Quick viewing(Text Mode)

Nanotechnology for Regenerative Medicine: Nanomaterials for Stem Cell Imaging

Nanotechnology for Regenerative Medicine: Nanomaterials for Stem Cell Imaging

REVIEW

Nanotechnology for regenerative : for imaging

Aniruddh Solanki1, Although stem cells hold great potential for the treatment of many injuries and John D Kim1 & degenerative diseases, several obstacles must be overcome before their therapeutic Ki-Bum Lee1,2,3† application can be realized. These include the development of advanced techniques to †Author for correspondence 1Rutgers, The State University understand and control functions of microenvironmental signals and novel methods to of New Jersey, Department of track and guide transplanted stem cells. The application of to stem cell Chemistry & Chemical biology would be able to address those challenges. This review details the current Biology, Piscataway, NJ 08854, USA challenges in , the current applications of in stem cell 2Institute for Advanced biology and further potential of nanotechnology approaches towards regenerative Materials, Devices & medicine, focusing mainly on magnetic - and quantum dot-based applications Nanotechnology, 607 Taylor in stem cell research. Road, Piscataway, NJ 08854, USA 3The Rutgers Stem Cell Why nanotechnology for microenvironments. Conventional experimental Research Center, Rutgers, regenerative medicine? The State University of New studies for specific cellular responses are typically Jersey, Piscataway, NJ 08854, The recent emergence of nanotechnology has set conducted on large cell populations, which inev- USA high expectations in biological science and medi- itably produce data measured from an inhomo- Tel.: +1 732 445 0281; cine; many scientists now predict that nanotech- geneous distribution of cellular responses. Unless Fax: +1 732 445 5312; E-mail: [email protected] nology can solve many key questions concerning cellular responses and processes are isolated from biological systems that transpire at the nanoscale. inhomogeneous signals at the single cell level, it , defined broadly as the approach of would be extremely difficult to elucidate the science and engineering at the nanometer scale intricate cellular systems and to analyze the com- towards biomedical applications, has been drawing plex dynamic signaling transductions. Further- considerable attention in the area of nanotechnol- more, conventional biomedical approaches reveal ogy [1]. Given that the sizes of functional elements very little concerning genotypic aspects that tran- in biology are in the nanometer scale range, it is scend into cell phenotypes. Thus, to better not surprising that nanomaterials interact with understand and control the responses of cells biological systems at the molecular level [2]. In towards external stimuli at the single cell or sin- addition, nanomaterials have novel electronic, gle molecule level, it is imperative to characterize optical, magnetic and structural properties that the full range of cell behaviors (e.g., self-renewal, cannot be obtained from either individual mole- differentiation, migration and apoptosis). cules or bulk materials. These unique features can Recently, stem cells have gained much atten- be tuned precisely to explore biological phenom- tion for the treatment of devastating injuries ena through numerous innovative techniques. and damage caused by degenerative diseases, One of the major goals of biology is to address the diabetes and aging [4]. Stem cells self-renew for spatial–temporal interactions of biomolecules at long periods of time and then further differenti- the cellular and integrated systems level [3]. How- ate into specialized cells and tissues on stimula- ever, to apply nanotechnology to biology and tion by appropriate microenvironmental cues. medicine, several conditions must be considered: They are typically categorized as embryonic • Nanomaterials must be designed to interact stem cells (ESCs) or tissue-specific adult stem with and cells without interfering cells, depending on their origin and differentia- with their biological activities tion capability. ESCs, which originate from the inner-cell mass of the blastocyst-stage embryo, • Nanomaterials must maintain their physical Keywords: magnetic are able to differentiate into all cell lineages nanoparticle, quantum dots, properties after surface modification regenerative medicine, stem found in the three primary germ layers of the cell imaging • Nanomaterials must be nontoxic embryo (e.g., endoderm, mesoderm and ecto- Cells are single living units of organisms that derm) [5]. Although it has been shown that part of receive the input signals from disease and injury human ESCs (hESCs) can differentiate into and then return the output signals to their many interesting cell types, such as cells of

10.2217/17435889.3.4.567 © 2008 Future Medicine Ltd ISSN 1743-5889 Nanomedicine (2008) 3(4), 567–578 567 REVIEW – Solanki, Kim & Lee

