Skip to main content

Cell surface engineering and application in cell delivery to heart diseases

Abstract

Cell-based therapy has expanded its influence in cancer immunotherapy, regenerative medicine, and tissue engineering. Due to their secretory functions, differentiation capabilities, specific homing effects through chemotaxis, distinctive therapeutic potentials, and ex vivo expandability, cells have become an attractive reagent for advanced therapeutic strategies. Therefore, the ability to modify cells and manipulate their functions according to intended therapeutic designs has been the central scientific interest in the field of biomedical research. Many innovative methods have been developed with genetic modification of cells being the most advanced cell surface engineering technique. Although genetic modification is a powerful tool, it has a limited applicability due to the permanent modifications made on cells. Alternatively, many endeavors have been made to develop surface engineering techniques that can circumvent the limitations of genetic modification. In this review, current methods of non-genetic cell surface modification, including chemical conjugations, polymeric encapsulation, hydrophobic insertion, enzymatic and metabolic addition, will be introduced. Moreover, cell surface engineering plausible for cardiac remodeling and the future prospective will be discussed at the end.

Background

Cell surface engineering to provide new characteristics and functions to cells has drawn continuous interest from researchers in biomedical science as cell therapy has emerged as a prominent therapeutic strategy equivalent and complementary to the conventional therapeutic approaches. Research endeavors over the past several decades have identified various types of cells as suitable living drugs and versatile drug carriers. In particular, stem cells, including mesenchymal stem cells (MSCs), hematopoietic stem cells (HSCs), and induced pluripotent stem cells (iPSCs), and immune cells, such as T-cells and Natural Killer (NK) cells, have been favored candidates for regenerative medicine and cell-based cancer immunotherapy, respectively. Both bone marrow-derived and adipose-derived MSCs readily isolated from the body are able to release cytokines and growth factors that can be utilized towards wound healing, treating cardiovascular diseases, and correcting neurological disorders [1,2,3,4,5,6,7]. Adoptive transfer of ex vivo cultured and activated immune cells isolated from cancer patients has shown refreshing clinical results [8, 9]. Unfortunately, these breakthrough discoveries in both regenerative medicine and cancer immunotherapy using cells as therapeutic reagents soon faced a common problem: the inability to control cellular functions to maximize the therapeutic benefits. MSCs directly injected into the myocardium showed low retention rate with only 0.44% of the transplanted MSCs remaining in the myocardium after 4 days of administration [10]. Moreover, systemic injection of MSCs on rat myocardial infarction (MI) models revealed less than 1% accumulation of MSCs in the ischemic myocardium [11]. To overcome the low retention rates and enhance the target homing effect, MSCs were genetically engineered to overexpress CXC chemokine receptor 4 (CXCR4), a receptor for stromal-derived factor-1 (SDF-1) expressed in injured myocardium [12]. The resulting genetically modified MSCs showed enhanced target homing effect and greater retention rate in the ischemic myocardium after the intravenous delivery. The developmental story of cell-based cancer immunotherapy is not so different from MSCs in regenerative medicine. Although the efficacy of adoptive transfer of tumor infiltrating lymphocytes (TILs) was examined over several decades, genetically engineered T cells expressing chimeric antigen receptors (CARs) rapidly replaced the application of TILs due to their high specificity, non-MHC-restricted recognition of tumor antigen, superior potency, and improved in vivo persistency [9, 13, 14].

Early attempts to control the cellular interactions and reprogramming the cellular functions focused on the ex vivo preconditioning [15, 16]. In this method, multiple stimuli, including pharmacological agents, cytokines, stimulatory ligands, and/or microenvironmental preconditioning, are challenged to the cells of interest in order to achieve enhanced cell survival, differentiation, paracrine effects, specificity, potency, and target homing effect. For instance, hypoxic conditioning increased the expression of pro-survival and pro-angiogenic factors on MSCs and improved their potential to repair the injured myocardium [17, 18]. Many ex vivo immune cell expansion and activation protocols also require addition of cytokines, such as interleukin (IL)-2, IL-12, IL-15, IL-18, and IL-2, to the culture media [15, 19]. Although preconditioning methods improved the in vivo cell retention and survival, they only allowed minimal gain of control to manipulate the cellular functions that is necessary to redirect cells for therapeutic purposes. As cell therapy continues to evolve, preconditioning methods have been integrated as essential protocols for the growth and maintenance of cells cultured in ex vivo conditions, and many creative methods have been developed to improve the therapeutic feasibility and effectiveness of cells.

Genetic engineering, currently the state-of-the-art modification techniques, has opened up new avenues to tailor preexisting cells to acquire specific therapeutic functions. The most celebrated example is the aforementioned CAR-T cells. Recently, the United States Food and Drug Administration (FDA) approved two CAR-T cells, Kymriah™ and Yescarta™, for the treatment of B cell precursor acute lymphoblastic leukemia (BCP-ALL) and large B cell lymphoma [20]. Both CAR-T cells are engineered to express CARs specific for CD19 expressed on normal and malignant B lineage cells. Genetic engineering also extends its application to modify MSCs by overexpressing receptors and proteins for regenerative medicine: CXCR4 to take advantage of SDF-1 chemotaxis; fibroblast growth factor-2 (FGF2) for improved viability after transplantation into injured myocardium; heme oxygenase-1 (HO-1) to improve cell survival, organ recovery, and function in injured heart; and vascular endothelial growth factor (VEGF) for angiogenesis and inhibition of progression of left ventricular hypertrophy [21, 22]. Undoubtedly, genetic engineering is a powerful tool to control the cellular function of cells; however, it has several drawbacks requiring profound consideration for incorporation into the therapeutic designs. The major drawback is the use of viral vectors to deliver therapeutic genes into the cells of interest [21, 23,24,25,26]. Viral vectors have higher risk of genetic integration that may lead to tumorigenesis and trigger immunogenic response [27]. Additional features introduced to cells through viral genetic engineering are permanent and irreversible, exacerbating the safety risk in clinical settings [28, 29]. Non-viral gene carriers alleviate the safety concerns; however, they show rather low transfection efficiency compared to viral vectors [30]. Because the success of genetic engineering heavily depends on the transduction/transfection efficiency, the resulting modified cells may show inconsistent and unpredictable therapeutic efficacy. This is because genetic engineering is not applicable to all types of cells, especially stem cells and slowly dividing cells.

Alternative to genetic engineering, non-genetic cell surface engineering techniques, such as covalent conjugation [31,32,33,34], electrostatic interactions [35,36,37], hydrophobic insertion [38,39,40,41,42,43], offer more transient and reversible modifications to control cellular functions. Instead of manipulating cells at the gene and protein level, these techniques modify the cell using the characteristics of lipids, proteins, and glycans present in the cell membrane [5, 29, 44, 45]. Because those are essential components for cells, non-genetic surface engineering techniques can potentially be applied to a broad range of cells from different origins. Through non-genetic cell surface engineering, biomaterials including proteins, surface receptors, antibodies, peptides, genetic materials, and protective polymers, have been used to endow specific functions to cells [31, 33, 34, 42, 44, 46,47,48]. Research areas that have benefitted from these cell surface modifications include (1) investigation of adding new functions, (2) reducing graft rejection for transplantation by masking the surface antigens, (3) creation of heterogeneous cluster of cells by cell-to-cell attachment, (4) enhancing immune effector functions, and (5) programing cell-to-cell interactions.

Ideal cell surface engineering methods should provide control over the fate and function of the modified cells without interfering with cell survival, proliferation, and cellular activities. Therefore, this review attempts to provide a concise guide on cell surface engineering techniques that meet the purpose of modifying the cell surface properties. The first section summarizes each type of non-genetic cell surface engineering technique with application on different cell types. In the following section, challenges and considerations of engineering the surface of living cells are discussed. Finally, example of cell surface engineering technique is presented as a promising method to redirect MSCs for cardiac diseases.

