Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 May 11.
Published in final edited form as: Adv Exp Med Biol. 2014;811:255–275. doi: 10.1007/978-94-017-8739-0_13

Stem Cells and Nanomaterials

Marie-Claude Hofmann 1
PMCID: PMC5425934  NIHMSID: NIHMS856733  PMID: 24683036

Abstract

Because of their ability to self-renew and differentiate into many cell types, stem cells offer the potential to be used for tissue regeneration and engineering. Much progress has recently been made in our understanding of the biology of stem cells and our ability to manipulate their proliferation and differentiation to obtain functional tissues. Similarly, nanomaterials have been recently developed that will accelerate discovery of mechanisms driving stem cell fate and their utilization in medicine. Nanoparticles have been developed that allow the labeling and tracking of stem cells and their differentiated phenotype within an organism. Nanosurfaces are engineered that mimic the extracellular matrix to which stem cells adhere and migrate. Scaffolds made of functionalized nanofibers can now be used to grow stem cells and regenerate damaged tissues and organs. However, the small scale of nanomaterials induces changes in their chemical and physical properties that might modify their interactions with cells and tissues, and render them toxic to stem cells. Therefore a thorough understanding of stem cell-nanomaterial interactions is still necessary not only to accelerate the success of medical treatments but also to ensure the safety of the tools provided by these novel technologies.

Keywords: Stem cells, Nanomaterials, Differentiation, Regenerative medicine, Toxicity

13.1 Introduction

Nanotechnology involves the fabrication and use of materials and devices on an atomic and molecular scale, with at least one dimension measuring from 1 to 100 nm [1]. Materials and tools created using nanotechnologies have at least two advantages. First, their minuscule sizes make them of interest in bioengineering and medicine, for example to build scaffolds for tissue engineering and to carry drugs that target specific cells and tissues [25]. Second, the fact that certain physical and chemical properties change as the size of the system decreases renders nanomaterials particularly useful in mechanical, chemical and electrical engineering, and ultimately life sciences [6]. Indeed nanotubes, nanowires, fullerene derivatives (buckyballs), and quantum dots are now used for the manufacturing of novel analytical tools for biotechnology [712]. Because of their novel properties, nanoscale materials can also be exploited to modulate cell proliferation or differentiation by influencing their attachment or manipulating their environment [1316]. This feature is particularly applicable for the modulation of stem cell fate in regeneration studies.

Stem cells are undifferentiated cells that have the dual ability to self-renew to maintain their own pool, or to differentiate into functional mature cells. During early mammalian embryogenesis, the inner cell mass (ICM) of the blastocyst is made of pluripotent cells, or embryonic stem cells (ES cells) that are able to proliferate and differentiate into all cell lineages that will eventually generate the fetal organs [17]. As these pluripotent stem cells continue to divide, they start to specialize and become multipotent stem cells. Multipotent stem cells are found in the fetus and the adult animal; they are less plastic than ES cells and are able to differentiate only into specific lineages. For example, mesenchymal stem cells (MSCs) isolated from adult bone marrow or cord blood can generate only bone, cartilage, adipocytes, cardiomyocytes, nerve cells and supporting cells such as stromal fibroblasts (Fig. 13.1) [19]. Adipose tissue-derived stem cells (ADSC) are similar to MSCs and are found in the stromal-vascular fraction of fat tissue [20]. Hematopoietic stem cells, found in the bone marrow, produce both the lymphoid and myeloid lineages and are responsible for maintaining blood cell production throughout life [21]. The intestinal crypts contain stem cells that self-renew to continuously regenerate the gut epithelium, but can also differentiate into enterocytes, enteroendocrine cells, goblet cells and Paneth cells with distinct functions [22, 23]. Similarly, skin stem cells self-renew and/or differentiate to produce keratinocytes, hair follicles, sebaceous glands and sweat glands [24]. While multipotent stem cells usually produce several, but restricted, cell types, some stem cells are unipotent and give rise to only one kind of mature cells. For example, spermatogonial stem cells (SSCs) of the testis ultimately produce only sperm cells [25]. However, SSCs have the unique property to revert to an ES cell-like state when cultured in the appropriate conditions, and might become some day a source of adult pluripotent stem cells for use in regenerative medicine [2629]. Induced pluripotent stem cells or iPS cells, are pluripotent stem cells derived from adult somatic cells, typically fibroblasts, by forcing the expression of pluripotent genes. In mice, these genes originally were OCT4, SOX2, c-MYC and KLF4 [3032]. However, about 16 % of chimeric mice obtained after blastocyst injection of the iPS cells died of tumors within 100 days after birth, presumably because of the oncogenic properties of c-MYC. Therefore, mouse iPS cells were later obtained by omitting c-MYC in the gene transfection cocktail [33]. In humans, efficient production of iPS cells was demonstrated by forced expression of OCT4, NANOG, SOX2 and LIN28 [34]. Expression of these genes reprograms the cells, which are then able to differentiate into tissue types of the three embryonic germline layers. Although progresses still need to be made to improve efficiency and ensure their safety, iPS cells will certainly be used in the future for tissue engineering purposes. Because stem cells constitute the building blocks for organ development and tissue repair, the past 15 years have seen growing interest in their biology and in the mechanisms that drive these cells into specialized differentiation programs. These cells are also surprisingly easy to isolate, culture and differentiate in vitro and in vivo, therefore the drive to translate findings into clinical therapeutics has led to an increase of their applications to regenerative medicine. Stem cells hold great promises for the treatment of Parkinson disease, congenital abnormalities and spinal cord injury, as well as liver and skin regeneration. While delivering on these expectations still needs extensive work, nanotechnology will without doubt help in visualization, fate tracking, and manipulating stem cells and their environment for building and repairing tissues [35, 36]. However, there is great concern about the health consequences of using nanomaterials due to their extremely small size, their high surface area and increased surface reactivity (i.e., redox ability) as compared to larger materials. This review will describe how the combination of nanotechnology and stem cell research will dramatically advance our ability to understand and control stem cell fate decisions. This technology will lead to novel stem cell-based therapeutics for the prevention, diagnosis, and treatment of human diseases, provided that the toxicity of nanomaterials is properly assessed and understood.

Fig. 13.1.

Fig. 13.1

Differentiation potential of mesenchymal stem cells (MSCs). MSC can differentiate in many tissues including bone, cartilage, adipocytes, cardiac cells and neurons (Adapted from Meregalli et al. [18])

13.2 Nanomaterials for Labeling and In Vivo Tracking of Stem Cells

Therapies using stem cell transplantations to regenerate tissues are currently being investigated to treat a multitude of degenerative disorders such as heart failures [37, 38], brain and spinal cord injuries [39, 40], diseases of the retina [41], liver failure [42], kidney dysfunction [43] and lower limb ischemia [44]. After transplantation, stem cells are expected to engraft, differentiate into specific cells or tissues in response to the surrounding microenvironment, and restore this tissue’s functional properties. Despite these advances, the mechanisms underlying stem cell proliferation, differentiation, migration and integration within the host tissue are still poorly understood. This lack of knowledge is mainly due to our previous inability to monitor the in vivo behavior of stem cells in long-term studies because metabolic degradation and reduced photostability of cell markers such as DiI (1,1′-dioctadecyl-3,3,3′3′-tetramethylindocarbocyanine perchlorate) or DiO (3,3-Dioctadecyloxacarbocyanine perchlorate) compromised the quality of the signal over time.

The past decade focused on discovering and improving novel approaches for labeling and tracking stem cells for tissue engineering. These methods are now increasingly replacing earlier methods and mainly utilize nanoparticles. The first attempt to label and track cells injected into mice made use of superparamagnetic iron oxide (SPIO) particles for labeling of cancer cells, as well as neural and mesenchymal stem cells (Fig. 13.2) [4548]. While the transplanted cells could be tracked with magnetic resonance imaging (MRI), poor resolution impaired the analysis of the labeled cells at the cellular level. Subsequently, various SPIOs and SPIONs (super-paramagnetic iron oxide nanoparticles) have been designed that carry specific alterations of their physical, biological and chemical properties to improve their functionality and MRI tracking [49]. In particular, sensitive, non-toxic SPIONs are now designed for future human therapeutic purposes. They can be dextran-coated to minimize aggregation [45, 50], or incubated with cathionic compounds such as poly-L-Lysine [51, 52], protamine sulfate [53] or Lipofectamine [54] to facilitate absorption by non-phagocytic cells. While the sensitivity of MRI is lower than that of single photon emission tomography (SPECT), absence of exposure to ionizing radiation and long imaging window offer an obvious advantage for human clinical studies. Indeed, the labeled stem cells should retain the marker and remain viable for months to allow their long-term follow-up, and assessment of tissue function in pre-clinical and clinical trials.

Fig. 13.2.

Fig. 13.2

Long-term monitoring of SPION-labeled mesenchymal stem cells (MSCs). Rat brain striata were injected bilaterally with quinolinic acid to induce neuronal death. After 1 h, SPION-labeled MSCs were injected into one side at the same location as quinolinic acid, while the other side received only saline. Hypointense signals (black spots) indicated the presence of MSCs in all analyzed periods. Seven days after cell injection, the black spots were also observed in the contralateral injured and non-transplanted striatum, and persisted for at least 60 days after lesion (DAL) and MSC injection (From Moraes et al. [45])

Tracking transplanted cells or stem cells in vivo to monitor their tissue distribution, viability and differentiation is of great importance for tissue engineering and cell therapy. Although labeling with SPIONs has become the most common procedure for longitudinal tracking of stem cells, other nanotracers have been recently developed. These include silica and titanium nanoparticles, fluorescent gold nanoclusters, nanodiamonds and quantum dots, which all have different degrees of toxicity [5559]. In order to improve biocompatibility, alternative strategies are being developed to provide tracers amenable to clinical translation. For example, nanomaterials with dual paramagnetic and fluorescent characteristics were recently synthetized. In 2005, Vuu and colleagues reported the construction of a dual-modality contrast agent, Gadolinium-Rhodamine nanoparticles (Gd-Rd-NPs), which they used to label and track a breast cancer cell line after injection into the hind limb of mice (Fig. 13.3) [60]. Gadolinium allowed detection of the labeled tumor cells by whole animal MRI while Rhodamine was used to visualize the cells in whole animal optical imaging or in tissue sections. More recently, fluorescent biocompatible nanoparticles were produced by polymerization of methyl methacrylate-Rhodamine complexes [61]. These nanoparticles were used to label and track human amniotic fluid cells (hAFCs), which are known to contain a significant number of stem cells with pluripotent properties [62]. Labeled hAFCs were injected into the lateral brain ventricles of mice and could be tracked for at least 14 days. Labeling was strong and reliable, indicating that these novel nanoparticles, which are highly biocompatible, are a promising tool for cell therapy.

Fig. 13.3.

