Skip to main content

Non-viral gene delivery to human mesenchymal stem cells: a practical guide towards cell engineering

Abstract

In recent decades, human mesenchymal stem cells (hMSCs) have gained momentum in the field of cell therapy for treating cartilage and bone injuries. Despite the tri-lineage multipotency, proliferative properties, and potent immunomodulatory effects of hMSCs, their clinical potential is hindered by donor variations, limiting their use in medical settings. To address this challenge, gene delivery technologies have emerged as a promising approach to modulate the phenotype and commitment of hMSCs towards specific cell lineages, thereby enhancing osteochondral repair strategies. This review provides a comprehensive overview of current non-viral gene delivery approaches used to engineer MSCs, highlighting key factors such as the choice of nucleic acid or delivery vector, transfection strategies, and experimental parameters. Additionally, it outlines various protocols and methods for qualitative and quantitative evaluation of their therapeutic potential as a delivery system in osteochondral regenerative applications. In summary, this technical review offers a practical guide for optimizing non-viral systems in osteochondral regenerative approaches.

Graphical Abstract

hMSCs constitute a key target population for gene therapy techniques. Nevertheless, there is a long way to go for their translation into clinical treatments. In this review, we remind the most relevant transfection conditions to be optimized, such as the type of nucleic acid or delivery vector, the transfection strategy, and the experimental parameters to accurately evaluate a delivery system. This survey provides a practical guide to optimizing non-viral systems for osteochondral regenerative approaches.

Highlights

- hMSCs-based therapies represent a promising strategy for osteochondral regeneration and repair;

- Non-viral gene delivery vectors are a promising and safe tool to engineer hMSCs ex vivo;

- There is a lack of standardized procedures to engineer hMSCs ex vivo;

- Optimization of key transfection parameters is needed to effectively engineer hMSCs.

Introduction

In the last decades, human mesenchymal stem cells (hMSCs) have gained momentum in several cell therapy applications for the treatment of cartilage and bone lesions [1,2,3,4]. hMSCs are multipotent stem cells that can be isolated and expanded from many tissues, such as bone marrow (hBMMSCs) [5], umbilical cord (hUCMSCs) [6], and adipose tissue (hAMSCs) [7], possess high capacity for self-renewal, display a potent immunomodulatory effect in vivo, and under appropriate culture conditions display a multi-lineage multipotency differentiation in vitro (i.e., they can differentiate into osteoblasts, adipocytes, chondrocytes, myocytes, and neurons). All these characteristics make hMSCs a very appealing tool for tissue engineering, regenerative medicine approaches [5], and the treatment of many diseases and disorders including cardiovascular diseases [8], autoimmunity [9], and cancer [7].

However, the aging of hMSCs, which relies on donor age, is a critical factor affecting cell therapy outcomes, primarily when used in cartilage tissue repair strategies [10]. In this context, the possibility to modulate the phenotype and commitment of hMSCs toward a selective cell lineage using gene delivery technologies has paved the way for the development of ever more effective osteochondral repair strategies [11]. To date, conventional approaches relying on the use of recombinant growth factors (GFs) have significant drawbacks such as the short half-life of proteins, and rapid body clearance. This implies repeated administrations to achieve and lengthen the therapeutic effect of the treatment [12]. In this scenario, gene delivery strategies based on the delivery of osteo or chondrogenic genes, such as those encoding for the insulin-like growth factor-1 (IGF-1) [13,14,15], transforming growth factor beta (TGF-β) [16,17,18,19], bone morphogenic proteins (BMPs) [20,21,22,23], runt-related transcription factors (RUNXs) [24,25,26], as well as genes encoding for the SOX family transcription factors [27,28,29] among others, did improve the regenerative potential of hMSCs (for comprehensive reviews, please refer to [30,31,32]). Although various approaches have proven promising in vitro and in vivo, the implementation of engineered cell therapies is still painfully slow. In this light, successful strategies to efficiently modulate the hMSCs’ behavior for clinical use are mandatory [33].

As a rule of thumb, an effective gene delivery strategy must be very efficient in transferring nucleic acids (NAs) to target cell populations allowing tighter control of gene expression over time while minimizing cytotoxicity and safety concerns arising from the overall process. This aspect holds particular importance when working with primary cell cultures, such as hMSCs, as they are known to be challenging to transfect. Indeed, these cells exhibit higher sensitivity and lower division rates compared to cell lines, making the successful delivery of genetic material more difficult [34]. So far, much effort has been devoted to devising suitable means to improve the delivery efficiency of NAs into target hMSCs. Such strategies are generally classified into two main categories based on the way used to transfer the genetic cargo into cells: gene delivery through physical methods and delivery mediated by vectors [35].

Generally, physical methods, including electroporation [36,37,38], sonoporation [39,40,41], magnetofection [42,43,44], and micro-injection [45,46,47], involve the application of mechanical or electrical forces to temporarily create pores in the lipid bilayer of the plasma membrane. This allows for the introduction of the (naked) NA payload. Therefore, physical methods are ideally suited for treating cells cultured as two-dimensional monolayers ex vivo. Overall, these methods are very effective in transferring genetic cargo to hMSCs, as they force the entry of NAs into cells through the transient disruption of the plasma membrane, instead of relying on endocytosis pathways [48]. Nonetheless, side effects related to high cytotoxicity make them somewhat inconvenient [49].

Another popular way to deliver NAs into target cells involves the use of vector-based delivery systems, commonly referred to as carriers. Their function consists in encapsulating and protecting the NAs within particles, significantly improving their delivery to target cells. These carriers can be classified into two main types: viral and non-viral vectors. Among them, viral vectors, that is, engineered viruses completely void of parent virus genes, harness the viral infection pathway while avoiding the subsequent viral protein expression upon transduction of host cells [50]. Although high transduction efficiency and stable gene expression may be achieved with viral carriers, they are still plagued by inherent issues such as the limited size of NAs that can be packed and delivered, random recombination (i.e., oncogenic potential), tropism, cytotoxicity, and immunogenicity [51]. These concerns have prompted the development of non-viral alternatives, providing scalable, robust, and cost-effective solutions.

Within the last few years, non-viral gene delivery vectors are witnessing a surge of interest, which has led to the recent development of effective products such as the Pfizer/BioNTech and Moderna COVID-19 vaccines [52]. The success of COVID-19 vaccines has given new momentum and legitimized the use of non-viral gene delivery strategies in clinics. Non-viral gene delivery vectors encompass various options, including cationic lipids (CLs) and polymers (CPs), which are able to spontaneously interact with NAs, forming complexes known as lipoplexes and polyplexes, respectively. These complexes serve as a vehicle to carry the genetic cargo into host cells [35, 53] through a process called transfection. In addition to CLs and CPs, there are other non-viral gene delivery carriers available, such as inorganic nanoparticles. Examples include calcium phosphate and mesoporous silica nanoparticles, which have demonstrated successful use in gene delivery (for a comprehensive review on this topic, please refer to [54, 55]).

Non-viral vectors have been extensively utilized in vitro over the past two decades to transfer target genes into hMSCs and control their behavior [31, 56] with some success. There is still a long way to go to find an effective way to engineer hMSCs via gene transfer. This is mainly due to the general lack of standardized test procedures for the screening and implementation of gene delivery strategies, thus largely contributing to the number of inconsistent findings. The variability in test protocols used to transfer NAs in vitro into patient-derived hMSCs using non-viral vectors has given rise to a very crowded and controversial body of literature that makes it hard to find the most suitable combination of factors and parameters to effectively transfect target cells.

In this scenario, the goal of this review is twofold. On one hand, it provides an overview of the current non-viral vectors and strategies used to deliver NAs to hMSCs in vitro to commit them toward bone and cartilage lineages. On the other hand, we sought to highlight the main protocols and parameters affecting the transfection of hMSCs that should be taken into account when performing in vitro research intended for osteochondral applications and to suggest novel ways to expedite and improve the gene transfer to hMSCs. In such a way, as a proof of concept, we conducted a series of transfection experiments using a 25 kDa branched polyethyleneimine (bPEI) as a transfection agent for a plasmid DNA (pDNA) encoding the luciferase reporter gene in primary cultures of MSCs. The purpose of these experiments was to provide the reader with valuable insights into the key factors affecting transfection efficacy in MSCs, including cell density, NA dose, and transfection duration. Besides, this literature survey is intended to point out the most prominent factors and parameters that may affect the transfection output and to provide tips on how to evaluate qualitatively and quantitatively the transfection outcomes in this cell type.

Non-viral gene delivery carriers for MSCs

Non-viral vectors offer distinct advantages over both viral counterparts and physical methods. Above all, they are renowned for their ease of use, along with the possibility to fine-tune their physicochemical properties. To design complexes that best fit the target hMSCs, a wide variety of polymers and lipids have been devised and optimized over years. Nevertheless, each has pros and cons, such that the ideal vector has not been identified yet. Herein, we seek to highlight the most prominent non-viral carriers and the progress made in improving their chemical features to achieve great gene transfer to hMSCs without excessive side effects.

➣ Cationic polymers (CPs)

Cationic polymers (CPs) (Fig. 1A) represent the first choice when dealing with the transfection of MSCs. Most of the strategies developed so far devised the use of gold standard polymeric vectors, namely the class of synthetic polyethyleneimines (PEIs) [23, 27, 28, 57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72], poly-L-lysines (PLLs) [73], and dendrimers such as polyamidoamines (PAMAMs) [74,75,76], as well as biopolymers such as chitosans [57, 77]. These macromolecules have some protonatable moieties, such as primary, secondary, and tertiary amines, which account for their interaction with NAs and the cell membrane. The advantages of polymer-based carriers rely on their ease of use and the possibility of fine-tuning some features, such as the molecular weight (Mw), branching degree, and chain length, to achieve a good trade-off between efficiency and inherent cytotoxicity of the resulting complexes [35, 78]. Nonetheless, the performances of CPs are affected by their limited ability to overcome plasma membranes and release NAs intracellularly. Part of these issues has been tackled with some success, such that some promising vectors are in the limelight [73, 79,80,81].

Fig. 1
figure 1

Schematic of the variables and factors affecting non-viral gene delivery to MSCs

Different research groups sought to improve effectiveness while reducing the toxicity of some CPs by adding hydrophobic moieties to the polymer. For instance, polyplexes made with PAMAM dendrimers functionalized with hydrocarbon chains of variable lengths showed improved cellular uptake and reduced cytotoxicity if compared to gold standard vectors such as 25 kDa bPEI [82,83,84]. Likewise, Uludağ’s group devised PLL functionalized with palmitic acid and PEI with linoleic acid to increase their hydrophobicity. The resulting particles reached the nuclear region more effectively than those obtained with gold standard transfection agents, such as Lipofectamine® and 25 kDa bPEI, leading to improved effectiveness and decreased cytotoxicity [85, 86].

Other strategies employed to reduce the cytotoxicity of polymer vectors involve the incorporation of biodegradable moieties. These include heterocyclic amines [87], 2,2-bis(hydroxymethyl)propionic acid (bis-MPA) [88], bioreducible disulfide bonds [20, 89, 90], hydrolysable ester bonds [58, 91, 92], β-cyclodextrin (β-CyD) moieties [93, 94]. These modifications have shown significant improvements in transfection efficiency, leading to robust expression levels in MSCs. It is worth noting that, in most cases mentioned above, transgene expression using these smart polymers was enhanced by up to 10 times compared to unmodified PEI, and approximately 2 to 5 times when compared to commercially sourced reagent like Lipofectamine 2000®. Therefore, these promising candidates offer a safe and effective means of delivering NAs to MSCs.

More recently, much effort has been devoted to designing non-viral gene delivery vectors featuring targeting ability. To this aim, cell-selective ligands, typically short oligopeptides that bind to cognate cell-surface receptors displayed by target cell populations, have been covalently tethered to polymeric carriers. Taking MSCs as a target, there is a vast literature on this subject. The most thoroughly investigated is the popular RGD tripeptide (arginine-glycine-aspartic acid), which has been used to improve the interaction between the non-viral vectors and the MSCs [81, 95]. Indeed, some transmembrane receptors, such as αVβ1, αVβ3, and αVβ5 integrins, are known to recognize and bind to the RGD consensus motif [96,97,98]. On the other hand, because such integrins are rather ubiquitous, the functionalization of polymers with RGD can give rise to off-target effects. Other peptides, which are more selective for MSCs, have thus been grafted to CPs and used in transfection with some success [99, 100]. In addition to targeting ad hoc-designed peptides, natural full-length polymers, such as hyaluronic acid (HA), can be used for this purpose. HA is a natural polysaccharide found in most body tissues that is capable of interacting with the CD44 cell-surface glycoprotein of many cell types [101]. It has been shown that hMSCs recognize HA through the CD44 and CD54 receptors [102,103,104]. As a result, the conjugation of HA to PEI was found to improve the transfection efficiency of the CP itself on MSCs [80, 89, 102].

➣ Cationic lipids (CLs)

Since the first report by Felgner and colleagues in 1987 [105], the use of CL-based carriers for gene delivery has bloomed. CLs are amphiphilic molecules consisting of three building blocks which are i) a protonatable headgroup connected to at least one ii) hydrophobic moiety through iii) a linker [53]. It is worth noting that the structure of CLs can be intentionally modified to enhance their interaction and delivery capabilities for various types of NAs, including pDNA as well as RNA in the form of small interfering RNA (siRNA) and micro RNA (miRNA) (Fig. 1B) [53, 106]. While highly effective in binding anionic NAs, CLs are frequently used in combination with ionic phospholipids or non-ionic surfactants to give supramolecular arrangements such as liposomes or niosomes (Fig. 1A), respectively, that allow for improving their delivery performances [107,108,109]. Nevertheless, despite the virtually endless list of vectors available, most gene delivery strategies devised for MSCs of different origins rely on the use of Lipofectamine® [20, 110,111,112,113,114,115,116,117,118,119,120], the gold standard, commercially-available lipid transfection reagent consisting of a 3:1 (w/w) mixture of the cationic lipid 2,3-dioleoyloxy-N-[2(sperminecarboxiamido)ethyl[-N,N-dimethyl-1-propaniminium trifluoroacetate (DOSPA) and the zwitterionic 1,2-dioleoyl-sn-glycerol-3-phosphoethanolamine (DOPE) [121]. The success of Lipofectamine® relies upon its ease of use and broad-spectrum activity. Lipofectamine®-based lipoplexes proved to efficiently treat in vitro either mouse-derived or human MSCs to express osteogenic and chondrogenic genes [47, 122,123,124,125,126]. Aside from very few exceptions, no CL other than Lipofectamine® has been used to transfect MSCs. In this scenario, there is still room for improvement, and the design of novel lipid-based mixtures with improved efficiency and reduced side effects may open the way for effective hMSCs manipulation [127].