heart, brain or bone [5], the therapeutic poten- Nanomaterials for molecular tial of hESCs has not been fully realized owing & cellular imaging to numerous restrictions, including biological Although nanoparticles can be synthesized from issues concerning immunogenicity and rejection various materials using several methods, the cou- and social issues concerning ethics and pling and functionalization of nanoparticles with morality [6,7]. Adult stem/progenitor cells (e.g., biomolecules should be carried out in controlled mesenchymal [MSCs], hematopoietic and neu- conditions, such as a specific salt concentration ral stem cells [NSCs]) reside in mature tissue or pH. For this purpose, interdisciplinary knowl- compartments and are known to function as the edge from molecular biology, bioorganic chemis- replication resources for cell renewal during nor- try, bioinorganic chemistry and surface mal homeostasis of tissue . In con- chemistry must be used to functionalize nano- trast to ESCs, adult stem cells can only particles with biomolecules. With significant proliferate for a few passages and their differen- advancements in synthetic and modification tiation ability is limited to certain cell types, methodologies, nanomaterials can be modified depending on where they are located (e.g., bone to desired sizes, shapes, compositions and prop- marrow, brain or epithelial tissues) [8]. erties [10,11]; they can then be functionalized Intrinsic regulators (e.g., growth factors and readily with biomolecules through combined signaling molecules) and cellular microenviron- methodologies from bioorganic, bioinorganic ments, such as extracellular matrices (ECMs), and surface chemistry. are two prime factors that have critical roles in the regulation of stem cell behaviors. To harness Magnetic nanomaterials: the unique potential of stem cells, it is important iron oxide nanoparticles to understand the functions of intrinsic regula- Inorganic nanoparticles, especially iron oxide tors and extracellular microenvironments during nanoparticles and quantum dots (QDs), are stem cell fate [9]. Furthermore, to fully achieve one of the most promising materials for stem the therapeutic promise of stem cells, several cell research because they can be synthesized critical issues (Box 1) need to be addressed. easily in large quantities from various materials Nanostructures and nanomaterials can inter- using relatively simple methods. The dimen- act intrinsically with biological systems at the sions of the nanoparticles can be tuned from single molecular level with high specificity. The one to a few hundred nanometers with a mono- unique properties of nanomaterials and nano- dispersed size distribution. Moreover, they can structures can be particularly useful in control- comprise different metals, metal oxides and ling intrinsic stem cell signals and in dissecting semiconducting materials, whose compositions the mechanisms underlying embryonic and and sizes are variable. adult stem cell behavior (Figure 1). Iron oxide nanoparticles can either bind to Herein, we have summarized nanotechnology the external cell membrane or can be internal- approaches for stem cell research and have fur- ized into the . Particles that are ther addressed some of the challenges concerning bound externally do not affect cell viability, these research efforts. Owing to the extensive although, they may interfere with cell-surface scope of the topic and space limitations, we have interactions or may simply detach from the cell focused primarily on cellular imaging from the membrane [12]. However, iron oxide nano- numerous applications of nanotechnology in particles that can be internalized within cells stem cell biology. have their surfaces modified to ensure high uptake efficiency with minimum deleterious effects on the cells [13]. For example, coating the Box 1. Critical issues for the therapeutic applications of surface of superparamagnetic iron oxide nano- stem cells. particles (SPIONs) with dextran or other poly- • The long-term behavior of transplanted stem cells in the target tissues mers enhances stability and solubility [14] and • The pluripotency/multipotency of stem cells to differentiate towards also prevents aggregation [15]. The coated SPI- homogeneous populations of specific cell types ONs are useful for tracking and studying • The control of transplanted stem cells to migrate to the correct stem/progenitor cells with MRI. In this regard, microenvironmental places magnetic iron oxide nanoparticles and their • The tracking of transplanted stem cells by labeling techniques composites are emerging as novel contrast • The optimal time period for stem cell-replacement for agents for MRI and are much more sensitive degenerative diseases than conventional gadolinium-based contrast

568 Nanomedicine (2008) 3(4) futurefuture sciencescience groupgroup Nanotechnology for regenerative medicine – REVIEW

Figure 1. Regulation of stem cell fate by microenvironmental signals and the corresponding applications of nanotechnology.

Signals/cues Nanotechnology approaches Cellular response

Soluble signals Self renewal

Growth factors Cytokines Molecular imaging Chemokines Signal Differentiation Biodetection Cell–cell interactions transduction Gene Cadherins expression Cell arrays Insoluble/physical Apoptosis signals Materials for Laminin 2D and 3D ECM Stem/progenitor cells Fibronectin Mechanical force ECM patterning Migration

Nanomedicine © Future Science Group Ltd (2008)

ECM: .

agents [16]. The use of SPIONs as in vivo cellu- within the cytoplasmic endosomes [21]. In addi- lar-imaging agents is increasing rapidly. Since tion, the gold-coated shell enhances MRI con- their unique properties enable precise control of trast significantly. More importantly, gold has size and composition, magnetic nanoparticles well-defined surface chemistry with thiol or offer great potential for highly specific MRI to amine moieties. This offers an attractive and track stem/progenitor cells. The major transfer convenient route for further functionalization of mechanism of nanoparticles through the cell the SPIONs with biomolecules through thiol- or membrane, to label stem cells, is endocytosis or, amine-coupling chemistry [22]. One of the more specifically, pinocytosis [17–19]. advantages of using SPIONs to label stem cells is Dextran-coated SPIONs, which are com- that the migration of stem cells after implanta- monly used to label stem cells, may be unfavora- tion can be detected noninvasively using MRI. ble to endocytosis, thus reducing their labeling The stem cells can be further retrieved from efficiency. Therefore, the stem cells would require excised tissues, such as spleen and bone marrow, higher concentrations of nanoparticles and addi- by using magnetic-sorting techniques [23]. tional transfection agents. In addition, several studies have found that iron oxide nanoparticles, Semiconductor nanomaterials: QDs which are dissolved within the cells, may increase In addition to magnetic nanoparticles, QDs are the formation of free hydroxyl radicals and reac- being used extensively for applications in cell tive oxygen species. These may have toxic effects, biology, such as cell labeling, cell tracking and such as an increase in the rate of apoptosis or cell in vivo imaging, owing to their potential in imag- death and alterations in cellular metabolism [20]. ing and detection applications. QDs are robust Moreover, dissolved Fe2+ ions from the dissolved fluorescent semiconducting nanocrystals with iron oxide nanoparticles may have potential toxic broad absorption spectra and narrow emission effects on the cells. To protect stem cells from the spectra (Figure 2) [24]. toxic effects of SPIONs and to track the behavior QDs overcome the limitations of conven- of stem cells successfully in vivo, the SPIONs can tional imaging methods, such as fluorescence be coated with gold. Coating the SPIONs with microscopy and differential interference con- gold provides an inert shell around the nanopar- trast microscopy. Conventional methods are ticles and protects them from rapid dissolution limited by a lack of quantitative data, high

futurefuture sciencescience groupgroup www.futuremedicine.com 569 REVIEW – Solanki, Kim & Lee

Figure 2. Excitation and emission spectra of QDs. Because absorption and emission spectra exhibit sensitive changes depending on particle size, a wide range of emission spectra from 2,000,000 ultraviolet to infrared can be obtained. There- ) -1 fore, unique emission spectra, due to synthesiz-

cm 1,600,000 -1 ing particles with different diameters, have been acquired [30–32]. Generally, QDs have a core 1,200,000 composed of heavy metals, such as CdSe or CdTe, with a surrounding ZnS shell. The thick- 800,000 ness of the shell can be tuned depending on the reaction time. Typically, the core/shell QDs 400,000 with sizes ranging from 2 to 8 nm in diameter are synthesized by changing reaction condi-