Fig. 13.3

In vivo optical imaging of mouse hind limb after inoculation with unlabeled tumor cells or gadolinium-rhodamine nanoparticle-labeled tumor cells. The picture shows a female mouse bearing unlabeled tumor (left limb) and Gd-Rd-NP-labeled tumor (right limb) imaged 7 days post subcutaneous inoculation (1 × 106 cells in each limb). The fluorescent signal is localized only to the labeled tumor site (From Vuu et al. [60])

Cancer stem cells (CSCs) comprise a small fraction of cancer cells and are believed by many to be at the origin of most tumors [63]. A number of studies suggest that CSCs are also at the origin of metastases. Identifying and tracking metastatic cancer cells is a significant task that can also be achieved using fluorescent-labeled nanoparticles. Indeed, efficacy of treatment can be radically improved through earlier detection of metastatic foci, which depends on sensitivity and specificity of the analytic tool. Soster and colleagues recently developed Rhodamine and/or Cy5 fluorescent silica-poly(ethylene glycol) nanoparticles (SPNs) coupled to two metastasis-specific peptides [64 ]. A poly(ethylene glycol) (PEG) shell embeds a dye-bound silica core, and presents carbonyl groups to enable covalent attachment of targeting ligands, such as antibodies, proteins, or short peptides. These nanoparticles were coupled to the peptides CGIYRLRS and CGVYSLRS that are specific for metastatic colon carcinoma. They were intravenously injected into SCID mice bearing a spleen primary tumor (site of injection of colon cancer cells) and hepatic metastases. Whole organ imaging and immunohistochemistry clearly indicated that the circulating nanoparticles left the tumor capillaries and homed nearby the tumor epithelial cells. This method might therefore provide a reliable and safe way to detect small metastases before surgery.

Despite this progress, multi-imaging methods are still needed to follow the fate of different cell types simultaneously in the host tissue. For example, in tissue engineering there is the need of imaging stem cells together with endothelial cells since vascularization of reconstructed tissues is crucial for stem cell survival, which depends on oxygen and nutrient supply. Simultaneous labeling of mesenchymal stem cells (MSCs) and endothelial cells in polysaccharides-based scaffolds was recently performed by Di Corato and colleagues (Fig. 13.4) [65]. Paramagnetic gadolinium nanoparticles were used to label MSCs while SPIONs marked endothelial cells. The cells were embedded in polysaccharide-based scaffolds that were implanted subcutaneously in the flank of mice. Visualization by high-resolution MRI demonstrated that the two cell types remained distinct, with cellular level of resolution within the mouse body. Importantly, the authors demonstrated that the labeling of the MSCs did not alter their ability to differentiate into the osteogenic, chondrogenic or adipocytic lineages. In summary, our ability to trace stem cells in vivo using nanoparticles has remarkably improved during the past 10 years. It is evident that manufacturing of more specific and efficient nanoparticles for the visualization of these cells was concomitant to improvement of high resolution imaging techniques. In addition to their reliability and efficiency, nanoparticles with potential use in regenerative medicine have become for the most part biocompatible.

Fig. 13.4.

Fig. 13.4

Simultaneous imaging of endothelial cells and mesenchymal stem cells (MSCs). Two contrast agents, iron oxide nanoparticles and gadolinium oxide nanoparticles were used to label endothelial cells and MSCs respectively. No impact on cell function, including their capacity for differentiation, was detected. The labeled cells were seeded together in a polysaccharide-based scaffold and visualized simultaneously by MRI. Labeled endothelial cells appeared in black while MSCs appeared in white in vitro and in vivo (From Di Corato et al. [65])

13.3 Nanomaterials for Manipulating Stem Cell Commitment

Directing the fate of stem cells in vivo is of paramount importance for the success of tissue regeneration and engineering. Extrinsic cues from the natural microenvironment of many stem cell types have been recently elucidated and include activation of the Wnt, Hedgehog and Notch pathways, binding of growth factors such as nerve growth factor (NGF) or bone morphogenetic protein (BMP) to specific membrane receptors, and composition of the extracellular matrix (ECM) [23, 6672]. Recently, nanomaterials have been manufactured with the intention to successfully mimic or even bypass the effect of biological molecules. For example, nanofibers can increase human umbilical cord blood stem cell expansion without inducing differentiation [73]. Incorporation of growth factors such as basic fibroblast growth factor (bFGF) during scaffold preparation by electrospinning techniques stimulates skin stem cells to accelerate wound healing [74]. Fibrin nanoscaffolds with different geometry of the fibrin structure can be prepared, which are amenable to encapsulation of growth factors [75, 76]. These growth factors can then be delivered to a target site to promote stem cell growth or differentiation where tissues need to be regenerated. Indeed, Fibrin scaffolds loaded with growth factors, such as vascular endothelial growth factor (VEGF) and nerve growth factor (NGF) stimulated blood vessel and nerve repair [77, 78 ]. Gene therapy using a bone morphogenetic (BMP) expression vector embedded into a Fibrin nanoscaffold allowed bone regeneration [79]. In addition to creating nanosystems able to deliver growth factors, many efforts have been directed at the construction of scaffolds mimicking the ECM for directing stem cell fate. Increasing evidence has revealed that the ECM itself through its geometry at the nanoscale level and interactions with cellular receptors such as integrins can modulate the shape and therefore gene expression and fate of stem cells [8085]. Mechanical forces and matrix elasticity are also of great importance [8688]. Indeed, the degree of substrate rigidity can regulate cell phenotype, which was demonstrated by the fact that an increase of matrix stiffness results in preosteoblast proliferation, while a decrease of stiffness promotes their differentiation [89]. Yet another feature of the extracellular matrix that can be mimicked using nanomaterials to direct stem cell fate is the variety of its topography, which is at the nanometer scale. Cell responses modulated by nanotopography include alignment, survival, motility, proliferation and differentiation [9093]. For example, human induced pluripotent stem cells (iPSCs) seeded onto nanostructured silicon substrates responded by elongating and aligning along the grating axis and expressed neuronal markers, while the same cells seeded on flat substrates spread randomly and conserved their pluripotent properties [94]. The same behavior can be obtained from human MSCs [90]. Interestingly, the dimension of TiO2 nanotubes, in particular their diameter, is instrumental in directing proliferation or differentiation of human MSCs into osteogenic lineages, although the size at which differentiation occurs seems controversial [9597]. Arrangement of nanopits is also instrumental in determining stem cell fate. In a study using primary human osteoblastic cells, square and hexagonal nanopit arrays perturbed the formation of mature focal adhesions and the cell-spreading process, while groove/ridge arrays promoted polarised morphology and focal adhesion complexes [98]. Similarly, a square array of nanopits can support self-renewal of human MSCs, while a slightly disorganized array will promote their osteogenic differentiation [99]. Further, the genetic profile of human primary MSCs cultured on arrays presenting nanopits and raised nanomounds indicated that these cells differentiate into bone more efficiently than in planar surfaces with dexamethasone, a glucocorticoid commonly used to direct the differentiation of MSC into osteoblasts [100]. In another study, neural progenitors embedded into a nanofiber matrix engineered to contain the laminin-specific cell-binding domain (IKVAV) differentiated preferentially into neurons [101 ]. Similarly, a nanofiber scaffold that contained RGD, a binding domain for cellular integrins in many ECM proteins, significantly improved MSCs differentiation into osteoblasts [102 ]. Altogether, these studies clearly demonstrate that mimicking the ECM by nanotopography and engineering adhesion peptides is able to influence human adult stem cell fate, in many cases without addition of growth or differentiation factors. Therefore nanomaterials can efficiently replace the ECM and facilitate tissue regeneration and engineering protocols.

13.4 Nanomaterials Supporting Stem Cells for Regenerative Medicine

Healing and regeneration of injured or lost tissues require complex interactions between stem cells, growth factors, and extracellular matrix. Scaffolds mimicking the extracellular matrix must be constructed to establish form and structure of the organ or tissue to be regenerated. Scaffolds must be physically stable in the implanted site, allow the homing, expansion and differentiation of stem cells, and be non-toxic and biodegradable.

13.4.1 Nanofibers

Since they reproduce the morphology and structure of the natural ECM, polymeric nanofibers are among the most suitable nanomaterials for tissue engineering applications. Scaffolds made of nanof bers are three-dimensional meshes formed through the non-covalent assembly of peptides with nanometer diameters, and are commonly used in attempts of regenerating bone, skin and nerve tissue (Fig. 13.5). The most common methods for the fabrication of polymeric nanofibers are self-assembly, phase separation and electrospinning [104]. Self-assembly and phase separation allows the creation of short nanofibers with very small diameters (about 10 nm). For example, the RADA16 peptide nanofiber material self-assembles into scaffolds that were used for the growth of hippocampal neurons that subsequently produced neurites and synapses [105, 106 ]. This material contains the amino acids alanine, lysine, and glutamate in repeated sequences of Arg-Ala-Asp-Ala (RADA). RADA16-I nanofibers scaffold has been used for brain tissue reconstruction in mice after trauma [107], and has also been found to promote regeneration in experimental spinal cord and brain injuries [108 ]. Stem cell embedding into the material is feasible [109111 ]. For example, Guo and colleagues embedded Schwann cells and neuroprogenitor cells (NPCs) inside a RADA16-I scaffold that integrated well within the injured spinal cord. They also demonstrated that the transplanted cells were able to survive and differentiate into neurons, astrocytes and oligodendrocytes [111]. In one of their study, Garreta and colleagues embedded embryoid bodies derived from ES cells in RADA16-I, and achieved differentiation using osteogenic medium [109]. Several functional motifs can be attached to RADA16-I, which are known to promote cell adhesion, differentiation or stem cell homing. In these conditions, neural stem cells adhered well to their matrix environment, survived and differentiated toward both the neuronal and glial phenotypes [110]. Because it is made essentially from amino acids, this type of scaffold is well tolerated and does not elicit detectable immune response or inflammation in animals. However, despite other advantages such as easiness of use and affordability, self-assembled nanofibrous scaffolds are limited in their ability to form pores wide enough to allow cell proliferation and migration since the pore size is usually between 5 and 200 nm [112 ]. In addition, nanofibers produced by self-assembly are very short and are often phagocytosed by the cells [113].

Fig. 13.5.

Fig. 13.5

Topography of different electrospun nanofibrous structures (From Reddy et al. [103])

Electrospinning is a reliable method to fabricate long continuous strands of nanofibers with a diameter ranging from nanometers to microns (50–1,000 nm) [104]. The nanofibers form a mesh with pore sizes ranging from several to tens of micrometers, which favors cell proliferation and migration [114]. Materials used in electros-pinning are natural or synthetic biopolymers or combination of both. They include poly(L-lactic acid), alginates, silicon or chitosan, sometimes in combination with collagen or gelatin [13, 115118]. ECM proteins/peptides and growth factors can also easily be incorporated into this type of scaffold [119]. Scaffolds made of electrospun nanofibers of different types have been widely used for bone and cartilage tissue engineering using MSCs [116, 120124]. Even adipose-derived stem cells (ADSCs) could proliferate and differentiate along the osteogenic pathway in absence of any induction medium when electrospun poly(L-lactic acid)/collagen nanofibers were engineered with a cell adhesion peptide and functionalized to retain calcium phosphate (hydroxyapatite) [123, 125]. This confirms the positive influence of cell-matrix interactions on stem cell survival and proliferation [126]. To increase the tensile strength of the scaffold, McCullen and colleagues encapsulated multiwalled carbon nanotubes (MWNTs) in poly (L-lactic acid) nanofibers [123]. Interestingly, their data indicated that addition of MWNTs significantly stimulated ADSCs proliferation. Electrospun poly(L-lactic acid)/gelatin fibers were also recently used as scaffolds to differentiate neural stem cells into motor neurons [127]. Also, poly-l-lactide (PLLA) and hybrid PLLA/ collagen (PLLA/Coll) scaffolds fabricated by electrospinning were recently used to differentiate MSCs into vascular endothelial cells [128].