On the other hand, niosomes have recently emerged as a promising alternative for the efficient transfection of MSCs. Broadly speaking, niosomes are self-assembled vesicles (Fig. 1A) consisting of a combination of CLs, non-ionic surfactants, helper lipids, and charge modifiers able to interact with NAs and form nioplexes [109, 128]. Experimental evidence demonstrated that niosomes possess higher stability and reduced cytotoxicity compared to their pure CL counterparts. Moreover, they are cheaper than their liposomal counterparts and easier to prepare [109, 128]. In a work by Pedraz’s group, niosomes composed of the CL 2,3-di(tetradecyloxy)propan-1-amine (DTPA) and the non-ionic surfactant polysorbate 80 were used to complex plasmid DNA (pDNA) and transfect mouse-derived MSCs with BMP-7 encoding gene, thus driving the osteogenic differentiation of MSCs with no detrimental effects [129]. Likewise, our group recently developed an effective strategy to transfect hMSCs by using nioplexes consisting of pDNA and a combination of 1,2-di-o-octadecenyl-3-trimethylammonium propane (DOTMA) as the CL, polysorbate 60 as the non-ionic surfactant and cholesterol as helper lipid [130]. The so-formed nioplexes displayed noticeable transfection efficiency and reduced cytotoxicity compared to Lipofectamine®-based lipoplexes. These, together with other works found in the literature [131,132,133], highlight the suitability of such kinds of materials as promising delivery systems for engineering MSCs.

Overall, the safe and effective delivery of therapeutic NAs stretches to MSCs is a hot topic that still represents a challenge for scientists. In this perspective, rational design approaches and the study of the structure–function relationship (SAR), that is, the intimate interconnection between the vector structure and its biological activity, of these delivery carriers will help elucidate the interplay between their efficiency and cytotoxicity.

Enviromental parameters affecting gene delivery to MSCs

As a matter of fact, the therapeutic potential of engineered hMSCs strongly depends on the transient or stable expression of the proteins of interest upon transfection [134]. Unfortunately, as hMSCs are hard-to-transfect cells, they are preferably engineered ex vivo and subsequently grafted in vivo [56, 134]. In vitro transfections are usually performed by seeding MSCs in monolayers, which are next challenged with lipoplexes or polyplexes directly added to the cell culture medium (i.e., bolus transfection). Although bolus transfection is the simplest way possible to deliver NAs to the cells, the efficiency of the overall process is hampered by the limited mass transport of complexes, stability issues related to their stay in the extracellular milieu, as well as the lack of physiological behavior of MSCs in such oversimplified in vitro culture systems. To overcome such drawbacks, other options such as reverse (or substrate-mediated) transfection strategies are now on the hype (Fig. 1C).

As a rule of thumb, the in vitro culture of MSCs mostly relies upon the use of matrices and scaffolds to mimic the in vivo conditions. In contrast to conventional bolus transfection, reverse transfection allows for a higher amount of NAs available per cell and closer control over the cell behavior through chemical and physical cues of the substrate itself [135]. In reverse transfection, NA-containing nanoparticles are immobilized or spotted on a flat (2D) surface or 3D scaffold by different techniques [59]. Cells are then seeded on top of it or embedded within the scaffold. In this light, with the aim to entice MSCs to internalize gene delivery complexes, improve the transfection efficiency and prolong the transgene expression, scientists sought to optimize the substrate properties and design ever more favorable cell-surface interfaces. Generally, 2D or 3D matrices displaying different stiffness, roughness, topography, and surface chemistry have been used to control the MSCs’ behavior and transfectability [136]. As a matter of fact, substrate stiffness per se has been found to induce the osteogenic or chondrogenic differentiation of MSCs via integrin-dependent signaling underpinning the mechano-induced MSCs differentiation [137,138,139]. Likewise, the substrate stiffness has proven to regulate some key mechanisms enabling the internalization of non-viral vectors, thus impacting their effectiveness as well [140,141,142].

Substrate topography is another determinant in hMSCs differentiation. Of note, the nanostructure provided by the substrate impact focal adhesion assembly at the cell-surface interface. As a consequence, cytoskeletal rearrangements occur. Because the cytoskeleton links the extracellular matrix (ECM) and the nucleus, it ultimately regulates gene expression and cell phenotype [60, 143,144,145]. Acting at the cell-membrane level, nanotopographical cues have thus been found to impact the internalization of nanoparticles as well [70, 146,147,148].

Besides, also the substrates’ surface chemistry plays a role in reverse gene delivery [149]. Segura’s group has extensively refined the physical and chemical cues of the substrate to improve the transfection of MSCs [141]. For instance, when mouse-derived MSCs were cultured on a combination of different ECM components, such as collagen type IV, fibronectin, and laminin, the cells did duplicate faster and display higher transgene expression with respect to cells cultured on uncoated tissue culture plates [150]. By the same token, other approaches took advantage of chemical and physical biomimetic environmental cues to extend the transgene expression in MSCs [141, 151,152,153].

Altogether, this body of evidence suggests a twofold effect of the substrate features, that is, their synergistic effect on the ultimate transfection efficiency and hMSCs phenotype regulation [31]. For instance, some studies suggest the use of softer substrates with an elastic modulus around 25 kPa to commit hMSCs toward a chondrogenic lineage [154,155,156]. Likewise, other works reported that stiffer substrates with an elastic modulus equal to a few hundred kPa were able to drive MSCs toward osteoblast-like phenotypes when combined with the delivery of complexes containing pDNA encoding the BMP-2 protein [157].

Another greatly overlooked approach relies on the modulation of cell response and improvement of non-viral gene delivery vector effectiveness through exogenous mechanical cues. Different mechanical stimuli have been found to regulate the gene transfer process by easing some steps of the delivery pathway, namely internalization, cytoplasmic transport, and nuclear import of complexes [136, 158]. Indeed, mechanical loading applied to cells is associated with specific cell responses, such as mechanoregulation of membrane trafficking [159,160,161] and cytoskeletal remodeling [162,163,164,165,166,167]. In this scenario, cyclic stretch [168,169,170], shear stress [61, 171, 172], and vibrational loading [62] may boost the transfection efficiency of non-viral gene delivery vectors by enhancing their uptake and intracellular trafficking. Even more interestingly, nanoscale vibrational loading alone has been used to trigger the osteogenic differentiation of hMSCs [63, 173].

Overall, this evidence suggests that the environmental cues to which hMSCs are exposed affect their phenotype, and thus their ability to internalize non-viral particles. Therefore, the use of exogenous stimuli, such as the one exerted by either the substrate or from mechanical loading, coupled with the non-viral gene delivery could represent a promising strategy to develop engineered MSCs with therapeutic potential.

Nevertheless, since there is no advancement without robust screening protocols, the following sections unveil how to harness the most relevant experimental features to improve the efficiency of a given transfectant with MSCs and highlight ways to analyze their efficiency in vitro.

In vitro transfection assays

How to set main transfection parameters

The efficiency of non-viral gene delivery strategies is highly dependent on several factors and conditions used to carry out transfections. Consequently, the optimization of the experimental conditions, such as the cell type and passage number, the cell density, the dose and type of NA to be transferred (Fig. 1B), the type of transfection reagent, and the transfection time are extremely relevant (Fig. 1D).

Broadly speaking, the susceptibility to transfection depends on the cell source, as MSCs can be isolated from different tissues, such as the bone marrow [1, 5, 17, 19, 27, 28, 34, 64,65,66,67, 113,114,115,116,117, 120, 132, 135, 141, 151, 152, 174,175,176,177,178,179,180,181,182,183,184,185,186] and the adipose tissue [34, 118, 187, 188], and from diverse sources such as human [1, 26,27,28, 34, 66,67,68, 86, 110, 112, 115, 119, 152, 153, 174, 176, 177, 181, 183, 187, 189,190,191,192,193,194], rat [5, 34, 64, 113, 116, 117, 120, 135, 151, 175, 178, 180, 184, 195], murine [20, 34, 118, 132, 141, 181, 182, 185, 196, 197], goat [34, 65, 188], porcine [29, 114, 179], rabbit [186], horse [198], and canine [198]. Therefore, because of the differences in the overall cell metabolism among MSCs coming from different tissues and species, it is difficult to compare the transfection outcomes found in the literature.

Another pivotal parameter affecting transfection outcomes is the cell passage. As a general rule, once extracted from the donor, MSCs are typically transfected between passages 1-to-8 to avoid unintended cell differentiation and aging [20, 26, 34, 65,66,67, 114, 151, 152, 174, 178, 181,182,183, 190, 192, 198]. Nevertheless, for achieving more reliable transfection values, it is recommended to avoid cell passages higher than 4.

Another crucial factor influencing transfection efficiency is the type of NA to be delivered. In the case of pDNA, it needs to penetrate the cell nucleus to be functional and express the desired protein. This process occurs either through the disassembly of the nuclear envelope in mitotic cells or via pore complexes in non-dividing cells [199]. In contrast, mRNA only requires the cytosolic transcriptional machinery to express the protein product, leading to higher transfection efficiencies compared to pDNA. However, the use of mRNAs entails higher immunogenicity and shorter duration of protein expression compared to pDNA [200]. On the other hand, siRNA and miRNA inhibit the expression of complementary RNAs, thereby modulating cell phenotype and achieving longer duration of expression compared to mRNA [31].

When performing in vitro transfections, both the cell density and the NA dose used to transfect cells significantly impact the gene transfer efficiency as this corresponds to the amount of NAs available on a per-cell basis.

Even little variations in a single parameter may give significantly different transfection outcomes, such that a proper trade-off between the NA dose delivered to cells and the cell density should be investigated anytime. To this purpose, a thorough survey of the literature employing non-viral vectors such as PEI [69, 86, 151, 174, 177, 179, 201, 202], Lipofectamine® [198], nano-hydroxyapatite (nHA) [19, 69, 152, 153, 175, 178], linear bioreducible poly(urethane amine) (SSPUA) [187], TransIT [21], and the multi-domain cell-penetrating peptide GAG-binding enhanced transduction (GET) [178], pointed out three main different subsets of MSCs densities used in transfection experiments, such as low (< 1 × 104 cells/cm2), medium (from 1 × 104 to 5 × 104 cells/cm2), and high cell density (from 5 × 104 to 2.5 × 105 cells/cm2). The same applies to the NA dose delivered to cells. In the latter case, experiments can be grouped into low (< 0.5 µg/cm2), medium (from 0.5 to 1.5 µg/cm2), and high NA dose (from 1.5 to 3 µg/cm2). Unfortunately, no exclusive relationship between these two parameters (cell density/cm2 and NA µg/cm2) can be established since they were not proportionally scaled in the different studies. Therefore, in order to compare the variety of conditions used by different authors, we normalized the NA dose as µg of plasmid per 105 cells and we found a linear fitting between cell densities and plasmid doses, falling most of the studies conditions in the intermedium range (Fig. 2).

Fig. 2
figure 2

Schematic representation of the correlation between the dose of NAs per 105 cells, ranging from low (from 0 to 1 µg/105 cells), medium (from 1 to 5 µg/105 cells), and high dose (from 5 to 20 µg/105 cells), and the cell density, ranging from low (from 0 to 1 × 104 cells/cm2), medium (from 1 to 5 × 104 cells/cm2), and high cell density (from 5 × 104 to 2.5 × 105 cells/cm2). The trend line (R2 = 0.79) represents the tendency of the data taken from articles (references [17, 19, 21, 69, 86, 110, 112, 113, 152, 153, 174,175,176, 178, 179, 187, 198, 201,202,203]), where the cell density (cells/cm2) and the dose (µg) are specified

Nonetheless, due to the variation in cell densities, NA doses and methods used to analyse transfection, it is not possible to make definitive conclusions regarding transfection efficiency. To shed more light on this issue, we herein provide some genuine results on MSCs and draw some conclusions on the interplay among the cell density, the NA dose, and the transfection time. Transfection studies were thus carried out by using the benchmark transfectant 25 kDa bPEI to complex the pDNA encoding the luciferase reporter gene luc2 (hereafter referred to as pluc) at the extremely effective cationic polymer amino groups (N) to the anionic NA phosphate moieties (P) ratio (N/P) of 10 [152, 174].

Transfections were carried out on bone marrow-derived-MSCs from two patients herein named #1 and #2, kept in culture, and transfected between passages 1 and 2. The transgene expression was evaluated 24, 48, and 72 h post-addition of polyplexes to the cells (Fig. 3).

Fig. 3
figure 3

Transfection efficiency, expressed as luciferase activity normalized to the total protein content (RLU/mg of protein), following transfection with bPEI/pluc complexes prepared at N/P 10 on hMSCs isolated from two patients (i.e., patient #1 and patient #2, right and left panels, respectively) as a function of the pDNA dose (0.1, 0.5, and 1 µg/cm2) and the cell density (5 × 103, 1.5 × 104, 5 × 104, and 1 × 105 cells/cm2). Luciferase activity was evaluated 24, 48, and 72 h post-transfection. Results are expressed as mean ± SD (n = 3, * p < 0.05)

In light of the results depicted in Fig. 3, we draw some conclusions on the role of the cell density and pDNA dose on the ultimate transfection efficiency of bPEI-based polyplexes and propose a guideline on how to design in vitro assays for MSCs’ transfection. Interestingly, regardless of the pDNA dose used, the cell seeding density of 1.5 × 104 cells/cm2 led to the highest levels of luciferase activity (up to a maximum 12.8-fold difference compared to any other cell density tested; p < 0.003). Therefore, we assume that the optimal MSCs density to carry out transfection assays should be set close to this value. It is worthy of note that transgene expression strongly depends on cell doubling [35], in the way that exogenous pDNA freely enters nuclei due to the nuclear cell membrane disruption occurring during mitotic events. Based on our results, we can speculate that cells seeded at a density of 1.5 × 104 cells/cm2 are in a high proliferative state, such that more pDNA is made available to the transcription machinery.

