exceptional biocompatibility in a homologous relationship, AMNPs represent a viable substitute for the administration of medicines, bioactive macromolecules, and gene-editing components, particularly in algae. Thus, future research on the size distribution and aggregation of NPs, as well as their surface properties, morphology, and dissolution rate, will shed light on how to regulate their release for gene editing.

AMNPs biosynthesis

Top-down and bottom-up approaches are typically used in classical NPs synthesis. The former refers to the transformation of bulk material into thinner crystallites, whilst the later refers to the production of particles by assembling ultra-small building blocks. In terms of effectiveness, tunability, and environmental friendliness, bottom-up technique is best for green synthesis of NPs [199, 200]. Following the natural bio-mineralization process, the major step in synthesising AMNPs is to capture target metal ions from the surrounding environment and enzymatically reduce them to nanoscale. AMNPs can be formed intracellularly or extracellularly, depending on the origins. The intracellular pathway is a very complex and dynamic biological environment in which positively charged metal ions are absorbed by electrostatic interactions to negatively charged cell walls. They were then transformed into metallic NPs by intracellular enzymes [201]. Algae-based metabolic pathways eliminate the need for pre-treatment and give AMNPs high colloidal stability, which benefite from increased biocompatibility and steric stabilisation of bioactive compounds [202, 203]. However, downstream separation of nanomaterials originated from the living cells is still challenging [204].

Extracellular NP synthesis, on the other hand, avoids the additional downstream processing steps and is thus more promising for a variety of applications [205, 206]. Typically, the biosynthesis of NPs comprised two steps: the preparation of algae extracts and metal precursor solution, followed by their incubation for the synthesis reaction [207]. Following incubation, AMNPs have a tendency to aggregate, giving them a thermodynamic stability with different sizes and shapes [207, 208]. The successful synthesis of NPs is indicated by the colour changes of cultured mixture. Despite extracellular synthesis of metallic NPs is easy to harvest in large-scale production, it may grow slower than that of intracellular biosynthesis under the circumstance of cell-free extract. The primary reason may be due to the binding of the active proteins with intracellularly produced NPs rather than extracellularly, which suggests that the active proteins have a special function distinct from cell-free synthesis. As a result, algae have been recognised as the ideal bio-based substrate for the extracellular synthesis of NPs due to their abundance of bioactive components [209]. Despite the fact that NPs have a wide range of uses, efficient, environmental friendly, and easy to scale up, but their synthesis mechanisms are not well understood [210].

Applications of microalgae-based NPs in delivery

The ideal drug delivery systems (DDSs) primarily use nanocarriers with the requirements of safety, effectiveness, and optimal bioavailability to improve the selectivity and targeting of various cargoes into specific cells [211]. However, the shortcomings of conventional NPs, such as their poor stability, high cost, time-consuming, and toxicity, are becoming increasingly apparent in tandem with the increasing demand for nano-DDSs [212, 213]. Many marine bioactive compounds have been employed as reducing and stabilising agents based on the extracellular synthesis of NPs to create metallic nano-DDSs with remarkable biocompatibility, biodegradability, minimal immunogenicity, and non-toxicity [214, 215] (Fig. 2b). Marine carbohydrates have been shown to be the suitable substrates for the construction of AMNPs as DDSs due to considerable advantages in terms of biodiversity, diverse biological activities, and ease of preparation [216, 217]. Bioinspired multifunctional DDSs have been synthesized through extracellular synthesis using the carbohydrates found in marine algae, such as fucoidan [218], resveratrol [219], porphyrin [220], carrageenan oligosaccharide [221], and chitosan [222, 223]. These carbohydrate-based NPs have an exceptional 60%–92% drug loading efficiency, enabling a promising use in the delivery of the CRISPR/Cas system. Nonetheless, the number and type of algae-derived carbohydrates NPs are limited only to a few metals, like AuNPs and AgNPs. Therefore, additional research should be done to determine the other active components from algae that could be used for various types of cargo [15]. In contrast to metallic NPs, marine carbohydrates-based DDSs may be created as carriers through self-assembly or covalent crosslinking and combine the benefits of nanoscale systems with the characteristics of carbohydrates, such as targeted delivery, high biocompatibility, structural modularity, and biodegradability [224].

Fig. 2
figure 2

Preparation of microalgae-based nanomaterials. a Purification and separation of diatom biosilica. b Biosynthesis of AMNPs. c EVs isolation by differential tangential flow filtration

Aside from that, extracellular vesicles (EVs) generated from microalgae and diatom biosilica are also efficient biological delivery nanomaterials. Diatom-derived biosilica has high specific surface area, excellent drug-holding capacities and biocompatibility, and tailorable surface functionalization for tunable properties because it is encased in a porous 3D nanopatterned silica structure made from biosilica that self-assembled into intricate porous shells [225]. As a result, biosilica can be employed as a sophisticated microcarrier for targeted distribution for therapeutic and medical imaging purposes which is a cost effective and environmental friendly process [226,227,228]. Biosilica has been created using pulverisation and the strong oxidising capabilities of piranha solution, with the advantages of being cost-effective, easy, quick, and environmental friendly [227] (Fig. 2a). Cargoes could be loaded both on the surface and inside the biosilica NPs, which have a substantial impact on the releasing kinetics of active compounds due to their hierarchical 3D silica porous architectures [229]. More importantly, increased surface modification activities will aid in the targeted delivery of cargoes and the synthesis of multifunctional smart biosilica NPs [230]. For example, chitosan molecules grafted biosilica demonstrated consistent pH responsiveness and notable drug load efficiency with about 90% [231]. EVs, on the other hand, have been widely exploited as the potent tools for cargoes delivery due to their intercellular communication capabilities [232]. Microalgae are an abundant and sustainable source of natural EVs with diverse bioactivities, therefore they are positioned to become competitive competitors in innovative delivery systems [233, 234] (Fig. 2c).

Microalgae-based biohybrid microrobots

Aside from the previously described microalgae-based nanocarriers, living microalgae-based delivery systems have also exhibited impressive delivery capabilities [235]. Microalgae-based biohybrid microrobots have a number of benefits over conventional synthetic NPs, such as effective and controllable drug delivery, improved stability and bioavailability of therapeutic agents, decreased toxicity and side effects, customizability, eco-friendliness, as well as cost-effectiveness [236, 237]. These microrobots were propelled by magnetic fields or light beams and had high propulsion speeds and phototactic guidance skills, allowing for precise navigation to tissues and difficult-to-reach bodily cavities [238]. As illustrated in Fig. 3, Chlamydomonas reinhardtii, Spirulina, and Chlorella are gaining popularity for use in the construction of microrobots [239, 240]. The modifications and cargo loading of microrobots have negligible influence on their motion, thereby paving the way for the development of living delivery vehicles with enormous potential. Through external magnetically actuated control [241,242,243,244,245], Fe3O4 magnetized Chlorella microrobot demonstrated a greater drug loading efficiency of 98.2% and excellent targeting delivery capabilities (90% target cell death) [246]. The superior drug loading efficiency may be attributed to unique permeation pretreatment and highly negative zeta potential, providing new insights into the preparation of other microalgae-based microrobots.

Fig. 3
figure 3

Preparation and application of microalgae-based biohybrid microrobots. ac represents the preparation process and application of microalgae-based biohybrid microrobots, including the Chlamydomonas reinhardtii (a), Spirulina (b), and Chlorella (c) etc.

Additionally, light-driven microalgal microrobots have shown great potential due to their rapid phototaxis response. Numerous tests have shown that biohybrid microrobots are resilient and can be directed towards light stimuli with powerful and precise motion. Although microalgal microrobots can be loaded with drugs with a high manufacturing yield of approximately 90% [247], the drug release effectiveness of microalgal microrobots is inadequate, ranging only from 5 to 10% [248]. Thus, improving drug release efficiency is a critical step towards expanding the use of microalgal microrobots. Microalgal microrobots have also displayed excellent delivery capabilities in local tissues without external targeted actuation. An example is the modification of Chlamydomonas reinhardtii with neutrophil membrane-coated and drug-loaded PNPs which allowed antibiotics to achieve about 90% drug release efficiency in the lungs over the first 20 h [235]. Once the issue of low drug release efficiency is resolved, microalgal microrobots will be able to deliver drugs in vivo in an effective, targeted, and safe ways. Furthermore, microalgal microrobots have outstanding fluorescence imaging capabilities that enable non-invasive tracking and real-time monitoring in vivo. They can also overcome hypoxia in tissue engineering and generate cytotoxic reactive oxygen through photodynamic treatment [249]. In summary, the development of microalgal microrobots has opened up new opportunities as a flexible platform for targeted distribution, fluorescence imaging, photothermal therapy, radiotherapy, and other biotechnology applications.

Current issues and future prospects

Current drawbacks of gene editing in microalgae

Although CRISPR/Cas systems have demonstrated great promise in the field of microalgae, their application may be hampered by a lack of understanding of microalgal biology, inefficient delivery of genetic materials, decreased expression of Cas9 due to unknown factors, low repair efficiency of the homology-directed repair (HDR) pathway, and other factors [250]. (1) Microalgae bioengineering is still in its infancy, with only a few fully sequenced genome species [251]. In the field of microalgal research, artificial intelligence (AI) has gained popularity and can be used to predict gene sequencing and editing. In this way, artificial intelligence (AI) technology fills the gap between commercial microalgae applications and genetic engineering [252, 253]. (2) It is believed that the cell wall of microalgal cells is the major barrier that reduces the delivery effectiveness of genetic materials during the introduction of macromolecules [254]. In comparison to conventional physical pretreatments like ultrasonication, high-pressure homogenization, bead milling, cryogenic grinding, and pulsed electric field, or mechanical pretreatments like acid, alkaline, and strong oxidant [255], some cutting-edge techniques, such as zinc oxide nanowire array microdevice system and optimised droplet electroporation, are significantly more effective for algal cells with rigid cell walls [256]. It is also necessary to use the right and appropriate delivery vector to increase the effectiveness of genetic materials' transport, and AMNPs have a particularly high potential for microalgae genetic editing with improved biocompatibility [15]. (3). Many approaches have been suggested to combat off-target effect of CRISPR/Cas system, for example reasonable design of sgRNA using different computational tools, like CRISPR-P 2.0 [257], E-CRISP [258], and CasFinder [259], further modification of sgRNA, application of anti-CRISPR proteins as well as appropriate editing conditions (time and temperature). Meanwhile, the cytotoxic effects of Cas9 nuclease could be mitigated by substituting it with Cas12a (4) The expression of Cas9 proteins could be reduced by unknown circumstances. Inserting introns into the Cas9 coding region could improve the efficacy of gene editing, which would significantly improve Cas9 expression [260]. (5) Due to competition from the NHEJ pathway, the HDR pathway is typically less effective in microalgae genome editing for DSBs repair in the host [261]. Given this, various approaches to improving HDR pathway repair efficiency have been suggested, including inhibition of NHEJ pathway [262], regulation of HDR-related factors [263], cell cycle synchronization [264], design of donor DNA template [265], and proximity of CRISPR component and donor DNA template [266].

Natural ingredient improvement

Currently, microalgae are potential sources of valuable bioactive compounds that can be used in different sectors, such as pharmaceutical, cosmetic, and nutraceutical industries [267]. Microalgae could replace higher plants due to its rapid growth, high biomass yield, minimal water use, daily harvest, lack of seasonal restrictions, and ease of culture in in environments with significant climate change [252]. However, most of the biomolecules are produced in relatively small amounts, making it difficult to meet industrial demands [268]. The CRISPR/Cas system is a promising way to boost the production of bioactive molecules with the benefits of pinpoint accuracy, high editing efficiency, multiplex knock-in/out capability, low cost, and short cycle [268, 269] (Fig. 4a). It is ideal to combine genetic and metabolic engineering with omic technologies (transcriptome analysis, proteomics, and liquid chromatography mass spectroscopy) for the purpose of identifying the dynamic molecular regulatory mechanisms and genetic networks involved in the various metabolic pathways in microalgae [256, 270]. Inducing targeted regulation and modification in microalgal cells could be used to precisely control the metabolic pathway and acquire the strains that can produce high-yield targeted compounds that are competitive with present commercial sources.