Extinction coefficient (m 0 tions, such as temperature, duration and lig- 460 560 660 760 860 ands. The unique photophysical properties of Wavelength (nm) QDs stem from their nanometer-scale size; by changing sizes and compositions, their optical 2,000,000 properties can be controlled precisely for many applications. For example, QDs can be used 1,600,000 effectively in multiplexing experiments in which multiple biological units can be labeled 1,200,000 simultaneously. Moreover, owing to their resist- ance to photobleaching, QDs have enabled sci- 800,000 entists to study live cells and complex mechanisms of biological processes in a real-time 400,000 manner [33,34]. Normalized fluorescence 0 Nanoparticle-based applications for 460 560 660 760 860 regenerative medicine Wavelength (nm) Magnetic nanoparticles for in vivo stem cell tracking

(A) Broad excitation spectra of QDs. (B) Narrow emission spectra of QDs. Transplanting various progenitor cells and stem QD: Quantum dot. cells for tissue regeneration is an extremely Reprinted with permission from BMC [24]. promising therapeutic strategy. One of the key factors in this approach is the availability of tech- background noise from labeled biomolecules niques that would enable long-term, noninvasive and a requirement for long observation times detection of transplanted stem/progenitor cells owing to photobleaching, which gradually leads and, at the same time, enable of their to a loss of signals [25]. However, QDs exhibit differentiation, survival and proliferation within extreme brightness and resistance to photo- the desired organs. Several techniques, such as bleaching, which permits the use of lasers with MRI, bioluminescence, positron emission tom- low intensities over long periods of time, thus ography and multiple photon microscopy, are making them extremely useful for live-cell now available for in vivo cellular imaging; of imaging. In addition, QDs offer many advan- these, MRI offers several advantages, such as tages, such as high fluorescent intensity from high resolution, speed, easy accessibility and 3D high quantum yields and high molar extinction capabilities [35,36]. In addition to providing coefficients, resistance to chemical degradation information regarding the transplanted stem and long fluorescence lifetimes (>10 ns). Multi- cells, a significant advantage of MRI is that it ple QDs with different emission wavelengths provides information regarding the surround- can be used in parallel for multiplex ing tissues (e.g., edema, lesion or inflamma- imaging [26–29]. The interesting optical proper- tion), which may have an effect on the fate of ties of QDs originate from the interactions grafted stem cells or may hinder the recovery of between electrons, holes and their local environ- damaged tissues [37]. Magnetic iron oxide nano- ments, which can be controlled precisely to gen- particles, whose sizes can be tuned precisely, erate desired emission and absorption spectra. offer great potential for MRI applications. The

570 Nanomedicine (2008) 3(4) futurefuture sciencescience groupgroup Nanotechnology for regenerative medicine – REVIEW

magnetic nanocrystals tend to behave as a single and polycationic transfection magnetic domain in which all nuclear spins agents. In addition to internalizing ligands, the couple to create a single large magnetic domain. SPIONs can be multifunctionalized using fluo- At certain temperatures and crystal sizes, these rescent and isotope labels. These multi- moments wander randomly (superparamag- functional nanoparticles (Figure 4) can be used to netic) or become locked in one direction, mak- combine methods, such as optical and nuclear ing the material ferromagnetic [38]. Magnetic imaging, with MRI to validate the cellular nanocrystals of differing compositions and sizes behavior in vivo. This was demonstrated aptly can be synthesized to generate ultrasensitive by Weissleder and coworkers [23]. The magnetic molecular images, as shown in Figure 3. nanoparticles used by this group consisted of Dextran and other polymer-coated SPIONs small (5 nm) monocrystalline superpara- are currently used in a number of biomedical magnetic iron oxide cores that were stabilized applications; for example, Endorem® (Geur- by coating with cross-linked aminated dextran. bet, France) is a commercially available con- The overall size of the nanoparticles further trast agent based on SPIONs surface coated increased to 45 nm. To modify the nano- with dextran [39]. It is a suitable particles with a fluorescent label, the internaliz- for labeling human MSCs (hMSCs) and ing ligands, fluorescein isothiocyanate- human ESCs (hESCs) as it does not need a derivatized HIV-TAT peptides, were attached transfection agent (which may damage the to the coat of aminated dextran. In addition, the stem cells) to facilitate its cellular uptake. Feri- SPIONs were further modified for concomitant dex® and Sinerem® are other commercially nuclear imaging by reacting the dextran coating available dextran-coated SPIONs that are com- with a chelator, diethylenetriamine penta-acetic bined with commercially available transfection acid, so as to label the nanoparticles with 111In agents, such as Fungene™, Superfect™ or isotope. The modified SPIONs, with a triple Lipofectamine [39,40]. The use of transfection label (magnetic, fluorescent and isotope), inter- agents at higher concentrations may increase nalized into hematopoietic stem and neural pro- toxicity and, at lower concentrations, may not genitor cells efficiently. The group further lead to sufficient cellular uptake [40]. Thus, the demonstrated that the labeled neural progenitor amount of transfection agent needed to cells retained their capability for differentiation enhance internalization is optimized carefully and the iron incorporation did not have any before combining it with SPIONs. The effect on viability and proliferation of hemat- amount also depends on the stem cell type to opoietic (CD34+) cells. In another study, dex- be labeled. tran-coated magnetic iron oxide nanoparticles Stem/progenitor cells can be labeled with with a core diameter of 4.6 ± 1.2 nm and an SPIONs by modifying their surfaces with inter- overall size, after coating with dextran, of nalizing ligands, such as the HIV-Tat peptide, 8–20 nm were attached covalently to OX-26, an

Figure 3. Neural stem cells labeled with iron oxide nanoparticles.