Since most of the studies employ transfection time windows, i.e., the time elapsing between the delivery of complexes to the cells in culture and the transgene expression analysis, between 24 and 72 h [20, 21, 26, 65, 68, 110, 118, 132, 177, 178, 182, 184, 185, 190, 204], or even longer [29, 181, 186], we sought also to elucidate the effect of this parameter on the transgene expression. Interestingly, irrespective of the cell density, the highest luciferase activity was reached 24 h-post polyplex delivery (up to a 10.0-fold difference compared to any other time point; p < 0.001). This is an interesting point since it means that maximum transgene expression of the luciferase gene occurs in this timeframe regardless of the cell density, NA dose, and cell source used. However, we cannot generalize the conclusion to every transgene of interest as each has specific expression patterns. This implies that good experimental practice should include the optimization of the transfection time depending on the transgene used.

Besides, the results reported in Fig. 3 suggest that the pDNA dose used to transfect cells has, to some extent, no significant influence on the effectiveness of bPEI-polyplexes and can be varied as long as it does not affect the viability of hMSCs, which was > 85% in any conditions (Figure S1).

How to evaluate the distribution and internalization pathways of complexes into transfected MSCs

To be effective, non-viral gene delivery vectors must be able to overcome many extra- and intracellular barriers, that is, rate-limiting steps which include the crossing of the cell membrane, the release from endo-lysosomes in the cytoplasm, and the nuclear import of NAs (when the delivery involves DNA). Therefore, the uptake and intracellular trafficking of complexes should be assessed especially when screening new reagents.

Overall, shedding light on the structure–activity relationship of the vector will allow the identification and optimization of essential parameters, such as the shape, size, and surface charge of nanoparticles, that are crucial for efficient gene delivery [205].

As for the cell uptake, protocols commonly used to assess the internalization and intracellular distribution of the complexes rely on the use of fluorescently labeled NAs. As summarized in Table 1, most works exploit cyanine dyes, as they cover a wide range of wavelengths depending on the length of the polyalkene bridge connecting the two nitrogen heterocyclics, hence providing versatility for diverse applications [19, 27, 66, 86, 116,117,118, 184]. Compared to other traditional dyes such as fluorescein [5, 27] and rhodamine [113, 189], cyanines exhibit enhanced water solubility and photostability, being less sensitive to pH changes, i.e. those occurring when complexes enter the endo-lysosomal compartment. Other molecules used to examine complexes uptake include fluorophores such as Dy547 [34], YOYO-1 [177, 187], TAMRA [185], and Texas Red [174]. While Dy547 and YOYO-1 increase their fluorescence once bound to NAs, TAMRA and Texas Red can be used to label either the vector, the NAs, or both of them, and allow acquiring further insight into the internalization process.

Table 1 Current strategies to evaluate internalization of complexes into MSCs including uptake and inhibition assays

Flow cytometry (FCM) is by far the most effective method to gather quantitative data on complex internalization by cells. To this aim, fluorescence-labeled gene delivery complexes are added to cell monolayers and incubated for a certain time. At given time points, cells are harvested, fixed, the fluorescent signal is measured using an FCM and compared to negative controls (i.e., untransfected cells) [27, 34, 66, 86, 116,117,118]. Alternatively, confocal microscopy (CSM) allows one to get insights into the in-cell distribution of the labeled complexes. Briefly, following transfection, cells are fixed, stained, and mounted on cover slides before being imaged using a fluorescent microscope. While the staining protocol depends on the specific targets, the most used procedures make use of nuclear dyes (e.g., 4,6-diamino-2-phenylindole—DAPI, Hoechst 34580, Hoechst 33258, and Hoechst 33342 that bind to DNA) and cytoplasmic stains (specific antibodies, e.g., anti-tubulin, or binders, e.g., phalloidin, coupled to fluorochromes, such as Alexa Fluor) [5, 19, 34, 184, 185, 187].

Most studies are performed to shed more light on the complexes uptake and internalization pathways. The endocytic routes can be thus selectively inhibited to assess the contribution of each pathway to the particle uptake. The inhibitors currently used in internalization assays comprise i) chlorpromazine to block clathrin-mediated endocytosis (CME) [113, 135, 187, 190], ii) genistein, filipin, or methyl-β-cyclodextrin to inhibit caveolae-mediated endocytosis (CvME) [110, 113, 187, 190] and iii) wortmannin [113, 187], or amiloride [110, 190] to suppress macropinocytosis, that is, the three main entry routes for non-viral gene delivery vectors [208]. It has been shown that particle size plays an essential role in the internalization pathway, and most particles with a diameter ranging from 50 to 500 nm are internalized through CME or CvME pathways. However, while CL-based particles are typically internalized via CME, polyplexes are usually internalized through both CME and CvME pathways [72]. To elucidate the role of each endocytic route on complexes uptake and transfection efficiency, cells are incubated with specific endocytosis inhibitors before carrying out transfection. The ultimate expression of a reporter gene (e.g., Green Fluorescent Protein (GFP) [113, 187, 190], luciferase (luc) [135]) or therapeutic transgene (e.g., BMP-2 [110]) is analyzed afterward and compared to that of cells transfected in the absence of such inhibitors.

Regardless of the internalization route, lipoplexes and polyplexes end up in vesicles called endosomes, which are another intracellular barrier toward effective gene transfer. Tracking the intracellular journey of complexes through the staining of the lysosomal compartment can help elucidate the intracellular fate of nanoparticles. The information gained can be used to design endosomal escape strategies. To improve transfection, some authors exploited the use of media additives, i.e., molecules exerting an effect by enhancing the transfection efficiency in MSCs. For instance, Pannier’s group introduced the use of glucocorticoids, such as dexamethasone (DEX), to improve transgene expression in MSCs. This strategy has proven successful for various NAs and has paved the way for its application in numerous fields. Additionally, DEX has shown pro-anabolic effects in MSCs chondrogenesis and is commonly used in the formulation of chondrogenic cell culture media. Specifically, the use of glucocorticoids was found to positively counteract the cytotoxic effect of non-viral vectors by decreasing cell-oxidative stress arising as a consequence of complexes’ delivery and preventing the decline of cell metabolism [209, 210]. Likewise, our group devised the use of sucrose as a lysosomotropic agent to enhance complexes escape from the endo-lysosomes, thus favoring their cytoplasmatic transport [211]. Briefly, MSCs were transfected with nioplexes made of DOTMA, polysorbate 60 and cholesterol, and complexing pDNA encoding the β-galactosidase reporter in a sucrose-enriched (i.e., 40 mM sucrose) culture medium. As a result, the transgene expression improved due to the rise in intracellular pDNA content [130], whereas cell viability was not affected.

A thorough understanding of the uptake and intracellular trafficking pathways of complexes is needed when dealing with new non-viral vectors. This issue becomes even more relevant when using hard-to-transfect cells, such as hMSCs. The combination of different experimental tools, such as fluorescence microscopy, FCM, and the use of endocytic inhibitors thus represents a straightforward way to design ever more effective delivery strategies to engineer hMSCs.

How to evaluate cell viability and transfection efficiency following transfection

The assessment of cell viability following transfection allows one to evaluate the cytotoxic effects of non-viral carriers. The oldest methods to assess cell viability comprised the use of dye exclusion assays to determine the number of live and dead cells through microscope analysis and FCM. The most used cell membrane-impermeable dyes are Trypan blue [112, 204], 7-AAD (7-aminoactinomycin D) [112], and propidium iodide (PI) [133, 212]. Novel and simpler viability assays based on the use of biochemical markers are now preferred (and preferable) to evaluate cell metabolic activity. Most common cell viability assays include MTT [17, 26, 27, 86, 110, 135, 141, 152, 178, 182, 185, 189, 192, 194], MTS [5, 34, 67, 68, 153, 176, 177], CCK-8 [20, 21, 34, 116, 184], Live/Dead assay [5, 19, 29, 67, 132, 141, 174, 175, 185, 191, 193, 197], resazurin assays [34, 113, 119, 179, 187], and DNA-based quantification kits [28, 34, 69, 114, 120, 188, 194] (Fig. 4).

Fig. 4
figure 4

Common colorimetric indicators used to evaluate the cytotoxicity of non-viral gene delivery carriers

Current methods for cell viability assessment, such as MTT, CCK-8, and MTS, rely on the intracellular reduction of tetrazolium salts into formazan dyes that are measured spectrophotometrically to evaluate viable cells. While the MTT assay requires an additional step to dissolve intracellular water-insoluble formazan crystals, MTS and CCK-8 comprise water-soluble formazan products and allow one to avoid the final solubilization step [213, 214].

Conversely, resazurin-based methods require reduction by cell dehydrogenases using NADH/NADPH as co-substrates. The most commonly resazurin-based methods used are Alamar Blue® [19, 113, 118, 179, 187, 196] and CellTiter-Blue® Cell Viability Assay [34, 119, 190]. In contrast to destructive assays, such as most tetrazolium salt-based assays, the last ones do not even interfere with cell metabolism.

Another assay frequently used to investigate cell viability is Live/Dead staining based on the simultaneous labeling of viable and dead cells by combining a membrane-permeable dye, such as Calcein-AM (green fluorescence), and a membrane-impermeable high-affinity DNA stain, such as ethidium homodimer-1 (red fluorescence) [213].

Besides, the side effects of transfection on cell viability can also be assessed by quantifying the nuclear DNA content in transfected cells. On this note, Quant-it Pico Green dsDNA Kit is one of the most reliable assays used for this purpose [28, 34, 120, 188, 194].

Assessing the transfection efficiency is the primary way to gauge the ability of a non-viral gene carrier to introduce exogenous NAs into a specific cell type and alter the expression of a target gene or genes. Depending on the transgene being expressed or the genetic sequence silenced, different methods are available to assess the transfection efficiency of non-viral vectors, including i) spectrophotometric measurements, ii) fluorescence microscopy, iii) FCM, iv) polymerase chain reaction (PCR), v) enzyme-linked immunosorbent assay (ELISA), Western Blot (WB), and vi) immunocyto/immunohistochemical assays (Fig. 5).

Fig. 5
figure 5

Schematic classification of the most popular methods and technologies used to gauge the transfection efficiency of non-viral gene delivery carriers

On the other hand, the most common screening methods to analyze transfection efficiency are based on the assessment of the activity of the luciferase enzyme (luc) encoded by the corresponding reporter gene [35, 96], and fluorescence microscopy and FCM analyses, if the reporter transgene delivered encodes for a fluorescent protein, such as GFP, red fluorescence protein (RFP), and yellow fluorescent protein (YFP) [215]. Luciferase assays rely on the enzymatic conversion of the luciferin substrate into oxyluciferin with concomitant emission of photons (luminescence) by transfected cells [34]. The assay is rapid, sensitive, and specific, as it is compatible with most existing viability tests [216]. Conversely, fluorescence microscopy allows to determine the transfection efficiency through image analysis of transfected vs. untransfected cells [217]. This technique is simple yet time-consuming and cumbersome. Hence, FCM allows for discrimination among non-transfected and transfected cells by manually gating the fluorescence signal of a cell subpopulation in a sample, providing the percentage of transfected cells over the total cell number. Of note, this technique is quantitative, sensitive, and enables the fast analysis of large cell numbers [217].

Whereas PCR is a widely used technology for the exponential amplification of a DNA fragment, quantitative PCR (qPCR) is the gold standard for the detection and quantification of a NA target. In brief, transfected cells are lysed, mRNAs are recovered, and reverse transcribed through RT-PCR to give a pool of complementary DNAs. Next, the NA stretch of interest is amplified and quantified using qPCR. More often, the NAs delivered in transfection assays are pDNAs encoding for osteo- or chondrogenic factors [18, 26,27,28, 34, 67, 117, 118, 120, 153, 174, 177, 179,180,181,182,183, 186, 191, 193,194,195,196,197, 206, 218], but it is fairly frequent to use micro-RNAs (miRNAs), that is, a class of noncoding RNAs inhibiting the translation of the mRNA involved in MSCs differentiation [116], such as miR-335 [66], miR26-a [181], miR-138 [117, 184], miR133a [116, 206], miR-218 [182], or miR-122 [180]. Even if qPCR is highly sensitive, it is a destructive technique requiring the disruption of the samples under investigation before analysis. This implies that it is not suited to carry out time-course experiments.

To probe the post-transfection expression of osteo- or chondrogenic markers other than mRNAs, ELISA or WB are routinely performed. ELISA is the most widely used method to unambiguously detect and quantify the concentration of a given protein, which relies on the binding affinity of an antibody-antigen pair [219]. In the case of soluble, secreted markers, cells are transfected and cultured for the desired timeframe. Culture medium aliquots are harvested at fixed times for ELISA analysis to quantify the expression of the target protein over time. Most common examples of proteins assessed by ELISA include BMP-2 [19, 21, 29, 34, 65, 69, 114, 115, 119, 153, 178, 179, 186, 190, 197], TGF-β1 [1, 17, 18, 114, 186], TGF-β3 [19, 153, 179], and vascular endothelial growth factor (VEGF) [69, 178, 181].

On the other hand, WB assay [20, 27, 28, 181, 182] relies on the specific interaction of antibodies with target antigens (osteo- or chondrogenic proteins) present in the culture media or intracellularly. WB allows separating proteins based on size through a molecular sieve (i.e., sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE)), transferring them onto a solid support, and identifying the protein of interest using specific antibodies labeled with a tag, such as horseradish peroxidase (HRP). WB is cheaper than ELISA on a per-sample basis, but it is a semi-quantitative (image processing), cumbersome, and time-consuming technique.

Other methods to assess the presence of specific proteins in cell monolayers or tissue sections are the immunocytochemical (ICC) and immunohistochemical (IHC) analyses. The main advantage of these techniques lies in the ability to detect transgene-encoded proteins while preserving the structural integrity of the samples, thus enabling the localization of the target antigen(s) in situ. Conversely, IHC and ICC are essentially qualitative. The operating principle is similar to that of WB and ELISA, which means taking advantage of the antibodies to recognize the target proteins and visualize them using chromogenic or fluorescent-based detections. In the former case, the detection is made possible by the enzymatic cleavage of a substrate to give a visible precipitate, whereas, in the latter one, a fluorophore is conjugated to the antibody specific to the target protein, and the signal is detected through fluorescence microscopy.

Standard protocols for IHC require the specimen fixation in paraformaldehyde (4—10% (v/v) in water), followed by its inclusion in paraffin or cryo-embedding media, such as optimal cutting temperature (OCT) compound, and sectioning. For the staining of paraffin-embedded sections, samples required to be first dewaxed in a xylene-ethanol gradient and rehydrated, while OCT-embedded samples required to warm the slides to room temperature to remove the embedding medium. On the other hand, ICC constitute the easiest and practical option, requiring only the fixation of the samples.