Fig. 4
figure 4

Extensive applications of gene editing microalgae. a Enhancing the production of natural ingredient in microalgae. b Extending the expression profile of microalgae systems. c Exploration of microalgae biofuel. d Enhancing the removal efficiency for wastewater and reduction in carbon dioxide emissions

Extending expression profile of microalgae systems

Production of recombinant biopharmaceuticals, including vaccines, antibodies, enzymes, hormones, antibiotics, cytokines, thrombolytic agents, and other proteins with medicinal value, is a rapidly growing market for disease prevention and treatment [271] (Fig. 4b). Typically, these proteins are produced by bacteria, yeast, fungus, mammalian cells, and insect, but they have drawbacks, such as expensive, poor yields, prone to contamination, fragile, and trigger immunogenic effects [272]. Eukaryotic microalgae, in contrast, have a shorter culture cycle and are less impacted by seasonal weather conditions in addition to being able to complete complex protein folding and modification to produce active proteins [271]. Both the nucleus and the chloroplast of microalgae can be genetically modified at the same time to produce biopharmaceuticals, providing a large-scale platform for recombinant protein production and industrial use [273].

Antibiotics

Antibiotic overuse and regulatory limits are driving an increase in research for antibiotic alternatives, particularly for antibiotic-resistant bacteria. Small oligopeptides known as antimicrobial peptides (AMPs) are present in plants, animals, and bacteria and serve as a general defence against pathogenic microorganisms [274, 275]. In comparison to other systems, the microalgae expression system can manufacture synthetic antibiotics that have the benefit of being flexible while protecting recombinant peptides within their cell walls from digestion [276]. More importantly, microalgae can effectively suppress the growth of bacterial strains through the interaction with their excretion products, such as fatty acids. [277]. Hence, C. reinhardtii and H. pluvialis have expressed additional antimicrobial and antifungal proteins (ToAMP4 or antimicrobial peptide piscidin-4 gene). Some of them were produced with yields up to 0.32% of total host soluble proteins with more potent antibacterial action against E. coli, S. enteritidis, S. aureus, and B. subtilis, despite the fact that expression had no effect on algal growth [278]. The future green synthesis of antibiotics with microalgae is obviously considerably facilitated by the use of gene editing techniques.

Human proteins

Recombinant human proteins could be created using the microalgal nucleus as a new expression system for the purpose of treating diseases where these proteins are deficient or where it is necessary to overexpress specific molecules, such as cytokines, growth factors, and human interferon [279, 280]. Aside from that, more health-promoting proteins, like the human growth hormone, growth factors hVEGF-165, hPDGF-B, and hSDF-1, have been successfully produced in the chloroplast system of C. reinhardtii, [281,282,283]. Chloroplast systems have higher levels of recombinant protein production when compared to nucleus systems. Additionally, the chloroplast system has the ability to express multiple transgenes, precise integration of foreign DNA to any locus, and the absence of gene silencing effects [275]. As a result, more algal-derived human proteins will be synthesised in the future, which could be employed as safe and economical biological agents to treat a variety of illness.

Vaccines and antigens

There are many benefits of using microalgae as an expression system for manufacturing antigens and antibodies, including their low cost, the capacity to perform post-translational modifications, and elimination of the need for protein extraction and purification. For instance, a direct vaccination against WSSV infection in shrimps could be created using transgenic Dunaliella salina that expresses the viral envelope protein VP28 [284]. In fact, the recombinant protein-based vaccinations are not just limited to aquaculture and have the potential to develop into animal feed as well as human food. For instance, biopharmaceuticals synthesize from microalgae can be employed in immunotherapy for diseases like cancer [285] and SARS-COV-2 [286]. A chimeric protein to treat breast cancer was expressed in the microalgae Schizochytrium sp. by fusing B-cell epitopes from the tumour related antigens, human epidermal growth factor receptor-2, mucin-like glycoprotein 1, Wilms' tumour antigen, and mammaglobin [287]. These epitopes serve as immunotherapy targets and aid in the synthesis of antibodies and CD8+/CD4+T cell, which kill the malignant cells [288,289,290]. Berndt et al. [291] use C. reinhardtii to produce recombinant SARS-CoV-2 spike RBD proteins that have a comparable affinity to those expressed in mammalian cells. The findings revealed that it is feasible to use algae to produce functional proteins [286]. In short, antigens and antibodies produced by microalgae could serve a wide variety of applications in the future, including the development of diagnostic tools and the treatment of human, plant, and animal diseases.

Exploration of microalgae biofuel

Microalgae are a viable resource for biofuel generation and the generation of biodiesel using microalgae biomass is 100 times more efficient than that using higher plants as feedstocks [292] (Fig. 4c). Furthermore, microalgae-based biofuels are compatible with existing gasoline engines, eliminating the need for engine modifications [293]. The viability of biofuel as an alternative to traditional fossil fuels will be determined primarily by its economic benefits. Unfortunately, the actual costs of microalgal biofuels are still deemed higher than conventional fossil oils, owing to limited storage lipids and carbohydrates yield in microalgae mass [294]. Numerous initiatives have been launched in the past to increase the production of microalgae biomass and biofuels, including the selection of suitable microalgae species [267], pretreatment of biomass [295] and application of nanotechnology [296]. However, this field still has several constraints that must be addressed with more robust editing tool, such as low carbon fixation efficiency, low lipid accumulation rate, and long cultivation period [297]. The CRISPR/Cas technology presents the best option for resolving the aforementioned problems in microalgae. Microalgae metabolic pathways, for example, can be altered by raising acetyl coenzyme A carboxylase, lowering enzymatic activity of phospholipase A2, or boosting lipid and carbohydrate production. Omic technologies can be used to identify the underlying dynamic molecular regulatory mechanisms and genetic networks involved in the metabolic pathways [298]. Gene-edited microalgal variations might be used in the future to produce more environmentally friendly and clean energy.

Wastewater treatment

An increasing number of pollutants found in wastewater effluents, such as pharmaceuticals, endocrine disruptors, industrial chemicals, heavy metals, pesticides, etc., are posing threat to human and environmental health [299]. Activated charcoal, flocculation, chemical precipitation, reverse osmosis, ultraviolet disinfection, ultrafiltration, electro-coagulation, and ion exchange are few examples of methods to treat wastewater [300]. However, these methods are not cost-effective since these methods demand a lot of energy and labour and they are unable to eliminate a variety of emerging pollutants in wastewater completely [301]. Microalgal-bioremediation systems have thus been shown to be an effective strategy in wastewater treatment processes to overcome the limitations of such methods. The advantages of microalgal-bioremediation [302] (Fig. 4d) are that it may effectively remove N and P through photosynthetic processes and CO2 sequestration. In the meantime, oxygen is released into the water, increasing the amount of dissolved oxygen [303]. Additionally, microalgae increase the pH and dissolved oxygen in the broth as a result of their photosynthetic activities, which reduces pathogen survival [304]. Microalgae have a variety of appealing advantages, but there are still several obstacles existing which are high turbidity, contamination by other microbes, and difficulties in harvesting microalgae biomass. These issues can be resolved using the methods including genomics, computational biology, proteomics, bioinformatics, molecular modelling, molecular dynamics simulation, and a specialised algorithm for pathways prediction [305]. These combined approaches may provide a fast understanding of pollutant binding, degradation, and absorption [306]. Based on this, more robust microalgal strains could be created using gene editing by altering the related genes that are responsible for the biosynthesis of specific enzymes, increasing the adaptability and biosorption ability. This would further improve the efficiency of wastewater treatment and lower operating costs.

CO2 sequestration

Large-scale human activity and excessive greenhouse gas emissions have caused enormous changes in the environment, including the greenhouse effect. These changes have resulted in global warming, climate change, and changes in other environmental aspects [307]. Critical climate change has sparked initiatives worldwide to cut greenhouse gas emissions, especially CO2, with the primary goal of keeping rises in global temperature to 2 ℃ [308]. CO2 sequestration is critical for managing climate stability and balancing CO2 level in the atmosphere [309]. CO2 sequestration by microalgae is the most promising approach for mitigating the consequences of greenhouse emissions [310] (Fig. 4d). Microalgae capture CO2 and provide various economic and ecological advantages over conventional terrestrial plants, such as rapid growth and sustainability [311], high photosynthetic efficiency, and cooperation with CO2 sequestration and wastewater treatment [312]. Additionally, the resulting microalgae biomass can be used to synthesis high-value biomolecules, such as carbohydrates and lipids to produce byproducts for example biofuels and as chemical precursors [313]. A better understanding of the microalgae metabolic pathway with the establishment of ideal genome editing platform has enabled researchers to increase the photosynthetic efficiency of microalgae chassis cells, optimise the microalgae culture conditions, and increase microalgae biomass and yield [314]. Engineered microalgal cell factories based on synthetic biology methods have made it feasible to increase economic efficiency, lessen the greenhouse effect, and protect the environment.

Conclusion

Target-specific gene editing methods have substantially helped the microalgae industry, enabling improvements in the fields of natural ingredients enhancement, expression profile extension, wastewater treatment, CO2 sequestration, and other related fields. Even though this field has made significant advancements, there are still some problems that prevent the widespread application of this cutting-edge technology. These problems include the difficulty of delivering editing components, lack of knowledge about microalgae biology, ambiguous molecular regulatory mechanisms and genetic networks associated with microalgae, and the ineffectiveness of gene editing tools. In this review, a variety of efficient approaches aimed at resolving those problems have been thoroughly discussed. The synthesis and use of novel AMNPs has the potential to increase the effectiveness of gene editing component delivery while minimising damage to microalgae cells. Due to their intrinsic anti-bacterial, anti-cancer, anti-viral, anti-pollution, and bio-remediation capabilities, AMNPs confer additional competitiveness in the gene editing of microalgae in the realms of biomedicine and environmental remediation. Following extensive study on AMNPs, gene editing in microalgae would offer a plethora of opportunities in a variety of industries.

Availability of data and materials

Not applicable.

Abbreviations

AMNPs:

Algal-mediated nanoparticles

CRISPR/Cas system:

Clustered regularly interspaced short palindromic repeats-associated system

ZFN:

Zinc finger nuclease

TALEN:

Transcription activator-like effector-based nuclease

NPs:

Nanoparticles

NP:

Nanoparticle

pDNA:

Plasmid DNA

RNPs:

Ribonucleoproteins

LNPs:

Lipid NPs

PNPs:

Polymeric nanoparticles

AuNPs:

Gold nanoparticles

ApoE:

Apolipoprotein E

SORT:

Selective organ targeting

PEI:

Polyethylenimine

PLGA:

Poly (lactic-co-glycolic-acid)

PBAE:

Poly (β-amino ester)

PEG:

Poly (ethylene–glycol)

NIR:

Near-infrared

GFP:

Green fluorescent protein

HSF:

Heat-shock factor

GNR:

Gold nanorod

DDSs:

Drug delivery systems

EVs:

Extracellular vehicles

PE:

Polyelectrolyte

DOX:

Doxorubicin

NHS:

N-hydroxysuccinimide

DBCO:

Dibenzocyclooctyne

HDR:

Homology-directed repair

AI:

Artificial intelligence

NHEJ:

Nonhomologous end joining

AMPs:

Antimicrobial peptides

GHGs:

Greenhouse gases

References

  1. Su H, Feng J, Lv J, Liu Q, Nan F, Liu X, et al. Molecular mechanism of lipid accumulation and metabolism of oleaginous chlorococcum sphacosum gd from soil under salt stress. Int J Mol Sci. 2021;22:1304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Da Ros PC, Silva CS, Silva-Stenico ME, Fiore MF, De Castro HF. Assessment of chemical and physico-chemical properties of cyanobacterial lipids for biodiesel production. Mar Drugs. 2013;11:2365–81.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Giri DD, Dwivedi H, Khalaf DAA, Pal DB, Otaibi AA, Areeshi MY, et al. Sustainable production of algae-bacteria granular consortia based biological hydrogen: new insights. Bioresour Technol. 2022;352: 127036.

    Article  CAS  PubMed  Google Scholar 

  4. Harahap BM, Ahring BK. Acetate production from syngas produced from lignocellulosic biomass materials along with gaseous fermentation of the syngas: a review. Microorganisms. 2023;11:995.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bayona-Morcillo PJ, Gomez-Serrano C, Gonzalez-Lopez CV, Massa D, Jimenez-Becker S. Effect of the application of hydrolysate of chlorella vulgaris extracted by different techniques on the growth of pelargonium x hortorum. Plants. 2022;11:191.

    Article  Google Scholar 

  6. Tsvetanova F, Yankov D. Bioactive compounds from red microalgae with therapeutic and nutritional value. Microorganisms. 2022;10:2290.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mallick N, Bagchi SK, Koley S, Singh AK. Progress and challenges in microalgal biodiesel production. Front Microbiol. 2016;7:1019.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Venkata Subhash G, Rajvanshi M, Navish Kumar B, Govindachary S, Prasad V, Dasgupta S. Carbon streaming in microalgae: extraction and analysis methods for high value compounds. Bioresour Technol. 2017;244:1304–16.