13.4.2 Nanotubes

Nanotubes are cylindrical structures made of a single or several layers of carbon or silicon atoms. Carbon nanotubes are known for their electrical conductive capacity and strong mechanical properties, and they can be modified to accommodate stem cell adhesion. Single-walled or multi-walled carbon nanotubes (SWNTs or MWNTs) are used in regenerative medicine alone or as complement to poly(L-lactic acid) fibers to provide tensile or mechanical strength. As mentioned above, addition of MWNTs to poly(L-lactic acid) nanofibers seem to increase adipose stem cell proliferation [123], but they can also improve chondrogenic differentiation of MSCs [129]. When nanoscaffolds made of poly-(L-lactic acid) and MWNTs were modified with poly-L-lysine to improve cell adhesion, the MSC-derived chondrocytes produced more glycosaminoglycans than the same cells in control scaffolds without nanotubes or poly-L-lysine [129]. Combination of MSCs with the latter nanomaterial is therefore very promising for cartilage regeneration, one of the most difficult tasks in regenerative medicine since this tissue is not vascularized. MWNTs could also be combined to chitosan or hydroxyapatite to successfully mimic bone tissue with acceptable pore size amenable to periosteal stem cell proliferation [130]. Single-walled carbon nanotubes (SWNTs) were also functionalized with poly lactic-co-glycolic acid to form a biocompatible substrate allowing proliferation and differentiation of pre-osteoblasts that successfully expressed mature osteoblast markers [131]. Single-walled and multi-walled carbon nanotubes are also increasingly used as scaffolds for neural stem cell growth and differentiation. Neural stem cells (NSCs) can be isolated from the mammalian brain, propagated in culture and transplanted into damaged sites if provided with the necessary substrate. Differentiation of NSCs on single-walled carbon nanotubes layers was recently demonstrated by Jan and Kotov [132]. The stem cells were seeded onto this substrate using differentiation medium without epidermal growth factor (EGF), which is necessary for their proliferation. In these conditions, the NSCs could differentiate into neurons, astrocytes, and oligodendrocytes, with expression of neural markers such as nestin, microtubule-associated protein 2 (MAP2), glial fibrillary acidic protein (GFAP), and the oligodendrocyte marker O4. The same authors demonstrated that thin films of single-walled carbon nanotubes mixed with laminin induced NSCs differentiation into a functional neural network with synaptic connections. In addition, the neurons were excitable [133]. Another study recently established that carbon nanotubes seeded with subventricular zone neural progenitor cells (NPCs) could effectively repair damaged neural tissue after induced stroke [134]. Further, enhanced myotube formation from myoblasts and differentiation of MSCs into cardiomyocytes were possible after seeding these cells on carbon nanotubes mixed with polyurethane or poly-(L-lactic acid), followed by electric stimulation [135, 136]. Therefore, functionalized carbon nanotubes seem to fulfill the essential requirement for the regeneration of damaged nerve and muscle tissues, which is biocompatibility, induction of stem cell differentiation and excitation of the differentiated cells.

13.4.3 Use of ES Cells on Nanoscaffolds

Because of their pluripotent properties, ES cells have also been used in nanoscaffolds to probe their biocompatibility and potential for driving stem cell fate. Since these cells reliably differentiate into cardiomyocytes, they have been seeded into nanoscaffolds predominantly to assess the influence of the material on cardiac tissue regeneration [137139]. Nanofibers made of biodegradable polyurethane, polyethylene glycol and polycaprolactone (PCL) alone or in combination could induce the formation of cardiomyocytes from ES cells. Also, nanofiber density seemed to play an important role [138]. Other applications using ES cells include the use of electrospun PCL nanoscaffolds to enhance their differentiation into neural lineages when cultured with retinoic acid [140]. In a study by Carlberg and colleagues, human ES cells cultured on electrospun fibrous polyurethane scaffolds differentiated predominantly into dopaminergic neurons without the need of hormones or growth factors [141]. Finally, human ES cells seeded into PCL scaffolds could be differentiated into adipocytes after induction with a hormone cocktail containing retinoic acid, insulin and triiodothyronine (T3) [142].

13.5 Stem Cell Toxicity by Nanomaterials

Because the physical and chemical properties of many compounds will change when their size is reduced to nanodimensions, nanomaterials are increasingly used for novel industrial, medical and military applications. This is particularly true for carbon- and metal-based nanotubes and nanoparticles. The small size of nanomaterials modifies their behavior to such extent that quantum physics rather than classical physics is needed to understand their properties. Characteristics of a material such as solubility, absorption, color, transparency, emission wavelength, conductivity, melting point and catalytic behavior will be drastically altered as the size is reduced [143]. It is therefore likely that these novel properties will change how the materials interact with living tissues [144]. For these reasons, there are concerns that certain nanomaterials might be harmful upon inhalation, ingestion or skin contact. Indeed, due to their very small size, nanomaterials can easily translocate across cell membranes and reach critical organelles such as mitochondria, the endoplasmic reticulum and possibly the nucleus [145148]. It has also been demonstrated that the clearance of particles is reduced as the particle size decreases, therefore the deleterious effect of nanosized compounds, if any, will last longer [149]. Because stem cells are particularly sensitive to toxicants, the use of nanomaterials in bioengineering and medicine is a concern if their toxicity is not assessed.

13.5.1 Stem Cell Toxicity of Carbon Nanotubes

While toxicity of carbon nanotubes on cells and tissues in vitro and in vivo has been reported as early as 2003 [150152], in 2007 the group of Y. Hong published for the first time the effects of MWNTs at the molecular level in ES cells [148]. Because stem cells are particularly sensitive to DNA damage, they have developed specialized mechanisms to preserve genomic integrity. One of these mechanisms is the expression and phosphorylation of the protein p53, which will arrest the cell cycle to allow DNA repair by the enzyme 8-oxoguanine-DNA glycosylase 1 (OGG1). The authors demonstrated that MWNTs significantly induce the expression and phosphorylation of p53, followed by an increase in expression of OGG1 in the nucleus and mitochondria, suggesting damages in the nuclear and mitochondrial DNA. Further, the authors demonstrated that DNA double-strand breaks occurred and that the frequency of mutations increased after repair. DNA alterations often happen in response to the formation of reactive oxygen species [153, 154]. A recent study testing the influence of SWNTs on myoblasts shows that these compounds also significantly alter DNA integrity [155].

Carbon nanotube manufacturing is rapidly increasing. Because of their fibrous-like shape and durability, there are concerns that their toxic properties may be similar to those of asbestosis or other fibrous materials [156]. Indeed, MWNTs induce lung inflammation and fibrosis in mice exposed to them by inhalation [157]. Therefore strict industrial hygiene measures should be taken to limit exposure during their manipulation. As mentioned above, efforts to functionalize carbon nanotubes have ensured their biocompatibility, especially when used with human NSCs or MSCs for tissue repair. Nevertheless their long-term toxicity after scaffold implantation into an animal body still needs to be assessed.

13.5.2 Stem Cell Toxicity of Metal-Based Nanoparticles

Metal and metal oxide nanoparticles (NPs) are increasingly used in medicine, consumer products, and industry. They are used for cell tracking within an animal, drug targeting, coating of medical devices, sunscreens, detergents, clothing, printer inks and fuel additives [158]. As for other nanomaterials, nanoparticles are defined as particulate matter with at least one dimension less than 100 nm. Most NP types in current use are metal-based NPs, such as nanosilver, zinc oxide, titanium dioxide, and iron oxide. Manufactured NPs have different properties than natural NPs because of their large surface area-to-volume ratio and size that is often smaller than 20 nm. These novel physical and chemical properties imply that their interactions with cells and organelles need to be critically analyzed since many undesirable adverse effects can be triggered. Metal NPs easily penetrate tissues, epithelia and cell membranes, and pass through the blood-brain and blood-testis barriers [159161]. Ingested nanoparticles also generate DNA alterations due to production of ROS in the bone marrow, liver and lung, which are not necessarily due to the release of free metal ions [162, 163]. Therefore these compounds are likely to interfere with the biology, viability and fate of germ line and somatic stem cells. Our group was first to assess the in vitro cytotoxicity of metal nanoparticle in germ line stem cells, or spermatogonial stem cells (SSCs) [164]. Because these cells maintain the production of sperm throughout life, preserving their viability and integrity is of paramount importance. Adverse effects on germ line stem cells maintenance, maturation and differentiation can inhibit fertility, cause cancer, and may have negative effects on the development and fertility of offspring. Our work demonstrated that nanoparticles induced production of reactive oxygen species (ROS), decrease of metabolism and apoptosis in these cells, and that the degree of toxicity depended on the metal used for nanoparticle fabrication. Silver nanoparticles were significantly more toxic than aluminum oxide nanoparticles while molybdenum nanoparticles had no deleterious effect on these stem cells (Fig. 13.6).

Fig. 13.6.

Fig. 13.6

Toxic effects of metal-based nanoparticles. The effect of silver and molybdenum nanoparticles on the metabolic activity of the C18–4 spermatogonial stem cells was evaluated after 48 h incubation. At the end of the incubation period, mitochondrial function was determined by the MTS reduction assay. (a) MTS reduction in presence of different concentrations of cadmium chloride and cadmium oxide. Cadmium chloride is a known toxicant and is used here as a positive control. (b) MTS reduction in presence of different concentrations of silver carbonate and silver nanoparticles (Ag—15 nm). (c) MTS reduction in presence of different concentrations of sodium molybdate and molybdenum nanoparticles. Overall silver nanoparticles were as toxic as cadmium chloride. Molybdenum nanoparticles were toxic only a high concentrations (From Braydich-Stolle et al. [164])

13.5.2.1 Toxicity of Zinc Oxide Nanoparticles

Deng and colleagues were among the first to test the effect of ZnO nanoparticles on NSCs in vitro [165]. They showed that at low concentrations (<12 ppm) ZnO nanoparticles had no effect on cell viability, but that apoptosis and necrosis increased at concentrations above 12 ppm. They also concluded that free Zn2+ ions were responsible for high doses effects. While scientists generally agree that ZnO nanoparticles are neither toxic to differentiated cells nor to stem cells, it was recently demonstrated that their in vitro effects depend on the number and type of cells seeded [166, 167]. For example, Taccola and colleagues provided evidence that ZnO nanoparticles selectively kill rapidly proliferating cells such as MSCs, but have no effect on the cells once they are differentiated along the osteogenic pathway [167]. Similarly ZnO nanoparticles seem to selectively kill cancer cells, possibly through ROS production leading to cell apoptosis, and is now used for certain therapies [168].