In all cases, the samples undergo a series of steps preparatory to the addition and incubation with antibodies and, eventually, the chromogenic substrate. Finally, the mounting medium is added to the slices, which are visualized through microscopy. IHC and ICC are suitable for different applications including the evaluation of the expression of specific transgenes, such as SOX9 [28] or NANOG [26]. To increase the number of details, IHC can be combined with other counterstaining, such as hematoxylin or eosin [1, 111, 114, 117, 176, 180, 183]. This traditional histological technique relies on the affinity of the different cell components for each dye based on their acid–base nature. Therefore, whereas hematoxylin stains in purplish blue basophilic organelles such as nuclei, eosin counterstains in pink the basic cell compartments such as the cytoplasm [5, 18, 23, 34, 69, 114, 118, 135, 153, 186, 188, 190, 206, 218].

How to induce and evaluate MSC differentiation

The inherent ability of MSCs to differentiate into mesoderm lineage cells, such as osteoblasts and chondrocytes, points out their great potential in osteochondral repair [220]. Despite their promising potential, the direct transplantation or intra-articular injection of MSCs often results in a mixture of hypertrophic, cartilaginous, and fibrous tissues [220]. Consequently, a significant body of research has been dedicated to gene delivery strategies aimed at committing MSCs to express specific proteins and factors, ultimately directing their differentiation toward a desired cell phenotype (Table 2). However, it is worth highlighting that high levels of transgene expression do not necessarily lead to increased MSCs differentiation. Therefore, the selection of an appropriate delivery vector greatly influences the bioactivity of the gene product and the attainment of the desired cell phenotype [179].

Table 2 Non-viral gene-delivery based therapeutic approaches for bone and/or cartilage repair

MSCs play a crucial role in the bone-healing process as they serve as precursors to osteoblasts and chondrocytes, and they likely also contribute to modulating the healing response. Thus, the osteogenesis of MSCs is regulated by various proteins, hormones, and GFs. Most of the osteo-reparative approaches focus on the overexpression of NAs encoding for protein GFs, such as BMP-2 alone [5, 20,21,22,23, 115, 175, 177, 190], or in combination with other related family members, such as BMP-6 [65], BMP-7 [65], or BMP-9 to promote MSCs osteogenesis [218]. Although BMP-2 has a significant osteogenic potential, its activity can be hindered by the presence of extracellular inhibitors and antagonists, such as Noggin, which exhibits a strong affinity for BMP-2 and prevents it from binding to its receptor. In order to enhance the effects of BMP-2, the use of siRNA against these regulators has therefore emerged as a promising approach. This technique aims to suppress the expression of these inhibitors, thus allowing for a more potent and effective action of BMP-2. Hence, an effective osteogenic differentiation of MSCs can be attained using siRNA against Noggin [34, 68, 118, 191]. Moreover, a variety of miRNAs, including anti miR-138, miR-20a, miR-218, and miR-133a [67, 68, 116, 117, 180, 183, 184, 191, 206], have been identified as potential targets to upregulate cell signaling pathways related to osteogenesis.

Specific culture conditions are also required to selectively induce osteo- or chondrogenic differentiation of MSCs after transfection with suitable therapeutic genes. Of note, most protocols in this regard require a minimum culture time of 14 days, and the use of media enriched with ascorbic acid, β-glycerophosphate, and dexamethasone [194].

Commonly assessed osteogenic markers comprise RUNX2, osterix (OSX), ALP, COL-I, osteocalcin (OCN), and a range of members from the BMP superfamily. Because ALP, BMPs, COL-I, RUNX2, and OSX are essential for osteoblast differentiation, they are considered early markers of osteogenesis. Instead, OCN and osteopontin (OPN) are deemed late cell differentiation markers as they are involved in mineralization and ECM synthesis. Since the expression levels of such biomarkers increase in the sequential steps of bone formation and regeneration, their monitoring over time employing analytical techniques such as qPCR [18, 20, 26, 67, 116, 117, 152, 177, 180, 183, 190, 202, 206, 218], ELISA [19, 21,22,23, 115, 178, 202, 203] or WB [20, 117, 180, 184, 191] is a valuable way to assess the effectiveness of a given osteogenic strategy.

The detection of biochemical markers associated with the differentiation process can be also performed through spectrophotometric measurements. Osteogenic markers can be assessed by measuring ALP expression and calcium deposition by the cells. ALP is the most recognized protein marker of early osteogenic differentiation due to its pivotal role in bone matrix formation and calcification [5, 18, 23, 65, 116,117,118]. Several kits are available to detect and quantify this protein. They rely on the use of substrates that are converted by the secreted ALP into colored, fluorescent, or luminescent products. Besides, to determine the calcium concentration within the physiological range, there are colorimetric kits (λ (Abs) = 575 nm) that detect alkaline phosphatase substrate (pNPP-Na hexahydrate) in cell samples.

In addition to the aforementioned techniques, IHC and ICC are mostly used to detect osteogenic differentiation, as they offer the possibility of investigating the distribution of an antigen over the different cell types thus allowing to gather insights on the tissue architecture. Specific histological stains for bone encompass Alizarin red [5, 18, 65, 68, 69, 116, 118, 174, 175, 190, 191, 202, 206, 218] and Sirius red [117, 180, 194], which stain in red–orange the calcium deposits and the collagen fibers, respectively.

The process of cartilage development begins with the condensation of MSCs, followed by their differentiation into chondrocytes, which secrete the cartilage matrix components such as proteoglycans and COL-II. Of note, it is important to maintain an appropriate balance of these markers during cartilage formation to prevent chondrocytes hypertrophy and matrix mineralization, as an imbalance can compromise the integrity of the cartilage tissue [221].

For this reason, common strategies to promote the chondrogenic differentiation of MSCs rely on the use of NAs encoding for members of the TGF-β superfamily, such as TGF-β1 or TGF-β3, which enhanced the expression of proteoglycans and COL-II. However, the main limitation of this factor is that often results in the expression of hypertrophic and osteogenic markers [222]. Additionally, the cartilage-specific transcription factor SOX9 holds potential in achieving better outcomes for cartilage regeneration, promoting chondrocytes proliferation and the cartilage matrix synthesis while minimizing hypertrophy [131].

Besides, the chondrogenic differentiation of MSCs is quite often conducted in 3D cultures (aggregate or matrix) for a minimum of 21 days, using media containing ascorbic acid, dexamethasone, insulin-transferrin-selenious acid mix, pyruvate, bovine serum albumin (BSA) and TGF-β [111, 194].

Gene markers of chondrogenesis include aggrecan (ACAN), COL-I, COL-II, COL-X, and the early chondrogenic factor SOX9. However, their expression patterns are specific to the cartilage type. For instance, the upregulation of ACAN and COL-II genes often occurs during articular cartilage repair, whereas the expression of COL-I (fibrocartilage marker) and COL-X (hypertrophic marker) might decrease (Fig. 6). Similar to the osteogenic processes, these markers can be evaluated by qPCR [27, 111, 120, 153, 194], ELISA [179, 188] or WB [27] techniques.

Fig. 6
figure 6

Most used therapeutic genes and markers to induce and evaluate osteochondral differentiation. Abbreviations: BMP-2, -6, -7, -9: bone morphogenic protein 2, 6, 7, 9; siRNA: small interfering RNA; miRNA: micro-RNA; TGF-β1 and -β3: transforming growth factor-β1 and -β3; SOX9: sex-determining region Y-type high mobility group box 9; RUNX2: core binding factor alpha 2; OSX: osterix; ALP: alkaline phosphatase; COL-I, -II, -X: type-I, -II, and -X: collagen; OCN: osteocalcin; ACAN: aggrecan

Common biochemical analysis of chondrogenesis involves the use of dyes such as dimethyl methylene blue (DMMB, λ Abs = 525 nm), which binds to sulfated GAGs enabling their prompt detection [1, 27, 114].

Histological analysis of cartilage phenotype usually relies on the staining of sulfated glycosaminoglycans (GAG) in the ECM with Alcian [27, 111, 135, 153, 179, 194] and Toluidine blue [111] (blue staining) or Safranin-O [1, 27, 111, 114, 135] (red staining). Nevertheless, the most reliable way to assess the osteo- or chondrogenic commitment of MSCs post-transfection is to combine at least two of the abovementioned methods.

Conclusions and future perspectives

The use of MSCs as therapeutic vehicles holds great promise for the treatment of osteochondral diseases. MSCs can be easily isolated from different sources and manipulated ex vivo to direct them toward osteo- or chondrogenic lineages. However, the current approaches, mostly relying on the use of soluble factors to drive MSCs differentiation, are somehow frustrating and ineffective. In this scenario, the control of MSCs phenotype using non-viral gene delivery strategies has garnered increased interest in recent years. The transient expression achieved through these systems aligns more closely with natural wound healing processes, making them particularly appealing [223].

While continuous effort in gene delivery research has led to the development of safe and fair efficient vectors, no standard procedure does exist for the ex vivo engineering of hMSCs, thus limiting the translation of MSCs-based therapies into the clinics. In light of this, this review aims at highlighting the main progress undertaken in non-viral gene delivery research for MSCs’ transfection and suggest ways to improve their efficacy in the target cells. Since gene transfer outcomes strongly depend on the cell response, we pointed out the use of exogenous environmental cues (either mechanical stimuli or substrate features), combined with the delivery of non-viral vectors, as a way to improve gene transfer efficiency and drive MSCs differentiation.

Besides, since there is no real advancement without robust models, we also provided readers with a practical guide and some genuine results for conducting screening studies on MSCs. This allows for the evaluation of MSCs’ commitment to osteo and chondrogenic phenotypes in vitro. As part of this approach, we provide an exemplification of how to optimize transfection conditions with the gold standard cationic polymer PEI in primary cultures of MSCs.

When focusing on MSC differentiation assays, it is crucial to select the most appropriate therapeutic gene based on the desired application. Overexpression of the GF BMP-2, along with the utilization of siRNA to prompt osteogenesis-related cell signaling pathways, emerged as a promising strategy. In addition, the overexpression of the transcription factor SOX9 appears to be a suitable approach for enhancing MSCs chondrogenesis, leading to the development of through a hyaline-like cartilage phenotype while reducing hypertrophy.

Overall, through this comprehensive review, our sincere hope is to establish a solid foundation for advancing research in MSCs therapy, paving the way for further breakthroughs in this field.

Availability of data and materials

Please contact author for data requests.

Abbreviations

7-AAD:

7-Aminoactinomycin D

ACAN:

Aggrecan

ALP:

Alkaline phosphatase

baf-A1:

Bafilomycin-A1

BMMSCs:

Bone marrow derived mesenchymal stem cells

BMP-2, -6, -7, -9:

Bone morphogenic protein-2, -6, -7, -9

bPEI:

Branched polyethyleneimine

CaP:

Calcium phosphate

cBMMSCs:

Caprine bone marrow derived mesenchymal stem cells

CC:

Cytochemical

cDNA:

Circular DNA

CLs:

Cationic lipids

CME:

Clathrin-mediated endocytosis

CPs:

Cationic polymers

COL-I:

Type-I collagen

COL-II:

Type-II collagen

COL-X:

Type-X collagen

CSM:

Confocal microscopy

CvME:

Caveolae-mediated endocytosis

Cy3:

Cyanine 3

Cy5:

Cyanine 5

DAPI:

4', 6-Diamidino-2-phenylindol

DEX:

Dexamethasone

DMMB:

Dimethyl methylene blue

DOPE:

1,2-Dioleoyl-sn-glycerol-3-phosphoethanolamine

DOSPA:

Cationic lipid 2,3-dioleoyloxy-N-[2(sperminecarboxiamido)ethyl[-N,N-dimethyl-1-propaniminium trifluoroacetate

DPMSCs:

Dental pulp mesenchymal stem cells

DPPSCs:

Dental pulp pluripotent stem cells

ECM:

Extracellular matrix

EGFP:

Enhanced green fluorescent protein

ELISA:

Enzyme-linked immunosorbent assay

FCM:

Flow cytometry

FITC:

Fluorescein isothiocyanate

FGF-2:

Fibroblast growth factor

GAG:

Glycosaminoglycans

GET:

GAG-binding enhanced transduction

GFs:

Growth factors

GFP:

Green fluorescent protein

gMSCs:

Goat mesenchymal stem cells

HA:

Hyaluronic acid

hAMSCs:

Human adipose mesenchymal stem cells

hBMMSCs:

Human bone marrow derived mesenchymal stem cells

hiPSCs:

Human induced pluripotent stem cells

hMSCs:

Human mesenchymal stem cells

hUCMSCs:

Human umbilical cord mesenchymal stem cells

ICC:

Immunocytochemistry

IGF-1:

Insulin-like growth factor-1

IHC:

Immunohistochemistry

luc:

Luciferase

MCM:

Mineral-coated microparticles

Me-β-CD:

Methyl-β-cyclodextrin

miRNA:

Micro-RNA

MYOD:

Myogenic differentiation protein

NAs:

Nucleic acids

nHA:

Hydroxyapatite nanoparticles

NPs:

Nanoparticles

OCN:

Osteocalcin

OCT3 and 4:

Octamer-binding transcription factor 3 and 4

OPN:

Osteopontin

OSX:

Osterix

PAA-BA:

Poly (amidoamine) with pendant aminobutyl group

PAMAM:

Polyamidoamines

PCR:

Polymerase chain reaction

pDNA:

Plasmid DNA

PEI:

Polyethyleneimine

PI:

Propidium iodide

PLGA:

Poly (DL-lactic-co-glycolic acid)

PLLs:

Poly-L-lysines

rAMSCs:

Rat adipose derived mesenchymal stem cells

RFP:

Red fluorescence protein

RITC:

Rhodamine isothiocyanate

rMSCs:

Rat mesenchymal stem cells

RUNX2:

Runt-related transcription factor 2

SAR:

Structure–function relationship

siRNA:

Small interfering RNA

SOX9:

Sex-determining region Y-type high mobility group box 9

TDMSCs:

Tonsil derived mesenchymal stem cells

TGF-β1 and -β3:

Transforming growth factor-β1 and -β3

UV:

Ultraviolet

VEGF:

Vascular endothelial growth factor

WB:

Western blot

YFP:

Yellow fluorescence protein

References

  1. Solorio LD, Vieregge EL, Dhami CD, Dang PN, Alsberg E. Engineered cartilage via self-assembled hMSC sheets with incorporated biodegradable gelatin microspheres releasing transforming growth factor-$β$1. J Control Release. 2012;158(2):224–32.