Non-genetic Surface Engineering

Covalent Conjugation

Covalent conjugation chemically, metabolically, or enzymatically attaches bioactive substances to the cell membrane [31, 33, 34, 48,49,50,51]. Chemical conjugation is the most straightforward method that takes advantage of surface-exposed functional groups on the membrane proteins as grafting points. Currently, N-hydroxyl-succinimidyl ester (NHS) groups [31, 33, 34, 48], maleimide [51], and pyridyldithiol [52, 53] are the most frequently used chemical cross-linkers (Fig. 1). The use of NHS-activated esters modifies exposed amine groups on the surface of bioactive molecules. Maleimide conjugated biomolecules can be selectively attached to surface-exposed thiol group, generating a non-cleavable thio-ether bond. In case degradable conjugation is desired, pyridyldithiol modified biomolecules can be attached to free thiols on the surface to create reducible disulfide bonds. The key advantage of chemical conjugation is the broad applicability. Biomaterials functionalized with cross-linkers can be used to modify variety of cells. Unlike the random modification through chemical conjugation, metabolic and enzymatic conjugation methods provide more selective attachment of biomaterials. Saxon et al. and Prescher et al. reported the use of sophisticated metabolic surface modification that takes advantage of unnatural sialic acid biosynthesis [49, 50]. Human cells undergo unnatural sialic acid biosynthesis when exposed to unnatural sugar N-α-azidoacetylmannosamine (ManNAz), an analog of the native sugar N-acetylmannosamine (Fig. 1). This process incorporates N-α-azidoacetyl sialic acid (SiaNAz), a metabolite of ManNAz, to the membrane glycoconjugates. The added azide groups further provide attachment points for biomaterials through Staudinger ligation [49, 50] or click-chemistry [54, 55]. Similar to metabolic conjugation, enzymatic conjugation also provides covalent attachment of biomaterials on a designated spot on the cell surface. As reported by Swee et al., transpeptidase sortase A from Staphylococcus aureus efficiently conjugates peptides or proteins with LPETG-motif to the N-terminal glycine exposed on the surface of different types of cells (Fig. 1) [56]. Although conjugated biomaterials gradually disappear over time, modifications installed through covalent conjugation are stable compare to other non-genetic surface engineering methods [33, 34, 40]. Moreover, the degree of modification is difficult to control with covalent conjugation, and higher degree of modification using bioactive molecules, both small or large, may cause significant physiological alterations, such as reduction of membrane mobility and diffusion kinetics to the modified cells [38, 44, 57].

Fig. 1
figure 1

Modes of non-genetic cell surface engineering techniques. (1) Incorporation of cross-linkers, such as NHS, Maleimide, or pyridyldithiol, allows cell surface modification with biomaterials through chemical covalent conjugation. Cell metabolism of unnatural sugar and enzymatic reactions can be exploited to attach functional groups on the cell surface. (2) Electrostatic interactions between the cell surface and the charged polymers such as PEI, PLL, PAA, and PSS can modify cells through layer-by-layer technique. Also, charged block-co-polymers, such as PLL-PEG, can modify the cell surface through electrostatic interaction. (3) Lipid-conjugated bioactive molecules or polymers with long alkyl chains can be embedded into the cell membrane through hydrophobic interaction. Abbreviations: NHS: N-hydroxyl-succinimidyl ester; ManNAz: N-α-azidoacetylmannosamine; PAA: Poly(acrylic acid); PEG: Poly(ethylene glycol); PEI: Poly(ethyleneimine); PLL: Poly-L-lysine; PSS: Poly(styrene) sulfate; PVA: Poly(vinyl alcohol); SiaNAz: N-α-azidoacetyl sialic acid

Electrostatic Interaction

Electrostatic interactions modify the cell surface by establishing self-assembled structures between the negatively charged cell surface and cationic polymers (Fig. 1). Cells initially modified with cationic polymers can be engineered again via a layer-by-layer technique by sequentially applying anionic and cationic polymers [35, 37, 58,59,60]. Because modified cells encapsulated by multiple polymeric layers can reduce molecular recognition, the electrostatic layer-by-layer approach has been often investigated in the cell transplantation research [37, 59]. Many cationic/anionic polymers and poly electrolytes, such as poly-L-lysine (PLL), poly(styrene) sulfate (PSS), poly(allylamine hydrochloride) (PAH), poly(diallydimethylammonium) chloride (PDADMAC or PDDA), poly(ethyleneimine) (PEI), polyphosphoric acid (PPP), and poly(acrylic acid) (PAA), and hyaluronic acid (HA) have been used to generate multiple layers on the cell membrane [35, 37, 58,59,60,61]. Thickness of the polymer layer can be controlled by changing the number of layers and the new surface properties of the modified cells rely on the polymer characteristics of the outermost layer. However, it should be noted that high charge density of cationic polymers significantly reduces the viability of modified cells [58, 62, 63]. To improve the cell viability after the surface modification, PLL-graft-poly(ethylene glycol) (PLL-g-PEG) can be introduced to coat the surface cells [59, 64]. Surface modification of PLL-g-PEG was further developed to incorporate functional groups, such as biotin, hydrazide, and azide, to capture streptavidin, aldehyde, and cyclooctyne [64]. The main advantage of surface engineering through electrostatic interaction is that cells are protected from the sheer stress and immune response by the non-invasive encapsulation. Biocompatibility of cationic polymers, however, should be resolved in order to be used in cell therapy.

Hydrophobic insertion

Amphiphilic polymers polymerized with long alkyl chains, such as phospholipid-conjugated PEGs and poly(vinyl alcohol) (PVA), provide noninvasive modifications of the cell surface through hydrophobic interaction (Fig. 1). Similarly, a large number of different cell types have been modified via hydrophobic interaction with lipid-conjugated biomaterials for specific function [38,39,40,41,42,43, 57, 65,66,67,68,69,70]. Most lipophilic membrane dyes currently available in the market, such as Dil, DiD, DiR, and DiO, are developed upon cell surface modification through hydrophobic interaction. Interaction of lipid-conjugated PEGs with lipid bilayers was examined by Yamamoto et al. using surface plasmon resonance (SPR) spectroscopy [71]. Lipids with different lengths of alkyl chains—1,2-dimyristoyl-sn-glycerol-3-phosphatidylethanolamine (DMPE, 14 carbons), 1,2-dipalmitoyl-sn-glycerol-3-phosphatidylethanolamine (DPPE, 16 carbons), and 1,2-distearoyl-sn-glycerol-3-phosphatidylethanolamine (DSPE, 18 carbons)—were conjugated with PEG (5 kDa) and applied onto the lipid bilayer. Out of all lipid-PEG conjugates, DMPE showed the most rapid incorporation to the membrane. Insertion of DPPE showed concentration-dependent behavior; however, incorporation of DSPE was only observed at high concentration. Dissociation of DMPE was more rapid compared to DPPE when modified lipid bilayer was washed with PBS. No dissociation was observed once DSPE was incorporated into the membrane. Thus, it was noted that longer hydrophobic chains reduce the incorporation rate and the dissociation rate of lipid molecules [71]. Interestingly, fluorescence of FITC-labeled lipid-PEGs was recovered in a few minutes in fluorescent recovery after photobleaching (FRAP) assay [71]. This observation indicates that lipid-PEGs embedded in the lipid bilayer were able to diffuse laterally within the lipid bilayer. Unlike covalent conjugation and electrostatic interaction, surface modification with hydrophobic insertion allows membrane-anchored bioactive molecules to participate in the dynamic movement of cell membrane. Most importantly, cells modified with lipid-conjugated biomaterials showed negligible toxicity, and the modified cells resumed normal cellular activities [65,66,67]. Instead of preparing the lipid-conjugatedmolecules, modification of cell surface can be achieved by liposomal fusion strategy [72]. Because liposomes are vesicles composed of lipids and lipid-conjugated molecules, large sections of the liposomes containing specialized lipids can be incorporated into the membrane without causing severe toxicity [72,73,74,75]. Fate of the lipid-conjugated bioactive molecules has not been fully understood, and the exclusion pathway requires further investigation; however, the endocytosis of membrane-anchored lipid-conjugated biomaterials has not been observed [76]. Lipid-conjugated biomolecules are believed to be released from the cells to the surroundings due to equilibrium differences [40]. Although molecules of interest must be hydrophobized by lipid or alkyl chain conjugation and the retention time on the surface is variable, hydrophobic insertion is an attractive surface engineering technology that offers rapid and nontoxic surface modification to virtually any type of cell.