13.5.2.2 Toxicity of Aluminum Oxide Nanoparticles

Aluminum oxide nanoparticles (ANPs) are widely used, and several studies have shown that ANPs negatively affect cellular morphology and cellular components. ANPs induce oxidative stress, which leads to apoptosis, DNA damage and protein degradation in vitro, while their ingestion affects neurobehavioral patterns [169171]. However, few studies have tested their toxicity specifically in stem cells. Only the recent investigation of Alshatwi and colleagues with MSCs indicated that ANPs might induce changes in cell morphology, decrease in cell viability and upregulation of apoptosis pathways. These effects were concentration-dependent but the physiological relevance of the doses used was not discussed [172].

13.5.2.3 Toxicity of Silver Nanoparticles

The anti-bacterial effects of silver have been known and exploited for centuries, in particular for the treatment of wounds and burns [173]. Silver is used as an antibacterial surface coating on medical devices such as stents, catheters and heart valves, and is non-toxic to human and animals in its bulk chemical form [174]. However increasing amounts of silver nanomaterials, in particular silver nanoparticles, are now finding their way into consumer products as additives to sprays, as surfactants for textiles and food containers, and as drinking water disinfectants to name a few applications. This trend is concerning because many studies have now demonstrated that the toxicity of nanosilver is much greater than that of most carbon-based and metal-based nanomaterials [143]. The cellular toxic effects of silver nanoparticles are size-dependent, and increase as the particle size decreases [175, 176]. They include reduction of mitochondrial function, increase of membrane leakage and increase of production of reactive oxygen species (ROS) [164]. They also can deplete cellular glutathione, an antioxidant [177]. As mentioned before, ROS cause DNA damages such as base oxidations, base changes or single and double-strand breaks that can lead to cell death or to mutations causing cancers. Recent studies in ES cells demonstrated that silver nanoparticles up-regulated expression and phosphorylation of the protein p53, induced DNA double-strand breaks, decreased cellular metabolism and promoted apoptosis [178]. In this work, silver nanoparticles with different surface chemistries were used: uncoated nanoparticles and particles coated with a polysaccharide to ensure biocompatibility and dispersion. Interestingly, in ES cells the coated silver nanoparticles produced more damages than the uncoated nanoparticles, but the cause remains unclear. Our group also recently investigated some of the mechanisms causing the toxic effects of silver nanoparticles in germ line stem cells [176]. Silver nanoparticles were hydrocarbon or polysaccharide-coated to study the influence of surface chemistry. We demonstrated that at low doses (<10 μg/ml), silver nanoparticles interfered with the glial cell line-derived neurotrophic factor (GDNF) signaling pathway, which is crucial for germ line stem cell self-renewal. More precisely, silver nanoparticles inhibited the phosphorylation of the membrane-associated FYN kinase downstream of the RET (REarranged during Transfection) transmembrane receptor after activation by GDNF. Silver nanoparticles did not interfere with GDNF binding to its receptor nor with receptor phosphorylation. Further, we demonstrated that silver nanoparticles reduced germ line stem cell viability and proliferation in a size- and concentration-dependent manner and that particle coating had no influence on the magnitude of the toxic effects. The effect of silver nanoparticles has since been tested on human MSCs [179]. When exposed to the dose of 10 μg/ ml, these cells experienced DNA damage and their viability decreased, as shown for other stem cell types, but their ability to migrate was not impaired. However, Samberg and colleagues recently demonstrated that silver nanoparticles exert minimal toxicity to human adipose tissue-derived stem cells (ADSCs) and do not impair their differentiation [180]. Therefore, for the same sizes and concentrations, stem cell effects of silver nanoparticles might be cell type-dependent, with germ line stem cells particularly sensitive to this potential toxicant.

Conclusion

Nanomaterials have found many applications in stem cell research including tracking, delivering, and controlling the fate of these cells for tissue engineering. These applications have already started to transform several areas of cell biology and medicine. However, strategies allowing large-scale stem cell expansion before their differentiation are still lacking. In addition, the creation of complex tissues is still in infancy and will need the ability to direct the growth and differentiation of different cell types in three dimensions within biocompatible scaffolds. Furthermore, toxicity of certain nanoscaffolds such as MWNTs is still a concern since coatings that ensure their biocompatibility might not be stable within the tissue to be repaired. Finally, as the demand for nanomaterials is increasing, potential inhalation during manufacturing or packing, and their accumulation in the environment should be carefully monitored to ensure human safety.