    Article  CAS  PubMed  Google Scholar 

  9. Hlavova M, Turoczy Z, Bisova K. Improving microalgae for biotechnology–from genetics to synthetic biology. Biotechnol Adv. 2015;33:1194–203.

    Article  CAS  PubMed  Google Scholar 

  10. Kurita T, Moroi K, Iwai M, Okazaki K, Shimizu S, Nomura S, et al. Efficient and multiplexable genome editing using platinum talens in oleaginous microalga, nannochloropsis oceanica nies-2145. Genes Cells. 2020;25:695–702.

    Article  CAS  PubMed  Google Scholar 

  11. Wang JY, Doudna JA. Crispr technology: a decade of genome editing is only the beginning. Science. 2023;379:eadd8643.

    Article  CAS  PubMed  Google Scholar 

  12. Coelho MA, De Braekeleer E, Firth M, Bista M, Lukasiak S, Cuomo ME, et al. Crispr guard protects off-target sites from cas9 nuclease activity using short guide rnas. Nat Commun. 2020;11:4132.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cheng H, Zhang F, Ding Y. Crispr/cas9 delivery system engineering for genome editing in therapeutic applications. Pharmaceutics. 2021;13:1649.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Feng S, Wang Z, Li A, Xie X, Liu J, Li S, et al. Strategies for high-efficiency mutation using the crispr/cas system. Front Cell Dev Biol. 2021;9: 803252.

    Article  PubMed  Google Scholar 

  15. Chaudhary R, Nawaz K, Khan AK, Hano C, Abbasi BH, Anjum S. An overview of the algae-mediated biosynthesis of nanoparticles and their biomedical applications. Biomolecules. 2020;10:1498.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gerken HG, Donohoe B, Knoshaug EP. Enzymatic cell wall degradation of chlorella vulgaris and other microalgae for biofuels production. Planta. 2013;237:239–53.

    Article  CAS  PubMed  Google Scholar 

  17. Hao X, Luo L, Jouhet J, Rebeille F, Marechal E, Hu H, et al. Enhanced triacylglycerol production in the diatom phaeodactylum tricornutum by inactivation of a hotdog-fold thioesterase gene using talen-based targeted mutagenesis. Biotechnol Biofuels. 2018;11:312.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sizova I, Greiner A, Awasthi M, Kateriya S, Hegemann P. Nuclear gene targeting in chlamydomonas using engineered zinc-finger nucleases. Plant J. 2013;73:873–82.

    Article  CAS  PubMed  Google Scholar 

  19. Vazquez-Dominguez I, Garanto A, Collin RWJ. Molecular therapies for inherited retinal diseases-current standing, opportunities and challenges. Genes. 2019;10:654.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ghribi M, Nouemssi SB, Meddeb-Mouelhi F, Desgagne-Penix I. Genome editing by crispr-cas: a game change in the genetic manipulation of chlamydomonas. Life. 2020;10:295.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sanchez-Baltasar R, Garcia-Torralba A, Nieto-Romero V, Page A, Molinos-Vicente A, Lopez-Manzaneda S, et al. Efficient and fast generation of relevant disease mouse models by in vitro and in vivo gene editing of zygotes. CRISPR J. 2022;5:422–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Naduthodi MIS, Sudfeld C, Avitzigiannis EK, Trevisan N, Van Lith E, Alcaide Sancho J, et al. Comprehensive genome engineering toolbox for microalgae nannochloropsis oceanica based on crispr-cas systems. ACS Synth Biol. 2021;10:3369–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Nymark M, Sharma AK, Sparstad T, Bones AM, Winge P. A crispr/cas9 system adapted for gene editing in marine algae. Sci Rep. 2016;6:24951.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hopes A, Nekrasov V, Kamoun S, Mock T. Editing of the urease gene by crispr-cas in the diatom thalassiosira pseudonana. Plant Methods. 2016;12:49.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Kim J, Chang KS, Lee S, Jin E. Establishment of a genome editing tool using crispr-cas9 in chlorella vulgaris utex395. Int J Mol Sci. 2021;22:480.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Farasat I, Kushwaha M, Collens J, Easterbrook M, Guido M, Salis HM. Efficient search, mapping, and optimization of multi-protein genetic systems in diverse bacteria. Mol Syst Biol. 2014;10:731.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Shin SE, Lim JM, Koh HG, Kim EK, Kang NK, Jeon S, et al. Crispr/cas9-induced knockout and knock-in mutations in chlamydomonas reinhardtii. Sci Rep. 2016;6:27810.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Baek K, Kim DH, Jeong J, Sim SJ, Melis A, Kim JS, et al. DNA-free two-gene knockout in chlamydomonas reinhardtii via crispr-cas9 ribonucleoproteins. Sci Rep. 2016;6:30620.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kao PH, Ng IS. Crispri mediated phosphoenolpyruvate carboxylase regulation to enhance the production of lipid in chlamydomonas reinhardtii. Bioresour Technol. 2017;245:1527–37.

    Article  CAS  PubMed  Google Scholar 

  30. Baek K, Yu J, Jeong J, Sim SJ, Bae S, Jin E. Photoautotrophic production of macular pigment in a chlamydomonas reinhardtii strain generated by using DNA-free crispr-cas9 rnp-mediated mutagenesis. Biotechnol Bioeng. 2018;115:719–28.

    Article  CAS  PubMed  Google Scholar 

  31. Ferenczi A, Chew YP, Kroll E, Von Koppenfels C, Hudson A, Molnar A. Mechanistic and genetic basis of single-strand templated repair at cas12a-induced DNA breaks in chlamydomonas reinhardtii. Nat Commun. 2021;12:6751.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kim J, Lee S, Baek K, Jin E. Site-specific gene knock-out and on-site heterologous gene overexpression in chlamydomonas reinhardtii via a crispr-cas9-mediated knock-in method. Front Plant Sci. 2020;11:306.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Li H, Shen CR, Huang CH, Sung LY, Wu MY, Hu YC. Crispr-cas9 for the genome engineering of cyanobacteria and succinate production. Metab Eng. 2016;38:293–302.

    Article  CAS  PubMed  Google Scholar 

  34. Wendt KE, Ungerer J, Cobb RE, Zhao H, Pakrasi HB. Crispr/cas9 mediated targeted mutagenesis of the fast growing cyanobacterium synechococcus elongatus utex 2973. Microb Cell Fact. 2016;15:115.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Gordon GC, Korosh TC, Cameron JC, Markley AL, Begemann MB, Pfleger BF. Crispr interference as a titratable, trans-acting regulatory tool for metabolic engineering in the cyanobacterium Synechococcus sp. Strain pcc 7002. Metab Eng. 2016;38:170–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Huang CH, Shen CR, Li H, Sung LY, Wu MY, Hu YC. Crispr interference (crispri) for gene regulation and succinate production in cyanobacterium s. Elongatus pcc 7942. Microb Cell Fact. 2016;15:196.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Yao L, Cengic I, Anfelt J, Hudson EP. Multiple gene repression in cyanobacteria using crispri. ACS Synth Biol. 2016;5:207–12.

    Article  CAS  PubMed  Google Scholar 

  38. Poliner E, Takeuchi T, Du ZY, Benning C, Farre EM. Nontransgenic marker-free gene disruption by an episomal crispr system in the oleaginous microalga, nannochloropsis oceanica ccmp1779. ACS Synth Biol. 2018;7:962–8.

    Article  CAS  PubMed  Google Scholar 

  39. Wang Q, Lu Y, Xin Y, Wei L, Huang S, Xu J. Genome editing of model oleaginous microalgae nannochloropsis spp. By crispr/cas9. Plant J. 2016;88:1071–81.

    Article  CAS  PubMed  Google Scholar 

  40. Liu X, Zhang D, Zhang J, Chen Y, Liu X, Fan C, et al. Overexpression of the transcription factor atlec1 significantly improved the lipid content of chlorella ellipsoidea. Front Bioeng Biotechnol. 2021;9: 626162.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Vingiani GM, De Luca P, Ianora A, Dobson ADW, Lauritano C. Microalgal enzymes with biotechnological applications. Mar Drugs. 2019;17:459.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Wang L, Yang L, Wen X, Chen Z, Liang Q, Li J, et al. Rapid and high efficiency transformation of chlamydomonas reinhardtii by square-wave electroporation. Biosci Rep. 2019;39:BSR20181210.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Nouemssi SB, Ghribi M, Beauchemin R, Meddeb-Mouelhi F, Germain H, Desgagne-Penix I. Rapid and efficient colony-pcr for high throughput screening of genetically transformed chlamydomonas reinhardtii. Life. 2020;10:186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Moosburner MA, Gholami P, Mccarthy JK, Tan M, Bielinski VA, Allen AE. Multiplexed knockouts in the model diatom phaeodactylum by episomal delivery of a selectable cas9. Front Microbiol. 2020;11:5.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Bolanos-Martinez OC, Mahendran G, Rosales-Mendoza S, Vimolmangkang S. Current status and perspective on the use of viral-based vectors in eukaryotic microalgae. Mar Drugs. 2022;20:434.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Brown LE, Sprecher SL, Keller LR. Introduction of exogenous DNA into chlamydomonas reinhardtii by electroporation. Mol Cell Biol. 1991;11:2328–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Lee S, Kim YY, Ahn HJ. Systemic delivery of crispr/cas9 to hepatic tumors for cancer treatment using altered tropism of lentiviral vector. Biomaterials. 2021;272: 120793.

    Article  CAS  PubMed  Google Scholar 

  48. Yao X, Lyu P, Yoo K, Yadav MK, Singh R, Atala A, et al. Engineered extracellular vesicles as versatile ribonucleoprotein delivery vehicles for efficient and safe crispr genome editing. J Extracell Vesicles. 2021;10: e12076.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ramakrishna S, Kwaku Dad AB, Beloor J, Gopalappa R, Lee SK, Kim H. Gene disruption by cell-penetrating peptide-mediated delivery of cas9 protein and guide rna. Genome Res. 2014;24:1020–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ren X, Wei C, Yan Q, Shan X, Wu M, Zhao X, et al. Optimization of a novel lipid extraction process from microalgae. Sci Rep. 2021;11:20221.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Manghwar H, Li B, Ding X, Hussain A, Lindsey K, Zhang X, et al. Crispr/cas systems in genome editing: Methodologies and tools for sgrna design, off-target evaluation, and strategies to mitigate off-target effects. Adv Sci. 2020;7:1902312.

    Article  CAS  Google Scholar 

  52. Tong S, Moyo B, Lee CM, Leong K, Bao G. Engineered materials for in vivo delivery of genome-editing machinery. Nat Rev Mater. 2019;4:726–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Shivram H, Cress BF, Knott GJ, Doudna JA. Controlling and enhancing crispr systems. Nat Chem Biol. 2021;17:10–9.

    Article  CAS  PubMed  Google Scholar 

  54. Liu R, Liang L, Freed EF, Gill RT. Directed evolution of crispr/cas systems for precise gene editing. Trends Biotechnol. 2021;39:262–73.

    Article  CAS  PubMed  Google Scholar 

  55. Kaczmarek JC, Kowalski PS, Anderson DG. Advances in the delivery of rna therapeutics: from concept to clinical reality. Genome Med. 2017;9:60.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021;20:101–24.

    Article  CAS  PubMed  Google Scholar 

  57. Hatit MZC, Lokugamage MP, Dobrowolski CN, Paunovska K, Ni H, Zhao K, et al. Species-dependent in vivo mrna delivery and cellular responses to nanoparticles. Nat Nanotechnol. 2022;17:310–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhang X, Zhang H, Gu J, Zhang J, Shi H, Qian H, et al. Engineered extracellular vesicles for cancer therapy. Adv Mater. 2021;33: e2005709.

    Article  PubMed  Google Scholar 

  59. Wang D, Zhang F, Gao G. Crispr-based therapeutic genome editing: strategies and in vivo delivery by aav vectors. Cell. 2020;181:136–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wilbie D, Walther J, Mastrobattista E. Delivery aspects of crispr/cas for in vivo genome editing. Acc Chem Res. 2019;52:1555–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Yu W, Mookherjee S, Chaitankar V, Hiriyanna S, Kim JW, Brooks M, et al. Nrl knockdown by aav-delivered crispr/cas9 prevents retinal degeneration in mice. Nat Commun. 2017;8:14716.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Niu Y, Shen B, Cui Y, Chen Y, Wang J, Wang L, et al. Generation of gene-modified cynomolgus monkey via cas9/rna-mediated gene targeting in one-cell embryos. Cell. 2014;156:836–43.