(A) Photomicrograph showing iron oxide nanoparticles in stem cells stained with Prussian blue and counterstained with neutral red. (B) Transmission-electron photomicrograph of stem cells showing the presence of iron oxide nanoparticles in nuclear and cell membranes. ©2006 Massachusetts Medical Society. All rights reserved [44].

futurefuture sciencescience groupgroup www.futuremedicine.com 571 REVIEW – Solanki, Kim & Lee

Figure 4. Multifunctional inorganic nanoparticles. that was similar to that of unlabeled stem cells. The cellular uptake of these composites is through a nonspecific process, thus Surface modification Magnetic materials offering a great opportunity to label a variety of stem cells without regard to their origin or Luminescent materials animal species. In a recent study, Kehr and coworkers labeled NSCs with gold-coated monocrystalline SPIONs [21]. The NSCs were infused into the spinal cord of rats and tracked O by means of MRI for over a month. The MRI MNP signals persisted 1 month postsurgery and the gold surface protected the nanoparticles from N Qds being digested by the glial macrophages. It was concluded that gold-coated SPIONs may rep- resent a class of superior MRI labels for long- O MNP term in vivo tracking of stem cells. In another Gold recent study, Zhu and coworkers labeled human NSCs with SPIONs using a nonlipo- somal lipid-based transfecting agent [43]. The labeled cells were then implanted in the region Dextran of brain damage in a patient suffering from brain trauma. The migration of NSCs from the site of injection to the border of the injured tis- Antibody sue of the brain was detected successfully [43]. One of the challenges of using SPIONs is the Nanomedicine © Future Science Group Ltd (2008) potential transfer of the contrast from the

labeled stem cells to other cell types, such as macrophages, which metabolize iron after antitransferrin receptor monoclonal antibody [41]. engulfing the stem cells. However, through The antibody-functionalized nanoparticles detailed studies and experimentation, Zhu and were used to label oligodendrocyte progenitor coworkers excluded the possibility that mag- cells by targeting the transferrin receptors on netic signals could have been generated by the cells. The progenitor cells were made macrophages engulfing the NSCs and thus highly magnetic by incubating them with iron concluded that the signals were indeed gener- oxide nanoparticles. Because the oligodendro- ated by the migrating stem cells and not by the cyte progenitor cells have been shown previ- engulfed stem cells. ously to myelinate large areas in the CNS Tracking of stem cell migration is not limited significantly [42], they were transplanted into to NSCs/progenitor cells. It is also possible to the spinal cord of myelin-deficient rats. After study the migration of stem cells labeled with neurotransplantation, these cells could be SPIONs in other systems, such as the cardio- tracked easily using MRI and the extent of vascular system. Regenerative medicine for car- myelination could be determined. The progen- diac diseases will have enormous therapeutic itor cells retain their capacity for myelination potential in the future for situations involving and migration fully in vivo [41]. ischemic cardiac injury, which involves irrevers- Another important example of transfecting ible cardiac damage. Bulte and cowork- agents are dendrimers. composites ersdemonstrated the potential of MRI in of iron oxide nanoparticles, also known as mag- tracking magnetically labeled MSCs in a swine netodendrimers, represent a versatile new class model of myocardial infarction [44] . The MSCs of contrast agent for MRI. They were devel- were labeled with dextran-coated SPIONs oped by Frank and coworkers and label mam- (Feridex®) to noninvasively track the quantity malian cells efficiently, including NSCs and and location of the MSCs after myocardial inf- MSCs [36]. They have an oligocrystalline struc- arction. The MRI tracking of the MSCs labeled ture of 7–8 nm. Labeling NSCs and MSCs with Feridex® was feasible and represents a with magnetodendrimers did not affect their preferred method for studying engraftment of growth rate and they exhibited a growth rate MSCs in myocardial infarction.

572 Nanomedicine (2008) 3(4) futurefuture sciencescience groupgroup Nanotechnology for regenerative medicine – REVIEW

QD imaging for stem cells cell populations labeled with QDs that exhibit Quantum dots or semiconductor nanocrystals different emission wavelengths at the same have opened doors to an array of diverse appli- time) is one of the biggest advantages of using cations in biological sciences, such as live moni- QDs for tracking stem cells in vivo. Wu and toring of physiological events taking place in coworkers successfully demonstrated in vivo cells by labeling specific cellular structures or multiplex imaging of mouse ESCs labeled with proteins with QDs having different colors, QDs [24]. They injected ESCs labeled with six monitoring cell migration, tracing cell lineage different QDs subcutaneously, having diverse and in vivo cell tracking [45–48]. Their unique emission wavelengths of 525, 565, 605, 655, photophysical properties coupled with their 705 and 800 nm, into various locations on the diverse biological applications make QDs back of athymic nude mice and detected the attractive nanoprobes for investigating stem cell labeled cells in vivo using a single excitation behavior (Figure 5). Furthermore, QDs are wavelength (425 nm), as shown in Figure 7. advantageous for studying dynamic changes They also concluded that, within the sensitivi- occurring in the membranes of stem/progenitor ties of the screening assays, the QDs did not cells. Functionalized QDs bind selectively to affect viability, proliferation or differentiation individual molecules on the cell surface and help capacity of the ESCs [24]. in tracking the motion of those individual mol- QDs can be used efficiently to label neural ecules. In a study by Cho and coworkers [49], stem and progenitor cells (NSPCs) in vivo and functionalized QDs were used to demonstrate can be used to study the migration and differ- changes in integrin dynamics during osteogenic entiation of NSPCs during mammalian devel- differentiation of human bone marrow-derived opment. However, direct QD labeling of progenitor cells. In this study, QDs conjugated NSPCs is a considerable challenge and not with integrin antibodies enabled precise optical many techniques to label QDs directly and effi- identification of integrin molecules, which led ciently exist. Haydar and coworkers developed to a detailed examination of the molecular novel in utero electroporation and ultrasound- dynamics of integrin molecules involved in guided delivery techniques to label the NSPCs osteogenic differentiation of the progenitor cells directly in vivo [46]. NSPCs labeled with QDs [49]. In stem cell-based therapy, it is extremely using the techniques described previously, were important to monitor the survival and location found to differentiate into three principle cell of stem cells after they are transplanted to the types: oligodendrocyte progenitors, astrocytes desired location. Transplanted stem cells, which and neurons. QDs were found in all three types may be either embryonic or adult stem cells, are of cells after differentiation. The cells were also expected to remodel and differentiate in found to migrate away from the site of injection, response to surrounding microenvironments, resulting in tissue regeneration and repair [43]. Figure 5. Confocal fluorescent image MSCs labeled with bright, photostable QDs of MSCs labeled with QDs and couple functionally with cardiomyocytes in colabeled with calcein. coculture, thus demonstrating the usefulness of QDs as labeling agents in culture (Figure 6) [50]. hMSCs were labeled with QDs biocon- jugated with arginine–glycine–aspartic acid peptide during self-replication and multilineage differentiations into chondrogenic, androgenic and adipogenic cells in a long-term labeling study. Human MSCs labeled with QDs remained as viable as the unlabeled hMSCs from the same subpopulation, thus suggesting 20 µm that QDs are useful probes from long-term labeling of stem cells [51].