Challenges and considerations of engineering the surface of living cells

Cell membrane dynamics

Cell membrane is in a dynamic state. It is subjected to undergo constant remodeling where most of its components—lipids and membrane proteins—are internalized, degraded, recycled, and replaced [77, 78]. The rate of these processes is highly dependent on the type of lipids and proteins and varies widely from hours to weeks [79]. Cell membrane lipids and proteins are routinely internalized through endocytosis, pinocytosis, and phagocytosis. Due to their size, type, and property, biomaterials that are chemically conjugated, electrostatically adsorbed, or hydrophobically embedded on the membrane, may internalize mostly through endocytosis [80]. The process of endocytosis is initiated as complementary ligands bind to surface receptors or as bioactive substances are absorbed on the cell membrane [81,82,83]. These events trigger invaginations of small areas containing the receptors and affected regions of cell membrane. Subsequently, the invaginated pockets are closed, and newly formed vesicles are transported to the intracellular compartments. During endocytosis, any molecules and materials on the invaginated cell membrane and in the proximal media will be taken up by the cells, resulting in the loss of desired functions installed via surface engineering. Therefore, surface engineering methods should consider cell membrane dynamics in order to improve the surface residence time of the desired biomaterials for prolonged therapeutic effects.

In vivo system

Unlike the in vitro experimental settings, in vivo environment is an integrated system of many complex mechanical and biochemical interactions. Transplanted or adoptively transferred surface-engineered therapeutic cells are exposed to sheer stress and hemodynamic forces that can strip off the installed surface modification [84]. Migration in the circulation and endothelial transmigration in the tissues, as demonstrated by leukocytes and stem cells, require extensive reshaping of the cell membrane [85, 86]. In the spleen, circulating cells are forced to enter the compact network of sinusoidal capillaries to eliminate damaged and aged cells [87]. In order to compensate for the mechanical stress from the in vivo environment, surface-engineered cells must display unaltered membrane flexibility and elasticity. Surface-engineered cells in blood circulation are also exposed to coagulation factors, the complement immune system, and inflammation mediators that drastically reduce duration of therapeutic effects [88, 89]. Macrophages and monocytes of innate immune defense system are often stimulated in response to the bioactive substances on surface-modified cells and subsequently eliminate them from the body by phagocytosis [90]. Immunogenic biomaterials, such as proteins synthesized from bacterial host and antibodies isolated from animals, are opsonized by neutralizing antibodies and are cleared by the innate immune system and complement activation [91,92,93,94]. Thus, cell surface modification, regardless of the methods employed, must not sacrifice the membrane flexibility and elasticity but rather provide new functionality in addition to the protection against mechanical and biological challenges for clinical applications.

Clinical translation

For clinical translation, surface-engineered cells must satisfy several fundamental principles of biocompatibility. Because cells are the most critical component of cell therapy, any modifications applied to the cell surface should not have detrimental effects on cell viability. At any stage of preparation, cell viability should be maintained by changes in pH, osmolality, temperature, pressure, degree of agitation, and exposure to organic solvent [84]. Surface modification should not become a physical barrier that blocks diffusion of necessary nutrients. This is particularity important for islet cell transplantation, where surface-modified islet cells secrete insulin in response to glucose levels [39, 66, 95]. Unless the purpose for surface engineering is to mask the surface antigens during transplantation or adoptive transfer of immune cells—for the sake of reducing the occurrence of graft-versus-host disease (GVHD)—surface proteins and receptors should be exposed on the surface without hindrance to bind growth factors and ligands to signal cell survival, proliferation, and activation. Moreover, surface engineered biomolecules should not reduce the membrane flexibility and elasticity, which are the essential properties of cell membrane that allows cell adhesion, migration, and signaling [96,97,98,99]. Lastly, the cost of surface engineering cells for therapeutic purposes must be affordable. Genetic engineered cells, such as CAR-T cells, can be finely tuned to provide personalized cell therapy for many cancers and diseases; however, the cost of treatment is extremely expensive, estimated at $25,000 per treatment [100]. Genetically engineered stem cells are also anticipated to be one of the most expensive treatment options. The high cost arises from the labor-intensive and time-consuming certified process to prepare genetically engineered cells. The surface modification methods discussed earlier have the potential to be applied as an alternative technology to genetic engineering and are more economical with rapid preparation of therapeutic cells.

Application of hydrophobic insertion for cardiac diseases

Stem cell delivery for cardiac injury

Cardiac injuries and diseases remain the most common cause of death globally with a higher annual death rate compared to any other causes [101]. The major reason of the highest mortality is that cardiac injury and diseases can progress rapidly, as seen in the cases of acute myocardial infarction (AMI) and MI. Conversely, these cardiac diseases often show lagging progress of cardiac remodeling that frustrates the recovery. Consequently, cardiac hypertrophy and myocardial fibrosis eventually prevail [102,103,104,105,106]. Heart failure and even death may result as a series of catastrophic processes, including cellular injury, mechanical dysfunction, and disruption of structural integrity, occur. Therefore, clinicians and scientists are burdened to develop therapeutic methods to repair and replace the injured cardiomyocytes or associated cells in the infarcted myocardium.

In terms of therapy designed for cardiac injuries and diseases, the treatment options to heal the infarcted cardiac tissue are extremely limited. Currently available therapies for AMI and MI, such as the treatments concentrated on reducing myocardial oxygen needs, extend survival by protecting the remaining cardiomyocytes without addressing the fundamental problem—the loss of cardiomyocytes [107, 108]. Several strategies of cardiac regeneration have emerged from decades of intensive research efforts. Although most of these strategies are still in the early stage, some are beginning to be clinically tested for practicality [109].

Current research in the field of cardiac diseases attempts to stimulate the endogenous regenerative mechanisms via cell-based therapies. Many have believed that regenerative therapies employing stem cells, especially MSCs, have enormous potential for clinical applications to treat cardiac diseases [110]. MSCs, being multipotent stem cells, can differentiate into several cell types, such as mesodermal lineage cells and myogenic lineage [111]. These MSC-based therapies for cardiac diseases are achieved by the intermingling of two major components: a cardiomyocyte source as a target for cardiac regeneration; and a non-myocardial tissue acting as a source for regeneration in an effective cardiac environment [112]. Besides the two major components, other influential factors, such as the type of stem cell being used, its proliferative and differentiation capacity, the targeting to localize the damaged site, the route and site of stem cell transplantation, survival capability of the engrafted cells and so forth, should be carefully tweaked to achieve a successful MSC-based therapy [112].

Regenerative medicine for heart diseases using stem cells has been controversial and readers’ discretion is strongly advised [113,114,115]. One of the most challenged idea is the existence of resident endogenous stem cells or cardiac progenitor cells (CPCs). The current views concur on the fact that c-Kit+ CPCs, once thought to show regenerative functions and ability to replace the lost cardiomyocytes due to the cardiac injury through differentiating into cardiomyocytes, are rare and have minimal cardiomyogenic potential [116]. However, the prevailing view on the rarity of resident endogenous stem cells should not discourage the idea of stem cell therapy towards cardiac injury and diseases since several preclinical have shown improved cardiac function after the transplantation of MSCs into infarcted heart and clinical studies have reported modest benefits for patients with ischemic cardiomyopathy [117,118,119,120] Evidence suggests that these beneficial recovery and protective effects are indirect contributions of MSCs through paracrine signaling [114, 121, 122]. The transplanted MSCs secrete growth factors, microRNA (miRNAs), immunomodulatory signals, and exosomes in order to promote prosurvival mechanism and encourage restorative effects in the injured myocardium [112, 114]. Although clinical results and mechanism of actions have not been clearly shown, it is difficult to preclude the therapeutic benefits of stem cell delivery for heart diseases due to lack of understanding. To reiterate, the ability of any treatment strategies to compensate for the loss of the functioning cardiomyocytes, even though it may not indicate the physical replacement of cardiomyocytes, is the essence of stem cell therapy for cardiac injury. One way to improve the clinical outcome of stem cell therapy is to develop a competent delivery method that can specifically target the disease site within the therapeutic time window. In this aspect, cell surface engineering offers the means to enhance the targeting effect of MSCs, or any discovered therapeutic cells or stem cells, without altering their native functions.