    Article  CAS  PubMed  Google Scholar 

  63. Xue W, Chen S, Yin H, Tammela T, Papagiannakopoulos T, Joshi NS, et al. Crispr-mediated direct mutation of cancer genes in the mouse liver. Nature. 2014;514:380–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Yan Y, Zhu X, Yu Y, Li C, Zhang Z, Wang F. Nanotechnology strategies for plant genetic engineering. Adv Mater. 2021;24:e2106945.

    Google Scholar 

  65. Chaverra-Rodriguez D, Macias VM, Hughes GL, Pujhari S, Suzuki Y, Peterson DR, et al. Targeted delivery of crispr-cas9 ribonucleoprotein into arthropod ovaries for heritable germline gene editing. Nat Commun. 2018;9:3008.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Yin H, Xue W, Chen S, Bogorad RL, Benedetti E, Grompe M, et al. Genome editing with cas9 in adult mice corrects a disease mutation and phenotype. Nat Biotechnol. 2014;32:551–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Pu Y, Yin H, Dong C, Xiang H, Wu W, Zhou B, et al. Sono-controllable and ros-sensitive crispr-cas9 genome editing for augmented/synergistic ultrasound tumor nanotherapy. Adv Mater. 2021;33: e2104641.

    Article  PubMed  Google Scholar 

  68. Wang HX, Li M, Lee CM, Chakraborty S, Kim HW, Bao G, et al. Crispr/cas9-based genome editing for disease modeling and therapy: Challenges and opportunities for nonviral delivery. Chem Rev. 2017;117:9874–906.

    Article  CAS  PubMed  Google Scholar 

  69. Seki A, Rutz S. Optimized rnp transfection for highly efficient crispr/cas9-mediated gene knockout in primary t cells. J Exp Med. 2018;215:985–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Fajrial AK, He QQ, Wirusanti NI, Slansky JE, Ding X. A review of emerging physical transfection methods for crispr/cas9-mediated gene editing. Theranostics. 2020;10:5532–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Song X, Liu C, Wang N, Huang H, He S, Gong C, et al. Delivery of crispr/cas systems for cancer gene therapy and immunotherapy. Adv Drug Deliv Rev. 2021;168:158–80.

    Article  CAS  PubMed  Google Scholar 

  72. Heckl D, Kowalczyk MS, Yudovich D, Belizaire R, Puram RV, Mcconkey ME, et al. Generation of mouse models of myeloid malignancy with combinatorial genetic lesions using crispr-cas9 genome editing. Nat Biotechnol. 2014;32:941–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Russell S, Bennett J, Wellman JA, Chung DC, Yu ZF, Tillman A, et al. Efficacy and safety of voretigene neparvovec (aav2-hrpe65v2) in patients with rpe65-mediated inherited retinal dystrophy: a randomised, controlled, open-label, phase 3 trial. Lancet. 2017;390:849–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Naldini L. Gene therapy returns to centre stage. Nature. 2015;526:351–60.

    Article  CAS  PubMed  Google Scholar 

  75. Shirley JL, De Jong YP, Terhorst C, Herzog RW. Immune responses to viral gene therapy vectors. Mol Ther. 2020;28:709–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Baum C, Kustikova O, Modlich U, Li Z, Fehse B. Mutagenesis and oncogenesis by chromosomal insertion of gene transfer vectors. Hum Gene Ther. 2006;17:253–63.

    Article  CAS  PubMed  Google Scholar 

  77. Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet. 2003;4:346–58.

    Article  CAS  PubMed  Google Scholar 

  78. Chen F, Alphonse M, Liu Q. Strategies for nonviral nanoparticle-based delivery of crispr/cas9 therapeutics. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2020;12: e1609.

    Article  PubMed  Google Scholar 

  79. Yin H, Song CQ, Dorkin JR, Zhu LJ, Li Y, Wu Q, et al. Therapeutic genome editing by combined viral and non-viral delivery of crispr system components in vivo. Nat Biotechnol. 2016;34:328–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Glass Z, Lee M, Li Y, Xu Q. Engineering the delivery system for crispr-based genome editing. Trends Biotechnol. 2018;36:173–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Liu C, Zhang L, Liu H, Cheng K. Delivery strategies of the crispr-cas9 gene-editing system for therapeutic applications. J Control Release. 2017;266:17–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Li L, Hu S, Chen X. Non-viral delivery systems for crispr/cas9-based genome editing: challenges and opportunities. Biomaterials. 2018;171:207–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Canatella PJ, Karr JF, Petros JA, Prausnitz MR. Quantitative study of electroporation-mediated molecular uptake and cell viability. Biophys J. 2001;80:755–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Bak RO, Dever DP, Reinisch A, Cruz Hernandez D, Majeti R, Porteus MH. Multiplexed genetic engineering of human hematopoietic stem and progenitor cells using crispr/cas9 and aav6. Elife. 2017;6:e27873.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Li L, He ZY, Wei XW, Gao GP, Wei YQ. Challenges in crispr/cas9 delivery: potential roles of nonviral vectors. Hum Gene Ther. 2015;26:452–62.

    Article  CAS  PubMed  Google Scholar 

  86. Mintzer MA, Simanek EE. Nonviral vectors for gene delivery. Chem Rev. 2009;109:259–302.

    Article  CAS  PubMed  Google Scholar 

  87. Pack DW, Hoffman AS, Pun S, Stayton PS. Design and development of polymers for gene delivery. Nat Rev Drug Discov. 2005;4:581–93.

    Article  CAS  PubMed  Google Scholar 

  88. Patra JK, Das G, Fraceto LF, Campos EVR, Rodriguez-Torres MDP, Acosta-Torres LS, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology. 2018;16:71.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Klochkov SG, Neganova ME, Nikolenko VN, Chen K, Somasundaram SG, Kirkland CE, et al. Implications of nanotechnology for the treatment of cancer: recent advances. Semin Cancer Biol. 2021;69:190–9.

    Article  CAS  PubMed  Google Scholar 

  90. Rajasekaran D, Srivastava J, Ebeid K, Gredler R, Akiel M, Jariwala N, et al. Combination of nanoparticle-delivered sirna for astrocyte elevated gene-1 (aeg-1) and all-trans retinoic acid (atra): An effective therapeutic strategy for hepatocellular carcinoma (hcc). Bioconjug Chem. 2015;26:1651–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Liang Y, Iqbal Z, Wang J, Xu L, Xu X, Ouyang K, et al. Cell-derived extracellular vesicles for crispr/cas9 delivery: engineering strategies for cargo packaging and loading. Biomater Sci. 2022;10:4095–106.

    Article  CAS  PubMed  Google Scholar 

  92. Ebeid K, Meng X, Thiel KW, Do AV, Geary SM, Morris AS, et al. Synthetically lethal nanoparticles for treatment of endometrial cancer. Nat Nanotechnol. 2018;13:72–81.

    Article  CAS  PubMed  Google Scholar 

  93. Gunawan C, Lim M, Marquis CP, Amal R. Nanoparticle-protein corona complexes govern the biological fates and functions of nanoparticles. J Mater Chem B. 2014;2:2060–83.

    Article  CAS  PubMed  Google Scholar 

  94. Pan L, He Q, Liu J, Chen Y, Ma M, Zhang L, et al. Nuclear-targeted drug delivery of tat peptide-conjugated monodisperse mesoporous silica nanoparticles. J Am Chem Soc. 2012;134:5722–5.

    Article  CAS  PubMed  Google Scholar 

  95. Degors IMS, Wang C, Rehman ZU, Zuhorn IS. Carriers break barriers in drug delivery: endocytosis and endosomal escape of gene delivery vectors. Acc Chem Res. 2019;52:1750–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Yaron PN, Holt BD, Short PA, Losche M, Islam MF, Dahl KN. Single wall carbon nanotubes enter cells by endocytosis and not membrane penetration. J Nanobiotechnology. 2011;9:45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Rouet R, Thuma BA, Roy MD, Lintner NG, Rubitski DM, Finley JE, et al. Receptor-mediated delivery of crispr-cas9 endonuclease for cell-type-specific gene editing. J Am Chem Soc. 2018;140:6596–603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol. 2015;33:941–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Donahue ND, Acar H, Wilhelm S. Concepts of nanoparticle cellular uptake, intracellular trafficking, and kinetics in nanomedicine. Adv Drug Deliv Rev. 2019;143:68–96.

    Article  CAS  PubMed  Google Scholar 

  100. De Sousa Almeida M, Susnik E, Drasler B, Taladriz-Blanco P, Petri-Fink A, Rothen-Rutishauser B. Understanding nanoparticle endocytosis to improve targeting strategies in nanomedicine. Chem Soc Rev. 2021;50:5397–434.

    Article  Google Scholar 

  101. Behzadi S, Serpooshan V, Tao W, Hamaly MA, Alkawareek MY, Dreaden EC, et al. Cellular uptake of nanoparticles: journey inside the cell. Chem Soc Rev. 2017;46:4218–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Khalil IA, Kogure K, Futaki S, Hama S, Akita H, Ueno M, et al. Octaarginine-modified multifunctional envelope-type nanoparticles for gene delivery. Gene Ther. 2007;14:682–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Xu S, Olenyuk BZ, Okamoto CT, Hamm-Alvarez SF. Targeting receptor-mediated endocytotic pathways with nanoparticles: rationale and advances. Adv Drug Deliv Rev. 2013;65:121–38.

    Article  CAS  PubMed  Google Scholar 

  104. Gilleron J, Querbes W, Zeigerer A, Borodovsky A, Marsico G, Schubert U, et al. Image-based analysis of lipid nanoparticle-mediated sirna delivery, intracellular trafficking and endosomal escape. Nat Biotechnol. 2013;31:638–46.

    Article  CAS  PubMed  Google Scholar 

  105. Sahay G, Querbes W, Alabi C, Eltoukhy A, Sarkar S, Zurenko C, et al. Efficiency of sirna delivery by lipid nanoparticles is limited by endocytic recycling. Nat Biotechnol. 2013;31:653–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Pei D. How do biomolecules cross the cell membrane? Acc Chem Res. 2022;55:309–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Varkouhi AK, Scholte M, Storm G, Haisma HJ. Endosomal escape pathways for delivery of biologicals. J Control Release. 2011;151:220–8.

    Article  CAS  PubMed  Google Scholar 

  108. Wojnilowicz M, Glab A, Bertucci A, Caruso F, Cavalieri F. Super-resolution imaging of proton sponge-triggered rupture of endosomes and cytosolic release of small interfering rna. ACS Nano. 2019;13:187–202.

    Article  CAS  PubMed  Google Scholar 

  109. Yuba E, Kanda Y, Yoshizaki Y, Teranishi R, Harada A, Sugiura K, et al. Ph-sensitive polymer-liposome-based antigen delivery systems potentiated with interferon-gamma gene lipoplex for efficient cancer immunotherapy. Biomaterials. 2015;67:214–24.

    Article  CAS  PubMed  Google Scholar 

  110. Akita H, Kudo A, Minoura A, Yamaguti M, Khalil IA, Moriguchi R, et al. Multi-layered nanoparticles for penetrating the endosome and nuclear membrane via a step-wise membrane fusion process. Biomaterials. 2009;30:2940–9.

    Article  CAS  PubMed  Google Scholar 

  111. Hu Y, Litwin T, Nagaraja AR, Kwong B, Katz J, Watson N, et al. Cytosolic delivery of membrane-impermeable molecules in dendritic cells using ph-responsive core-shell nanoparticles. Nano Lett. 2007;7:3056–64.

    Article  CAS  PubMed  Google Scholar 

  112. Manganiello MJ, Cheng C, Convertine AJ, Bryers JD, Stayton PS. Diblock copolymers with tunable ph transitions for gene delivery. Biomaterials. 2012;33:2301–9.

    Article  CAS  PubMed  Google Scholar 

  113. Semple SC, Akinc A, Chen J, Sandhu AP, Mui BL, Cho CK, et al. Rational design of cationic lipids for sirna delivery. Nat Biotechnol. 2010;28:172–6.

    Article  CAS  PubMed  Google Scholar 

  114. Nakase I, Kobayashi S, Futaki S. Endosome-disruptive peptides for improving cytosolic delivery of bioactive macromolecules. Biopolymers. 2010;94:763–70.

    Article  CAS  PubMed  Google Scholar 

  115. Yang J, Bahreman A, Daudey G, Bussmann J, Olsthoorn RC, Kros A. Drug delivery via cell membrane fusion using lipopeptide modified liposomes. ACS Cent Sci. 2016;2:621–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Selby LI, Cortez-Jugo CM, Such G, Such G, Johnston APR. Nanoescapology: Progress toward understanding the endosomal escape of polymeric nanoparticles. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2017;9:e1452.