References

  • 1.Cui D, Gao H. Advance and prospect of bionanomaterials. Biotechnol Prog. 2003;19(3):683–692. doi: 10.1021/bp025791i. [DOI] [PubMed] [Google Scholar]
  • 2.Rajangam T, An SS. Fibrinogen and fibrin based micro and nano scaffolds incorporated with drugs, proteins, cells and genes for therapeutic biomedical applications. Int J Nanomedicine. 2013;8:3641–3662. doi: 10.2147/IJN.S43945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Liu W, Thomopoulos S, Xia Y. Electrospun nanofibers for regenerative medicine. Adv Healthc Mater. 2012;1(1):10–25. doi: 10.1002/adhm.201100021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ozpolat B, Sood AK, Lopez-Berestein G. Nanomedicine based approaches for the delivery of siRNA in cancer. J Intern Med. 2010;267(1):44–53. doi: 10.1111/j.1365-2796.2009.02191.x. [DOI] [PubMed] [Google Scholar]
  • 5.Duncan R. Polymer conjugates as anticancer nanomedicines. Nat Rev Cancer. 2006;6(9):688–701. doi: 10.1038/nrc1958. [DOI] [PubMed] [Google Scholar]
  • 6.Uskokovic V. Entering the era of nanoscience: time to be so small. J Biomed Nanotechnol. 2013;9(9):1441–1470. doi: 10.1166/jbn.2013.1642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Pandey PC, Chauhan DS, Singh V. Role of nanostructured networks as analytical tools for biological systems. Front Biosci (Elite Ed) 2013;5:622–642. doi: 10.2741/e644. [DOI] [PubMed] [Google Scholar]
  • 8.Vilela D, Garoz J, Colina A, Gonzalez MC, Escarpa A. Carbon nanotubes press-transferred on PMMA substrates as exclusive transducers for electrochemical microfluidic sensing. Anal Chem. 2012;84(24):10838–10844. doi: 10.1021/ac303049x. [DOI] [PubMed] [Google Scholar]
  • 9.Ramulu TS, Venu R, Sinha B, Lim B, Jeon SJ, Yoon SS, Kim CG. Nanowires array modified electrode for enhanced electrochemical detection of nucleic acid. Biosens Bioelectron. 2013;40(1):258–264. doi: 10.1016/j.bios.2012.07.034. [DOI] [PubMed] [Google Scholar]
  • 10.Kruss S, Hilmer AJ, Zhang J, Reuel NF, Mu B, Strano MS. Carbon nanotubes as optical biomedical sensors. Adv Drug Deliv Rev. 2013;65(15):1933–1950. doi: 10.1016/j.addr.2013.07.015. [DOI] [PubMed] [Google Scholar]
  • 11.Riegler J, Nann T. Application of luminescent nanocrystals as labels for biological molecules. Anal Bioanal Chem. 2004;379(7–8):913–919. doi: 10.1007/s00216-004-2706-y. [DOI] [PubMed] [Google Scholar]
  • 12.Lisdat F, Schafer D, Kapp A. Quantum dots on electrodes – new tools for bioelectroanalysis. Anal Bioanal Chem. 2013;405(11):3739–3752. doi: 10.1007/s00216-013-6789-1. [DOI] [PubMed] [Google Scholar]
  • 13.Premnath P, Tan B, Venkatakrishnan K. Direct patterning of free standing three dimensional silicon nanofibrous network to facilitate multi-dimensional growth of fibroblasts and osteoblasts. J Biomed Nanotechnol. 2013;9(11):1875–1881. doi: 10.1166/jbn.2013.1689. [DOI] [PubMed] [Google Scholar]
  • 14.Martinelli V, Cellot G, Toma FM, Long CS, Caldwell JH, Zentilin L, Giacca M, Turco A, Prato M, Ballerini L, Mestroni L. Carbon nanotubes promote growth and spontaneous electrical activity in cultured cardiac myocytes. Nano Lett. 2012;12(4):1831–1838. doi: 10.1021/nl204064s. [DOI] [PubMed] [Google Scholar]
  • 15.Solanki A, Chueng ST, Yin PT, Kappera R, Chhowalla M, Lee KB. Axonal alignment and enhanced neuronal differentiation of neural stem cells on graphene-nanoparticle hybrid structures. Adv Mater. 2013;25(38):5477–5482. doi: 10.1002/adma.201302219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Sheets K, Wunsch S, Ng C, Nain AS. Shape-dependent cell migration and focal adhesion organization on suspended and aligned nanofiber scaffolds. Acta Biomater. 2013;9(7):7169–7177. doi: 10.1016/j.actbio.2013.03.042. [DOI] [PubMed] [Google Scholar]
  • 17.Pelton TA, Bettess MD, Lake J, Rathjen J, Rathjen PD. Developmental complexity of early mammalian pluripotent cell populations in vivo and in vitro. Reprod Fertil Dev. 1998;10(7–8):535–549. doi: 10.1071/rd98084. [DOI] [PubMed] [Google Scholar]
  • 18.Meregalli M, Farini A, Torrente Y. Mesenchymal stem cells as muscle reservoir. J Stem Cell Res Ther. 2011;1:105. [Google Scholar]
  • 19.Bobis S, Jarocha D, Majka M. Mesenchymal stem cells: characteristics and clinical applications. Folia Histochem Cytobiol. 2006;44(4):215–230. [PubMed] [Google Scholar]
  • 20.Konno M, Hamabe A, Hasegawa S, Ogawa H, Fukusumi T, Nishikawa S, Ohta K, Kano Y, Ozaki M, Noguchi Y, Sakai D, Kudoh T, Kawamoto K, Eguchi H, Satoh T, Tanemura M, Nagano H, Doki Y, Mori M, Ishii H. Adipose-derived mesenchymal stem cells and regenerative medicine. Dev Growth Differ. 2013;55(3):309–318. doi: 10.1111/dgd.12049. [DOI] [PubMed] [Google Scholar]
  • 21.Wilkinson AC, Gottgens B. Transcriptional regulation of haematopoietic stem cells. Adv Exp Med Biol. 2013;786:187–212. doi: 10.1007/978-94-007-6621-1_11. [DOI] [PubMed] [Google Scholar]
  • 22.Clevers H. The intestinal crypt, a prototype stem cell compartment. Cell. 2013;154(2):274–284. doi: 10.1016/j.cell.2013.07.004. [DOI] [PubMed] [Google Scholar]
  • 23.Yeung TM, Chia LA, Kosinski CM, Kuo CJ. Regulation of self-renewal and differentiation by the intestinal stem cell niche. Cell Mol Life Sci. 2011;68(15):2513–2523. doi: 10.1007/s00018-011-0687-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Blanpain C, Fuchs E. Epidermal homeostasis: a balancing act of stem cells in the skin. Nat Rev Mol Cell Biol. 2009;10(3):207–217. doi: 10.1038/nrm2636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Phillips BT, Gassei K, Orwig KE. Spermatogonial stem cell regulation and spermatogenesis. Philos Trans R Soc Lond B Biol Sci. 2010;365(1546):1663–1678. doi: 10.1098/rstb.2010.0026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kanatsu-Shinohara M, Inoue K, Lee J, Yoshimoto M, Ogonuki N, Miki H, Baba S, Kato T, Kazuki Y, Toyokuni S, Toyoshima M, Niwa O, Oshimura M, Heike T, Nakahata T, Ishino F, Ogura A, Shinohara T. Generation of pluripotent stem cells from neonatal mouse testis. Cell. 2004;119(7):1001–1012. doi: 10.1016/j.cell.2004.11.011. [DOI] [PubMed] [Google Scholar]
  • 27.Simon L, Ekman GC, Kostereva N, Zhang Z, Hess RA, Hofmann MC, Cooke PS. Direct transdifferentiation of stem/progenitor spermatogonia into reproductive and nonreproductive tissues of all germ layers. Stem Cells. 2009;27(7):1666–1675. doi: 10.1002/stem.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kossack N, Meneses J, Shefi S, Nguyen HN, Chavez S, Nicholas C, Gromoll J, Turek PJ, Reijo-Pera RA. Isolation and characterization of pluripotent human spermatogonial stem cell-derived cells. Stem Cells. 2009;27(1):138–149. doi: 10.1634/stemcells.2008-0439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Golestaneh N, Kokkinaki M, Pant D, Jiang J, DeStefano D, Fernandez-Bueno C, Rone JD, Haddad BR, Gallicano GI, Dym M. Pluripotent stem cells derived from adult human testes. Stem Cells Dev. 2009;18(8):1115–1126. doi: 10.1089/scd.2008.0347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  • 31.Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature. 2007;448(7151):313–317. doi: 10.1038/nature05934. [DOI] [PubMed] [Google Scholar]
  • 32.Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, Hochedlinger K, Bernstein BE, Jaenisch R. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature. 2007;448(7151):318–324. doi: 10.1038/nature05944. [DOI] [PubMed] [Google Scholar]
  • 33.Nakagawa M, Koyanagi M, Tanabe K, Takahashi K, Ichisaka T, Aoi T, Okita K, Mochiduki Y, Takizawa N, Yamanaka S. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol. 2008;26(1):101–106. doi: 10.1038/nbt1374. [DOI] [PubMed] [Google Scholar]
  • 34.Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R, Slukvin II, Thomson JA. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318(5858):1917–1920. doi: 10.1126/science.1151526. [DOI] [PubMed] [Google Scholar]
  • 35.Jiang J, Papoutsakis ET. Stem-cell niche based comparative analysis of chemical and nanomechanical material properties impacting ex vivo expansion and differentiation of hematopoietic and mesenchymal stem cells. Adv Healthc Mater. 2013;2(1):25–42. doi: 10.1002/adhm.201200169. [DOI] [PubMed] [Google Scholar]
  • 36.Bressan E, Carraro A, Ferroni L, Gardin C, Sbricoli L, Guazzo R, Stellini E, Roman M, Pinton P, Sivolella S, Zavan B. Nanotechnology to drive stem cell commitment. Nanomedicine (Lond) 2013;8(3):469–486. doi: 10.2217/nnm.13.12. [DOI] [PubMed] [Google Scholar]
  • 37.Schachinger V, Assmus B, Britten MB, Honold J, Lehmann R, Teupe C, Abolmaali ND, Vogl TJ, Hofmann WK, Martin H, Dimmeler S, Zeiher AM. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction: final one-year results of the TOPCARE-AMI Trial. J Am Coll Cardiol. 2004;44(8):1690–1699. doi: 10.1016/j.jacc.2004.08.014. [DOI] [PubMed] [Google Scholar]
  • 38.Sanganalmath SK, Bolli R. Cell therapy for heart failure: a comprehensive overview of experimental and clinical studies, current challenges, and future directions. Circ Res. 2013;113(6):810–834. doi: 10.1161/CIRCRESAHA.113.300219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Tsukamoto A, Uchida N, Capela A, Gorba T, Huhn S. Clinical translation of human neural stem cells. Stem Cell Res Ther. 2013;4(4):102. doi: 10.1186/scrt313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Mothe AJ, Tator CH. Review of transplantation of neural stem/progenitor cells for spinal cord injury. Int J Dev Neurosci. 2013;31(7):701–713. doi: 10.1016/j.ijdevneu.2013.07.004. [DOI] [PubMed] [Google Scholar]
  • 41.Ramsden CM, Powner MB, Carr AJ, Smart MJ, da Cruz L, Coffey PJ. Stem cells in retinal regeneration: past, present and future. Development. 2013;140(12):2576–2585. doi: 10.1242/dev.092270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ordovas L, Park Y, Verfaillie CM. Stem cells and liver engineering. Biotechnol Adv. 2013;31(7):1094–1107. doi: 10.1016/j.biotechadv.2013.07.002. [DOI] [PubMed] [Google Scholar]
  • 43.Yokote S, Yokoo T. Stem cells in kidney regeneration. Curr Med Chem. 2012;19(35):6009–6017. [PubMed] [Google Scholar]
  • 44.Hart CA, Tsui J, Khanna A, Abraham DJ, Baker DM. Stem cells of the lower limb: their role and potential in management of critical limb ischemia. Exp Biol Med. 2013;238(10):1118–1126. doi: 10.1177/1535370213503275. [DOI] [PubMed] [Google Scholar]
  • 45.Moraes L, Vasconcelos-dos-Santos A, Santana FC, Godoy MA, Rosado-de-Castro PH, Jasmin, Azevedo-Pereira RL, Cintra WM, Gasparetto EL, Santiago MF, Mendez-Otero R. Neuroprotective effects and magnetic resonance imaging of mesenchymal stem cells labeled with SPION in a rat model of Huntington’s disease. Stem Cell Res. 2012;9(2):143–155. doi: 10.1016/j.scr.2012.05.005. [DOI] [PubMed] [Google Scholar]