    Article  Google Scholar 

  2. De Bari C, Roelofs AJ. Stem cell-based therapeutic strategies for cartilage defects and osteoarthritis. Curr Opin Pharmacol. 2018;40:74–80.

    Article  Google Scholar 

  3. Kangari P, Talaei-Khozani T, Razeghian-Jahromi I, Razmkhah M. Mesenchymal stem cells: amazing remedies for bone and cartilage defects. Stem Cell Res Ther. 2020;11(1):492.

    Article  Google Scholar 

  4. Zheng W, Li H, Hu K, Li L, Bei M. Chondromalacia patellae: current options and emerging cell therapies. Stem Cell Res Ther. 2021;12(1):412.

    Article  Google Scholar 

  5. Liang Z, Luo Y, Lv Y. Mesenchymal stem cell-derived microvesicles mediate BMP2 gene delivery and enhance bone regeneration. J Mater Chem part B. 2020;8(30):6378–89.

    Article  Google Scholar 

  6. López-Seijas J, Miranda-Balbuena D, Iglesias-Fente A, et al. Development of new non-viral systems for genetic modification of senescent cells. Mol Ther Nucleic Acids. 2023;32:302–17.

    Article  Google Scholar 

  7. Ho YK, Woo JY, Tu GXE, Deng LW, Too HP. A highly efficient non-viral process for programming mesenchymal stem cells for gene directed enzyme prodrug cancer therapy. Sci Rep. 2020;10(1):14257.

    Article  Google Scholar 

  8. Alam P, Haile B, Arif M, et al. Inhibition of senescence-associated genes Rb1 and Meis2 in adult cardiomyocytes results in cell cycle reentry and cardiac repair post-myocardial infarction. J Am Heart Assoc. 2019;8(15):12089.

    Article  Google Scholar 

  9. Gonzalez-Rey E, Gonzalez MA, Varela N, et al. Human adipose-derived mesenchymal stem cells reduce inflammatory and T cell responses and induce regulatory T cells in vitro in rheumatoid arthritis. Ann Rheum Dis. 2010;69(01):241–8.

    Article  Google Scholar 

  10. Dimmeler S, Leri A. Aging and disease as modifiers of efficacy of cell therapy. Circ Res. 2008;102(11):1319–30.

    Article  Google Scholar 

  11. Pawitan JA, Bui TA, Mubarok W, et al. Enhancement of the therapeutic capacity of mesenchymal stem cells by genetic modification: a systematic review. Front Cell Dev Biol. 2020;8: 587776.

    Article  Google Scholar 

  12. Partridge KA, Oreffo ROC. Gene delivery in bone tissue engineering: progress and prospects using viral and nonviral strategies. Tissue Eng. 2004;10(1–2):295–307.

    Article  Google Scholar 

  13. Capito RM, Spector M. Collagen scaffolds for nonviral IGF-1 gene delivery in articular cartilage tissue engineering. Gene Ther. 2007;14(9):721–32.

    Article  Google Scholar 

  14. Cucchiarini M, Madry H. Overexpression of human IGF-I via direct rAAV-mediated gene transfer improves the early repair of articular cartilage defects in vivo. Gene Ther. 2014;21(9):811–9.

    Article  Google Scholar 

  15. Maihöfer J, Madry H, Rey-Rico A, et al. Hydrogel-guided, rAAV-Mediated IGF-I overexpression enables long-term cartilage repair and protection against perifocal osteoarthritis in a large-animal full-thickness chondral defect model at one year in vivo. Adv Mater. 2021;33(16): e2008451.

    Article  Google Scholar 

  16. Ziv R, Steinhardt Y, Pelled G, Gazit D, Rubinsky B. Micro-electroporation of mesenchymal stem cells with alternating electrical current pulses. Biomed Microdevices. 2009;11(1):95–101.

    Article  Google Scholar 

  17. Deng W, Fu M, Cao Y, et al. Angelica sinensis polysaccharide nanoparticles as novel non-viral carriers for gene delivery to mesenchymal stem cells. Nanomedicine. 2013;9(8):1181–91.

    Article  Google Scholar 

  18. Pan H, Zheng Q, Yang S, et al. A novel peptide-modified and gene-activated biomimetic bone matrix accelerating bone regeneration. J Biomed Mater Res part A. 2014;102(8):2864–74.

    Article  Google Scholar 

  19. Gonzalez-fernandez T, Tierney EG, Cunniffe GM, O’Brien FJ, Kelly DJ. Gene delivery of TGF-β3 and BMP2 in an MSC-laden alginate hydrogel for articular cartilage and endochondral bone tissue engineering. Tissue Eng. 2016;22(1):776–87.

    Article  Google Scholar 

  20. Jeong H, Lee ES, Jung G, et al. Bioreducible-cationic poly(amido amine)s for enhanced gene delivery and osteogenic differentiation of tonsil-derived mesenchymal stem cells. J Biomed Nanotechnol. 2016;12(5):1023–34.

    Article  Google Scholar 

  21. Hsieh MK, Wu CJ, Chen CC, et al. BMP-2 gene transfection of bone marrow stromal cells to induce osteoblastic differentiation in a rat calvarial defect model. Mater Sci Eng part C. 2018;91:806–16.

    Article  Google Scholar 

  22. Loozen LD, Kruyt MC, Kragten AHM, et al. BMP-2 gene delivery in cell-loaded and cell-free constructs for bone regeneration. PLoS ONE. 2019;14(7): e0220028.

    Article  Google Scholar 

  23. Loozen LD, Wegman F, Öner FC, Dhert WJA, Alblas J. Porous bioprinted constructs in BMP-2 non-viral gene therapy for bone tissue engineering. J Mater Chem part B. 2013;1(48):6619–26.

    Article  Google Scholar 

  24. Zhao Z, Zhao M, Xiao G, Franceschi RT. Gene transfer of the Runx2 transcription factor enhances osteogenic activity of bone marrow stromal cells in vitro and in vivo. Mol Ther. 2005;12(2):247–53.

    Article  Google Scholar 

  25. Lee SJ, Kang SW, Do HJ, et al. Enhancement of bone regeneration by gene delivery of BMP2/Runx2 bicistronic vector into adipose-derived stromal cells. Biomaterials. 2010;31(21):5652–9.

    Article  Google Scholar 

  26. Núñez-Toldrà R, Dosta P, Montori S, Ramos V, Atari M, Borrós S. Improvement of osteogenesis in dental pulp pluripotent-like stem cells by oligopeptide-modified poly($β$-amino ester)s. Acta Biomater. 2017;53:152–64.

    Article  Google Scholar 

  27. Kim JMJHJM, Park JS, Yang HN, et al. The use of biodegradable PLGA nanoparticles to mediate SOX9 gene delivery in human mesenchymal stem cells (hMSCs) and induce chondrogenesis. Biomaterials. 2011;32(1):268–78.

    Article  Google Scholar 

  28. Park JS, Yang HN, Woo DG, et al. Chondrogenesis of human mesenchymal stem cells mediated by the combination of SOX trio SOX5, 6, and 9 genes complexed with PEI-modified PLGA nanoparticles. Biomaterials. 2011;32(14):3679–88.

    Article  Google Scholar 

  29. Gonzalez-Fernandez T, Rathan S, Hobbs C, et al. Pore-forming bioinks to enable spatio-temporally defined gene delivery in bioprinted tissues. J Control Release. 2019;301(1):13–27.

    Article  Google Scholar 

  30. Yoon JK, Kang ML, Park JH, et al. Direct control of stem cell behavior using biomaterials and genetic factors. Stem Cells Int. 2018;2018:8642989.

    Article  Google Scholar 

  31. Hamann A, Nguyen A, Pannier AK. Nucleic acid delivery to mesenchymal stem cells: a review of nonviral methods and applications. J Biol Eng. 2019;13(1):7.

    Article  Google Scholar 

  32. Madry H, Venkatesan JK, Carballo-pedrares N, Rey-rico A, Cucchiarini M. Scaffold-mediated gene delivery for osteochondral repair. Pharmaceutics. 2020;12(10):1–23.

    Article  Google Scholar 

  33. Miceli V, Bulati M, Iannolo G, Zito G, Gallo A, Conaldi PG. Therapeutic properties of mesenchymal stromal/stem cells: the need of cell priming for cell-free therapies in regenerative medicine. Int J Mol Sci. 2021;22(2):1–20.

    Article  Google Scholar 

  34. Ramasubramanian A, Shiigi S, Lee KG, Yang F. Non-viral delivery of inductive and suppressive genes to adipose-derived stem cells for osteogenic differentiation. Pharm Res. 2011;28(2):227–39.

    Google Scholar 

  35. Bono N, Ponti F, Mantovani D, Candiani G. Non-viral in vitro gene delivery: it is now time to set the bar! Pharmaceutics. 2020;12(2):183.

    Article  Google Scholar 

  36. Haleem-Smith H, Derfoul A, Okafor C, et al. Optimization of high-efficiency transfection of adult human mesenchymal stem cells in vitro. Mol Biotechnol. 2005;30(1):9–19.

    Article  Google Scholar 

  37. Kim HJ, Im GI. Electroporation-mediated transfer of SOX trio genes (SOX-5, SOX-6, and SOX-9) to enhance the chondrogenesis of mesenchymal stem cells. Stem Cells Dev. 2011;20(12):2103–14.

    Article  Google Scholar 

  38. Flanagan M, Gimble JM, Yu G, Xia X, Bunnell BA, Li S. Competitive DNA transfection formulation via electroporation for human adipose stem cells and mesenchymal stem cells. Biol Proced Online. 2012;14(1):7.

    Article  Google Scholar 

  39. Otani K, Yamahara K, Ohnishi S, Obata H, Kitamura S, Nagaya N. Nonviral delivery of siRNA into mesenchymal stem cells by a combination of ultrasound and microbubbles. J Control release. 2009;133(2):146–53.

    Article  Google Scholar 

  40. Li P, Gao Y, Liu Z, et al. DNA transfection of bone marrow stromal cells using microbubble-mediated ultrasound and polyethylenimine: an in vitro study. Cell Biochem Biophys. 2013;66(3):775–86.

    Article  Google Scholar 

  41. Feichtinger GA, Hofmann AT, Slezak P, et al. Sonoporation increases therapeutic efficacy of inducible and constitutive BMP2/7 in vivo gene delivery. Hum Gene Ther Methods. 2014;25(1):57–71.

    Article  Google Scholar 

  42. Fouriki A, Dobson J. Oscillating magnet array-based nanomagnetic gene transfection of human mesenchymal stem cells. Nanomedicine (Lond). 2014;9(7):989–97.

    Article  Google Scholar 

  43. Zhang TY, Wu JH, Xu QH, et al. Design of magnetic gene complexes as effective and serum resistant gene delivery systems for mesenchymal stem cells. Int J Pharm. 2017;520(1–2):1–13.

    Google Scholar 

  44. Delyagina E, Schade A, Scharfenberg D, et al. Improved transfection in human mesenchymal stem cells: effective intracellular release of pDNA by magnetic polyplexes. Nanomedicine (Lond). 2014;9(7):999–1017.

    Article  Google Scholar 

  45. Tsulaia TV, Prokopishyn NL, Yao A, et al. Glass needle-mediated microinjection of macromolecules and transgenes into primary human mesenchymal stem cells. J Biomed Sci. 2003;10(3):328–36.

    Article  Google Scholar 

  46. Han SW, Nakamura C, Kotobuki N, et al. High-efficiency DNA injection into a single human mesenchymal stem cell using a nanoneedle and atomic force microscopy. Nanomedicine. 2008;4(3):215–25.

    Article  Google Scholar 

  47. Guan Z, Chen S, Pan F, Fan L, Sun D. Effects of gene delivery approaches on differentiation potential and gene function of mesenchymal stem cells. IEEE Trans Biomed Eng. 2022;69(1):83–95.

    Article  Google Scholar 

  48. Mehier-Humbert S, Guy RH. Physical methods for gene transfer: improving the kinetics of gene delivery into cells. Adv Drug Deliv Rev. 2005;57(5):733–53.

    Article  Google Scholar 

  49. Mellott AJ, Forrest ML, Detamore MS. Physical non-viral gene delivery methods for tissue engineering. Ann Biomed Eng. 2012;41(3):446–68.

    Article  Google Scholar 

  50. Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet. 2003;4(5):346–58.

    Article  Google Scholar 

  51. Uludag H, Ubeda A, Ansari A. At the intersection of biomaterials and gene therapy: progress in non-viral delivery of nucleic acids. Front Bioeng Biotechnol. 2019;7(1):1–21.

    Google Scholar 

  52. Kisby T, Yilmazer A, Kostarelos K. Reasons for success and lessons learnt from nanoscale vaccines against COVID-19. Nat Nanotechnol. 2021;16(8):843–50.

    Article  Google Scholar 

  53. Ponti F, Campolungo M, Melchiori C, Bono N, Candiani G. Cationic lipids for gene delivery: many players, one goal. Chem Phys Lipids. 2021;235: 105032.

    Article  Google Scholar 

  54. Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 2020 202 20(2), 101–124 (2020).

  55. Loh XJ, Lee TC, Dou Q, Deen GR. Utilising inorganic nanocarriers for gene delivery. Biomater Sci. 2016;4(1):70–86.

    Article  Google Scholar 

  56. Santos JL, Pandita D, Rodrigues JP, Pego AL, Granja P, Tomas H. Non-viral gene delivery to mesenchymal stem cells: methods, strategies and application in bone tissue engineering and regeneration. Curr Gene Ther. 2011;11(1):46–57.

    Article  Google Scholar 

  57. Corsi K, Chellat F, Yahia L, Fernandes JC. Mesenchymal stem cells, MG63 and HEK293 transfection using chitosan-DNA nanoparticles. Biomaterials. 2003;24(7):1255–64.

    Article  Google Scholar 

  58. Yang F, Green JJ, Dinio T, et al. Gene delivery to human adult and embryonic cell-derived stem cells using biodegradable nanoparticulate polymeric vectors. Gene Ther. 2009;16(4):533–46.

    Article  Google Scholar 

  59. Jang JH, Bengali Z, Houchin TL, Shea LD. Surface adsorption of DNA to tissue engineering scaffolds for efficient gene delivery. J Biomed Mater Res part A. 2006;77(1):50–8.