QDs also elucidate the mechanisms involved The QDs are distributed in the perinuclear region in mechanical integration of stem cells to the within the stem cells. As the MSCs proliferate, surrounding tissues and their differentiation the QDs remain bright and are easy to detect. MSC: ; QD: Quantum dot. into specific cell lineages in vivo [45]. In addi- Reprinted with permission from BMC [51]. tion, multiplex imaging (i.e., tracking different futurefuture sciencescience groupgroup www.futuremedicine.com 573 REVIEW – Solanki, Kim & Lee

Figure 6. TEM images of QD-labeled MSCs.

nm 2 µm 500 nm nm

(A) Low magnification of MSC showing QD aggregates (arrow) in endosomal vesicles surrounding the nuclear membrane. (B) High magnification of single-enlarged vesicle showing presence of individual QDs (arrow). MSC: Mesenchymal stem cell; QD: Quantum dot; TEM: Transmission-electron microscopy. Reprinted with permission from BMC [51].

suggesting that neither the QDs nor the in vivo could be solved by coating the QDs and making labeling techniques had any effect on migration them biologically inert [55]. Larger molecules, such and differentiation of the NSPCs. Furthermore, as proteins (e.g., streptavidin and bovine serum their method demonstrated a lack of toxicity albumin), further slow the photo-oxidation of the and good tolerance of NSPCs for QDs, partic- core [56]. Bioconjugation of QDs with biomole- ularly during early embryonic mammalian cules, such as arginine–glycine–aspartic acid, did development [46]. not show any toxic effect on hMSCs as compared Despite having unique optical properties and a with unlabeled hMSCs [51]. In a dose-dependent host of advantages over the conventional tracking study involving the labeling of MSCs with QDs, it agents, toxicity is a primary concern for the appli- was observed that if the exposure of QDs to MSCs cation of QDs in biology. Stem/progenitor cells was optimized and limited to low concentrations, tend to be extremely sensitive and thus toxicity is a then the QDs were not significantly toxic [50]. primary determinant in deciding whether QDs would be feasible for stem cell tracking, especially Other challenges & opportunities in vivo. Some literature studies do suggest that Integration of nanotechnology and stem cell QDs are nontoxic; nevertheless, recent data show research should provide new opportunities for sci- that cytotoxicity is dependent on the phys- entists to address fundamental questions of stem ico–chemical properties, dose and exposure con- cell biology at the single-molecule level. Although centrations [52]. Although the mechanism of many nanoparticle-based applications currently cytotoxicity is not yet clearly known and is under garner much discussion, other important appli- thorough investigation, concerns regarding the cations of nanotechnology, regarding the regula- toxicity of QDs have been raised because they are tion of stem cell fate, are also being developed. used for cell-tracking studies in live animals. QDs These applications include microenvironmental contain heavy metals, such as cadmium and sele- engineering and gene manipulation. nium, and the cytotoxicity is observed owing to the presence of Cd2+ and Se2- ions [53,54]. Toxic- Surface engineering stem ity can be considerably reduced by coating the cell microenvironment core made of CdSe with a shell of a material, Stem cells normally reside within specific extra- such as ZnS, which reduces toxicity significantly cellular microenvironments that are typically by blocking the oxidation of CdSe by air [55]. referred to as stem cell niches [57,58], which are Although the toxicity may not be critical at the comprised of a complex mixture of soluble and low concentrations optimized for labeling, it insoluble ECM and signal molecules. It is well could be detrimental for embryo development at known that morphogenetic signaling molecules higher concentrations. Nevertheless, the problem and ECM components can control stem cell

574 Nanomedicine (2008) 3(4) futurefuture sciencescience groupgroup Nanotechnology for regenerative medicine – REVIEW

behaviors. For example, a variety of factors that Gene manipulation of stem cells regulate stem cell development have been explored using nanomaterials in the context of stem cell fate regulation [58,59]. Gene delivery has an important role in recogniz- These factors include cadherins, laminins and ing the potential of regenerative medicine. To morphometric families. Even though sev- manipulate the expression level of key genes in eral combinatorial high-throughput screening stem cells, several biomolecules, such as gene methods probing the effect of soluble signal mole- vectors, siRNA, proteins and small molecules, cules on stem cell differentiation have been have been developed. Because several transcrip- reported, similar approaches for screening the tion factors that regulate stem cell differentiation effect of insoluble cues are limited owing to techni- into specific cell types have been demonstrated, cal difficulties. Only a few types of ECM arrays gene delivery could be an immensely powerful fabricated by conventional spot-array techniques tool for specific differentiation of stem cells. The have been used to probe stem cell differentiation development of safe and efficient gene delivery and migration behaviors [60]. There is plenty of systems, which can lead to high levels of gene room for improvement in this approach in terms expression within stem cells, is an urgent of pattern density, recognition sensitivity and small requirement for the effective implementation of sample-volume requirement. Regarding this chal- regenerative medicine. Recently, Akaike and lenge, several soft micro/nanolithographic tools, coworkers developed a biofunctionalized inor- such as microcontact printing and dip-pen nano- ganic, apatite nanoparticle-based gene delivery lithography, are potentially useful. Both methods system, which showed high affinity for mouse have been used successfully to generate ECM pat- surface and led to acceler- terns on different surfaces at the micro- or nano- ated trans-gene delivery [64]. Apatite nano- scale. With microcontact printing [61] and dip-pen particles by themselves are inefficient in [62,63], single stem cells and their transfecting the ESCs; however, when function- behavior on combinatorial ECM nanoarrays can alized with biomolecules, such as fibronectin and be studied. Key scientific issues, such as the effects E-cadherin chimera, the hybrid nanoparticle sys- of ECM composition and temporal–spatial effects tem shows enhanced trans-gene delivery, which of ECM materials on stem cell differentiation, can is notably higher than that of a commercially be further investigated. It is critical to address these available lipofection system [64]. Another nano- issues to enhance the feasibility of using stem cells material-based novel approach to gene delivery for therapeutic purposes. was demonstrated by Miyake and coworkers [65].