  • 46.Frank JA, Zywicke H, Jordan EK, Mitchell J, Lewis BK, Miller B, Bryant LH, Jr, Bulte JW. Magnetic intracellular labeling of mammalian cells by combining (FDA-approved) superparamagnetic iron oxide MR contrast agents and commonly used transfection agents. Acad Radiol. 2002;9(Suppl 2):S484–S487. doi: 10.1016/s1076-6332(03)80271-4. [DOI] [PubMed] [Google Scholar]
  • 47.Kraitchman DL, Heldman AW, Atalar E, Amado LC, Martin BJ, Pittenger MF, Hare JM, Bulte JW. In vivo magnetic resonance imaging of mesenchymal stem cells in myocardial infarction. Circulation. 2003;107(18):2290–2293. doi: 10.1161/01.CIR.0000070931.62772.4E. [DOI] [PubMed] [Google Scholar]
  • 48.Bulte JW, Douglas T, Witwer B, Zhang SC, Strable E, Lewis BK, Zywicke H, Miller B, van Gelderen P, Moskowitz BM, Duncan ID, Frank JA. Magnetodendrimers allow endosomal magnetic labeling and in vivo tracking of stem cells. Nat Biotechnol. 2001;19(12):1141–1147. doi: 10.1038/nbt1201-1141. [DOI] [PubMed] [Google Scholar]
  • 49.Wang YX, Xuan S, Port M, Idee JM. Recent advances in superparamagnetic iron oxide nanoparticles for cellular imaging and targeted therapy research. Curr Pharm Des. 2013;19(37):6575–6593. doi: 10.2174/1381612811319370003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Molday RS, MacKenzie D. Immunospecific ferromagnetic iron-dextran reagents for the labeling and magnetic separation of cells. J Immunol Methods. 1982;52(3):353–367. doi: 10.1016/0022-1759(82)90007-2. [DOI] [PubMed] [Google Scholar]
  • 51.Babic M, Horak D, Trchova M, Jendelova P, Glogarova K, Lesny P, Herynek V, Hajek M, Sykova E. Poly(L-lysine)-modified iron oxide nanoparticles for stem cell labeling. Bioconjug Chem. 2008;19(3):740–750. doi: 10.1021/bc700410z. [DOI] [PubMed] [Google Scholar]
  • 52.Arbab AS, Bashaw LA, Miller BR, Jordan EK, Bulte JW, Frank JA. Intracytoplasmic tagging of cells with ferumoxides and transfection agent for cellular magnetic resonance imaging after cell transplantation: methods and techniques. Transplantation. 2003;76(7):1123–1130. doi: 10.1097/01.TP.0000089237.39220.83. [DOI] [PubMed] [Google Scholar]
  • 53.Jasmin, Torres AL, Jelicks L, de Carvalho AC, Spray DC, Mendez-Otero R. Labeling stem cells with superparamagnetic iron oxide nanoparticles: analysis of the labeling efficacy by microscopy and magnetic resonance imaging. Methods Mol Biol. 2012;906:239–252. doi: 10.1007/978-1-61779-953-2_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Frank JA, Miller BR, Arbab AS, Zywicke HA, Jordan EK, Lewis BK, Bryant LH, Jr, Bulte JW. Clinically applicable labeling of mammalian and stem cells by combining superparamagnetic iron oxides and transfection agents. Radiology. 2003;228(2):480–487. doi: 10.1148/radiol.2281020638. [DOI] [PubMed] [Google Scholar]
  • 55.Wang HH, Lin CA, Lee CH, Lin YC, Tseng YM, Hsieh CL, Chen CH, Tsai CH, Hsieh CT, Shen JL, Chan WH, Chang WH, Yeh HI. Fluorescent gold nanoclusters as a biocompatible marker for in vitro and in vivo tracking of endothelial cells. ACS Nano. 2011;5(6):4337–4344. doi: 10.1021/nn102752a. [DOI] [PubMed] [Google Scholar]
  • 56.Rehor I, Vilimova V, Jendelova P, Kubicek V, Jirak D, Herynek V, Kapcalova M, Kotek J, Cerny J, Hermann P, Lukes I. Phosphonate-titanium dioxide assemblies: platform for multimodal diagnostic-therapeutic nanoprobes. J Med Chem. 2011;54(14):5185–5194. doi: 10.1021/jm200449y. [DOI] [PubMed] [Google Scholar]
  • 57.Fang CY, Vaijayanthimala V, Cheng CA, Yeh SH, Chang CF, Li CL, Chang HC. The exocytosis of fluorescent nanodiamond and its use as a long-term cell tracker. Small. 2011;7(23):3363–3370. doi: 10.1002/smll.201101233. [DOI] [PubMed] [Google Scholar]
  • 58.Lee JK, Chun SY, Im JY, Jin HK, Kwon TG, Bae JS. Specific labeling of neurogenic, endothelial, and myogenic differentiated cells derived from human amniotic fluid stem cells with silica-coated magnetic nanoparticles. J Vet Med Sci. 2012;74(8):969–975. doi: 10.1292/jvms.12-0016. [DOI] [PubMed] [Google Scholar]
  • 59.Chen Y. Quantum dots for labeling live cells. Methods Mol Biol. 2012;906:193–198. doi: 10.1007/978-1-61779-953-2_14. [DOI] [PubMed] [Google Scholar]
  • 60.Vuu K, Xie J, McDonald MA, Bernardo M, Hunter F, Zhang Y, Li K, Bednarski M, Guccione S. Gadolinium-rhodamine nanoparticles for cell labeling and tracking via magnetic resonance and optical imaging. Bioconjug Chem. 2005;16(4):995–999. doi: 10.1021/bc050085z. [DOI] [PubMed] [Google Scholar]
  • 61.Cova L, Bigini P, Diana V, Sitia L, Ferrari R, Pesce RM, Khalaf R, Bossolasco P, Ubezio P, Lupi M, Tortarolo M, Colombo L, Giardino D, Silani V, Morbidelli M, Salmona M, Moscatelli D. Biocompatible fluorescent nanoparticles for in vivo stem cell tracking. Nanotechnology. 2013;24(24):245603. doi: 10.1088/0957-4484/24/24/245603. [DOI] [PubMed] [Google Scholar]
  • 62.Cananzi M, Atala A, De Coppi P. Stem cells derived from amniotic fluid: new potentials in regenerative medicine. Reprod Biomed Online. 2009;18(Suppl 1):17–27. doi: 10.1016/s1472-6483(10)60111-3. [DOI] [PubMed] [Google Scholar]
  • 63.Visvader JE, Lindeman GJ. Cancer stem cells: current status and evolving complexities. Cell Stem Cell. 2012;10(6):717–728. doi: 10.1016/j.stem.2012.05.007. [DOI] [PubMed] [Google Scholar]
  • 64.Soster M, Juris R, Bonacchi S, Genovese D, Montalti M, Rampazzo E, Zaccheroni N, Garagnani P, Bussolino F, Prodi L, Marchio S. Targeted dual-color silica nanoparticles provide univocal identification of micrometastases in preclinical models of colorectal cancer. Int J Nanomedicine. 2012;7:4797–4807. doi: 10.2147/IJN.S33825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Di Corato R, Gazeau F, Le Visage C, Fayol D, Levitz P, Lux F, Letourneur D, Luciani N, Tillement O, Wilhelm C. High-resolution cellular MRI: gadolinium and iron oxide nanoparticles for in-depth dual-cell imaging of engineered tissue constructs. ACS Nano. 2013;7(9):7500–7512. doi: 10.1021/nn401095p. [DOI] [PubMed] [Google Scholar]
  • 66.Fakhry M, Hamade E, Badran B, Buchet R, Magne D. Molecular mechanisms of mesenchymal stem cell differentiation towards osteoblasts. World J Stem Cells. 2013;5(4):136–148. doi: 10.4252/wjsc.v5.i4.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Blank U, Karlsson G, Karlsson S. Signaling pathways governing stem-cell fate. Blood. 2008;111(2):492–503. doi: 10.1182/blood-2007-07-075168. [DOI] [PubMed] [Google Scholar]
  • 68.Hofmann MC. Gdnf signaling pathways within the mammalian spermatogonial stem cell niche. Mol Cell Endocrinol. 2008;288(1–2):95–103. doi: 10.1016/j.mce.2008.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Garcia TX, Hofmann MC. NOTCH signaling in Sertoli cells regulates gonocyte fate. Cell Cycle. 2013;12(16):2538–2545. doi: 10.4161/cc.25627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nagano MC, Yeh JR. The identity and fate decision control of spermatogonial stem cells: where is the point of no return? Curr Top Dev Biol. 2013;102:61–95. doi: 10.1016/B978-0-12-416024-8.00003-9. [DOI] [PubMed] [Google Scholar]
  • 71.Fuchs E. Finding one’s niche in the skin. Cell Stem Cell. 2009;4(6):499–502. doi: 10.1016/j.stem.2009.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Watt FM, Huck WT. Role of the extracellular matrix in regulating stem cell fate. Nat Rev Mol Cell Biol. 2013;14(8):467–473. doi: 10.1038/nrm3620. [DOI] [PubMed] [Google Scholar]
  • 73.Das H, Abdulhameed N, Joseph M, Sakthivel R, Mao HQ, Pompili VJ. Ex vivo nanofiber expansion and genetic modification of human cord blood-derived progenitor/stem cells enhances vasculogenesis. Cell Transplant. 2009;18(3):305–318. doi: 10.3727/096368909788534870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yang Y, Xia T, Zhi W, Wei L, Weng J, Zhang C, Li X. Promotion of skin regeneration in diabetic rats by electrospun core-sheath fibers loaded with basic fibroblast growth factor. Biomaterials. 2011;32(18):4243–4254. doi: 10.1016/j.biomaterials.2011.02.042. [DOI] [PubMed] [Google Scholar]
  • 75.Janmey PA, Winer JP, Weisel JW. Fibrin gels and their clinical and bioengineering applications. J R Soc Interface. 2009;6(30):1–10. doi: 10.1098/rsif.2008.0327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ahmed TA, Dare EV, Hincke M. Fibrin: a versatile scaffold for tissue engineering applications. Tissue Eng Part B Rev. 2008;14(2):199–215. doi: 10.1089/ten.teb.2007.0435. [DOI] [PubMed] [Google Scholar]
  • 77.Ehrbar M, Metters A, Zammaretti P, Hubbell JA, Zisch AH. Endothelial cell proliferation and progenitor maturation by fibrin-bound VEGF variants with differential susceptibilities to local cellular activity. J Control Release. 2005;101(1–3):93–109. doi: 10.1016/j.jconrel.2004.07.018. [DOI] [PubMed] [Google Scholar]
  • 78.Bhang SH, Jeon O, Choi CY, Kwon YH, Kim BS. Controlled release of nerve growth factor from fibrin gel. J Biomed Mater Res A. 2007;80(4):998–1002. doi: 10.1002/jbm.a.31050. [DOI] [PubMed] [Google Scholar]
  • 79.Lee TC, Ho JT, Hung KS, Chen WF, Chung YH, Yang YL. Bone morphogenetic protein gene therapy using a fibrin scaffold for a rabbit spinal-fusion experiment. Neurosurgery. 2006;58(2):373–380. doi: 10.1227/01.NEU.0000199725.03186.F6. discussion 373–380. [DOI] [PubMed] [Google Scholar]
  • 80.Sun Y, Chen CS, Fu J. Forcing stem cells to behave: a biophysical perspective of the cellular microenvironment. Annu Rev Biophys. 2012;41:519–542. doi: 10.1146/annurev-biophys-042910-155306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Guilak F, Cohen DM, Estes BT, Gimble JM, Liedtke W, Chen CS. Control of stem cell fate by physical interactions with the extracellular matrix. Cell Stem Cell. 2009;5(1):17–26. doi: 10.1016/j.stem.2009.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Huebsch N, Arany PR, Mao AS, Shvartsman D, Ali OA, Bencherif SA, Rivera-Feliciano J, Mooney DJ. Harnessing traction-mediated manipulation of the cell/matrix interface to control stem-cell fate. Nat Mater. 2010;9(6):518–526. doi: 10.1038/nmat2732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Trappmann B, Gautrot JE, Connelly JT, Strange DG, Li Y, Oyen ML, Cohen Stuart MA, Boehm H, Li B, Vogel V, Spatz JP, Watt FM, Huck WT. Extracellular-matrix tethering regulates stem-cell fate. Nat Mater. 2012;11(7):642–649. doi: 10.1038/nmat3339. [DOI] [PubMed] [Google Scholar]