    Article  Google Scholar 

  60. Watari S, Hayashi K, Wood JA, et al. Modulation of osteogenic differentiation in hMSCs cells by submicron topographically-patterned ridges and grooves. Biomaterials. 2012;33(1):128–36.

    Article  Google Scholar 

  61. Miyazaki H, Ogura M, Sakaguchi Y, Hasegawa T, Atobe S, Terai K. Mechanism of jet injector-induced plasmid DNA uptake: Contribution of shear stress and endocytosis. Int J Pharm. 2021;609: 121200.

    Article  Google Scholar 

  62. Ponti F, Bono N, Russo L, Bigini P, Mantovani D, Candiani G. Vibropolyfection: coupling polymer-mediated gene delivery to mechanical stimulation to enhance transfection of adherent cells. J Nanobiotechnology. 2022;20(1):363.

    Article  Google Scholar 

  63. Childs PG, Boyle CA, Pemberton GD, et al. Use of nanoscale mechanical stimulation for control and manipulation of cell behaviour. Acta Biomater. 2016;34:159–68.

    Article  Google Scholar 

  64. Moore DL. Isolation of enteroviruses from capillary blood specimens. J Pediatr. 1986;108(1):166.

    Article  Google Scholar 

  65. Loozen LD, Vandersteen A, Kragten AHM, et al. Bone formation by heterodimers through non-viral gene delivery of BMP-2/6 and BMP-2/7. Eur Cell Mater. 2018;35:195–208.

    Article  Google Scholar 

  66. Schade A, Delyagina E, Scharfenberg D, et al. Innovative strategy for MicroRNA delivery in human mesenchymal stem cells via magnetic nanoparticles. Int J Mol Sci. 2013;14(6):10726.

    Article  Google Scholar 

  67. Carthew J, Donderwinkel I, Shrestha S, Truong VX, Forsythe JS, Frith JE. In situ miRNA delivery from a hydrogel promotes osteogenesis of encapsulated mesenchymal stromal cells. Acta Biomater. 2020;101(1):249–61.

    Article  Google Scholar 

  68. Huynh CT, Nguyen MK, Naris M, Tonga GY, Rotello VM, Alsberg E. Light-triggered RNA release and induction of hMSC osteogenesis via photodegradable, dual-crosslinked hydrogels. Nanomedicine. 2016;11(12):1535–50.

    Article  Google Scholar 

  69. Curtin CM, Tierney EG, Mcsorley K, Cryan SA, Duffy GP, O’Brien FJ. Combinatorial gene therapy accelerates bone regeneration: non-viral dual delivery of VEGF and BMP2 in a collagen-nanohydroxyapatite scaffold. Adv Healthc Mater. 2015;4(2):223–7.

    Article  Google Scholar 

  70. Garg PK, Pandey S, Kim HN, et al. Synergistic effects of hyperosmotic polymannitol based non-viral vectors and nanotopographical cues for enhanced gene delivery. RSC Adv. 2016;6(112):111233–8.

    Article  Google Scholar 

  71. Xiang S, Zhang X, Xiang S, Zhang X. Cellular uptake mechanism of non-viral gene delivery and means for improving transfection efficiency. in: gene therapy - tools and potential applications (Chap. 4). IntechOpen, 71–90 (2013).

  72. Rejman J, Conese M, Hoekstra D. Gene transfer by means of lipo- and polyplexes: role of clathrin and caveolae-mediated endocytosis. J Liposome Res. 2006;16(3):237–47.

    Article  Google Scholar 

  73. Walsh DP, Murphy RD, Panarella A, et al. Bioinspired star-shaped poly(l-lysine) polypeptides: efficient polymeric nanocarriers for the delivery of DNA to mesenchymal stem cells. Mol Pharm. 2018;15(5):1878–91.

    Article  Google Scholar 

  74. Gheisari Y, Soleimani M, Azadmanesh K, Zeinali S. Multipotent mesenchymal stromal cells: optimization and comparison of five cationic polymer-based gene delivery methods. Cytotherapy. 2008;10(8):815–23.

    Article  Google Scholar 

  75. Santos JL, Oramas E, Pêgo AP, Granja PL, Tomás H. Osteogenic differentiation of mesenchymal stem cells using PAMAM dendrimers as gene delivery vectors. J Control release. 2009;134(2):141–8.

    Article  Google Scholar 

  76. Shakhbazau A, Shcharbin D, Petyovka N, et al. Non-virally modified human mesenchymal stem cells produce ciliary neurotrophic factor in biodegradable fibrin-based 3D scaffolds. J Pharm Sci. 2012;101(4):1546–54.

    Article  Google Scholar 

  77. Malakooty Poor E, Baghaban Eslaminejad M, Gheibi N, Bagheri F, Atyabi F. Chitosan-pDNA nanoparticle characteristics determine the transfection efficacy of gene delivery to human mesenchymal stem cells. Artif Cells Nanomed Biotechnol. 2014;42(6):376–84.

    Article  Google Scholar 

  78. Mintzer MA, Simanek EE. Nonviral vectors for gene delivery. Chem Rev. 2009;109(2):259–302.

    Article  Google Scholar 

  79. Benoit DSW, Boutin ME. Controlling mesenchymal stem cell gene expression using polymer-mediated delivery of siRNA. Biomacromol. 2012;13(11):3841–9.

    Article  Google Scholar 

  80. Park JS, Yi SW, Kim HJ, Park KH. Receptor-mediated gene delivery into human mesenchymal stem cells using hyaluronic acid-shielded polyethylenimine/pDNA nanogels. Carbohydr Polym. 2016;136:791–802.

    Article  Google Scholar 

  81. Pandita D, Santos JL, Rodrigues J, Pêgo AP, Granja PL, Tomás H. Gene delivery into mesenchymal stem cells: a biomimetic approach using RGD nanoclusters based on poly(amidoamine) dendrimers. Biomacromol. 2011;12(2):472–81.

    Article  Google Scholar 

  82. Santos JL, Oliveira H, Pandita D, et al. Functionalization of poly(amidoamine) dendrimers with hydrophobic chains for improved gene delivery in mesenchymal stem cells. J Control Release. 2010;144(1):55–64.

    Article  Google Scholar 

  83. Song H, Wang G, He B, et al. Cationic lipid-coated PEI/DNA polyplexes with improved efficiency and reduced cytotoxicity for gene delivery into mesenchymal stem cells. Int J Nanomedicine. 2012;7:4637–48.

    Google Scholar 

  84. Ayatollahi S, Hashemi M, Kazemi Oskuee R, et al. Synthesis of efficient gene delivery systems by grafting pegylated alkylcarboxylate chains to PAMAM dendrimers: Evaluation of transfection efficiency and cytotoxicity in cancerous and mesenchymal stem cells. J Biomater Appl. 2015;30(5):632–48.

    Article  Google Scholar 

  85. Clements BA, Incani V, Kucharski C, Lavasanifar A, Ritchie B, Uludaǧ H. A comparative evaluation of poly-L-lysine-palmitic acid and Lipofectamine 2000 for plasmid delivery to bone marrow stromal cells. Biomaterials. 2007;28(31):4693–704.

    Article  Google Scholar 

  86. Bahadur RKCC, Cezary K, Hasan U. Additive nanocomplexes of cationic lipopolymers for improved non-viral gene delivery to mesenchymal stem cells. J Mater Chem part B. 2015;3(19):3972–82.

    Article  Google Scholar 

  87. Salmasi Z, Shier WT, Hashemi M, et al. Heterocyclic amine-modified polyethylenimine as gene carriers for transfection of mammalian cells. Eur J Pharm Biopharm. 2015;96:76–88.

    Article  Google Scholar 

  88. Lancelot A, González-Pastor R, Concellón A, Sierra T, Martín-Duque P, Serrano JL. DNA Transfection to Mesenchymal Stem Cells Using a Novel Type of Pseudodendrimer Based on 2,2-Bis(hydroxymethyl)propionic Acid. Bioconjug Chem. 2017;28(4):1135–50.

    Article  Google Scholar 

  89. Yeh PH, Sun JS, Wu HC, Hwang LH, Wang TW. Stimuli-responsive HA-PEI nanoparticles encapsulating endostatin plasmid for stem cell gene therapy. RSC Adv. 2013;3(31):12922–32.

    Article  Google Scholar 

  90. Kim H, Nam K, Nam JP, et al. VEGF therapeutic gene delivery using dendrimer type bio-reducible polymer into human mesenchymal stem cells (hMSCs). J Control Release. 2015;220(Pt A):222–8.

    Article  Google Scholar 

  91. Tzeng SY, Hung BP, Grayson WL, Green JJ. Cystamine-terminated poly(beta-amino ester)s for siRNA delivery to human mesenchymal stem cells and enhancement of osteogenic differentiation. Biomaterials. 2012;33(32):8142–51.

    Article  Google Scholar 

  92. El Jundi A, Morille M, Bettache N, et al. Degradable double hydrophilic block copolymers and tripartite polyionic complex micelles thereof for small interfering ribonucleic acids (siRNA) delivery. J Colloid Interface Sci. 2020;580:449–59.

    Article  Google Scholar 

  93. Tong H, Wang C, Huang Y, et al. Polyethylenimine600-β-cyclodextrin: a promising nanopolymer for nonviral gene delivery of primary mesenchymal stem cells. Int J Nanomedicine. 2013;8:1935–46.

    Article  Google Scholar 

  94. Lai WF, Tang GP, Wang X, et al. Cyclodextrin-PEI-tat polymer as a vector for plasmid DNA delivery to placenta mesenchymal stem cells. Bionanoscience. 2011;1(3):89–96.

    Article  Google Scholar 

  95. Hujaya SD, Manninen A, Kling K, Wagner JB, Vainio SJ, Liimatainen H. Self-assembled nanofibrils from RGD-functionalized cellulose nanocrystals to improve the performance of PEI/DNA polyplexes. J Colloid Interface Sci. 2019;553:71–82.

    Article  Google Scholar 

  96. Kunath K, Merdan T, Hegener O, Häberlein H, Kissel T. Integrin targeting using RGD-PEI conjugates for in vitro gene transfer. J Gene Med. 2003;5(7):588–99.

    Article  Google Scholar 

  97. Juliano RL, Ming X, Nakagawa O, Xu R, Yoo H. Integrin targeted delivery of gene therapeutics. Theranostics. 2011;1:211–9.

    Article  Google Scholar 

  98. Pezzoli D, Tarsini P, Melone L, Candiani G. RGD-derivatized PEI-PEG copolymers: influence of the degree of substitution on the targeting behavior. J Drug Deliv Sci Technol. 2017;37:115–22.

    Article  Google Scholar 

  99. Santos JL, Pandita D, Rodrigues J, et al. Receptor-mediated gene delivery using PAMAM dendrimers conjugated with peptides recognized by mesenchymal stem cells. Mol Pharm. 2010;7(3):763–74.

    Article  Google Scholar 

  100. Beloor J, Ramakrishna S, Nam K, et al. Effective gene delivery into human stem cells with a cell-targeting Peptide-modified bioreducible polymer. Small. 2015;11(17):2069–79.

    Article  Google Scholar 

  101. Almalik A, Karimi S, Ouasti S, et al. Hyaluronic acid (HA) presentation as a tool to modulate and control the receptor-mediated uptake of HA-coated nanoparticles. Biomaterials. 2013;34(21):5369–80.

    Article  Google Scholar 

  102. Saraf A, Hacker MC, Sitharaman B, Grande-Allen KJ, Barry MA, Mikos AG. Synthesis and conformational evaluation of a novel gene delivery vector for human mesenchymal stem cells. Biomacromol. 2008;9(3):818–27.

    Article  Google Scholar 

  103. Zhu H, Mitsuhashi N, Klein A, et al. The role of the hyaluronan receptor CD44 in mesenchymal stem cell migration in the extracellular matrix. Stem Cells. 2006;24(4):928–35.

    Article  Google Scholar 

  104. Lisignoli G, Cristino S, Piacentini A, Cavallo C, Caplan AI, Facchini A. Hyaluronan-based polymer scaffold modulates the expression of inflammatory and degradative factors in mesenchymal stem cells: involvement of Cd44 and Cd54. J Cell Physiol. 2006;207(2):364–73.

    Article  Google Scholar 

  105. Felgner PL, Gadek TR, Holm M, et al. Lipofection: a highly efficient, lipid-mediated DNA-transfection procedure. Proc Natl Acad Sci U S A. 1987;84(21):7413–7.

    Article  Google Scholar 

  106. Pezzoli D, Chiesa R, De Nardo L, Candiani G. We still have a long way to go to effectively deliver genes! J Appl Biomater Funct Mater. 2012;10(2):82–91.

    Google Scholar 

  107. Wasungu L, Hoekstra D. Cationic lipids, lipoplexes and intracellular delivery of genes. J Control release. 2006;116(2):255–64.

    Article  Google Scholar 

  108. Simões S, Filipe A, Faneca H, et al. Cationic liposomes for gene delivery. Expert Opin Drug Deliv. 2005;2(2):237–54.

    Article  Google Scholar 

  109. Marianecci C, Di Marzio L, Rinaldi F, et al. Niosomes from 80s to present: the state of the art. Adv Colloid Interface Sci. 2014;205:187–206.

    Article  Google Scholar 

  110. Vanegas Sáenz JR, Tenkumo T, Kamano Y, et al. Amiloride-enhanced gene transfection of octa-arginine functionalized calcium phosphate nanoparticles. PLoS ONE. 2017;12(11): e0188347.

    Article  Google Scholar 

  111. Rim YA, Nam Y, Park N, et al. Chondrogenic differentiation from induced pluripotent stem cells using non-viral minicircle vectors. Cells. 2020;9(3):582.

    Article  Google Scholar 

  112. Dwivedi PP, Anderson PJ, Powell BC. Development of an efficient, non-viral transfection method for studying gene function and bone growth in human primary cranial suture mesenchymal cells reveals that the cells respond to BMP2 and BMP3. BMC Biotechnol. 2012;12:45.

    Article  Google Scholar 

  113. Zhang C, Jin R, Zhao P, Lin C. A family of cationic polyamides for in vitro and in vivo gene transfection. Acta Biomater. 2015;22:120–30.

    Article  Google Scholar 

  114. McMillan A, Nguyen MK, Gonzalez-Fernandez T, et al. Dual non-viral gene delivery from microparticles within 3D high-density stem cell constructs for enhanced bone tissue engineering. Biomaterials. 2018;161:240–55.