    Article  Google Scholar 

  117. Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. Nat Mater. 2013;12:991–1003.

    Article  CAS  PubMed  Google Scholar 

  118. Massignani M, Lopresti C, Blanazs A, Madsen J, Armes SP, Lewis AL, et al. Controlling cellular uptake by surface chemistry, size, and surface topology at the nanoscale. Small. 2009;5:2424–32.

    Article  CAS  PubMed  Google Scholar 

  119. Su X, Fricke J, Kavanagh DG, Irvine DJ. In vitro and in vivo mrna delivery using lipid-enveloped ph-responsive polymer nanoparticles. Mol Pharm. 2011;8:774–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Wu H, Zhu L, Torchilin VP. Ph-sensitive poly(histidine)-peg/dspe-peg co-polymer micelles for cytosolic drug delivery. Biomaterials. 2013;34:1213–22.

    Article  CAS  PubMed  Google Scholar 

  121. Zhao ZX, Gao SY, Wang JC, Chen CJ, Zhao EY, Hou WJ, et al. Self-assembly nanomicelles based on cationic mpeg-pla-b-polyarginine(r15) triblock copolymer for sirna delivery. Biomaterials. 2012;33:6793–807.

    Article  CAS  PubMed  Google Scholar 

  122. Dimitrov DS. Virus entry: Molecular mechanisms and biomedical applications. Nat Rev Microbiol. 2004;2:109–22.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Givens BE, Naguib YW, Geary SM, Devor EJ, Salem AK. Nanoparticle-based delivery of crispr/cas9 genome-editing therapeutics. AAPS J. 2018;20:108.

    Article  PubMed  Google Scholar 

  124. Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the crispr-cas9 system. Nat Protoc. 2013;8:2281–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Shalem O, Sanjana NE, Zhang F. High-throughput functional genomics using crispr-cas9. Nat Rev Genet. 2015;16:299–311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Zuckermann M, Hovestadt V, Knobbe-Thomsen CB, Zapatka M, Northcott PA, Schramm K, et al. Somatic crispr/cas9-mediated tumour suppressor disruption enables versatile brain tumour modelling. Nat Commun. 2015;6:7391.

    Article  CAS  PubMed  Google Scholar 

  127. Mali P, Yang L, Esvelt KM, Aach J, Guell M, Dicarlo JE, et al. Rna-guided human genome engineering via cas9. Science. 2013;339:823–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ, et al. In vivo genome editing using staphylococcus aureus cas9. Nature. 2015;520:186–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Chen S, Sanjana NE, Zheng K, Shalem O, Lee K, Shi X, et al. Genome-wide crispr screen in a mouse model of tumor growth and metastasis. Cell. 2015;160:1246–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Shen B, Zhang W, Zhang J, Zhou J, Wang J, Chen L, et al. Efficient genome modification by crispr-cas9 nickase with minimal off-target effects. Nat Methods. 2014;11:399–402.

    Article  CAS  PubMed  Google Scholar 

  131. Chang N, Sun C, Gao L, Zhu D, Xu X, Zhu X, et al. Genome editing with rna-guided cas9 nuclease in zebrafish embryos. Cell Res. 2013;23:465–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Shen B, Zhang J, Wu H, Wang J, Ma K, Li Z, et al. Generation of gene-modified mice via cas9/rna-mediated gene targeting. Cell Res. 2013;23:720–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Liang X, Potter J, Kumar S, Zou Y, Quintanilla R, Sridharan M, et al. Rapid and highly efficient mammalian cell engineering via cas9 protein transfection. J Biotechnol. 2015;208:44–53.

    Article  CAS  PubMed  Google Scholar 

  134. Kim S, Kim D, Cho SW, Kim J, Kim JS. Highly efficient rna-guided genome editing in human cells via delivery of purified cas9 ribonucleoproteins. Genome Res. 2014;24:1012–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Hendel A, Bak RO, Clark JT, Kennedy AB, Ryan DE, Roy S, et al. Chemically modified guide rnas enhance crispr-cas genome editing in human primary cells. Nat Biotechnol. 2015;33:985–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Woo JW, Kim J, Kwon SI, Corvalan C, Cho SW, Kim H, et al. DNA-free genome editing in plants with preassembled crispr-cas9 ribonucleoproteins. Nat Biotechnol. 2015;33:1162–4.

    Article  CAS  PubMed  Google Scholar 

  137. Crudele JM, Chamberlain JS. Cas9 immunity creates challenges for crispr gene editing therapies. Nat Commun. 2018;9:3497.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Zhang S, Shen J, Li D, Cheng Y. Strategies in the delivery of cas9 ribonucleoprotein for crispr/cas9 genome editing. Theranostics. 2021;11:614–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Lee B, Lee K, Panda S, Gonzales-Rojas R, Chong A, Bugay V, et al. Nanoparticle delivery of crispr into the brain rescues a mouse model of fragile x syndrome from exaggerated repetitive behaviours. Nat Biomed Eng. 2018;2:497–507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Wei T, Cheng Q, Farbiak L, Anderson DG, Langer R, Siegwart DJ. Delivery of tissue-targeted scalpels: Opportunities and challenges for in vivo crispr/cas-based genome editing. ACS Nano. 2020;14:9243–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Wei T, Cheng Q, Min YL, Olson EN, Siegwart DJ. Systemic nanoparticle delivery of crispr-cas9 ribonucleoproteins for effective tissue specific genome editing. Nat Commun. 2020;11:3232.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov. 2019;18:175–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Han JP, Kim M, Choi BS, Lee JH, Lee GS, Jeong M, et al. In vivo delivery of crispr-cas9 using lipid nanoparticles enables antithrombin gene editing for sustainable hemophilia a and b therapy. Sci Adv. 2022;8:6901.

    Article  Google Scholar 

  144. Miao L, Zhang Y, Huang L. Mrna vaccine for cancer immunotherapy. Mol Cancer. 2021;20:41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Cheng X, Lee RJ. The role of helper lipids in lipid nanoparticles (lnps) designed for oligonucleotide delivery. Adv Drug Deliv Rev. 2016;99:129–37.

    Article  CAS  PubMed  Google Scholar 

  146. Kowalski PS, Rudra A, Miao L, Anderson DG. Delivering the messenger: advances in technologies for therapeutic mrna delivery. Mol Ther. 2019;27:710–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Liu Q, Chen F, Hou L, Shen L, Zhang X, Wang D, et al. Nanocarrier-mediated chemo-immunotherapy arrested cancer progression and induced tumor dormancy in desmoplastic melanoma. ACS Nano. 2018;12:7812–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Akinc A, Querbes W, De S, Qin J, Frank-Kamenetsky M, Jayaprakash KN, et al. Targeted delivery of rnai therapeutics with endogenous and exogenous ligand-based mechanisms. Mol Ther. 2010;18:1357–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Fenton OS, Olafson KN, Pillai PS, Mitchell MJ, Langer R. Advances in biomaterials for drug delivery. Adv Mater. 2018;20:e1705328.

    Article  Google Scholar 

  150. Fonseca-Santos B, Gremiao MP, Chorilli M. Nanotechnology-based drug delivery systems for the treatment of alzheimer’s disease. Int J Nanomedicine. 2015;10:4981–5003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and challenges of liposome assisted drug delivery. Front Pharmacol. 2015;6:286.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Gillmore JD, Gane E, Taubel J, Kao J, Fontana M, Maitland ML, et al. Crispr-cas9 in vivo gene editing for transthyretin amyloidosis. N Engl J Med. 2021;385:493–502.

    Article  CAS  PubMed  Google Scholar 

  153. Qiu M, Glass Z, Chen J, Haas M, Jin X, Zhao X, et al. Lipid nanoparticle-mediated codelivery of cas9 mrna and single-guide rna achieves liver-specific in vivo genome editing of angptl3. Proc Natl Acad Sci USA. 2021;118:e2020401118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Akinc A, Maier MA, Manoharan M, Fitzgerald K, Jayaraman M, Barros S, et al. The onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs. Nat Nanotechnol. 2019;14:1084–7.

    Article  CAS  PubMed  Google Scholar 

  155. Bottger R, Pauli G, Chao PH, Al Fayez N, Hohenwarter L, Li SD. Lipid-based nanoparticle technologies for liver targeting. Adv Drug Deliv Rev. 2020;154–155:79–101.

    Article  PubMed  Google Scholar 

  156. Kenjo E, Hozumi H, Makita Y, Iwabuchi KA, Fujimoto N, Matsumoto S, et al. Low immunogenicity of lnp allows repeated administrations of crispr-cas9 mrna into skeletal muscle in mice. Nat Commun. 2021;12:7101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Cheng Q, Wei T, Farbiak L, Johnson LT, Dilliard SA, Siegwart DJ. Selective organ targeting (sort) nanoparticles for tissue-specific mrna delivery and crispr-cas gene editing. Nat Nanotechnol. 2020;15:313–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Cui Z, Zeng C, Huang F, Yuan F, Yan J, Zhao Y, et al. Cas13d knockdown of lung protease ctsl prevents and treats sars-cov-2 infection. Nat Chem Biol. 2022;18:1056–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Lin S, Staahl BT, Alla RK, Doudna JA. Enhanced homology-directed human genome engineering by controlled timing of crispr/cas9 delivery. Elife. 2014;3: e04766.

    Article  PubMed  PubMed Central  Google Scholar 

  160. Zou L, Chen F, Bao J, Wang S, Wang L, Chen M, et al. Preparation, characterization, and anticancer efficacy of evodiamine-loaded plga nanoparticles. Drug Deliv. 2016;23:908–16.

    Article  PubMed  Google Scholar 

  161. Wang Y, Ma B, Abdeen AA, Chen G, Xie R, Saha K, et al. Versatile redox-responsive polyplexes for the delivery of plasmid DNA, messenger rna, and crispr-cas9 genome-editing machinery. ACS Appl Mater Interfaces. 2018;10:31915–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Chen F, Zhang J, Wang L, Wang Y, Chen M. Tumor ph(e)-triggered charge-reversal and redox-responsive nanoparticles for docetaxel delivery in hepatocellular carcinoma treatment. Nanoscale. 2015;7:15763–79.

    Article  CAS  PubMed  Google Scholar 

  163. Ryu N, Kim MA, Park D, Lee B, Kim YR, Kim KH, et al. Effective pei-mediated delivery of crispr-cas9 complex for targeted gene therapy. Nanomedicine. 2018;14:2095–102.

    Article  CAS  PubMed  Google Scholar 

  164. Liu Y, Cao ZT, Xu CF, Lu ZD, Luo YL, Wang J. Optimization of lipid-assisted nanoparticle for disturbing neutrophils-related inflammation. Biomaterials. 2018;172:92–104.

    Article  CAS  PubMed  Google Scholar 

  165. Luo YL, Xu CF, Li HJ, Cao ZT, Liu J, Wang JL, et al. Macrophage-specific in vivo gene editing using cationic lipid-assisted polymeric nanoparticles. ACS Nano. 2018;12:994–1005.

    Article  CAS  PubMed  Google Scholar 

  166. Zhang Y, Shen S, Zhao G, Xu CF, Zhang HB, Luo YL, et al. In situ repurposing of dendritic cells with crispr/cas9-based nanomedicine to induce transplant tolerance. Biomaterials. 2019;217: 119302.

    Article  CAS  PubMed  Google Scholar 

  167. Zhang X, Jin H, Huang X, Chaurasiya B, Dong D, Shanley TP, et al. Robust genome editing in adult vascular endothelium by nanoparticle delivery of crispr-cas9 plasmid DNA. Cell Rep. 2022;38: 110196.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Liu Q, Zhao K, Wang C, Zhang Z, Zheng C, Zhao Y, et al. Multistage delivery nanoparticle facilitates efficient crispr/dcas9 activation and tumor growth suppression in vivo. Adv Sci. 2019;6:1801423.

    Article  Google Scholar 

  169. Li Q, Lv X, Tang C, Yin C. Co-delivery of doxorubicin and crispr/cas9 or rnai-expressing plasmid by chitosan-based nanoparticle for cancer therapy. Carbohydr Polym. 2022;287: 119315.

    Article  CAS  PubMed  Google Scholar 

  170. Wan T, Pan Q, Ping Y. Microneedle-assisted genome editing: a transdermal strategy of targeting nlrp3 by crispr-cas9 for synergistic therapy of inflammatory skin disorders. Sci Adv. 2021;7:eabe2888.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Farbiak L, Cheng Q, Wei T, Alvarez-Benedicto E, Johnson LT, Lee S, et al. All-in-one dendrimer-based lipid nanoparticles enable precise hdr-mediated gene editing in vivo. Adv Mater. 2021;33: e2006619.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Park J, Fong PM, Lu J, Russell KS, Booth CJ, Saltzman WM, et al. Pegylated plga nanoparticles for the improved delivery of doxorubicin. Nanomedicine. 2009;5:410–8.