  • 84.Kong HJ, Boontheekul T, Mooney DJ. Quantifying the relation between adhesion ligand-receptor bond formation and cell phenotype. Proc Natl Acad Sci U S A. 2006;103(49):18534–18539. doi: 10.1073/pnas.0605960103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chu C, Schmidt JJ, Carnes K, Zhang Z, Kong HJ, Hofmann MC. Three-dimensional synthetic niche components to control germ cell proliferation. Tissue Eng Part A. 2009;15(2):255–262. doi: 10.1089/ten.tea.2008.0100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Simmons CA, Matlis S, Thornton AJ, Chen S, Wang CY, Mooney DJ. Cyclic strain enhances matrix mineralization by adult human mesenchymal stem cells via the extracellular signal-regulated kinase (ERK1/2) signaling pathway. J Biomech. 2003;36(8):1087–1096. doi: 10.1016/s0021-9290(03)00110-6. [DOI] [PubMed] [Google Scholar]
  • 87.Gwak SJ, Bhang SH, Kim IK, Kim SS, Cho SW, Jeon O, Yoo KJ, Putnam AJ, Kim BS. The effect of cyclic strain on embryonic stem cell-derived cardiomyocytes. Biomaterials. 2008;29(7):844–856. doi: 10.1016/j.biomaterials.2007.10.050. [DOI] [PubMed] [Google Scholar]
  • 88.Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126(4):677–689. doi: 10.1016/j.cell.2006.06.044. [DOI] [PubMed] [Google Scholar]
  • 89.Kong HJ, Polte TR, Alsberg E, Mooney DJ. FRET measurements of cell-traction forces and nanoscale clustering of adhesion ligands varied by substrate stiffness. Proc Natl Acad Sci U S A. 2005;102(12):4300–4305. doi: 10.1073/pnas.0405873102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Yim EK, Pang SW, Leong KW. Synthetic nanostructures inducing differentiation of human mesenchymal stem cells into neuronal lineage. Exp Cell Res. 2007;313(9):1820–1829. doi: 10.1016/j.yexcr.2007.02.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.McMurray RJ, Gadegaard N, Tsimbouri PM, Burgess KV, McNamara LE, Tare R, Murawski K, Kingham E, Oreffo RO, Dalby MJ. Nanoscale surfaces for the long-term maintenance of mesenchymal stem cell phenotype and multipotency. Nat Mater. 2011;10(8):637–644. doi: 10.1038/nmat3058. [DOI] [PubMed] [Google Scholar]
  • 92.Lee MR, Kwon KW, Jung H, Kim HN, Suh KY, Kim K, Kim KS. Direct differentiation of human embryonic stem cells into selective neurons on nanoscale ridge/groove pattern arrays. Biomaterials. 2010;31(15):4360–4366. doi: 10.1016/j.biomaterials.2010.02.012. [DOI] [PubMed] [Google Scholar]
  • 93.Chen W, Villa-Diaz LG, Sun Y, Weng S, Kim JK, Lam RH, Han L, Fan R, Krebsbach PH, Fu J. Nanotopography influences adhesion, spreading, and self-renewal of human embryonic stem cells. ACS Nano. 2012;6(5):4094–4103. doi: 10.1021/nn3004923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Pan F, Zhang M, Wu G, Lai Y, Greber B, Scholer HR, Chi L. Topographic effect on human induced pluripotent stem cells differentiation towards neuronal lineage. Biomaterials. 2013;34(33):8131–8139. doi: 10.1016/j.biomaterials.2013.07.025. [DOI] [PubMed] [Google Scholar]
  • 95.Oh S, Brammer KS, Li YS, Teng D, Engler AJ, Chien S, Jin S. Stem cell fate dictated solely by altered nanotube dimension. Proc Natl Acad Sci U S A. 2009;106(7):2130–2135. doi: 10.1073/pnas.0813200106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Park J, Bauer S, von der Mark K, Schmuki P. Nanosize and vitality: TiO2 nanotube diameter directs cell fate. Nano Lett. 2007;7(6):1686–1691. doi: 10.1021/nl070678d. [DOI] [PubMed] [Google Scholar]
  • 97.Park J, Bauer S, Schlegel KA, Neukam FW, von der Mark K, Schmuki P. TiO2 nanotube surfaces: 15 nm–an optimal length scale of surface topography for cell adhesion and differentiation. Small. 2009;5(6):666–671. doi: 10.1002/smll.200801476. [DOI] [PubMed] [Google Scholar]
  • 98.Biggs MJ, Richards RG, Gadegaard N, Wilkinson CD, Oreffo RO, Dalby MJ. The use of nanoscale topography to modulate the dynamics of adhesion formation in primary osteoblasts and ERK/ MAPK signalling in STRO-1+ enriched skeletal stem cells. Biomaterials. 2009;30(28):5094–5103. doi: 10.1016/j.biomaterials.2009.05.049. [DOI] [PubMed] [Google Scholar]
  • 99.Dalby MJ, Gadegaard N, Tare R, Andar A, Riehle MO, Herzyk P, Wilkinson CD, Oreffo RO. The control of human mesenchymal cell differentiation using nanoscale symmetry and disorder. Nat Mater. 2007;6(12):997–1003. doi: 10.1038/nmat2013. [DOI] [PubMed] [Google Scholar]
  • 100.Dalby MJ, Andar A, Nag A, Affrossman S, Tare R, McFarlane S, Oreffo RO. Genomic expression of mesenchymal stem cells to altered nanoscale topographies. J R Soc Interface. 2008;5(26):1055–1065. doi: 10.1098/rsif.2008.0016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Silva GA, Czeisler C, Niece KL, Beniash E, Harrington DA, Kessler JA, Stupp SI. Selective differentiation of neural progenitor cells by high-epitope density nanofibers. Science. 2004;303(5662):1352–1355. doi: 10.1126/science.1093783. [DOI] [PubMed] [Google Scholar]
  • 102.Hosseinkhani H, Hosseinkhani M, Tian F, Kobayashi H, Tabata Y. Osteogenic differentiation of mesenchymal stem cells in self-assembled peptide-amphiphile nanofibers. Biomaterials. 2006;27(22):4079–4086. doi: 10.1016/j.biomaterials.2006.03.030. [DOI] [PubMed] [Google Scholar]
  • 103.Jayarama Reddy V, Radhakrishnan S, Ravichandran R, Mukherjee S, Balamurugan R, Sundarrajan S, Ramakrishna S. Nanofibrous structured bio-mimetic strategies for skin tissue regeneration. Wound Repair Regen. 2013;21(1):1–16. doi: 10.1111/j.1524-475X.2012.00861.x. [DOI] [PubMed] [Google Scholar]
  • 104.Lu T, Li Y, Chen T. Techniques for fabrication and construction of three-dimensional scaffolds for tissue engineering. Int J Nanomedicine. 2013;8:337–350. doi: 10.2147/IJN.S38635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zhang S, Holmes T, Lockshin C, Rich A. Spontaneous assembly of a self-complementary oligopeptide to form a stable macroscopic membrane. Proc Natl Acad Sci U S A. 1993;90(8):3334–3338. doi: 10.1073/pnas.90.8.3334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Holmes TC, de Lacalle S, Su X, Liu G, Rich A, Zhang S. Extensive neurite outgrowth and active synapse formation on self-assembling peptide scaffolds. Proc Natl Acad Sci U S A. 2000;97(12):6728–6733. doi: 10.1073/pnas.97.12.6728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Leung GK, Wang YC, Wu W. Peptide nanofiber scaffold for brain tissue reconstruction. Methods Enzymol. 2012;508:177–190. doi: 10.1016/B978-0-12-391860-4.00009-4. [DOI] [PubMed] [Google Scholar]
  • 108.Ellis-Behnke RG, Liang YX, You SW, Tay DK, Zhang S, So KF, Schneider GE. Nano neuro knitting: peptide nanofiber scaffold for brain repair and axon regeneration with functional return of vision. Proc Natl Acad Sci U S A. 2006;103(13):5054–5059. doi: 10.1073/pnas.0600559103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Garreta E, Genove E, Borros S, Semino CE. Osteogenic differentiation of mouse embryonic stem cells and mouse embryonic fibroblasts in a three-dimensional self-assembling peptide scaffold. Tissue Eng. 2006;12(8):2215–2227. doi: 10.1089/ten.2006.12.2215. [DOI] [PubMed] [Google Scholar]
  • 110.Gelain F, Bottai D, Vescovi A, Zhang S. Designer self-assembling peptide nanofiber scaffolds for adult mouse neural stem cell 3-dimensional cultures. PLoS One. 2006;1:e119. doi: 10.1371/journal.pone.0000119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Guo J, Su H, Zeng Y, Liang YX, Wong WM, Ellis-Behnke RG, So KF, Wu W. Reknitting the injured spinal cord by self-assembling peptide nanofiber scaffold. Nanomedicine. 2007;3(4):311–321. doi: 10.1016/j.nano.2007.09.003. [DOI] [PubMed] [Google Scholar]
  • 112.Zhang S, Holmes TC, DiPersio CM, Hynes RO, Su X, Rich A. Self-complementary oligopeptide matrices support mammalian cell attachment. Biomaterials. 1995;16(18):1385–1393. doi: 10.1016/0142-9612(95)96874-y. [DOI] [PubMed] [Google Scholar]
  • 113.Beniash E, Hartgerink JD, Storrie H, Stendahl JC, Stupp SI. Self-assembling peptide amphiphile nanofiber matrices for cell entrapment. Acta Biomater. 2005;1(4):387–397. doi: 10.1016/j.actbio.2005.04.002. [DOI] [PubMed] [Google Scholar]
  • 114.Lao L, Wang Y, Zhu Y, Zhang Y, Gao C. Poly(lactide-co-glycolide)/hydroxyapatite nanofibrous scaffolds fabricated by electrospinning for bone tissue engineering. J Mater Sci Mater Med. 2011;22(8):1873–1884. doi: 10.1007/s10856-011-4374-8. [DOI] [PubMed] [Google Scholar]
  • 115.Leung V, Hartwell R, Elizei SS, Yang H, Ghahary A, Ko F. Postelectrospinning modifications for alginate nanofiber-based wound dressings. J Biomed Mater Res Part B. 2013 doi: 10.1002/jbm.b.33028. (in press) [DOI] [PubMed] [Google Scholar]
  • 116.Xin X, Hussain M, Mao JJ. Continuing differentiation of human mesenchymal stem cells and induced chondrogenic and osteogenic lineages in electrospun PLGA nanofiber scaffold. Biomaterials. 2007;28(2):316–325. doi: 10.1016/j.biomaterials.2006.08.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Wang W, Itoh S, Matsuda A, Aizawa T, Demura M, Ichinose S, Shinomiya K, Tanaka J. Enhanced nerve regeneration through a bilayered chitosan tube: the effect of introduction of glycine spacer into the CYIGSR sequence. J Biomed Mater Res A. 2008;85(4):919–928. doi: 10.1002/jbm.a.31522. [DOI] [PubMed] [Google Scholar]
  • 118.Sarkar SD, Farrugia BL, Dargaville TR, Dhara S. Chitosan-collagen scaffolds with nano/ microfibrous architecture for skin tissue engineering. J Biomed Mater Res A. 2013;101(12):3482–3492. doi: 10.1002/jbm.a.34660. [DOI] [PubMed] [Google Scholar]
  • 119.Sun F, Zhou H, Lee J. Various preparation methods of highly porous hydroxyapatite/polymer nanoscale biocomposites for bone regeneration. Acta Biomater. 2011;7(11):3813–3828. doi: 10.1016/j.actbio.2011.07.002. [DOI] [PubMed] [Google Scholar]
  • 120.Li C, Vepari C, Jin HJ, Kim HJ, Kaplan DL. Electrospun silk-BMP-2 scaffolds for bone tissue engineering. Biomaterials. 2006;27(16):3115–3124. doi: 10.1016/j.biomaterials.2006.01.022. [DOI] [PubMed] [Google Scholar]
  • 121.Shih YR, Chen CN, Tsai SW, Wang YJ, Lee OK. Growth of mesenchymal stem cells on electrospun type I collagen nanofibers. Stem Cells. 2006;24(11):2391–2397. doi: 10.1634/stemcells.2006-0253. [DOI] [PubMed] [Google Scholar]
  • 122.Li WJ, Tuli R, Huang X, Laquerriere P, Tuan RS. Multilineage differentiation of human mesenchymal stem cells in a three-dimensional nanofibrous scaffold. Biomaterials. 2005;26(25):5158–5166. doi: 10.1016/j.biomaterials.2005.01.002. [DOI] [PubMed] [Google Scholar]
  • 123.McCullen SD, Stevens DR, Roberts WA, Clarke LI, Bernacki SH, Gorga RE, Loboa EG. Characterization of electrospun nanocomposite scaffolds and biocompatibility with adipose-derived human mesenchymal stem cells. Int J Nanomedicine. 2007;2(2):253–263. [PMC free article] [PubMed] [Google Scholar]