    Article  Google Scholar 

  115. Wegman F, Bijenhof A, Schuijff L, Öner FC, Dhert WJAA, Alblas J. Osteogenic differentiation as a result of BMP-2 plasmid DNA based gene therapy in vitro and in vivo. Eur Cells Mater. 2011;21(1):230–42.

    Article  Google Scholar 

  116. Wu K, Xu J, Liu M, et al. Induction of osteogenic differentiation of stem cells via a lyophilized microRNA reverse transfection formulation on a tissue culture plate. Int J Nanomedicine. 2013;8:1595–607.

    Google Scholar 

  117. Yan J, Zhang C, Zhao Y, et al. Non-viral oligonucleotide antimiR-138 delivery to mesenchymal stem cell sheets and the effect on osteogenesis. Biomaterials. 2014;35(27):7734–49.

    Article  Google Scholar 

  118. Lakhani CM. Delivery of siRNA via cationic Sterosomes to enhance osteogenic differentiation of mesenchymal stem cells. Physiol Behav. 2019;176(3):139–48.

    Google Scholar 

  119. Yu X, Murphy WL. 3-D scaffold platform for optimized non-viral transfection of multipotent stem cells. J Mater Chem part B. 2014;2(46):8193.

    Google Scholar 

  120. Chen L, Long D, Huang S, et al. Evaluation of a novel poly(amidoamine) with pendant aminobutyl group on the cellular properties of transfected bone marrow mesenchymal stem cells. J Biomed Mater Res part A. 2018;106(3):686–97.

    Article  Google Scholar 

  121. Dalby B, Cates S, Harris A, et al. Advanced transfection with Lipofectamine 2000 reagent: primary neurons, siRNA, and high-throughput applications. Methods. 2004;33(2):95–103.

    Article  Google Scholar 

  122. Tsuchiya H, Kitoh H, Sugiura F, Ishiguro N. Chondrogenesis enhanced by overexpression of sox9 gene in mouse bone marrow-derived mesenchymal stem cells. Biochem Biophys Res Commun. 2003;301(2):338–43.

    Article  Google Scholar 

  123. Hoare M, Greiser U, Schu S, et al. Enhanced lipoplex-mediated gene expression in mesenchymal stem cells using reiterated nuclear localization sequence peptides. J Gene Med. 2010;12(2):207–18.

    Google Scholar 

  124. Ribeiro SC, Mendes R, Madeira C, Monteiro GA, da Silva CL, Cabral JM. A quantitative method to evaluate mesenchymal stem cell lipofection using real-time PCR. Biotechnol Prog. 2010;26(5):1501–4.

    Article  Google Scholar 

  125. Boura JS, Dos SF, Gimble JM, et al. Direct head-to-head comparison of cationic liposome-mediated gene delivery to mesenchymal stem/stromal cells of different human sources: a comprehensive study. Hum Gene Ther Methods. 2013;24(1):38–48.

    Article  Google Scholar 

  126. de Carvalho TG, Pellenz FM, Laureano A, et al. A simple protocol for transfecting human mesenchymal stem cells. Biotechnol Lett. 2018;40(3):617–22.

    Article  Google Scholar 

  127. Buck J, Grossen P, Cullis PR, Huwyler J, Witzigmann D. Lipid-based DNA therapeutics: hallmarks of non-viral gene delivery. ACS Nano. 2019;13(4):3754–82.

    Article  Google Scholar 

  128. Grijalvo S, Puras G, Zárate J, et al. Cationic niosomes as non-viral vehicles for nucleic acids: challenges and opportunities in gene delivery. Pharmaceutics. 2019;11(2):50.

    Article  Google Scholar 

  129. Attia N, Mashal M, Grijalvo S, et al. Stem cell-based gene delivery mediated by cationic niosomes for bone regeneration. Nanomedicine. 2018;14(2):521–31.

    Article  Google Scholar 

  130. Carballo-Pedrares N, Kattar A, Concheiro A, Alvarez-Lorenzo C, Rey-Rico A. Niosomes-based gene delivery systems for effective transfection of human mesenchymal stem cells. Mater Sci Eng C. 2021;128:112307.

    Article  Google Scholar 

  131. Carballo-Pedrares N, Sanjurjo-Rodriguez C, Señarís J, et al. Chondrogenic differentiation of human mesenchymal stem cells via SOX9 delivery in cationic niosomes. Pharmaceutics. 2022;14(11):2327.

    Article  Google Scholar 

  132. Villate-Beitia I, Truong NF, Gallego I, et al. Hyaluronic acid hydrogel scaffolds loaded with cationic niosomes for efficient non-viral gene delivery. RSC Adv. 2018;8(56):31934–42.

    Article  Google Scholar 

  133. Vado Y, Puras G, Rosique M, et al. Design and validation of a process based on cationic niosomes for gene delivery into novel urine-derived mesenchymal stem cells. Pharmaceutics. 2021;13(5):696.

    Article  Google Scholar 

  134. Raisin S, Belamie E, Morille M. Non-viral gene activated matrices for mesenchymal stem cells based tissue engineering of bone and cartilage. Biomaterials. 2016;104:223–37.

    Article  Google Scholar 

  135. He CX, Li N, Hu YL, et al. Effective gene delivery to mesenchymal stem cells based on the reverse transfection and three-dimensional cell culture system. Pharm Res. 2011;28(7):1577–90.

    Article  Google Scholar 

  136. Graceffa V. Physical and mechanical cues affecting biomaterial-mediated plasmid DNA delivery: insights into non-viral delivery systems. J Genet Eng Biotechnol. 2021;19(1):90.

    Article  Google Scholar 

  137. Navarrete RO, Lee EM, Smith K, et al. Substrate stiffness controls osteoblastic and chondrocytic differentiation of mesenchymal stem cells without exogenous stimuli. PLoS One. 2017;12(1):e0170312.

    Article  Google Scholar 

  138. Pek YS, Wan ACA, Ying JY. The effect of matrix stiffness on mesenchymal stem cell differentiation in a 3D thixotropic gel. Biomaterials. 2010;31(3):385–91.

    Article  Google Scholar 

  139. Zhao W, Li X, Liu X, Zhang N, Wen X. Effects of substrate stiffness on adipogenic and osteogenic differentiation of human mesenchymal stem cells. Mater Sci Eng part C. 2014;40:316–23.

    Article  Google Scholar 

  140. Kong HJ, Liu J, Riddle K, Matsumoto T, Leach K, Mooney DJ. Non-viral gene delivery regulated by stiffness of cell adhesion substrates. Nat Mater. 2005;4(6):460–4.

    Article  Google Scholar 

  141. Gojgini S, Tokatlian T, Segura T. Utilizing cell-matrix interactions to modulate gene transfer to stem cells inside hyaluronic acid hydrogels. Mol Pharm. 2011;8(5):1582–91.

    Article  Google Scholar 

  142. Ledo AM, Vining KH, Alonso MJ, Garcia-Fuentes M, Mooney DJ. Extracellular matrix mechanics regulate transfection and SOX9-directed differentiation of mesenchymal stem cells. Acta Biomater. 2020;110:153–63.

    Article  Google Scholar 

  143. Tsimbouri P, Gadegaard N, Burgess K, et al. Nanotopographical effects on mesenchymal stem cell morphology and phenotype. J Cell Biochem. 2014;115(2):380–90.

    Article  Google Scholar 

  144. Dalby MJ, Gadegaard N, Oreffo ROC. Harnessing nanotopography and integrin-matrix interactions to influence stem cell fate. Nat Mater. 2014;13(6):558–69.

    Article  Google Scholar 

  145. Yim EKF, Darling EM, Kulangara K, Guilak F, Leong KW. Nanotopography-induced changes in focal adhesions, cytoskeletal organization, and mechanical properties of human mesenchymal stem cells. Biomaterials. 2010;31(6):1299–306.

    Article  Google Scholar 

  146. McMurray RJ, Gadegaard N, Tsimbouri PM, et al. Nanoscale surfaces for the long-term maintenance of mesenchymal stem cell phenotype and multipotency. Nat Mater. 2011;10(8):637–44.

    Article  Google Scholar 

  147. Teo BKK, Goh SH, Kustandi TS, Loh WW, Low HY, Yim EKF. The effect of micro and nanotopography on endocytosis in drug and gene delivery systems. Biomaterials. 2011;32(36):9866–75.

    Article  Google Scholar 

  148. Kulangara K, Yang Y, Yang J, Leong KW. Nanotopography as modulator of human mesenchymal stem cell function. Biomaterials. 2012;33(20):4998–5003.

    Article  Google Scholar 

  149. Phillips JE, Petrie TA, Creighton FP, García AJ. Human mesenchymal stem cell differentiation on self-assembled monolayers presenting different surface chemistries. Acta Biomater. 2010;6(1):12–20.

    Article  Google Scholar 

  150. Dhaliwal A, Lam J, Maldonado M, Lin C, Segura T. Extracellular matrix modulates non-viral gene transfer to mouse mesenchymal stem cells. Soft Matter. 2012;8(5):1451–9.

    Article  Google Scholar 

  151. Tierney EG, Duffy GP, Hibbitts AJ, Cryan SA, O’Brien FJ. The development of non-viral gene-activated matrices for bone regeneration using polyethyleneimine (PEI) and collagen-based scaffolds. J Control release. 2012;158(2):304–11.

    Article  Google Scholar 

  152. Tierney EG, McSorley K, Hastings CL, et al. High levels of ephrinB2 over-expression increases the osteogenic differentiation of human mesenchymal stem cells and promotes enhanced cell mediated mineralisation in a polyethyleneimine-ephrinB2 gene-activated matrix. J Control release. 2013;165(3):173–82.

    Article  Google Scholar 

  153. Lee YH, Wu HC, Yeh CW, et al. Enzyme-crosslinked gene-activated matrix for the induction of mesenchymal stem cells in osteochondral tissue regeneration. Acta Biomater. 2017;63:210–26.

    Article  Google Scholar 

  154. Zhan X. Effect of matrix stiffness and adhesion ligand density on chondrogenic differentiation of mesenchymal stem cells. J Biomed Mater Res part A. 2020;108(3):675–83.

    Article  Google Scholar 

  155. Mohammed M, Lai TS, Lin HC. Substrate stiffness and sequence dependent bioactive peptide hydrogels influence the chondrogenic differentiation of human mesenchymal stem cells. J Mater Chem part B. 2021;9(6):1676–85.

    Article  Google Scholar 

  156. Zhou Y, Qiu J, Wan L, Li J. The effect of matrix stiffness on the chondrogenic differentiation of mesenchymal stem cells. J Mol Histol. 2022;53(5):805–16.

    Article  Google Scholar 

  157. Chu C, Kong H. Interplay of cell adhesion matrix stiffness and cell type for non-viral gene delivery. Acta Biomater. 2012;8(7):2612–9.

    Article  Google Scholar 

  158. Hadi A, Rastgoo A, Haghighipour N, Bolhassani A, Asgari F, Soleymani S. Enhanced gene delivery in tumor cells using chemical carriers and mechanical loadings. PLoS One. 2018;13(12):e0209199.

    Article  Google Scholar 

  159. Joseph JG, Liu AP. Mechanical Regulation of Endocytosis: New Insights and Recent Advances. Adv. Biosyst. 4(5) (2020).

  160. Ferguson JP, Huber SD, Willy NM, et al. Mechanoregulation of clathrin-mediated endocytosis. J Cell Sci. 2017;130(21):3631–6.

    Google Scholar 

  161. Sinha B, Köster D, Ruez R, et al. Cells respond to mechanical stress by rapid disassembly of caveolae. Cell. 2011;144(3):402–13.

    Article  Google Scholar 

  162. Lim SM, Trzeciakowski JP, Sreenivasappa H, Dangott LJ, Trache A. RhoA-induced cytoskeletal tension controls adaptive cellular remodeling to mechanical signaling. Integr Biol (Camb). 2012;4(6):615–27.

    Article  Google Scholar 

  163. Fletcher DA, Mullins RD. Cell mechanics and the cytoskeleton. Nature. 2010;463(7280):485–92.

    Article  Google Scholar 

  164. Kaazempur Mofrad MR, Abdul-Rahim NA, Karcher H, Mack PJ, Yap B, Kamm RD. Exploring the molecular basis for mechanosensation, signal transduction, and cytoskeletal remodeling. Acta Biomater. 2005;1(3):281–93.

    Article  Google Scholar 

  165. Vining KH, Mooney DJ. Mechanical forces direct stem cell behaviour in development and regeneration. Nat Rev Mol cell Biol. 2017;18(12):728–42.

    Article  Google Scholar 

  166. Martino F, Perestrelo AR, Vinarský V, Pagliari S, Forte G. Cellular Mechanotransduction: From Tension to Function. Front. Physiol. 2018;9.

  167. Jaalouk DE, Lammerding J. Mechanotransduction gone awry. Nat Rev Mol cell Biol. 2009;10(1):63–73.

    Article  Google Scholar 

  168. Geiger RC, Taylor W, Glucksberg MR, Dean DA. Cyclic stretch-induced reorganization of the cytoskeleton and its role in enhanced gene transfer. Gene Ther. 2006;13(8):725–31.

    Article  Google Scholar 

  169. Taylor W, Gokay KE, Capaccio C, Davis E, Glucksberg M, Dean DA. The effects of cyclic stretch on gene transfer in alveolar epithelial cells. Mol Ther. 2003;7(4):542–9.

    Article  Google Scholar 

  170. Kaufman CD, Geiger RC, Dean DA. Electroporation- and mechanical ventilation-mediated gene transfer to the lung. Gene Ther. 2010;17(9):1098–104.

    Article  Google Scholar 

  171. Harris SS, Giorgio TD. Convective flow increases lipoplex delivery rate to in vitro cellular monolayers. Gene Ther. 2005;12(6):512–20.

    Article  Google Scholar 

  172. Takeda KM, Yamasaki Y, Dirisala A, et al. Effect of shear stress on structure and function of polyplex micelles from poly(ethylene glycol)-poly(l-lysine) block copolymers as systemic gene delivery carrier. Biomaterials. 2017;126:31–8.

    Article  Google Scholar 

  173. Nikukar H, Reid S, Tsimbouri PM, Riehle MO, Curtis ASG, Dalby MJ. Osteogenesis of mesenchymal stem cells by nanoscale mechanotransduction. ACS Nano. 2013;7(3):2758–67.