Figure 7. Multiplex imaging by QDs.

10,000

1000

QD525 QD565 100

QD605 QD655

10 QD705 QD800

Total signal background/exposure time (ms) signal background/exposure Total 1 525 565 605 655 705 800 QD

(A) ESCs labeled with QD 525, 565, 605, 655, 705 and 800 injected subcutaneously in the back of athymic nude mice immediately after labeling. The image was taken with a single excitation wavelength straight after injection. (B) Quantified fluorescent signal intensities of QDs. ESC: Embryonic stem cell; QD: Quantum dot. Reprinted with permission from BMC [24].

futurefuture sciencescience groupgroup www.futuremedicine.com 575 REVIEW – Solanki, Kim & Lee

In this approach, gold nanoparticles, 20 nm in medicine requires several considerations. First, diameter and conjugated with a DNA–poly- appropriate imaging methods should be ethylenimine complex, were patterned on a designed carefully by considering the specific solid surface (glass) and used as nanoscaffolds biological questions regarding stem cells that for the delivery of DNA into hMSCs through need to be addressed because each imaging reverse transfection. The authors claimed that technique has its unique set of advantages and this method of delivering genes efficiently from disadvantages. Second, nanomaterials might a solid surface to stem cells might be useful in have undesirable on stem cells owing the development of tools for in to their composition, size and physical proper- regenerative medicine [65]. ties. For instance, the cytotoxicity of QDs is a potential limitation for the application of Conclusions molecular probes for both cellular and clinical In this review, we have summarized nanoparticle- use in vitro and in vivo. Finally, nanotechnology based approaches for stem cell imaging. Consid- approaches for regenerative medicine essentially ering that nanomaterials intrinsically enable cel- necessitate synergetic effort and interdiscipli- lular and molecular imaging with high sensitivity nary expertise from biology, chemistry and engi- and high spatial resolution, it is not surprising neering. In particular, this approach would be that a growing number of imaging techniques beneficial to elucidate the complex cellular spa- based on nanoprobes are beginning to have an tial–temporal dynamics and signaling pathways impact on stem cell-based and research. in more effective ways. Addressing the chal- Good examples of these include stem cell track- lenges ahead would accelerate the development ing using magnetic nanoparticles and QD-based of nanotechnology approaches toward regenera- imaging of stem cell interactions with other tive medicine and facilitate the therapeutic microenvironmental cues. In addition, other application of stem cells. nanotechnology applications, such as surface engineering and drug delivery, have huge poten- Acknowledgement tial to further address challenges in the area of The authors gratefully acknowledge helpful comments from regenerative medicine. Collectively, advance- David Wang and Birju Shah. ments in nanotechnology enable the modulation of stem cell signaling pathways and improvement Financial & competing interests disclosure in their therapeutic applications. The authors have no relevant affiliations or financial involvement with any organization or entity with a finan- Future perspectives cial interest in or financial conflict with the subject matter The application of nanotechnology in regenera- or materials discussed in the manuscript. This includes tive medicine has already begun to revolutionize employment, consultancies, honoraria, stock ownership or several areas of stem cell research and will con- options, expert testimony, grants or patents received or tinue having great impacts on regenerative med- pending, royalties. icine. However, recognizing the optimum No writing assistance was utilized in the production of potential of nanotechnology in regenerative this manuscript.

Executive summary

• To apply nanotechnology to stem cell biology, several conditions must be considered: nanomaterials must be designed to interact with proteins and cells without perturbing their biological activities; nanomaterials must maintain their physical properties after the surface conjugation chemistry; and nanomaterials must be biocompatible and nontoxic.

• Magnetic iron oxide nanoparticles, the size of which can be tuned precisely, are used to label stem cells and offer great potential for tracking them in vivo using MRI to generate ultrasensitive images.

• Quantum dots, with their unique photophysical properties and resistance to photobleaching, can be used for multiplex imaging of stem cells in vitro and in vivo.

• Combinational exracellular matrix micro/nanoarrays, generated by soft-lithography, have great potential in studying and controlling the behavior of single stem cells.

• Nanomaterial-based gene delivery for manipulating stem cells has a vital role in recognizing the potential of regenerative medicine.