SDF-1/CXCR4 on MSCs

At the ischemic sites, MSCs can secrete arteriogenic cytokines, such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), placental growth factor (PIGF), and monocyte chemoattractant protein-1 (MCP-1), to repair the damaged tissues [123, 124]. Thus, many have strived to design a method that allows intravenously infused MSCs to target the injured myocardium. Unfortunately, a negligible number of MSCs migrated to the ischemic myocardium when a large amount of MSCs were intravenously infused [11]. Poor migration of MSCs is related to the loss of CXCR4 expression [125]. Ex vivo expansion of MSCs is necessary to generate a therapeutically relevant number of cells; however, MSCs express heterogeneous CXCR4 with significantly reduced affinity to their corresponding ligands, SDF-1, during the expansion. Moreover, Rombouts et al. have reported that ex vivo expansion of MSCs results in the loss of CXCR4 expression on MSCs [126]. This effect ultimately reduces the chemotaxis of MSCs along the chemokine gradient to specific sites. Systematic administration of MSCs should therefore be improved with a reliable targeting method to enhance therapeutic efficacy.

Immediately after myocardial infarction, injured cardiomyocytes up-regulate SDF-1 expression to recruit stem cells for repair [127, 128]. Although many studies have stated that migration of CXCR4+ bone marrow stem cells along the SDF-1 concentration gradient is critical for cardiac recovery [127, 129, 130], it has been suggested that the responsiveness to SDF-1 in these cells may mature over 4-7 days after MI [131, 132]. Conversely, expression of SDF-1 in the heart starts to decline 4-7 days after the ischemic injury [127]. Thus, expanding autologous MSCs—which takes several weeks—for the treatment of MI is not ideal due to the shallow therapeutic window of SDF-1 expression.

Previously, CXCR4 expression on MSCs had been induced by hypoxic culture conditions, addition of cytokine cocktails, and viral gene transduction. However, these methods are now discouraged due to the lengthy generation time and risk of altering the MSC properties [12, 133,134,135]. In order to exploit the SDF-1 gradient for targeted delivery of MSCs to the MI site, pre-expanded MSCs should be rapidly modified with the targeting moiety. Cell surface engineering using the hydrophobic insertions provides an excellent solution to enhance the homing of MSCs to the injured myocardium. Because it noninvasively engineers cells and readily modifies the cell membrane with therapeutic molecules containing lipophilic anchors [136], cell modification by hydrophobic insertion allows instantaneous generation of specialized therapeutic MSCs without a detrimental effect. To demonstrate the feasibility, MSCs were surface-engineered with recombinant CXCR4 (rCXCR4) hydrophobized with DMPE-PEGs (Fig. 2) [43]. In less than 10 min of incubating pre-expanded MSCs with rCXCR4-PEG-DMPE, specialized MSCs were generated. These cells exhibited a recovered response to SDF-1 with a two-fold improvement of migration ability toward the concentration gradient of SDF-1. Thus, cell surface engineering of MSCs with rCXCR4-PEG-DMPE will be explored with a focus on approaches that further enhance the therapeutic potential of MSCs for regenerative medicine.

Fig. 2
figure 2

Schematic representation of surface-engineered MSCs for cardiac regeneration. Ex vivo cultured and expanded MSCs were surface engineered through hydrophobic insertion to incorporate rCXCR4 on their membrane. Hydrophobic insertion generated homogeneous MSCs modified with hydrophobized rCXCR4 within a short time. Systemically infused rCXCR4-modified MSCs can migrate to the ischemic myocardium by taking advantage of SDF-1 concentration gradient. Abbreviations: MSCs: mesenchymal stem cells; PEG: Poly(ethylene glycol); rCXCR4: Recombinant CXC chemokine receptor 4; SDF-1: Stromal-derived factor-1

Conclusion

Cell therapy has advanced to the point where it aims to provide treatments for tissue degeneration, chronic inflammation, autoimmunity, genetic disorders, cancer, and infectious diseases [84]. Because the efficacy of cell therapy heavily depends on manipulating the fate and function of therapeutic cells, innovative strategies are continuously being introduced to enhance cell survival, boost native behaviors, add new functions, and improve therapeutic effects. Genetic modification has the advantage of expressing heterologous proteins in cells; however, the expression of desired protein heavily depends on the amount of genetic materials internalized by the cells and the efficiency of protein synthesis of the targeted cells. Nonetheless, viral gene transfer limits the application of genetically modified cells for therapies due to safety and economical concerns, including the use of viral vectors, expensive production cost, and extensive generation time. Non-genetic engineering allows for more creative designs to redirect cells for therapeutic purposes. Both synthetic and natural biomaterials can be incorporated onto the cell surface through covalent conjugation, electrostatic interaction, and hydrophobic interaction in order to provide unique properties and functionalities to cells. Although covalent conjugation and electrostatic interaction provide stable surface modification, the degree of modification is difficult to control. Excessive modification may disrupt the membrane integrity, resulting in severe cytotoxicity. Compared to other surface engineering methods, hydrophobic interaction is a safer membrane modification method that noninvasively modifies the cell surface by inserting lipid-conjugated molecules into the membrane. Despite limited understanding of the fate of lipid conjugated bioactive substances, surface engineering with hydrophobic interaction is an attractive technique because it can be applied to virtually any cell. Non-genetic cell surface engineering to improve their therapeutic potentials is still in its infancy, suggesting each technology should be further tailored to overcome the disadvantage and meet the specific demands of clinical application.

Abbreviations

AMI:

Acute myocardial infarction

BCP-ALL:

B cell precursor acute lymphoblastic leukemia

bFGF:

Basic fibroblast growth factor

CARs:

Chimeric antigen receptors

CD19:

Cluster of differentiation 19

CXCR4:

CXC chemokine receptor 4

DiD:

1,1’-dioctadecyl-3,3,3’,3’- tetramethylindodicarbocyanine, 4-chlorobenzenesulfonate salt

Dil:

(1,1’-dioctadecyl-3,3,3’ ,3’-tetramethylindocarbocyanine perchlorate)

DiO:

3,3’ -dioctadecyloxacarbocyanine, perchlorate

DiR:

1,1’-dioctadecyltetramethyl indotricarbocyanine Iodide

DMPE:

1,2-dimyristoyl-sn-glycerol-3-phosphatidylethanolamine

DPPE:

1,2-dipalmitoyl-sn-glycerol-3-phosphatidylethanolamine

DSPE:

1,2-distearoyl-sn-glycerol-3-phosphatidylethanolamine

FDA:

Food and Drug Administration

FGF2:

Fibroblast growth factor-2

FITC:

Fluorescein isothiocyanate

FRAP:

Fluorescent recovery after photobleaching

GVHD:

Graft-versus-host disease

HA:

Hyaluronic acid

HO-1:

Heme oxygenase-1

HSCs:

Hematopoietic stem cells

IL:

Interleukin

iPSCs:

Induced pluripotent stem cells

ManNAz:

N-α-azidoacetylmannosamine

MCP-1:

Monocyte chemoattractant protein-1

MHC:

Major histocompatibility complex

MI:

Myocardial infarction

MSCs:

Mesenchymal stem cells

NHS:

N-hydroxyl-succinimidyl ester

NK cells:

Natural Killer cells

PAA:

Poly(acrylic acid)

PAH:

Poly(allylamine hydrochloride)

PDADMAC or PDDA:

Poly(diallydimethylammonium) chloride

PEG:

Poly(ethylene glycol)

PEI:

Poly(ethyleneimine)

PIGF:

Placental growth factor (PIGF)

PLL:

Poly-L-lysine

PLL-g-PEG:

PLL-graft-poly(ethylene glycol)

PPP:

Polyphosphoric acid

PSS:

Poly(styrene) sulfate

PVA:

Poly(vinyl alcohol)

rCXCR4:

Recombinant CXC chemokine receptor 4

SDF-1:

Stromal-derived factor-1

SiaNAz:

N-α-azidoacetyl sialic acid

SPR:

Surface plasmon resonance

TILs:

Tumor infiltrating lymphocytes

VEGF:

Vascular endothelial growth factor

References

  1. Han J, Park J, Kim BS. Integration of mesenchymal stem cells with nanobiomaterials for the repair of myocardial infarction. Adv Drug Deliv Rev. 2015;95:15–28.

    Article  Google Scholar 

  2. Lindvall O, Kokaia Z. Stem cells for the treatment of neurological disorders. Nature. 2006;441(7097):1094–6.

    Article  Google Scholar 

  3. Marfia G, Navone SE, Di Vito C, Ughi N, Tabano S, Miozzo M, Tremolada C, Bolla G, Crotti C, Ingegnoli F, et al. Mesenchymal stem cells: potential for therapy and treatment of chronic non-healing skin wounds. Organogenesis. 2015;11(4):183–206.