    Article  Google Scholar 

  174. Yang HN, Park JS, Jeon SY, Park KH. Carboxymethylcellulose (CMC) formed nanogels with branched poly(ethyleneimine) (bPEI) for inhibition of cytotoxicity in human MSCs as a gene delivery vehicles. Carbohydr Polym. 2015;122(1):265–75.

    Article  Google Scholar 

  175. Curtin CM, Cunniffe GM, Lyons FG, et al. Innovative collagen nano-hydroxyapatite scaffolds offer a highly efficient non-viral gene delivery platform for stem cell-mediated bone formation. Adv Mater. 2012;24(6):749–54.

    Article  Google Scholar 

  176. Elangovan S, D’Mello SR, Hong L, et al. The enhancement of bone regeneration by gene activated matrix encoding for platelet derived growth factor. Biomaterials. 2014;35(2):737–47.

    Article  Google Scholar 

  177. Acri TM, Laird NZ, Geary SM, Salem AK, Shin K. Effects of calcium concentration on nonviral gene delivery to bone marrow-derived stem cells. J Tissue Eng Regen Med. 2019;13(12):2256–65.

    Article  Google Scholar 

  178. Raftery RM, Walsh DP, Blokpoel Ferreras L, et al. Highly versatile cell-penetrating peptide loaded scaffold for efficient and localised gene delivery to multiple cell types: From development to application in tissue engineering. Biomaterials. 2019;216: 119277.

    Article  Google Scholar 

  179. Gonzalez-Fernandez T, Sathy BN, Hobbs C, et al. Mesenchymal stem cell fate following non-viral gene transfection strongly depends on the choice of delivery vector. Acta Biomater. 2017;55:226–38.

    Article  Google Scholar 

  180. Shao D, Wang C, Sun Y, Cui L. Effects of oral implants with miR-122-modified cell sheets on rat bone marrow mesenchymal stem cells. Mol Med Rep. 2018;17(1):1537–44.

    Google Scholar 

  181. Li Y, Fan L, Liu S, et al. The promotion of bone regeneration through positive regulation of angiogenic–osteogenic coupling using microRNA-26a. Biomaterials. 2013;34(21):5048–58.

    Article  Google Scholar 

  182. Li Q, Hu Z, Rong X, Chang B, Liu X. Multifunctional polyplex micelles for efficient microRNA delivery and accelerated osteogenesis. Nanoscale. 2021;13(28):12198–211.

    Article  Google Scholar 

  183. Menciá Castanõ I, Curtin CM, Duffy GP, O’Brien FJ. Next generation bone tissue engineering: non-viral miR-133a inhibition using collagen-nanohydroxyapatite scaffolds rapidly enhances osteogenesis. Sci Rep. 2016;6(1):1–10.

    Article  Google Scholar 

  184. Wu G, Feng C, Hui G, et al. Improving the osteogenesis of rat mesenchymal stem cells by chitosan-based-microRNA nanoparticles. Carbohydr Polym. 2016;138:49–58.

    Article  Google Scholar 

  185. Raisin S, Morille M, Bony C, Noël D, Devoisselle JM, Belamie E. Tripartite polyionic complex (PIC) micelles as non-viral vectors for mesenchymal stem cell siRNA transfection. Biomaterials. 2017;5(9):1910–21.

    Article  Google Scholar 

  186. Chen J, Chen H, Li P, et al. Simultaneous regeneration of articular cartilage and subchondral bone in vivo using MSCs induced by a spatially controlled gene delivery system in bilayered integrated scaffolds. Biomaterials. 2011;32(21):4793–805.

    Article  Google Scholar 

  187. Ye Y, Jin R, Hu X, Zhuang J, Xia W, Lin C. Bioreducible poly(urethane amine)s for robust nucleic acid transfection in stem cells. Biomater Sci. 2019;7(8):3510–8.

    Article  Google Scholar 

  188. Sun XD, Jeng L, Bolliet C, Olsen BR, Spector M. Non-viral endostatin plasmid transfection of mesenchymal stem cells via collagen scaffolds. Biomaterials. 2009;30(6):1222–31.

    Article  Google Scholar 

  189. Shan CL, Huang B, You J, et al. High efficiency intracellular transport of cationic peptide stearate for gene delivery in tumor cells and multipotent stem cells. J Biomed Nanotechnol. 2014;10(11):3231–43.

    Article  Google Scholar 

  190. Khalil AS, Yu X, Dang PN, Alsberg E, Murphy WL. A microparticle approach for non-viral gene delivery within 3D human mesenchymal stromal cell aggregates. Acta Biomater. 2019;95:408–17.

    Article  Google Scholar 

  191. Nguyen MK, Jeon O, Krebs MD, Schapira D, Alsberg E. Sustained localized presentation of RNA interfering molecules from in situ forming hydrogels to guide stem cell osteogenic differentiation. Biomaterials. 2014;35(24):6278–86.

    Article  Google Scholar 

  192. Li Y, Yang C, Khan M, et al. Nanostructured PEG-based hydrogels with tunable physical properties for gene delivery to human mesenchymal stem cells. Biomaterials. 2012;33(27):6533–41.

    Article  Google Scholar 

  193. Yang HY, Van Ee RJ, Timmer K, et al. A novel injectable thermoresponsive and cytocompatible gel of poly(N-isopropylacrylamide) with layered double hydroxides facilitates siRNA delivery into chondrocytes in 3D culture. Acta Biomater. 2015;23:214–28.

    Article  Google Scholar 

  194. Ledo AM, Senra A, Rilo-Alvarez H, et al. mRNA-activated matrices encoding transcription factors as primers of cell differentiation in tissue engineering. Biomaterials. 2020;247(1): 120016.

    Article  Google Scholar 

  195. Shido R, Sumita Y, Hara M, et al. Gene-activated matrix harboring a miR20a-expressing plasmid promotes rat cranial bone augmentation. Regen Biomater. 2021;8(2):rbaa060.

    Article  Google Scholar 

  196. Nicholas Dias, Yung Peng RK. Controlled and sustained delivery of siRNA/NPs from hydrogels expedites bone fracture healing. Physiol Behav. 2017;176(3):139–148.

  197. Munsell EV, Kurpad DS, Freeman TA, Sullivan MO. Histone-targeted gene transfer of bone morphogenetic protein-2 enhances mesenchymal stem cell chondrogenic differentiation. Acta Biomater. 2018;71:156–67.

    Article  Google Scholar 

  198. Tidd N, Michelsen J, Hilbert B, Quinn JC. Minicircle mediated gene delivery to canine and equine mesenchymal stem cells. Int J Mol Sci. 2017;18(4):819.

    Article  Google Scholar 

  199. Bai H, Lester GMS, Petishnok LC, Dean DA. Cytoplasmic transport and nuclear import of plasmid DNA. Biosci Rep. 2017;37(6) .

  200. Carballo-Pedrares N, Fuentes-Boquete I, Díaz-Prado S, Rey-Rico A. Hydrogel-based localized nonviral gene delivery in regenerative medicine approaches—an overview. Pharmaceutics. 2020;12(8):752.

    Article  Google Scholar 

  201. King WJ, Kouris NA, Choi S, Ogle BM, Murphy WL. Environmental parameters influence non-viral transfection of human mesenchymal stem cells for tissue engineering applications. Cell Tissue Res. 2012;347(3):689–99.

    Article  Google Scholar 

  202. Chakka LRJ, Vislisel J, Vidal CMP, Biz MT, Salem AK, Cavalcanti BN. Application of BMP-2/FGF-2 gene–activated scaffolds for dental pulp capping. Clin Oral Investig. 2020;24(12):4427–37.

    Article  Google Scholar 

  203. Hacobian ARA, Posa-Markaryan K, Sperger S, et al. Improved osteogenic vector for non-viral gene therapy. Eur Cell Mater. 2016;31:191–204.

    Article  Google Scholar 

  204. Hausburg F, Müller P, Voronina N, Steinhoff G, David R. Protocol for MicroRNA transfer into adult bone marrow-derived hematopoietic stem cells to enable cell engineering combined with magnetic targeting. J Vis Exp. 2018;2018(136):57474.

    Google Scholar 

  205. Foroozandeh P, Aziz AA. Insight into Cellular Uptake and Intracellular Trafficking of Nanoparticles. Nanoscale Res Lett. 2018;13(1) .

  206. Castaño IM, Raftery RM, Chen G, et al. Rapid bone repair with the recruitment of CD206 + M2-like macrophages using non-viral scaffold-mediated miR-133a inhibition of host cells. Acta Biomater. 2020;109:267–79.

    Article  Google Scholar 

  207. Castaño IM, Curtin CM, Shaw G, Mary Murphy J, Duffy GP, O’Brien FJ. A novel collagen-nanohydroxyapatite microRNA-activated scaffold for tissue engineering applications capable of efficient delivery of both miR-mimics and antagomiRs to human mesenchymal stem cells. J Control Release. 2015;200:42–51.

    Article  Google Scholar 

  208. Xiang S, Tong H, Shi Q, et al. Uptake mechanisms of non-viral gene delivery. J Control release. 2012;158(3):371–8.

    Article  Google Scholar 

  209. Kelly AM, Plautz SA, Zempleni J, Pannier AK. Glucocorticoid cell priming enhances transfection outcomes in adult human mesenchymal stem cells. Mol Ther. 2016;24(2):331–41.

    Article  Google Scholar 

  210. Hamann A, Broad K, Nguyen A, Pannier AK. Mechanisms of unprimed and dexamethasone-primed nonviral gene delivery to human mesenchymal stem cells. Biotechnol Bioeng. 2019;116(2):427–43.

    Article  Google Scholar 

  211. Ciftci K, Levy RJ. Enhanced plasmid DNA transfection with lysosomotropic agents in cultured fibroblasts. Int J Pharm. 2001;218(1–2):81–92.

    Article  Google Scholar 

  212. Oieni J, Lolli A, D’Atri D, et al. Nano-ghosts: Novel biomimetic nano-vesicles for the delivery of antisense oligonucleotides. J Control Release. 2021;333:28–40.

    Article  Google Scholar 

  213. Stoddart MJ, Louis KS, Siegel AC, Kupcsik L, Czekanska EM. Chapters 1–5: Mammalian cell viability. Methods Mol Biol. 2011;740:1–27.

    Article  Google Scholar 

  214. Präbst K, Engelhardt H, Ringgeler S, Hübner H. Basic colorimetric proliferation assays: MTT, WST, and Resazurin. Methods Mol Biol. 2017;1601:1–17.

    Article  Google Scholar 

  215. Raup A, Jérôme V, Freitag R, Synatschke C V., Müller AHE. Promoter, transgene, and cell line effects in the transfection of mammalian cells using PDMAEMA-based nano-stars. Biotechnol. reports (Amsterdam, Netherlands). 2016;11, 53–61 .

  216. Shcharbin D, Pedziwiatr E, Blasiak J, Bryszewska M. How to study dendriplexes II: transfection and cytotoxicity. J Control Release. 2010;141(2):110–27.

    Article  Google Scholar 

  217. Marjanovič I, Kandušer M, Miklavčič D, Keber MM, Pavlin M. Comparison of flow cytometry, fluorescence microscopy and spectrofluorometry for analysis of gene electrotransfer efficiency. J Membr Biol. 2014;247(12):1259–67.

    Article  Google Scholar 

  218. Khorsand B, Elangovan S, Hong L, Dewerth A, Kormann MSD, Salem AK. A comparative study of the bone regenerative effect of chemically modified RNA encoding BMP-2 or BMP-9. AAPS J. 2017;19(2):438–46.

    Article  Google Scholar 

  219. Aydin S. A short history, principles, and types of ELISA, and our laboratory experience with peptide/protein analyses using ELISA. Peptides. 2015;72:4–15.

    Article  Google Scholar 

  220. Arshi A, Petrigliano FA, Williams RJ, Jones KJ. Stem cell treatment for knee articular cartilage defects and osteoarthritis. Curr Rev Musculoskelet Med. 2020;13(1):20–7.

    Article  Google Scholar 

  221. Madry H, Cucchiarini M. Gene therapy for human osteoarthritis: Principles and clinical translation. Expert Opin Biol Ther. 2016;16(3):331–46.

    Article  Google Scholar 

  222. Almalki SG, Agrawal DK. Key transcription factors in the differentiation of mesenchymal stem cells. Differentiation. 2016;92(1–2):41–51.

    Article  Google Scholar 

  223. Bonadio J, Smiley E, Patil P, Goldstein S. Localized, direct plasmid gene delivery in vivo: prolonged therapy results in reproducible tissue regeneration. Nat Med. 1999;5(7):753–9.

    Article  Google Scholar 

Download references

Acknowledgements

We thank Biobanco de A Coruña from SERGAS for providing biological samples

Funding

ARR acknowledges MICINN [PID2021-128461OB-100; RYC2018-025617-I;] and Xunta de Galicia [ED431F2021/10] for the funding. JLS acknowledges MICINN [FPU20/06176] for her pre-doctoral fellowship grant and NCP acknowledges Inditex-UDC 2021grant for her pre-doctoral stay funding.

Author information

Authors and Affiliations

Authors

Contributions

NCP – Natalia Carballo-Pedrares; FP – Federica Ponti; JLS – Junquera López-Seijas; DMB – Diego Miranda-Balbuena; NB – Nina Bono; GC – Gabriele Candiani; ARR – Ana Rey-Rico. NCP and FP contributed equally to this work. Conceptualization: GC, ARR. Methodology: NCP, FP, GC, ARR. Experimental assays: NCP, JLS, DMB. Writing—original draft: NCP, FP, JLS, DMB, NB, GC, ARR. Writing—review & editing: NB, GC, ARR. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Gabriele Candiani or Ana Rey-Rico.

Ethics declarations

Ethics approval and consent to participate

Human bone marrow aspirates were obtained from the proximal femur of patients undergoing hip arthroplasty (n = 2) provided by the Biobanco of A Coruña from SERGAS. The study was approved by the Comité de Ética de Investigación da Coruña (CEIC) (25 March 2019, 2021/425). All patients provided informed consent before inclusion in the study.

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.

Supplementary Information

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

Carballo-Pedrares, N., Ponti, F., Lopez-Seijas, J. et al. Non-viral gene delivery to human mesenchymal stem cells: a practical guide towards cell engineering. J Biol Eng 17, 49 (2023). https://doi.org/10.1186/s13036-023-00363-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13036-023-00363-7

Keywords