    Article  CAS  PubMed  Google Scholar 

  173. Faure AC, Dufort S, Josserand V, Perriat P, Coll JL, Roux S, et al. Control of the in vivo biodistribution of hybrid nanoparticles with different poly(ethylene glycol) coatings. Small. 2009;5:2565–75.

    Article  CAS  PubMed  Google Scholar 

  174. Rao NV, Ko H, Lee J, Park JH. Recent progress and advances in stimuli-responsive polymers for cancer therapy. Front Bioeng Biotechnol. 2018;6:110.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Wang S, Liu Q, Li L, Urban MW. Recent advances in stimuli-responsive commodity polymers. Macromol Rapid Commun. 2021;42: e2100054.

    Article  PubMed  Google Scholar 

  176. Li L, Yang Z, Zhu S, He L, Fan W, Tang W, et al. A rationally designed semiconducting polymer brush for nir-ii imaging-guided light-triggered remote control of crispr/cas9 genome editing. Adv Mater. 2019;31: e1901187.

    Article  PubMed  Google Scholar 

  177. Xie R, Wang X, Wang Y, Ye M, Zhao Y, Yandell BS, et al. Ph-responsive polymer nanoparticles for efficient delivery of cas9 ribonucleoprotein with or without donor DNA. Adv Mater. 2022;34: e2110618.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Luther DC, Huang R, Jeon T, Zhang X, Lee YW, Nagaraj H, et al. Delivery of drugs, proteins, and nucleic acids using inorganic nanoparticles. Adv Drug Deliv Rev. 2020;156:188–213.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Goddard ZR, Marin MJ, Russell DA, Searcey M. Active targeting of gold nanoparticles as cancer therapeutics. Chem Soc Rev. 2020;49:8774–89.

    Article  CAS  PubMed  Google Scholar 

  180. Nihongaki Y, Kawano F, Nakajima T, Sato M. Photoactivatable crispr-cas9 for optogenetic genome editing. Nat Biotechnol. 2015;33:755–60.

    Article  CAS  PubMed  Google Scholar 

  181. Wirth J, Garwe F, Meyer R, Csaki A, Stranik O, Fritzsche W. Plasmonically enhanced electron escape from gold nanoparticles and their polarization-dependent excitation transfer along DNA nanowires. Nano Lett. 2014;14:3809–16.

    Article  CAS  PubMed  Google Scholar 

  182. Zhang Z, Wang L, Wang J, Jiang X, Li X, Hu Z, et al. Mesoporous silica-coated gold nanorods as a light-mediated multifunctional theranostic platform for cancer treatment. Adv Mater. 2012;24:1418–23.

    Article  CAS  PubMed  Google Scholar 

  183. Chen X, Chen Y, Xin H, Wan T, Ping Y. Near-infrared optogenetic engineering of photothermal nanocrispr for programmable genome editing. Proc Natl Acad Sci USA. 2020;117:2395–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Peng H, Le C, Wu J, Li XF, Zhang H, Le XC. A genome-editing nanomachine constructed with a clustered regularly interspaced short palindromic repeats system and activated by near-infrared illumination. ACS Nano. 2020;14:2817–26.

    Article  CAS  PubMed  Google Scholar 

  185. Huang L, Zhou M, Abbas G, Li C, Cui M, Zhang XE, et al. A cancer cell membrane-derived biomimetic nanocarrier for synergistic photothermal/gene therapy by efficient delivery of crispr/cas9 and gold nanorods. Adv Healthc Mater. 2022;11: e2201038.

    Article  PubMed  Google Scholar 

  186. Ma L, Yin L, Li X, Chen S, Peng L, Liu G, et al. A smartphone-based visual biosensor for crispr-cas powered sars-cov-2 diagnostics. Biosens Bioelectron. 2022;195: 113646.

    Article  CAS  PubMed  Google Scholar 

  187. Lopez-Valls M, Escalona-Noguero C, Rodriguez-Diaz C, Pardo D, Castellanos M, Milan-Rois P, et al. Cascade: Naked eye-detection of sars-cov-2 using cas13a and gold nanoparticles. Anal Chim Acta. 2022;1205: 339749.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Zhang WS, Pan J, Li F, Zhu M, Xu M, Zhu H, et al. Reverse transcription recombinase polymerase amplification coupled with crispr-cas12a for facile and highly sensitive colorimetric sars-cov-2 detection. Anal Chem. 2021;93:4126–33.

    Article  CAS  PubMed  Google Scholar 

  189. Fan M, Han Y, Gao S, Yan H, Cao L, Li Z, et al. Ultrasmall gold nanoparticles in cancer diagnosis and therapy. Theranostics. 2020;10:4944–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Zhang P, Guo Z, Ullah S, Melagraki G, Afantitis A, Lynch I. Nanotechnology and artificial intelligence to enable sustainable and precision agriculture. Nat Plants. 2021;7:864–76.

    Article  PubMed  Google Scholar 

  191. Wu D, Zhou J, Creyer MN, Yim W, Chen Z, Messersmith PB, et al. Phenolic-enabled nanotechnology: Versatile particle engineering for biomedicine. Chem Soc Rev. 2021;50:4432–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Mousavi SM, Hashemi SA, Ghasemi Y, Atapour A, Amani AM, Savar Dashtaki A, et al. Green synthesis of silver nanoparticles toward bio and medical applications: review study. Artif Cells Nanomed Biotechnol. 2018;46:S855–72.

    Article  CAS  PubMed  Google Scholar 

  193. Zhang D, Ma XL, Gu Y, Huang H, Zhang GW. Green synthesis of metallic nanoparticles and their potential applications to treat cancer. Front Chem. 2020;8:799.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Alnadhari S, Al-Enazi NM, Alshehrei F, Ameen F. A review on biogenic synthesis of metal nanoparticles using marine algae and its applications. Environ Res. 2021;194: 110672.

    Article  CAS  PubMed  Google Scholar 

  195. Fawcett D, Verduin JJ, Shah M, Sharma SB, Poinern GEJ. A review of current research into the biogenic synthesis of metal and metal oxide nanoparticles via marine algae and seagrasses. J Nanosci. 2017;2017:8013850.

    Google Scholar 

  196. Jacob JM, Ravindran R, Narayanan M, Samuel SM, Pugazhendhi A, Kumar G. Microalgae: a prospective low cost green alternative for nanoparticle synthesis. Curr Opin Environ Sci Health. 2021;20: 100163.

    Article  Google Scholar 

  197. Pal S, Tak YK, Song JM. Does the antibacterial activity of silver nanoparticles depend on the shape of the nanoparticle? A study of the gram-negative bacterium escherichia coli. Appl Environ Microbiol. 2007;73:1712–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Makarov VV, Love AJ, Sinitsyna OV, Makarova SS, Yaminsky IV, Taliansky ME, et al. “Green” nanotechnologies: Synthesis of metal nanoparticles using plants. Acta Naturae. 2014;6:35–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Tran TV, Nguyen DTC, Kumar PS, Din ATM, Jalil AA, Vo DN. Green synthesis of zro2 nanoparticles and nanocomposites for biomedical and environmental applications: a review. Environ Chem Lett. 2022;20:1309–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Mohd Yusof H, Mohamad R, Zaidan UH, Abdul Rahman NA. Microbial synthesis of zinc oxide nanoparticles and their potential application as an antimicrobial agent and a feed supplement in animal industry: a review. J Anim Sci Biotechnol. 2019;10:57.

    Article  PubMed  PubMed Central  Google Scholar 

  201. Hulkoti NI, Taranath TC. Biosynthesis of nanoparticles using microbes-a review. Colloids Surf B Biointerfaces. 2014;121:474–83.

    Article  CAS  PubMed  Google Scholar 

  202. Dahoumane SA, Mechouet M, Wijesekera K, Filipe CDM, Sicard C, Bazylinski DA, et al. Algae-mediated biosynthesis of inorganic nanomaterials as a promising route in nanobiotechnology-a review. Green Chem. 2017;19:552–87.

    Article  CAS  Google Scholar 

  203. Rahman A, Kumar S, Bafana A, Lin J, Dahoumane SA, Jeffryes C. A mechanistic view of the light-induced synthesis of silver nanoparticles using extracellular polymeric substances of chlamydomonas reinhardtii. Molecules. 2019;24:3506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Zhang ZW, Chen J, Yang QL, Lan K, Yan ZY, Chen JQ. Eco-friendly intracellular microalgae synthesis of fluorescent cdse qds as a sensitive nanoprobe for determination of imatinib. Sensor Actuat B-Chem. 2018;263:625–33.

    Article  CAS  Google Scholar 

  205. Gahlawat G, Choudhury AR. A review on the biosynthesis of metal and metal salt nanoparticles by microbes. RSC Adv. 2019;9:12944–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Patil MP, Kim GD. Marine microorganisms for synthesis of metallic nanoparticles and their biomedical applications. Colloids Surf B Biointerfaces. 2018;172:487–95.

    Article  CAS  PubMed  Google Scholar 

  207. Sharma A, Sharma S, Sharma K, Chetri SPK, Vashishtha A, Singh P, et al. Algae as crucial organisms in advancing nanotechnology: a systematic review. J Appl Phycol. 2016;28:1759–74.

    Article  CAS  Google Scholar 

  208. Prasad TNVKV, Kambala VSR, Naidu R. Phyconanotechnology: synthesis of silver nanoparticles using brown marine algae cystophora moniliformis and their characterisation. J Appl Phycol. 2013;25:177–82.

    Article  CAS  Google Scholar 

  209. Siddiqi KS, Husen A. Fabrication of metal and metal oxide nanoparticles by algae and their toxic effects. Nanoscale Res Lett. 2016;11:363.

    Article  PubMed  PubMed Central  Google Scholar 

  210. Alijani HQ, Pourseyedi S, Torkzadeh Mahani M, Khatami M. Green synthesis of zinc sulfide (zns) nanoparticles using stevia rebaudiana bertoni and evaluation of its cytotoxic properties. J Mol Struct. 2019;1175:214–8.

    Article  CAS  Google Scholar 

  211. Yun YH, Lee BK, Park K. Controlled drug delivery: Historical perspective for the next generation. J Control Release. 2015;219:2–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Bozzuto G, Molinari A. Liposomes as nanomedical devices. Int J Nanomedicine. 2015;10:975–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Svenson S. The dendrimer paradox–high medical expectations but poor clinical translation. Chem Soc Rev. 2015;44:4131–44.

    Article  CAS  PubMed  Google Scholar 

  214. Sun Y, Ma XL, Hu H. Marine polysaccharides as a versatile biomass for the construction of nano drug delivery systems. Mar Drugs. 2021;19:345.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Chen XY, Zhao X, Gao YY, Yin JQ, Bai MY, Wang FH. Green synthesis of gold nanoparticles using carrageenan oligosaccharide and their in vitro antitumor activity. Mar Drugs. 2018;16:277.

    Article  PubMed  PubMed Central  Google Scholar 

  216. Manivasagan P, Oh J. Marine polysaccharide-based nanomaterials as a novel source of nanobiotechnological applications. Int J Biol Macromol. 2016;82:315–27.

    Article  CAS  PubMed  Google Scholar 

  217. Cardoso MJ, Costa RR, Mano JF. Marine origin polysaccharides in drug delivery systems. Mar Drugs. 2016;14:34.

    Article  PubMed  PubMed Central  Google Scholar 

  218. Manivasagan P, Bharathiraja S, Bui NQ, Jang B, Oh YO, Lim IG, et al. Doxorubicin-loaded fucoidan capped gold nanoparticles for drug delivery and photoacoustic imaging. Int J Biol Macromol. 2016;91:578–88.

    Article  CAS  PubMed  Google Scholar 

  219. Tomoaia G, Horovitz O, Mocanu A, Nita A, Avram A, Racz CP, et al. Effects of doxorubicin mediated by gold nanoparticles and resveratrol in two human cervical tumor cell lines. Colloids Surf B Biointerfaces. 2015;135:726–34.

    Article  CAS  PubMed  Google Scholar 

  220. Venkatpurwar V, Shiras A, Pokharkar V. Porphyran capped gold nanoparticles as a novel carrier for delivery of anticancer drug: in vitro cytotoxicity study. Int J Pharm. 2011;409:314–20.

    Article  CAS  PubMed  Google Scholar 

  221. Chen X, Han W, Zhao X, Tang W, Wang F. Epirubicin-loaded marine carrageenan oligosaccharide capped gold nanoparticle system for ph-triggered anticancer drug release. Sci Rep. 2019;9:6754.