    Article  Google Scholar 

  4. Murphy KC, Whitehead J, Falahee PC, Zhou D, Simon SI, Leach JK. Multifactorial Experimental Design to Optimize the Anti-Inflammatory and Proangiogenic Potential of Mesenchymal Stem Cell Spheroids. Stem Cells. 2017;35(6):1493–504.

    Article  Google Scholar 

  5. Park J, Andrade B, Seo Y, Kim MJ, Zimmerman SC, Kong H. Engineering the Surface of Therapeutic “Living” Cells. Chem Rev. 2018;118(4):1664–90.

    Article  Google Scholar 

  6. Segers VF, Lee RT. Stem-cell therapy for cardiac disease. Nature. 2008;451(7181):937–42.

    Article  Google Scholar 

  7. Strauer BE, Brehm M, Zeus T, Kostering M, Hernandez A, Sorg RV, Kogler G, Wernet P. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation. 2002;106(15):1913–8.

    Article  Google Scholar 

  8. Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nature reviews Cancer. 2008;8(4):299–308.

    Article  Google Scholar 

  9. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348(6230):62–8.

    Article  Google Scholar 

  10. Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation. 2002;105(1):93–8.

    Article  Google Scholar 

  11. Barbash IM, Chouraqui P, Baron J, Feinberg MS, Etzion S, Tessone A, Miller L, Guetta E, Zipori D, Kedes LH, et al. Systemic delivery of bone marrow-derived mesenchymal stem cells to the infarcted myocardium: feasibility, cell migration, and body distribution. Circulation. 2003;108(7):863–8.

    Article  Google Scholar 

  12. Cheng Z, Ou L, Zhou X, Li F, Jia X, Zhang Y, Liu X, Li Y, Ward CA, Melo LG, Kong D. Targeted migration of mesenchymal stem cells modified with CXCR4 gene to infarcted myocardium improves cardiac performance. Mol Ther. 2008;16(3):571–9.

    Article  Google Scholar 

  13. Yee C. Adoptive T cell therapy: Addressing challenges in cancer immunotherapy. J Transl Med. 2005;3(1):17.

    Article  Google Scholar 

  14. Barrett DM, Grupp SA, June CH. Chimeric Antigen Receptor- and TCR-Modified T Cells Enter Main Street and Wall Street. J Immunol. 2015;195(3):755–61.

    Article  Google Scholar 

  15. Redeker A, Arens R. Improving Adoptive T Cell Therapy: The Particular Role of T Cell Costimulation, Cytokines, and Post-Transfer Vaccination. Front Immunol. 2016;7:345.

    Article  Google Scholar 

  16. Yu SP, Wei Z, Wei L. Preconditioning strategy in stem cell transplantation therapy. Transl Stroke Res. 2013;4(1):76–88.

    Article  Google Scholar 

  17. Hu X, Yu SP, Fraser JL, Lu Z, Ogle ME, Wang JA, Wei L. Transplantation of hypoxia-preconditioned mesenchymal stem cells improves infarcted heart function via enhanced survival of implanted cells and angiogenesis. J Thorac Cardiovasc Surg. 2008;135(4):799–808.

    Article  Google Scholar 

  18. Sart S, Ma T, Li Y. Preconditioning stem cells for in vivo delivery. Biores Open Access. 2014;3(4):137–49.

    Article  Google Scholar 

  19. Granzin M, Wagner J, Kohl U, Cerwenka A, Huppert V, Ullrich E. Shaping of Natural Killer Cell Antitumor Activity by Ex Vivo Cultivation. Front Immunol. 2017;8:458.

    Article  Google Scholar 

  20. Zheng PP, Kros JM, Li J. Approved CAR T cell therapies: ice bucket challenges on glaring safety risks and long-term impacts. Drug Discov Today. 2018;23(6):1175–82.

    Article  Google Scholar 

  21. Hodgkinson CP, Gomez JA, Mirotsou M, Dzau VJ. Genetic engineering of mesenchymal stem cells and its application in human disease therapy. Human gene therapy. 2010;21(11):1513–26.

    Article  Google Scholar 

  22. Nowakowski A, Walczak P, Lukomska B, Janowski M. Genetic Engineering of Mesenchymal Stem Cells to Induce Their Migration and Survival. Stem Cells Int. 2016;2016:4956063.

    Article  Google Scholar 

  23. Kershaw MH, Westwood JA, Darcy PK. Gene-engineered T cells for cancer therapy. Nat Rev Cancer. 2013;13(8):525–41.

    Article  Google Scholar 

  24. Tey SK. Adoptive T-cell therapy: adverse events and safety switches. Clin Transl Immunology. 2014;3(6):e17.

    Article  Google Scholar 

  25. Glienke W, Esser R, Priesner C, Suerth JD, Schambach A, Wels WS, Grez M, Kloess S, Arseniev L, Koehl U. Advantages and applications of CAR-expressing natural killer cells. Front Pharmacol. 2015;6:21.

    Article  Google Scholar 

  26. Nowakowski A, Andrzejewska A, Janowski M, Walczak P, Lukomska B. Genetic engineering of stem cells for enhanced therapy. Acta Neurobiol Exp (Wars). 2013;73(1):1–18.

    Google Scholar 

  27. Nayak S, Herzog RW. Progress and prospects: immune responses to viral vectors. Gene Ther. 2010;17(3):295–304.

    Article  Google Scholar 

  28. Bonifant CL, Jackson HJ, Brentjens RJ, Curran KJ. Toxicity and management in CAR T-cell therapy. Mol Ther Oncolytics. 2016;3:16011.

    Article  Google Scholar 

  29. Csizmar CM, Petersburg JR, Wagner CR. Programming Cell-Cell Interactions through Non-genetic Membrane Engineering. Cell Chem Biol. 2018;25(8):931–40.

    Article  Google Scholar 

  30. Nayerossadat N, Maedeh T, Ali PA. Viral and nonviral delivery systems for gene delivery. Adv Biomed Res. 2012;1:27.

    Article  Google Scholar 

  31. Contreras JL, Xie D, Mays J, Smyth CA, Eckstein C, Rahemtulla FG, Young CJ, Anthony Thompson J, Bilbao G, Curiel DT, Eckhoff DE. A novel approach to xenotransplantation combining surface engineering and genetic modification of isolated adult porcine islets. Surgery. 2004;136(3):537–47.

    Article  Google Scholar 

  32. Stabler CL, Sun XL, Cui W, Wilson JT, Haller CA, Chaikof EL. Surface re-engineering of pancreatic islets with recombinant azido-thrombomodulin. Bioconjug Chem. 2007;18(6):1713–5.

    Article  Google Scholar 

  33. Cabric S, Sanchez J, Lundgren T, Foss A, Felldin M, Kallen R, Salmela K, Tibell A, Tufveson G, Larsson R, et al. Islet surface heparinization prevents the instant blood-mediated inflammatory reaction in islet transplantation. Diabetes. 2007;56(8):2008–15.

    Article  Google Scholar 

  34. Yun Lee D, Hee Nam J, Byun Y. Functional and histological evaluation of transplanted pancreatic islets immunoprotected by PEGylation and cyclosporine for 1 year. Biomaterials. 2007;28(11):1957–66.

    Article  Google Scholar 

  35. Germain M, Balaguer P, Nicolas JC, Lopez F, Esteve JP, Sukhorukov GB, Winterhalter M, Richard-Foy H, Fournier D. Protection of mammalian cell used in biosensors by coating with a polyelectrolyte shell. Biosens Bioelectron. 2006;21(8):1566–73.

    Article  Google Scholar 

  36. Elbert DL, Herbert CB, Hubbell JA. Thin polymer layers formed by polyelectrolyte multilayer techniques on biological surfaces. Langmuir. 1999;15(16):5355–62.

    Article  Google Scholar 

  37. Krol S, del Guerra S, Grupillo M, Diaspro A, Gliozzi A, Marchetti P. Multilayer nanoencapsulation. New approach for immune protection of human pancreatic islets. Nano Lett. 2006;6(9):1933–9.

    Article  Google Scholar 

  38. Rabuka D, Forstner MB, Groves JT, Bertozzi CR. Noncovalent cell surface engineering: incorporation of bioactive synthetic glycopolymers into cellular membranes. J Am Chem Soc. 2008;130(18):5947–53.