576 Nanomedicine (2008) 3(4) futurefuture sciencescience groupgroup Nanotechnology for regenerative medicine – REVIEW

Bibliography 17. Lu CW, Hung Y, Hsiao JK et al.: 32. Nie SM: Luminescent quantum dots for 1. Moghimi SM, Hunter AC, Murray JC: Bifunctional magnetic silica nanoparticles for multiplexed optical tagging of genes, Nanomedicine: current status and future highly efficient human stem cell labeling. proteins, and cells. Cytometry 25–25 (2002). prospects. FASEB J. 19(3), 311–330 (2005). Nano Lett. 7(1), 149–154 (2007). 33. Cui B, Wu C, Chen L: One at a time, live 2. Bogunia-Kubik K, Sugisaka M: From 18. Rogers WJ, Meyer CH, Kramer CM: tracking of NGF axonal transport using molecular biology to nanotechnology and Technology insight: in vivo cell tracking by quantum dots. Proc. Natl Acad. Sci. USA nanomedicine. Biosystems 65(2–3), 123–138 use of MRI. Nat. Clin. Pract. Cardiovasc. 104(34), 13666–13671 (2007). (2002). Med. 3(10), 554–562 (2006). 34. Maysinger D, Behrendt M, 3. Emerich DF, Thanos CG: Nanotechnology 19. Gupta AK, Curtis AS: Lactoferrin and Lalancette-Hébert M, Kriz J: Real-time and medicine. Expert Opin. Biol. Ther. 3(4), ceruloplasmin derivatized superparamagnetic imaging of astrocyte response to quantum 655–663 (2003). iron oxide nanoparticles for targeting cell dots: in vivo screening model system for 4. Slack JMW: Stem cells in epithelial tissues. surface receptors. Biomaterials 25(15), biocompatibility of nanoparticles. Nano Science 287(5457), 1431–1433 (2000). 3029–3040 (2004). Lett. 7(8), 2513–2520 (2007). 5. Smith AG: Embryo-derived stem cells: of 20. Emerit JC, Beaumont C, Trivin F: Trivin, 35. Chemaly ER, Yoneyama R, Frangioni JV, mice and men. Annu. Rev. Cell Dev. Biol. 17, iron metabolism, free radicals, and oxidative Hajjar RJ: Tracking stem cells in the 435–462 (2001). injury. Biomed. Pharmacother. 55(6), cardiovascular system. Trends Cardiovasc. 6. Bradley JA, Bolton EM, Pedersen RA: Stem 333–339 (2001). Med. 15(8), 297–302 (2005). cell medicine encounters the immune 21. Wang FH, Lee IH, Holmström N et al.: 36. Bulte JW, Douglas T, Witwer B: system. Nat. Rev. Immunol. 2(11), 859–871 Magnetic resonance tracking of nanoparticle Magnetodendrimers allow endosomal (2002). labelled neural stem cells in a rat’s spinal cord. magnetic labeling and in vivo tracking of 7. Thomson AW, Lu L, Murase N, Nanotechnology 17(8), 1911–1915 (2006). stem cells. Nat. Biotechnol. 19(12), Demetris AJ, Rao AS, Starzl TE: 22. Niemeyer CM, Ceyhan B: DNA-directed 1141–1147 (2001). Microchimerism, dendritic cell progenitors functionalization of with 37. Syková E, Jendelová P: Migration, fate and and transplantation tolerance. Stem Cells proteins. Angewandte Chemie-International in vivo imaging of adult stem cells in the 13(6), 622–639 (1995). Ed. 40, 3685–3688 (2001). CNS. Cell Death Differ. 14, 1336–1342 8. Robey PG: Stem cells near the century mark. 23. Lewin M, Carlesso N, Tung CH: Tat (2007). J. Clin. Invest. 105(11), 1489–1491 (2000). peptide-derivatized magnetic nanoparticles 38. Thorek DL, Chen AK, Czupryna J, 9. Odorico JS, Kaufman DS, Thomson JA: allow in vivo tracking and recovery of Tsourkas A: Superparamagnetic iron oxide Multilineage differentiation from human progenitor cells. Nat. Biotechnol. 18, nanoparticle probes for molecular imaging. embryonic stem cell lines. Stem Cells 19(3), 410–414 (2000). Ann. Biomed. Eng. 34(1), 23–38 (2006). 193–204 (2001). 24. Lin S, Xie X, Patel MR: Quantum dot 39. Corot C, Robert P, Idée JM, Port M: Recent 10. Chan WC, Maxwell DJ, Gao X, Bailey RE, imaging for embryonic stem cells. BMC advances in iron oxide nanocrystal Han M, Nie S: Luminescent quantum dots Biotechnol. 7, 67 (2007). technology for medical imaging. Adv. Drug for multiplexed biological detection and 25. Wu WW, Li AD: Optically switchable Deliv. Rev. 58(14), 1471–1504 (2006). imaging. Curr. Opin. Biotechnol. 13(1), nanoparticles for biological imaging. 40. Bulte JWM, Kraitchman DL: Iron oxide 40–46 (2002). Nanomed. 2(4), 523–531 (2007). MR contrast agents for molecular and 11. Moriarty P: Nanostructured materials. Rep. 26. Gao X, Yang L, Petros JA, Marshall FF, cellular imaging. NMR Biomed. 17, Prog. Phys. 64(3), 297–381 (2001). Simons JW, Nie S: In vivo molecular and 484–499 (2004). 12. Bulte JWM, Kraitchman DL: Monitoring cellular imaging with quantum dots. Curr. 41. Bulte JW, Zhang S, van Gelderen P: cell therapy using iron oxide MR contrast Opin. Biotechnol. 16(1), 63–72 (2005). Neurotransplantation of magnetically agents. Curr. Pharm. Biotechnol. 5(6), 27. Michalet X, Pinaud FF, Bentolila LA: labeled oligodendrocyte progenitors: 567–584 (2004). Quantum dots for live cells, in vivo imaging, magnetic resonance tracking of cell 13. Bulte JW, Kraitchman DL: Iron oxide MR and diagnostics. Science 307(5709), 538–544 migration and myelination. Proc. Natl Acad. contrast agents for molecular and cellular (2005). Sci. USA 96(26), 15256–15261 (1999). imaging. NMR Biomed. 17, 484–499 28. Nie S, Xing Y, Kim GJ, Simons JW: 42. Duncan ID, Milward EA: Glial-cell (2004). Nanotechnology applications in cancer. transplants – experimental therapies of 14. Dutz S, Wilfried A, Rudolf H et al.: Annu. Rev. Biomed. Eng. 9, 257–288 (2007). myelin diseases. Brain Pathol. 5(3), 301–310 Influence of dextran coating on the magnetic 29. Rhyner MN, Smith AM, Gao X, Mao H, (1995). behaviour of iron oxide nanoparticles. Yang L, Nie S: Quantum dots and 43. Zhu J, Zhou L, XingWu F: Tracking neural J. Magn. Magn. Mater. 311(1), 51–54 multifunctional nanoparticles: new contrast stem cells in patients with brain trauma. (2007). agents for tumor imaging. Nanomed. 1(2), N. Engl. J. Med. 355(22), 2376–2378 (2006). 15. Lawaczeck R, Menzel M, Pietsch H: 209–217 (2006). 44. Kraitchman DL, Heldman AW, Atalar E Superparamagnetic iron oxide particles: 30. Gao XH, Chan WCW, Nie SM: Quantum- et al.: In vivo magnetic resonance imaging of contrast media for magnetic resonance dot nanocrystals for ultrasensitive biological mesenchymal stem cells in myocardial imaging. App. Organometallic Chem. 18(10), labeling and multicolor optical encoding. infarction. Circulation 107, 2290–2293 506–513 (2004). J. Biomed. Opt. 7(4), 532–537 (2002). (2003). 16. Chemaly ER, Yoneyama R, Frangioni JV, 31. Gao XH, Nie SM: Molecular profiling of 45. Rosen AB, Kelly DJ, Schuldt AJ: Finding Hajjar RJ. Tracking stem cells in the single cells and tissue specimens with fluorescent needles in the cardiac haystack: cardiovascular system. Trends Cardiovasc. quantum dots. Trends Biotechnol. 21(9), tracking human mesenchymal stem cells Med. 15(8), 297–302 (2005). 371–373 (2003). labeled with quantum dots for quantitative