    Article  PubMed  PubMed Central  Google Scholar 

  222. Salem DS, Sliem MA, El-Sesy M, Shouman SA, Badr Y. Improved chemo-photothermal therapy of hepatocellular carcinoma using chitosan-coated gold nanoparticles. J Photochem Photobiol B. 2018;182:92–9.

    Article  CAS  PubMed  Google Scholar 

  223. Manivasagan P, Bharathiraja S, Bui NQ, Lim IG, Oh J. Paclitaxel-loaded chitosan oligosaccharide-stabilized gold nanoparticles as novel agents for drug delivery and photoacoustic imaging of cancer cells. Int J Pharm. 2016;511:367–79.

    Article  CAS  PubMed  Google Scholar 

  224. George A, Shah PA, Shrivastav PS. Natural biodegradable polymers based nano-formulations for drug delivery: a review. Int J Pharm. 2019;561:244–64.

    Article  CAS  PubMed  Google Scholar 

  225. Hussein HA, Abdullah MA. Anticancer compounds derived from marine diatoms. Mar Drugs. 2020;18:356.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Tang F, Li L, Chen D. Mesoporous silica nanoparticles: synthesis, biocompatibility and drug delivery. Adv Mater. 2012;24:1504–34.

    Article  CAS  PubMed  Google Scholar 

  227. Uthappa UT, Brahmkhatri V, Sriram G, Jung HY, Yu J, Kurkuri N, et al. Nature engineered diatom biosilica as drug delivery systems. J Control Release. 2018;281:70–83.

    Article  CAS  PubMed  Google Scholar 

  228. Delasoie J, Zobi F. Natural diatom biosilica as microshuttles in drug delivery systems. Pharmaceutics. 2019;11:537.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Aw MS, Simovic S, Yu Y, Addai-Mensah J, Losic D. Porous silica microshells from diatoms as biocarrier for drug delivery applications. Powder Technol. 2012;223:52–8.

    Article  CAS  Google Scholar 

  230. Delasoie J, Rossier J, Haeni L, Rothen-Rutishauser B, Zobi F. Slow-targeted release of a ruthenium anticancer agent from vitamin b12 functionalized marine diatom microalgae. Dalton Trans. 2018;47:17221–32.

    Article  CAS  PubMed  Google Scholar 

  231. Sasirekha R, Sheena TS, Sathiya Deepika M, Santhanam P, Townley HE, Jeganathan K, et al. Surface engineered amphora subtropica frustules using chitosan as a drug delivery platform for anticancer therapy. Mater Sci Eng C Mater Biol Appl. 2019;94:56–64.

    Article  CAS  PubMed  Google Scholar 

  232. Fu P, Zhang J, Li H, Mak M, Xu W, Tao Z. Extracellular vesicles as delivery systems at nano-/micro-scale. Adv Drug Deliv Rev. 2021;179: 113910.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Paterna A, Rao E, Adamo G, Raccosta S, Picciotto S, Romancino D, et al. Isolation of extracellular vesicles from microalgae: a renewable and scalable bioprocess. Front Bioeng Biotechnol. 2022;10: 836747.

    Article  PubMed  PubMed Central  Google Scholar 

  234. Adamo G, Fierli D, Romancino DP, Picciotto S, Barone ME, Aranyos A, et al. Nanoalgosomes: Introducing extracellular vesicles produced by microalgae. J Extracell Vesicles. 2021;10: e12081.

    Article  PubMed  PubMed Central  Google Scholar 

  235. Zhang F, Zhuang J, Li Z, Gong H, De Avila BE, Duan Y, et al. Nanoparticle-modified microrobots for in vivo antibiotic delivery to treat acute bacterial pneumonia. Nat Mater. 2022;21:1324–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Sun L, Yu Y, Chen Z, Bian F, Ye F, Sun L, et al. Biohybrid robotics with living cell actuation. Chem Soc Rev. 2020;49:4043–69.

    Article  CAS  PubMed  Google Scholar 

  237. Chen QW, Qiao JY, Liu XH, Zhang C, Zhang XZ. Customized materials-assisted microorganisms in tumor therapeutics. Chem Soc Rev. 2021;50:12576–615.

    Article  CAS  PubMed  Google Scholar 

  238. Xin H, Zhao N, Wang Y, Zhao X, Pan T, Shi Y, et al. Optically controlled living micromotors for the manipulation and disruption of biological targets. Nano Lett. 2020;20:7177–85.

    Article  CAS  PubMed  Google Scholar 

  239. Dawiec-Lisniewska A, Podstawczyk D, Bastrzyk A, Czuba K, Pacyna-Iwanicka K, Okoro OV, et al. New trends in biotechnological applications of photosynthetic microorganisms. Biotechnol Adv. 2022;59: 107988.

    Article  CAS  PubMed  Google Scholar 

  240. Zhang D, Zhong D, Ouyang J, He J, Qi Y, Chen W, et al. Microalgae-based oral microcarriers for gut microbiota homeostasis and intestinal protection in cancer radiotherapy. Nat Commun. 2022;13:1413.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Li M, Wu J, Lin D, Yang J, Jiao N, Wang Y, et al. A diatom-based biohybrid microrobot with a high drug-loading capacity and ph-sensitive drug release for target therapy. Acta Biomater. 2022;154:443–53.

    Article  CAS  PubMed  Google Scholar 

  242. Liu L, Wu J, Chen B, Gao J, Li T, Ye Y, et al. Magnetically actuated biohybrid microswimmers for precise photothermal muscle contraction. ACS Nano. 2022;16:6515–26.

    Article  CAS  PubMed  Google Scholar 

  243. Yan X, Zhou Q, Vincent M, Deng Y, Yu J, Xu J, et al. Multifunctional biohybrid magnetite microrobots for imaging-guided therapy. Sci Robot. 2017;2: eaaq155.

    Article  Google Scholar 

  244. Yasa O, Erkoc P, Alapan Y, Sitti M. Microalga-powered microswimmers toward active cargo delivery. Adv Mater. 2018;30: e1804130.

    Article  PubMed  Google Scholar 

  245. Zhong DN, Li WL, Qi YC, He J, Zhou M. Photosynthetic biohybrid nanoswimmers system to alleviate tumor hypoxia for fl/pa/mr imaging-guided enhanced radio-photodynamic synergetic therapy. Adv Funct Mater. 2020;30: 1910395.

    Article  CAS  Google Scholar 

  246. Gong D, Celi N, Zhang D, Cai J. Magnetic biohybrid microrobot multimers based on chlorella cells for enhanced targeted drug delivery. ACS Appl Mater Interfaces. 2022;14:6320–30.

    Article  CAS  PubMed  Google Scholar 

  247. Akolpoglu MB, Dogan NO, Bozuyuk U, Ceylan H, Kizilel S, Sitti M. High-yield production of biohybrid microalgae for on-demand cargo delivery. Adv Sci. 2020;7:2001256.

    Article  CAS  Google Scholar 

  248. Shchelik IS, Molino JVD, Gademann K. Biohybrid microswimmers against bacterial infections. Acta Biomater. 2021;136:99–110.

    Article  CAS  PubMed  Google Scholar 

  249. Zhong D, Zhang D, Xie T, Zhou M. Biodegradable microalgae-based carriers for targeted delivery and imaging-guided therapy toward lung metastasis of breast cancer. Small. 2020;16: e2000819.

    Article  PubMed  Google Scholar 

  250. Wang Y, Huang C, Zhao W. Recent advances of the biological and biomedical applications of crispr/cas systems. Mol Biol Rep. 2022;49:7087–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Tan FHP, Nadir N, Sudesh K. Microalgal biomass as feedstock for bacterial production of pha: advances and future prospects. Front Bioeng Biotechnol. 2022;10: 879476.

    Article  PubMed  PubMed Central  Google Scholar 

  252. Sreenikethanam A, Raj S, Gugulothu P, Bajhaiya AK. Genetic engineering of microalgae for secondary metabolite production: recent developments, challenges, and future prospects. Front Bioeng Biotechnol. 2022;10: 836056.

    Article  PubMed  PubMed Central  Google Scholar 

  253. Schmidt TJN, Berarducci B, Konstantinidou S, Raffa V. Crispr/cas9 in the era of nanomedicine and synthetic biology. Drug Discov Today. 2023;28: 103375.

    Article  CAS  PubMed  Google Scholar 

  254. Wang D, Li Y, Hu X, Su W, Zhong M. Combined enzymatic and mechanical cell disruption and lipid extraction of green alga neochloris oleoabundans. Int J Mol Sci. 2015;16:7707–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  255. Ren X, Liu Y, Fan C, Hong H, Wu W, Zhang W, et al. Production, processing, and protection of microalgal n-3 pufa-rich oil. Foods. 2022;11:1215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Guiheneuf F, Khan A, Tran LS. Genetic engineering: a promising tool to engender physiological, biochemical, and molecular stress resilience in green microalgae. Front Plant Sci. 2016;7:400.

    Article  PubMed  PubMed Central  Google Scholar 

  257. Liu H, Ding Y, Zhou Y, Jin W, Xie K, Chen LL. Crispr-p 2.0: an improved crispr-cas9 tool for genome editing in plants. Mol Plant. 2017;10:530–2.

    Article  CAS  PubMed  Google Scholar 

  258. Heigwer F, Kerr G, Boutros M. E-crisp: fast crispr target site identification. Nat Methods. 2014;11:122–3.

    Article  CAS  PubMed  Google Scholar 

  259. Abby SS, Neron B, Menager H, Touchon M, Rocha EP. Macsyfinder: a program to mine genomes for molecular systems with an application to crispr-cas systems. PLoS ONE. 2014;9: e110726.

    Article  PubMed  PubMed Central  Google Scholar 

  260. Dasgupta I, Flotte TR, Keeler AM. Crispr/cas-dependent and nuclease-free in vivo therapeutic gene editing. Hum Gene Ther. 2021;32:275–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  261. Liu M, Rehman S, Tang X, Gu K, Fan Q, Chen D, et al. Methodologies for improving hdr efficiency. Front Genet. 2018;9:691.

    Article  CAS  PubMed  Google Scholar 

  262. Maruyama T, Dougan SK, Truttmann MC, Bilate AM, Ingram JR, Ploegh HL. Increasing the efficiency of precise genome editing with crispr-cas9 by inhibition of nonhomologous end joining. Nat Biotechnol. 2015;33:538–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  263. Paulsen BS, Mandal PK, Frock RL, Boyraz B, Yadav R, Upadhyayula S, et al. Ectopic expression of rad52 and dn53bp1 improves homology-directed repair during crispr-cas9 genome editing. Nat Biomed Eng. 2017;1:878–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  264. Ferrari S, Jacob A, Beretta S, Unali G, Albano L, Vavassori V, et al. Efficient gene editing of human long-term hematopoietic stem cells validated by clonal tracking. Nat Biotechnol. 2020;38:1298–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  265. Renaud JB, Boix C, Charpentier M, De Cian A, Cochennec J, Duvernois-Berthet E, et al. Improved genome editing efficiency and flexibility using modified oligonucleotides with talen and crispr-cas9 nucleases. Cell Rep. 2016;14:2263–72.

    Article  CAS  PubMed  Google Scholar 

  266. Ma M, Zhuang F, Hu X, Wang B, Wen XZ, Ji JF, et al. Efficient generation of mice carrying homozygous double-floxp alleles using the cas9-avidin/biotin-donor DNA system. Cell Res. 2017;27:578–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  267. Khan MI, Shin JH, Kim JD. The promising future of microalgae: current status, challenges, and optimization of a sustainable and renewable industry for biofuels, feed, and other products. Microb Cell Fact. 2018;17:36.

    Article  PubMed  PubMed Central  Google Scholar 

  268. Deguchi M, Kane S, Potlakayala S, George H, Proano R, Sheri V, et al. Metabolic engineering strategies of industrial hemp (cannabis sativa l.): a brief review of the advances and challenges. Front Plant Sci. 2020;11:580621.

    Article  PubMed  PubMed Central  Google Scholar 

  269. Yi Z, Xu M, Magnusdottir M, Zhang Y, Brynjolfsson S, Fu W. Photo-oxidative stress-driven mutagenesis and adaptive evolution on the marine diatom phaeodactylum tricornutum for enhanced carotenoid accumulation. Mar Drugs. 2015;13:6138–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Fu W, Nelson DR, Mystikou A, Daakour S, Salehi-Ashtiani K. Advances in microalgal research and engineering development. Curr Opin Biotechnol. 2019;59:157–64.