    Article  Google Scholar 

  39. Teramura Y, Kaneda Y, Iwata H. Islet-encapsulation in ultra-thin layer-by-layer membranes of poly(vinyl alcohol) anchored to poly(ethylene glycol)-lipids in the cell membrane. Biomaterials. 2007;28(32):4818–25.

    Article  Google Scholar 

  40. Teramura Y, Kaneda Y, Totani T, Iwata H. Behavior of synthetic polymers immobilized on a cell membrane. Biomaterials. 2008;29(10):1345–55.

    Article  Google Scholar 

  41. Totani T, Teramura Y, Iwata H. Immobilization of urokinase on the islet surface by amphiphilic poly(vinyl alcohol) that carries alkyl side chains. Biomaterials. 2008;29(19):2878–83.

    Article  Google Scholar 

  42. Tomita U, Yamaguchi S, Sugimoto Y, Takamori S, Nagamune T. Poly(ethylene glycol)-Lipid-Conjugated Antibodies Enhance Dendritic Cell Phagocytosis of Apoptotic Cancer Cells. Pharmaceuticals (Basel). 2012;5(5):405–16.

    Article  Google Scholar 

  43. Won YW, Patel AN, Bull DA. Cell surface engineering to enhance mesenchymal stem cell migration toward an SDF-1 gradient. Biomaterials. 2014;35(21):5627–35.

    Article  Google Scholar 

  44. Teramura Y, Iwata H. Cell surface modification with polymers for biomedical studies. Soft Matter. 2010;6(6):1081–91.

    Article  Google Scholar 

  45. Abbina S, Siren EMJ, Moon H, Kizhakkedathu JN. Surface Engineering for Cell-Based Therapies: Techniques for Manipulating Mammalian Cell Surfaces. ACS Biomat Sci Eng. 2017.

  46. Zhao WA, Teo GSL, Kumar N, Karp JM. Chemistry and material science at the cell surface. Mater Today. 2010;13(4):14–21.

    Article  Google Scholar 

  47. Carlsten M, Childs RW. Genetic Manipulation of NK Cells for Cancer Immunotherapy: Techniques and Clinical Implications. Front Immunol. 2015;6:266.

    Article  Google Scholar 

  48. Lee DY, Lee S, Nam JH, Byun Y. Minimization of immunosuppressive therapy after islet transplantation: combined action of heme oxygenase-1 and PEGylation to islet. Am J Transplant. 2006;6(8):1820–8.

    Article  Google Scholar 

  49. Saxon E, Bertozzi CR. Cell surface engineering by a modified Staudinger reaction. Science. 2000;287(5460):2007–10.

    Article  Google Scholar 

  50. Prescher JA, Dube DH, Bertozzi CR. Chemical remodelling of cell surfaces in living animals. Nature. 2004;430(7002):873–7.

    Article  Google Scholar 

  51. Huang B, Abraham WD, Zheng Y, Bustamante Lopez SC, Luo SS, Irvine DJ. Active targeting of chemotherapy to disseminated tumors using nanoparticle-carrying T cells. Sci Transl Med. 2015;7(291):291ra294.

    Article  Google Scholar 

  52. Digilio G, Menchise V, Gianolio E, Catanzaro V, Carrera C, Napolitano R, Fedeli F, Aime S. Exofacial protein thiols as a route for the internalization of Gd(III)-based complexes for magnetic resonance imaging cell labeling. J Med Chem. 2010;53(13):4877–90.

    Article  Google Scholar 

  53. Torres AG, Gait MJ. Exploiting cell surface thiols to enhance cellular uptake. Trends Biotechnol. 2012;30(4):185–90.

    Article  Google Scholar 

  54. Hong V, Steinmetz NF, Manchester M, Finn MG. Labeling live cells by copper-catalyzed alkyne--azide click chemistry. Bioconjug Chem. 2010;21(10):1912–6.

    Article  Google Scholar 

  55. Baskin JM, Prescher JA, Laughlin ST, Agard NJ, Chang PV, Miller IA, Lo A, Codelli JA, Bertozzi CR. Copper-free click chemistry for dynamic in vivo imaging. Proc Natl Acad Sci U S A. 2007;104(43):16793–7.

    Article  Google Scholar 

  56. Swee LK, Lourido S, Bell GW, Ingram JR, Ploegh HL. One-step enzymatic modification of the cell surface redirects cellular cytotoxicity and parasite tropism. ACS Chem Biol. 2015;10(2):460–5.

    Article  Google Scholar 

  57. Paulick MG, Forstner MB, Groves JT, Bertozzi CR. A chemical approach to unraveling the biological function of the glycosylphosphatidylinositol anchor. Proc Natl Acad Sci U S A. 2007;104(51):20332–7.

    Article  Google Scholar 

  58. Chanana M, Gliozzi A, Diaspro A, Chodnevskaja I, Huewel S, Moskalenko V, Ulrichs K, Galla HJ, Krol S. Interaction of polyelectrolytes and their composites with living cells. Nano Lett. 2005;5(12):2605–12.

    Article  Google Scholar 

  59. Wilson JT, Cui W, Chaikof EL. Layer-by-layer assembly of a conformal nanothin PEG coating for intraportal islet transplantation. Nano Lett. 2008;8(7):1940–8.

    Article  Google Scholar 

  60. Martinez JS, Keller TC 3rd, Schlenoff JB. Cytotoxicity of free versus multilayered polyelectrolytes. Biomacromolecules. 2011;12(11):4063–70.

    Article  Google Scholar 

  61. Veerabadran NG, Goli PL, Stewart-Clark SS, Lvov YM, Mills DK. Nanoencapsulation of stem cells within polyelectrolyte multilayer shells. Macromol Biosci. 2007;7(7):877–82.

    Article  Google Scholar 

  62. Westman EH, Ek M, Enarsson LE, Wagberg L. Assessment of antibacterial properties of polyvinylamine (PVAm) with different charge densities and hydrophobic modifications. Biomacromolecules. 2009;10(6):1478–83.

    Article  Google Scholar 

  63. Lv H, Zhang S, Wang B, Cui S, Yan J. Toxicity of cationic lipids and cationic polymers in gene delivery. J Control Release. 2006;114(1):100–9.

    Article  Google Scholar 

  64. Wilson JT, Krishnamurthy VR, Cui W, Qu Z, Chaikof EL. Noncovalent cell surface engineering with cationic graft copolymers. J Am Chem Soc. 2009;131(51):18228–9.

    Article  Google Scholar 

  65. Miura S, Teramura Y, Iwata H. Encapsulation of islets with ultra-thin polyion complex membrane through poly(ethylene glycol)-phospholipids anchored to cell membrane. Biomaterials. 2006;27(34):5828–35.

    Article  Google Scholar 

  66. Fujita E, Teramura Y, Shiraga T, Yoshioka S, Iwatsubo T, Kawamura A, Kamimura H. Pharmacokinetics and tissue distribution of tacrolimus (FK506) after a single or repeated ocular instillation in rabbits. J Ocul Pharmacol Ther. 2008;24(3):309–19.

    Article  Google Scholar 

  67. Agudelo CA, Teramura Y, Iwata H. Cryopreserved agarose-encapsulated islets as bioartificial pancreas: a feasibility study. Transplantation. 2009;87(1):29–34.

    Article  Google Scholar 

  68. Arima Y, Teramura Y, Takiguchi H, Kawano K, Kotera H, Iwata H. Surface plasmon resonance and surface plasmon field-enhanced fluorescence spectroscopy for sensitive detection of tumor markers. Methods Mol Biol. 2009;503:3–20.

    Article  Google Scholar 

  69. Lim KS, Lee DY, Valencia GM, Won YW, Bull DA. Cell surface-engineering to embed targeting ligands or tracking agents on the cell membrane. Biochem Biophys Res Commun. 2017;482(4):1042–7.

    Article  Google Scholar 

  70. Itagaki T, Arima Y, Kuwabara R, Kitamura N, Iwata H. Interaction between cells and poly(ethylene glycol)-lipid conjugates. Colloids Surf B Biointerfaces. 2015;135:765–73.

    Article  Google Scholar 

  71. Yamamoto T, Teramura Y, Itagaki T, Arima Y, Iwata H. Interaction of poly(ethylene glycol)-conjugated phospholipids with supported lipid membranes and their influence on protein adsorption. Sci Technol Adv Mater. 2016;17(1):677–84.

    Article  Google Scholar 

  72. Pulsipher A, Griffin ME, Stone SE, Brown JM, Hsieh-Wilson LC. Directing neuronal signaling through cell-surface glycan engineering. J Am Chem Soc. 2014;136(19):6794–7.