futurefuture sciencescience groupgroup www.futuremedicine.com 577 REVIEW – Solanki, Kim & Lee

in vivo three-dimensional fluorescence 52. Jamieson T, Bakhshi R, Petrova D, 60. Lutolf MP, Hubbell JA: Synthetic analysis. Stem Cells 25(8), 2128–2138 Pocock R, Imani M, Seifalian AM: biomaterials as instructive extracellular (2007). Biological applications of quantum dots. microenvironments for morphogenesis in 46. Slotkin JR, Chakrabarti L, Dai HN et al.: Biomaterials 28, 4717–4732 (2007). . Nat. Biotechnol. 23(1), In vivo quantum dot labeling of mammalian 53. Ipe BI, Lehnig M, Niemeyer CM: On the 47–55 (2005). stem and progenitor cells. Dev. Dyn. 236, generation of free radical species from 61. Whitesides GM, Ostuni E, Takayama S, 3393–3401 (2007). quantum dots. Small 1(7), 706–709 Jiang X, Ingber DE: Soft lithography in 47. Wu JC: Comparison of imaging techniques (2005). biology and biochemistry. Annu. Rev. for tracking cardiac stem cell therapy. J. Nucl. 54. Kirchner C, Liedl T, Kudera S et al.: Biomed. Eng. 3, 335–373 (2001). Med. 48, 1916–1919 (2007). Cytotoxicity of colloidal CdSe and 62. Piner RD, Zhu J, Xu F, Hong S, 48. Arbab AS, Pandit SD, Anderson SA et al.: CdSe/ZnS nanoparticles. Nano Lett. 5(2), Mirkin CA: “Dip-pen” nanolithography. Magnetic resonance imaging and confocal 331–338 (2005). Science 283(5402), 661–663 (1999). microscopy studies of magnetically labeled 55. Medintz IL, Uyeda HT, Goldman ER, 63. Lee KB, Park SJ, Mirkin CA, Smith JC, endothelial progenitor cells trafficking to sites Mattoussi H: Quantum dot bioconjugates Mrksich M: Protein nanoarrays generated of tumor angiogenesis. Stem Cells 24(3), for imaging labelling and sensing. Nat. by dip-pen nanolithography. Science 671–678 (2006). Mater. 4, 435–446 (2005). 295(5560), 1702–1705 (2002). 49. Chen H, Titushkin I, Stroscio M, Cho M: 56. Alivisatos AP, Gu WW, Larabell C: 64. Kutsuzawa K, Chowdhury EH, Altered membrane dynamics of quantum Quantum dots as cellular probes. Annu. Nagaoka M, Maruyama K, Akiyama Y, dot-conjugated integrins during osteogenic Rev. Biomed. Eng. 7, 55–76 (2005). Akaike T: Surface functionalization of differentiation of human bone marrow 57. Fuchs E, Tumbar T, Guasch G: inorganic nano-crystals with fibronectin derived progenitor cells. Biophys. J. Socializing with the neighbors: stem cells and E-cadherin chimera synergistically 92(1399–1408), 1399 (2007). and their niche. Cell 116(6), 769–778 accelerates trans-gene delivery into 50. Muller-Borer BJ, Collins MC, Gunst PR, (2004). embryonic stem cells. Biochem. Cascio WE, Kypson AP: Quantum dot 58. Moore KA, Lemischka IR: Stem cells and Biophys. Res. Commun. 350, 514–520 labeling of mesenchymal stem cells. their niches. Science 311(5769), (2006). J. Nanobiotechnol. 5(9), PAGES? (2007). 1880–1885 (2006). 65. Uchimura E, YamadaS, Uebersax L, 51. Shah BS, Clark PA, Moioli EK, Stroscio MA, 59. Flaim CJ, Chien S, Bhatia SN: FujitaS, Miyake M, Miyake J: Method for Mao JJ: Labeling of mesenchymal stem cells An extracellular matrix microarray for reverse transfection using gold colloid as a by bioconjugated quantum dots. Nano Lett. probing . Nat. nano-scaffold. J. Biosci. Bioeng. 103(1), 7, 3071–3079 (2007). Methods 2(2), 119–125 (2005). 101–103 (2006).

578 Nanomedicine (2008) 3(4) futurefuture sciencescience groupgroup