    Article  CAS  PubMed  Google Scholar 

  271. Rosales-Mendoza S, Solis-Andrade KI, Marquez-Escobar VA, Gonzalez-Ortega O, Banuelos-Hernandez B. Current advances in the algae-made biopharmaceuticals field. Expert Opin Biol Ther. 2020;20:751–66.

    Article  CAS  PubMed  Google Scholar 

  272. Kesik-Brodacka M. Progress in biopharmaceutical development. Biotechnol Appl Biochem. 2018;65:306–22.

    Article  CAS  PubMed  Google Scholar 

  273. Lingg N, Zhang P, Song Z, Bardor M. The sweet tooth of biopharmaceuticals: importance of recombinant protein glycosylation analysis. Biotechnol J. 2012;7:1462–72.

    Article  CAS  PubMed  Google Scholar 

  274. Reddy KV, Yedery RD, Aranha C. Antimicrobial peptides: Premises and promises. Int J Antimicrob Agents. 2004;24:536–47.

    Article  CAS  PubMed  Google Scholar 

  275. Griesbeck C, Kobl I, Heitzer M. Chlamydomonas reinhardtii: a protein expression system for pharmaceutical and biotechnological proteins. Mol Biotechnol. 2006;34:213–23.

    Article  CAS  PubMed  Google Scholar 

  276. Walmsley AM, Arntzen CJ. Plants for delivery of edible vaccines. Curr Opin Biotechnol. 2000;11:126–9.

    Article  CAS  PubMed  Google Scholar 

  277. Lopez-Pacheco IY, Rodas-Zuluaga LI, Cuellar-Bermudez SP, Hidalgo-Vazquez E, Molina-Vazquez A, Araujo RG, et al. Revalorization of microalgae biomass for synergistic interaction and sustainable applications: Bioplastic generation. Mar Drugs. 2022;20:601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  278. Marinescu M, Popa CV. Pyridine compounds with antimicrobial and antiviral activities. Int J Mol Sci. 2022;23:5659.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Lai CL, Lau JY, Wu PC, Ngan H, Chung HT, Mitchell SJ, et al. Recombinant interferon-alpha in inoperable hepatocellular carcinoma: a randomized controlled trial. Hepatology. 1993;17:389–94.

    Article  CAS  PubMed  Google Scholar 

  280. Li H, Liu Q, Cui K, Liu J, Ren Y, Shi D. Expression of biologically active human interferon alpha 2b in the milk of transgenic mice. Transgenic Res. 2013;22:169–78.

    Article  CAS  PubMed  Google Scholar 

  281. Jarquin-Cordero M, Chavez MN, Centeno-Cerdas C, Bohne AV, Hopfner U, Machens HG, et al. Towards a biotechnological platform for the production of human pro-angiogenic growth factors in the green alga chlamydomonas reinhardtii. Appl Microbiol Biotechnol. 2020;104:725–39.

    Article  CAS  PubMed  Google Scholar 

  282. Centeno-Cerdas C, Jarquin-Cordero M, Chavez MN, Hopfner U, Holmes C, Schmauss D, et al. Development of photosynthetic sutures for the local delivery of oxygen and recombinant growth factors in wounds. Acta Biomater. 2018;81:184–94.

    Article  CAS  PubMed  Google Scholar 

  283. Chavez MN, Schenck TL, Hopfner U, Centeno-Cerdas C, Somlai-Schweiger I, Schwarz C, et al. Towards autotrophic tissue engineering: photosynthetic gene therapy for regeneration. Biomaterials. 2016;75:25–36.

    Article  CAS  PubMed  Google Scholar 

  284. Feng S, Feng W, Zhao L, Gu H, Li Q, Shi K, et al. Preparation of transgenic dunaliella salina for immunization against white spot syndrome virus in crayfish. Arch Virol. 2014;159:519–25.

    Article  CAS  PubMed  Google Scholar 

  285. Hernandez-Ramirez J, Wong-Arce A, Gonzalez-Ortega O, Rosales-Mendoza S. Expression in algae of a chimeric protein carrying several epitopes from tumor associated antigens. Int J Biol Macromol. 2020;147:46–52.

    Article  CAS  PubMed  Google Scholar 

  286. Chia WY, Kok H, Chew KW, Low SS, Show PL. Can algae contribute to the war with covid-19? Bioengineered. 2021;12:1226–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  287. Aurisicchio L, Peruzzi D, Koo G, Wei WZ, La Monica N, Ciliberto G. Immunogenicity and therapeutic efficacy of a dual-component genetic cancer vaccine cotargeting carcinoembryonic antigen and her2/neu in preclinical models. Hum Gene Ther. 2014;25:121–31.

    Article  CAS  PubMed  Google Scholar 

  288. Kufe DW. Muc1-c oncoprotein as a target in breast cancer: activation of signaling pathways and therapeutic approaches. Oncogene. 2013;32:1073–81.

    Article  CAS  PubMed  Google Scholar 

  289. Rafiq S, Purdon TJ, Daniyan AF, Koneru M, Dao T, Liu C, et al. Optimized t-cell receptor-mimic chimeric antigen receptor t cells directed toward the intracellular wilms tumor 1 antigen. Leukemia. 2017;31:1788–97.

    Article  CAS  PubMed  Google Scholar 

  290. Soysal SD, Muenst S, Kan-Mitchell J, Huarte E, Zhang X, Wilkinson-Ryan I, et al. Identification and translational validation of novel mammaglobin-a cd8 t cell epitopes. Breast Cancer Res Treat. 2014;147:527–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  291. Lin TW, Huang PH, Liao BH, Chao TL, Tsai YM, Chang SC, et al. Tag-free sars-cov-2 receptor binding domain (rbd), but not c-terminal tagged sars-cov-2 rbd, induces a rapid and potent neutralizing antibody response. Vaccines. 2022;10:1839.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  292. Georgianna DR, Mayfield SP. Exploiting diversity and synthetic biology for the production of algal biofuels. Nature. 2012;488:329–35.

    Article  CAS  PubMed  Google Scholar 

  293. Gilmour DJ. Microalgae for biofuel production. Adv Appl Microbiol. 2019;109:1–30.

    Article  CAS  PubMed  Google Scholar 

  294. Wen X, Du K, Wang Z, Peng X, Luo L, Tao H, et al. Effective cultivation of microalgae for biofuel production: a pilot-scale evaluation of a novel oleaginous microalga graesiella sp. Wbg-1. Biotechnol Biofuels. 2016;9:123.

    Article  PubMed  PubMed Central  Google Scholar 

  295. Blatti JL, Beld J, Behnke CA, Mendez M, Mayfield SP, Burkart MD. Manipulating fatty acid biosynthesis in microalgae for biofuel through protein-protein interactions. PLoS ONE. 2012;7: e42949.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  296. Frleta R, Popovic M, Smital T, Simat V. Comparison of growth and chemical profile of diatom skeletonema grevillei in bioreactor and incubation-shaking cabinet in two growth phases. Mar Drugs. 2022;20:697.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  297. Ward VCA, Rehmann L. Fast media optimization for mixotrophic cultivation of chlorella vulgaris. Sci Rep. 2019;9:19262.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  298. Luo L, Ren H, Pei X, Xie G, Xing D, Dai Y, et al. Simultaneous nutrition removal and high-efficiency biomass and lipid accumulation by microalgae using anaerobic digested effluent from cattle manure combined with municipal wastewater. Biotechnol Biofuels. 2019;12:218.

    Article  PubMed  PubMed Central  Google Scholar 

  299. Goodman JM, Boone-Heinonen J, Richardson DM, Andrea SB, Messer LC. Analyzing policies through a dohad lens: What can we learn? Int J Environ Res Public Health. 2018;15:2906.

    Article  PubMed  PubMed Central  Google Scholar 

  300. Andrade-Guel M, Cabello-Alvarado C, Bartolo-Perez P, Medellin-Banda DI, Avila-Orta CA, Cruz-Ortiz B, et al. Surface modification of tio(2)/zno nanoparticles by organic acids with enhanced methylene blue and rhodamine b dye adsorption properties. RSC Adv. 2022;12:28494–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  301. Forootanfar H, Rezaei S, Zeinvand-Lorestani H, Tahmasbi H, Mogharabi M, Ameri A, et al. Studies on the laccase-mediated decolorization, kinetic, and microtoxicity of some synthetic azo dyes. J Environ Health Sci Eng. 2016;14:7.

    Article  PubMed  PubMed Central  Google Scholar 

  302. Srivastava A, Seo SH, Ko SR, Ahn CY, Oh HM. Bioflocculation in natural and engineered systems: Current perspectives. Crit Rev Biotechnol. 2018;38:1176–94.

    Article  CAS  PubMed  Google Scholar 

  303. Savchenko O, Xing J, Yang X, Gu Q, Shaheen M, Huang M, et al. Algal cell response to pulsed waved stimulation and its application to increase algal lipid production. Sci Rep. 2017;7:42003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  304. Fuchs T, Arnold ND, Garbe D, Deimel S, Lorenzen J, Masri M, et al. A newly designed automatically controlled, sterilizable flat panel photobioreactor for axenic algae culture. Front Bioeng Biotechnol. 2021;9: 697354.

    Article  PubMed  PubMed Central  Google Scholar 

  305. Ranjbar S, Malcata FX. Is genetic engineering a route to enhance microalgae-mediated bioremediation of heavy metal-containing effluents? Molecules. 2022;27:1473.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  306. Goveas LC, Nayak S, Vinayagam R, Loke Show P, Selvaraj R. Microalgal remediation and valorisation of polluted wastewaters for zero-carbon circular bioeconomy. Bioresour Technol. 2022;365: 128169.

    Article  CAS  PubMed  Google Scholar 

  307. Gondi R, Kavitha S, Yukesh Kannah R, Parthiba Karthikeyan O, Kumar G, Kumar Tyagi V, et al. Algal-based system for removal of emerging pollutants from wastewater: a review. Bioresour Technol. 2022;344: 126245.

    Article  CAS  PubMed  Google Scholar 

  308. Ravutsov M, Mitrev Y, Shestakova P, Lazarova H, Simeonov S, Popova M. Co(2) adsorption on modified mesoporous silicas: the role of the adsorption sites. Nanomaterials. 2021;11:2831.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  309. Rekker S, Ives MC, Wade B, Webb L, Greig C. Measuring corporate paris compliance using a strict science-based approach. Nat Commun. 2022;13:4441.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  310. Chen Y, Xu C, Vaidyanathan S. Microalgae: a robust “green bio-bridge” between energy and environment. Crit Rev Biotechnol. 2018;38:351–68.

    Article  CAS  PubMed  Google Scholar 

  311. Cheah WY, Show PL, Chang JS, Ling TC, Juan JC. Biosequestration of atmospheric co2 and flue gas-containing co2 by microalgae. Bioresour Technol. 2015;184:190–201.

    Article  CAS  PubMed  Google Scholar 

  312. Li L, Huang J, Almutairi AW, Lan X, Zheng L, Lin Y, et al. Integrated approach for enhanced bio-oil recovery from disposed face masks through co-hydrothermal liquefaction with spirulina platensis grown in wastewater. Biomass Convers Biorefin. 2021;25:1–12.

    Google Scholar 

  313. Solovchenko AKhozin-Goldberg I. High-co2 tolerance in microalgae: possible mechanisms and implications for biotechnology and bioremediation. Biotechnol Lett. 2013;35:1745–52.

    Article  Google Scholar 

  314. Choi KR, Jang WD, Yang D, Cho JS, Park D, Lee SY. Systems metabolic engineering strategies: Integrating systems and synthetic biology with metabolic engineering. Trends Biotechnol. 2019;37:817–37.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (No. U1804112), the Basic Research Project of the Key Research Program of Colleges and Universities in Henan Province (23ZX005), and the Natural Science Foundation of Henan Province (232300421164).

Author information

Authors and Affiliations

Authors

Contributions

All authors took part in writing, reviewing, and editing the manuscript. SYF, XX, and JJL wrote the manuscript. AFL, QQW and DDG prepared the figures. SXL and ZLW created the table. TG and YLL collected and organized the literature. JZ modified the paper. PLS and DYYT reviewed and edited the paper. All authors reviewed the manuscript and approved it for publication.

Corresponding authors

Correspondence to Shuying Feng, Tao Guo, Jin Zhou or Pau Loke Show.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Feng, S., Xie, X., Liu, J. et al. A potential paradigm in CRISPR/Cas systems delivery: at the crossroad of microalgal gene editing and algal-mediated nanoparticles. J Nanobiotechnol 21, 370 (2023). https://doi.org/10.1186/s12951-023-02139-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-023-02139-z

Keywords