    Article  Google Scholar 

  73. Dutta D, Pulsipher A, Luo W, Yousaf MN. Synthetic chemoselective rewiring of cell surfaces: generation of three-dimensional tissue structures. J Am Chem Soc. 2011;133(22):8704–13.

    Article  Google Scholar 

  74. Csiszar A, Hersch N, Dieluweit S, Biehl R, Merkel R, Hoffmann B. Novel fusogenic liposomes for fluorescent cell labeling and membrane modification. Bioconjug Chem. 2010;21(3):537–43.

    Article  Google Scholar 

  75. Sarkar D, Vemula PK, Zhao W, Gupta A, Karnik R, Karp JM. Engineered mesenchymal stem cells with self-assembled vesicles for systemic cell targeting. Biomaterials. 2010;31(19):5266–74.

    Article  Google Scholar 

  76. Inui O, Teramura Y, Iwata H. Retention dynamics of amphiphilic polymers PEG-lipids and PVA-Alkyl on the cell surface. ACS Appl Mater Interfaces. 2010;2(5):1514–20.

    Article  Google Scholar 

  77. van Meer G, Voelker DR, Feigenson GW. Membrane lipids: where they are and how they behave. Nat Rev Mol Cell Biol. 2008;9(2):112–24.

    Article  Google Scholar 

  78. Sprong H, van der Sluijs P, van Meer G. How proteins move lipids and lipids move proteins. Nat Rev Mol Cell Biol. 2001;2(7):504–13.

    Article  Google Scholar 

  79. Vellai T, Takacs-Vellai K. Regulation of Protein Turnover by Longevity Pathways. Protein Metabol Homeostasis Aging. 2010;694:69–80.

    Article  Google Scholar 

  80. Grant BD, Donaldson JG. Pathways and mechanisms of endocytic recycling. Nat Rev Mol Cell Biol. 2009;10(9):597–608.

    Article  Google Scholar 

  81. Adler AF, Leong KW. Emerging links between surface nanotechnology and endocytosis: impact on nonviral gene delivery. Nano Today. 2010;5(6):553–69.

    Article  Google Scholar 

  82. McMahon HT, Boucrot E. Molecular mechanism and physiological functions of clathrin-mediated endocytosis. Nat Rev Mol Cell Biol. 2011;12(8):517–33.

    Article  Google Scholar 

  83. Kumari S, Mg S, Mayor S. Endocytosis unplugged: multiple ways to enter the cell. Cell Res. 2010;20(3):256–75.

    Article  Google Scholar 

  84. Stephan MT, Irvine DJ. Enhancing Cell therapies from the Outside In: Cell Surface Engineering Using Synthetic Nanomaterials. Nano Today. 2011;6(3):309–25.

    Article  Google Scholar 

  85. Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. 2007;7(9):678–89.

    Article  Google Scholar 

  86. Katritsis D, Kaiktsis L, Chaniotis A, Pantos J, Efstathopoulos EP, Marmarelis V. Wall shear stress: theoretical considerations and methods of measurement. Prog Cardiovasc Dis. 2007;49(5):307–29.

    Article  Google Scholar 

  87. Mebius RE, Kraal G. Structure and function of the spleen. Nat Rev Immunol. 2005;5(8):606–16.

    Article  Google Scholar 

  88. Dobrovolskaia MA, Aggarwal P, Hall JB, McNeil SE. Preclinical studies to understand nanoparticle interaction with the immune system and its potential effects on nanoparticle biodistribution. Mol Pharm. 2008;5(4):487–95.

    Article  Google Scholar 

  89. Nilsson B, Korsgren O, Lambris JD, Ekdahl KN. Can cells and biomaterials in therapeutic medicine be shielded from innate immune recognition? Trends Immunol. 2010;31(1):32–8.

    Article  Google Scholar 

  90. Wang JQ, Sui MH, Fan WM. Nanoparticles for Tumor Targeted Therapies and Their Pharmacokinetics. Curr Drug Metab. 2010;11(2):129–41.

    Article  Google Scholar 

  91. Dunkelberger JR, Song WC. Complement and its role in innate and adaptive immune responses. Cell Res. 2010;20(1):34–50.

    Article  Google Scholar 

  92. Harding FA, Stickler MM, Razo J, DuBridge RB. The immunogenicity of humanized and fully human antibodies: residual immunogenicity resides in the CDR regions. MAbs. 2010;2(3):256–65.

    Article  Google Scholar 

  93. Gorbet MB, Sefton MV. Biomaterial-associated thrombosis: roles of coagulation factors, complement, platelets and leukocytes. Biomaterials. 2004;25(26):5681–703.

    Article  Google Scholar 

  94. Sperling C, Fischer M, Maitz MF, Werner C. Blood coagulation on biomaterials requires the combination of distinct activation processes. Biomaterials. 2009;30(27):4447–56.

    Article  Google Scholar 

  95. Takemoto N, Teramura Y, Iwata H. Islet surface modification with urokinase through DNA hybridization. Bioconjug Chem. 2011;22(4):673–8.

    Article  Google Scholar 

  96. Lingwood D, Simons K. Lipid rafts as a membrane-organizing principle. Science. 2010;327(5961):46–50.

    Article  Google Scholar 

  97. Lautenschlager F, Paschke S, Schinkinger S, Bruel A, Beil M, Guck J. The regulatory role of cell mechanics for migration of differentiating myeloid cells. Proc Natl Acad Sci U S A. 2009;106(37):15696–701.

    Article  Google Scholar 

  98. Novo C, Fonseca E, Freitas AA. Altered fatty acid membrane composition modifies lymphocyte localization in vivo. Cell Immunol. 1987;106(2):387–96.

    Article  Google Scholar 

  99. Curtis AS, Chandler C, Picton N. Cell surface lipids and adhesion. III. The effects on cell adhesion of changes in plasmalemmal lipids. J Cell Sci. 1975;18(3):375–84.

    Google Scholar 

  100. Klingemann H, Boissel L, Toneguzzo F. Natural Killer Cells for Immunotherapy - Advantages of the NK-92 Cell Line over Blood NK Cells. Front Immunol. 2016;7:91.

    Article  Google Scholar 

  101. Doppler SA, Deutsch MA, Lange R, Krane M. Cardiac regeneration: current therapies-future concepts. J Thorac Dis. 2013;5(5):683–97.

    Google Scholar 

  102. Galli A, Lombardi F. Postinfarct Left Ventricular Remodelling: A Prevailing Cause of Heart Failure. Cardiol Res Pract. 2016;2016:2579832.

    Article  Google Scholar 

  103. Cohn JN, Ferrari R, Sharpe N. Cardiac remodeling--concepts and clinical implications: a consensus paper from an international forum on cardiac remodeling. Behalf of an International Forum on Cardiac Remodeling. J Am Coll Cardiol. 2000;35(3):569–82.

    Article  Google Scholar 

  104. Sutton MG, Sharpe N. Left ventricular remodeling after myocardial infarction: pathophysiology and therapy. Circulation. 2000;101(25):2981–8.

    Article  Google Scholar 

  105. Talman V, Ruskoaho H. Cardiac fibrosis in myocardial infarction-from repair and remodeling to regeneration. Cell Tissue Res. 2016;365(3):563–81.

    Article  Google Scholar 

  106. Prabhu SD, Frangogiannis NG. The Biological Basis for Cardiac Repair After Myocardial Infarction: From Inflammation to Fibrosis. Circ Res. 2016;119(1):91–112.

    Article  Google Scholar 

  107. Jadczyk T, Faulkner A, Madeddu P. Stem cell therapy for cardiovascular disease: the demise of alchemy and rise of pharmacology. Br J Pharmacol. 2013;169(2):247–68.

    Article  Google Scholar 

  108. Prat-Vidal C, Galvez-Monton C, Puig-Sanvicens V, Sanchez B, Diaz-Guemes I, Bogonez-Franco P, Perea-Gil I, Casas-Sola A, Roura S, Llucia-Valldeperas A, et al. Online monitoring of myocardial bioprosthesis for cardiac repair. Int J Cardiol. 2014;174(3):654–61.

    Article  Google Scholar 

  109. Lin Z, Pu WT. Strategies for cardiac regeneration and repair. Sci Transl Med. 2014;6(239):239rv231.

    Article  Google Scholar 

  110. Hare JM, Chaparro SV. Cardiac regeneration and stem cell therapy. Curr Opin Organ Transplant. 2008;13(5):536–42.

    Article  Google Scholar 

  111. Short B, Brouard N, Occhiodoro-Scott T, Ramakrishnan A, Simmons PJ. Mesenchymal stem cells. Arch Med Res. 2003;34(6):565–71.

    Article  Google Scholar 

  112. Singh A, Singh A, Sen D. Mesenchymal stem cells in cardiac regeneration: a detailed progress report of the last 6 years (2010-2015). Stem Cell Res Ther. 2016;7(1):82.

    Article  Google Scholar 

  113. van Berlo JH, Kanisicak O, Maillet M, Vagnozzi RJ, Karch J, Lin SC, Middleton RC, Marban E, Molkentin JD. c-kit+ cells minimally contribute cardiomyocytes to the heart. Nature. 2014;509(7500):337–41.

    Article  Google Scholar 

  114. Hashimoto H, Olson EN, Bassel-Duby R. Therapeutic approaches for cardiac regeneration and repair. Nat Rev Cardiol. 2018;15(10):585–600.

    Article  Google Scholar 

  115. Cai CL, Molkentin JD. The Elusive Progenitor Cell in Cardiac Regeneration: Slip Slidin’ Away. Circ Res. 2017;120(2):400–6.

    Article  Google Scholar 

  116. Tzahor E, Poss KD. Cardiac regeneration strategies: Staying young at heart. Science. 2017;356(6342):1035–9.

    Article  Google Scholar 

  117. Quevedo HC, Hatzistergos KE, Oskouei BN, Feigenbaum GS, Rodriguez JE, Valdes D, Pattany PM, Zambrano JP, Hu Q, McNiece I, et al. Allogeneic mesenchymal stem cells restore cardiac function in chronic ischemic cardiomyopathy via trilineage differentiating capacity. Proc Natl Acad Sci U S A. 2009;106(33):14022–7.

    Article  Google Scholar 

  118. Amado LC, Saliaris AP, Schuleri KH, St John M, Xie JS, Cattaneo S, Durand DJ, Fitton T, Kuang JQ, Stewart G, et al. Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarction. Proc Natl Acad Sci U S A. 2005;102(32):11474–9.

    Article  Google Scholar 

  119. Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP, Suncion VY, Tracy M, Ghersin E, Johnston PV, Brinker JA, et al. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA. 2012;308(22):2369–79.

    Article  Google Scholar 

  120. Mathiasen AB, Qayyum AA, Jorgensen E, Helqvist S, Fischer-Nielsen A, Kofoed KF, Haack-Sorensen M, Ekblond A, Kastrup J. Bone marrow-derived mesenchymal stromal cell treatment in patients with severe ischaemic heart failure: a randomized placebo-controlled trial (MSC-HF trial). Eur Heart J. 2015;36(27):1744–53.

    Article  Google Scholar 

  121. Roura S, Galvez-Monton C, Mirabel C, Vives J, Bayes-Genis A. Mesenchymal stem cells for cardiac repair: are the actors ready for the clinical scenario? Stem Cell Res Ther. 2017;8(1):238.

    Article  Google Scholar 

  122. Chen TS, Lai RC, Lee MM, Choo AB, Lee CN, Lim SK. Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs. Nucleic Acids Res. 2010;38(1):215–24.

    Article  Google Scholar 

  123. Kinnaird T, Stabile E, Burnett MS, Shou M, Lee CW, Barr S, Fuchs S, Epstein SE. Local delivery of marrow-derived stromal cells augments collateral perfusion through paracrine mechanisms. Circulation. 2004;109(12):1543–9.

    Article  Google Scholar 

  124. Kinnaird T, Stabile E, Burnett MS, Lee CW, Barr S, Fuchs S, Epstein SE. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circ Res. 2004;94(5):678–85.

    Article  Google Scholar 

  125. Wynn RF, Hart CA, Corradi-Perini C, O'Neill L, Evans CA, Wraith JE, Fairbairn LJ, Bellantuono I. A small proportion of mesenchymal stem cells strongly expresses functionally active CXCR4 receptor capable of promoting migration to bone marrow. Blood. 2004;104(9):2643–5.

    Article  Google Scholar 

  126. Rombouts WJ, Ploemacher RE. Primary murine MSC show highly efficient homing to the bone marrow but lose homing ability following culture. Leukemia. 2003;17(1):160–70.

    Article  Google Scholar 

  127. Askari AT, Unzek S, Popovic ZB, Goldman CK, Forudi F, Kiedrowski M, Rovner A, Ellis SG, Thomas JD, DiCorleto PE, et al. Effect of stromal-cell-derived factor 1 on stem-cell homing and tissue regeneration in ischaemic cardiomyopathy. Lancet. 2003;362(9385):697–703.

    Article  Google Scholar 

  128. Abbott JD, Huang Y, Liu D, Hickey R, Krause DS, Giordano FJ. Stromal cell-derived factor-1alpha plays a critical role in stem cell recruitment to the heart after myocardial infarction but is not sufficient to induce homing in the absence of injury. Circulation. 2004;110(21):3300–5.

    Article  Google Scholar 

  129. Tang YL, Qian K, Zhang YC, Shen L, Phillips MI. Mobilizing of haematopoietic stem cells to ischemic myocardium by plasmid mediated stromal-cell-derived factor-1alpha (SDF-1alpha) treatment. Regul Pept. 2005;125(1-3):1–8.

    Article  Google Scholar 

  130. Misao Y, Takemura G, Arai M, Ohno T, Onogi H, Takahashi T, Minatoguchi S, Fujiwara T, Fujiwara H. Importance of recruitment of bone marrow-derived CXCR4+ cells in post-infarct cardiac repair mediated by G-CSF. Cardiovasc Res. 2006;71(3):455–65.

    Article  Google Scholar 

  131. Schachinger V, Erbs S, Elsasser A, Haberbosch W, Hambrecht R, Holschermann H, Yu J, Corti R, Mathey DG, Hamm CW, et al. Intracoronary bone marrow-derived progenitor cells in acute myocardial infarction. N Engl J Med. 2006;355(12):1210–21.

    Article  Google Scholar 

  132. Penn MS. Importance of the SDF-1:CXCR4 axis in myocardial repair. Circ Res. 2009;104(10):1133–5.

    Article  Google Scholar 

  133. Ghadge SK, Muhlstedt S, Ozcelik C, Bader M. SDF-1alpha as a therapeutic stem cell homing factor in myocardial infarction. Pharmacol Ther. 2011;129(1):97–108.

    Article  Google Scholar 

  134. Shi M, Li J, Liao L, Chen B, Li B, Chen L, Jia H, Zhao RC. Regulation of CXCR4 expression in human mesenchymal stem cells by cytokine treatment: role in homing efficiency in NOD/SCID mice. Haematologica. 2007;92(7):897–904.

    Article  Google Scholar 

  135. Liu H, Xue W, Ge G, Luo X, Li Y, Xiang H, Ding X, Tian P, Tian X. Hypoxic preconditioning advances CXCR4 and CXCR7 expression by activating HIF-1alpha in MSCs. Biochem Biophys Res Commun. 2010;401(4):509–15.

    Article  Google Scholar 

  136. Chung HA, Kato K, Itoh C, Ohhashi S, Nagamune T. Casual cell surface remodeling using biocompatible lipid-poly(ethylene glycol)(n): development of stealth cells and monitoring of cell membrane behavior in serum-supplemented conditions. J Biomed Mater Res A. 2004;70(2):179–85.

    Article  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This publication was supported by National Heart, Lung, and Blood Institute of the National Institutes of Health under award number R01HL138242.

Availability of data and materials

Data sharing not applicable to this article as no datasets were generated or analyzed during the current study.

Author information

Authors and Affiliations

Authors

Contributions

Y-WW and DYL designed this review. DYL primarily wrote the draft and BHC, MJ, and ASK wrote parts of the draft. Y-WW, DYL, BHC, MJ, ASK, and DAB revised the draft and prepared the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Young-Wook Won.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

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

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, D.Y., Cha, BH., Jung, M. et al. Cell surface engineering and application in cell delivery to heart diseases. J Biol Eng 12, 28 (2018). https://doi.org/10.1186/s13036-018-0123-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13036-018-0123-6

Keywords