  • 124.Shafiee A, Soleimani M, Chamheidari GA, Seyedjafari E, Dodel M, Atashi A, Gheisari Y. Electrospun nanofiber-based regeneration of cartilage enhanced by mesenchymal stem cells. J Biomed Mater Res A. 2011;99(3):467–478. doi: 10.1002/jbm.a.33206. [DOI] [PubMed] [Google Scholar]
  • 125.Ravichandran R, Venugopal JR, Sundarrajan S, Mukherjee S, Ramakrishna S. Precipitation of nanohydroxyapatite on PLLA/PBLG/Collagen nanofibrous structures for the differentiation of adipose derived stem cells to osteogenic lineage. Biomaterials. 2012;33(3):846–855. doi: 10.1016/j.biomaterials.2011.10.030. [DOI] [PubMed] [Google Scholar]
  • 126.Brizzi MF, Tarone G, Defilippi P. Extracellular matrix, integrins, and growth factors as tailors of the stem cell niche. Curr Opin Cell Biol. 2012;24(5):645–651. doi: 10.1016/j.ceb.2012.07.001. [DOI] [PubMed] [Google Scholar]
  • 127.Binan L, Tendey C, De Crescenzo G, El Ayoubi R, Ajji A, Jolicoeur M. Differentiation of neuronal stem cells into motor neurons using electrospun poly-L-lactic acid/gelatin scaffold. Biomaterials. 2014;35(2):664–674. doi: 10.1016/j.biomaterials.2013.09.097. [DOI] [PubMed] [Google Scholar]
  • 128.Jia L, Prabhakaran MP, Qin X, Ramakrishna S. Stem cell differentiation on electrospun nanofibrous substrates for vascular tissue engineering. Mater Sci Eng C Mater Biol Appl. 2013;33(8):4640–4650. doi: 10.1016/j.msec.2013.07.021. [DOI] [PubMed] [Google Scholar]
  • 129.Holmes B, Castro NJ, Li J, Keidar M, Zhang LG. Enhanced human bone marrow mesenchymal stem cell functions in novel 3D cartilage scaffolds with hydrogen treated multi-walled carbon nanotubes. Nanotechnology. 2013;24(36):365102. doi: 10.1088/0957-4484/24/36/365102. [DOI] [PubMed] [Google Scholar]
  • 130.Fonseca-Garcia A, Mota-Morales JD, Quintero-Ortega IA, Garcia-Carvajal ZY, Martínez-López V, Ruvalcaba E, Solis L, Ibarra C, Gutiérrez MC, Terrones M, Sanchez IC, del Monte F, Velasquillo MC, Luna-Bárcenas G. Effect of doping in carbon nanotubes on the viability of biomimetic chitosan-carbon nanotubes-hydroxyapatite scaffolds. J Biomed Mater Res Part A. 2013 doi: 10.1002/jbm.a.34893. (in press) [DOI] [PubMed] [Google Scholar]
  • 131.Gupta A, Woods MD, Illingworth KD, Niemeier R, Schafer I, Cady C, Filip P, El-Amin SF., III Single walled carbon nanotube composites for bone tissue engineering. J Orthop Res. 2013;31(9):1374–1381. doi: 10.1002/jor.22379. [DOI] [PubMed] [Google Scholar]
  • 132.Jan E, Kotov NA. Successful differentiation of mouse neural stem cells on layer-by-layer assembled single-walled carbon nanotube composite. Nano Lett. 2007;7(5):1123–1128. doi: 10.1021/nl0620132. [DOI] [PubMed] [Google Scholar]
  • 133.Kam NW, Jan E, Kotov NA. Electrical stimulation of neural stem cells mediated by humanized carbon nanotube composite made with extracellular matrix protein. Nano Lett. 2009;9(1):273–278. doi: 10.1021/nl802859a. [DOI] [PubMed] [Google Scholar]
  • 134.Moon SU, Kim J, Bokara KK, Kim JY, Khang D, Webster TJ, Lee JE. Carbon nanotubes impregnated with subventricular zone neural progenitor cells promotes recovery from stroke. Int J Nanomedicine. 2012;7:2751–2765. doi: 10.2147/IJN.S30273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Sirivisoot S, Harrison BS. Skeletal myotube formation enhanced by electrospun polyurethane carbon nanotube scaffolds. Int J Nanomedicine. 2011;6:2483–2497. doi: 10.2147/IJN.S24073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Mooney E, Mackle JN, Blond DJ, O’Cearbhaill E, Shaw G, Blau WJ, Barry FP, Barron V, Murphy JM. The electrical stimulation of carbon nanotubes to provide a cardiomimetic cue to MSCs. Biomaterials. 2012;33(26):6132–6139. doi: 10.1016/j.biomaterials.2012.05.032. [DOI] [PubMed] [Google Scholar]
  • 137.Parrag IC, Zandstra PW, Woodhouse KA. Fiber alignment and coculture with fibroblasts improves the differentiated phenotype of murine embryonic stem cell-derived cardiomyocytes for cardiac tissue engineering. Biotechnol Bioeng. 2012;109(3):813–822. doi: 10.1002/bit.23353. [DOI] [PubMed] [Google Scholar]
  • 138.Gupta MK, Walthall JM, Venkataraman R, Crowder SW, Jung DK, Yu SS, Feaster TK, Wang X, Giorgio TD, Hong CC, Baudenbacher FJ, Hatzopoulos AK, Sung HJ. Combinatorial polymer electrospun matrices promote physiologically-relevant cardiomyogenic stem cell differentiation. PLoS One. 2011;6(12):e28935. doi: 10.1371/journal.pone.0028935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Prabhakaran MP, Mobarakeh LG, Kai D, Karbalaie K, Nasr-Esfahani MH, Ramakrishna S. Differentiation of embryonic stem cells to cardiomyocytes on electrospun nanofibrous substrates. J Biomed Mater Res Part B. 2013 doi: 10.1002/jbm.b.33022. (in press) [DOI] [PubMed] [Google Scholar]
  • 140.Xie J, Willerth SM, Li X, Macewan MR, Rader A, Sakiyama-Elbert SE, Xia Y. The differentiation of embryonic stem cells seeded on electrospun nanofibers into neural lineages. Biomaterials. 2009;30(3):354–362. doi: 10.1016/j.biomaterials.2008.09.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Carlberg B, Axell MZ, Nannmark U, Liu J, Kuhn HG. Electrospun polyurethane scaffolds for proliferation and neuronal differentiation of human embryonic stem cells. Biomed Mater. 2009;4(4):045004. doi: 10.1088/1748-6041/4/4/045004. [DOI] [PubMed] [Google Scholar]
  • 142.Kang X, Xie Y, Powell HM, James Lee L, Belury MA, Lannutti JJ, Kniss DA. Adipogenesis of murine embryonic stem cells in a three-dimensional culture system using electrospun polymer scaffolds. Biomaterials. 2007;28(3):450–458. doi: 10.1016/j.biomaterials.2006.08.052. [DOI] [PubMed] [Google Scholar]
  • 143.Schrand AM, Dai L, Schlager JJ, Hussain SM. Toxicity testing of nanomaterials. Adv Exp Med Biol. 2012;745:58–75. doi: 10.1007/978-1-4614-3055-1_5. [DOI] [PubMed] [Google Scholar]
  • 144.Nel A, Xia T, Madler L, Li N. Toxic potential of materials at the nanolevel. Science. 2006;311(5761):622–627. doi: 10.1126/science.1114397. [DOI] [PubMed] [Google Scholar]
  • 145.Foley S, Crowley C, Smaihi M, Bonfils C, Erlanger BF, Seta P, Larroque C. Cellular localisation of a water-soluble fullerene derivative. Biochem Biophys Res Commun. 2002;294(1):116–119. doi: 10.1016/S0006-291X(02)00445-X. [DOI] [PubMed] [Google Scholar]
  • 146.Christen V, Fent K. Silica nanoparticles and silver-doped silica nanoparticles induce endoplasmatic reticulum stress response and alter cytochrome P4501A activity. Chemosphere. 2012;87(4):423–434. doi: 10.1016/j.chemosphere.2011.12.046. [DOI] [PubMed] [Google Scholar]
  • 147.Zhang R, Piao MJ, Kim KC, Kim AD, Choi JY, Choi J, Hyun JW. Endoplasmic reticulum stress signaling is involved in silver nanoparticles-induced apoptosis. Int J Biochem Cell Biol. 2012;44(1):224–232. doi: 10.1016/j.biocel.2011.10.019. [DOI] [PubMed] [Google Scholar]
  • 148.Zhu L, Chang DW, Dai L, Hong Y. DNA damage induced by multiwalled carbon nanotubes in mouse embryonic stem cells. Nano Lett. 2007;7(12):3592–3597. doi: 10.1021/nl071303v. [DOI] [PubMed] [Google Scholar]
  • 149.Ferin J, Oberdorster G, Penney DP. Pulmonary retention of ultrafine and fine particles in rats. Am J Respir Cell Mol Biol. 1992;6(5):535–542. doi: 10.1165/ajrcmb/6.5.535. [DOI] [PubMed] [Google Scholar]
  • 150.Service RF. Nanomaterials show signs of toxicity. Nanomaterials show signs of toxicity. Science. 2003;300(5617):243. doi: 10.1126/science.300.5617.243a. [DOI] [PubMed] [Google Scholar]
  • 151.Shvedova AA, Castranova V, Kisin ER, Schwegler-Berry D, Murray AR, Gandelsman VZ, Maynard A, Baron P. Exposure to carbon nanotube material: assessment of nanotube cytotoxicity using human keratinocyte cells. J Toxicol Environ Health A. 2003;66(20):1909–1926. doi: 10.1080/713853956. [DOI] [PubMed] [Google Scholar]
  • 152.Lam CW, James JT, McCluskey R, Hunter RL. Pulmonary toxicity of single-wall carbon nanotubes in mice 7 and 90 days after intratracheal instillation. Toxicol Sci. 2004;77(1):126–134. doi: 10.1093/toxsci/kfg243. [DOI] [PubMed] [Google Scholar]
  • 153.Cheng C, Muller KH, Koziol KK, Skepper JN, Midgley PA, Welland ME, Porter AE. Toxicity and imaging of multi-walled carbon nanotubes in human macrophage cells. Biomaterials. 2009;30(25):4152–4160. doi: 10.1016/j.biomaterials.2009.04.019. [DOI] [PubMed] [Google Scholar]
  • 154.Moller P, Jacobsen NR, Folkmann JK, Danielsen PH, Mikkelsen L, Hemmingsen JG, Vesterdal LK, Forchhammer L, Wallin H, Loft S. Role of oxidative damage in toxicity of particulates. Free Radic Res. 2010;44(1):1–46. doi: 10.3109/10715760903300691. [DOI] [PubMed] [Google Scholar]
  • 155.Cancino J, Paino IM, Micocci KC, Selistre-de-Araujo HS, Zucolotto V. In vitro nanotoxicity of single-walled carbon nanotube-dendrimer nano-complexes against murine myoblast cells. Toxicol Lett. 2013;219(1):18–25. doi: 10.1016/j.toxlet.2013.02.009. [DOI] [PubMed] [Google Scholar]
  • 156.Pacurari M, Castranova V, Vallyathan V. Single- and multi-wall carbon nanotubes versus asbestos: are the carbon nanotubes a new health risk to humans? J Toxicol Environ Health A. 2010;73(5):378–395. doi: 10.1080/15287390903486527. [DOI] [PubMed] [Google Scholar]
  • 157.Snyder-Talkington BN, Dymacek J, Porter DW, Wolfarth MG, Mercer RR, Pacurari M, Denvir J, Castranova V, Qian Y, Guo NL. System-based identification of toxicity pathways associated with multi-walled carbon nanotube-induced pathological responses. Toxicol Appl Pharmacol. 2013;272(2):476–489. doi: 10.1016/j.taap.2013.06.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Tourinho PS, van Gestel CA, Lofts S, Svendsen C, Soares AM, Loureiro S. Metal-based nanoparticles in soil: fate, behavior, and effects on soil invertebrates. Environ Toxicol Chem. 2012;31(8):1679–1692. doi: 10.1002/etc.1880. [DOI] [PubMed] [Google Scholar]
  • 159.Sharma HS, Sharma A. Neurotoxicity of engineered nanoparticles from metals. CNS Neurol Disord Drug Targets. 2012;11(1):65–80. doi: 10.2174/187152712799960817. [DOI] [PubMed] [Google Scholar]
  • 160.Lan Z, Yang WX. Nanoparticles and spermatogenesis: how do nanoparticles affect spermatogenesis and penetrate the blood-testis barrier. Nanomedicine (Lond) 2012;7(4):579–596. doi: 10.2217/nnm.12.20. [DOI] [PubMed] [Google Scholar]
  • 161.Oberdorster G, Elder A, Rinderknecht A. Nanoparticles and the brain: cause for concern? J Nanosci Nanotechnol. 2009;9(8):4996–5007. doi: 10.1166/jnn.2009.gr02. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Moller P, Folkmann JK, Danielsen PH, Jantzen K, Loft S. Oxidative stress generated damage to DNA by gastrointestinal exposure to insoluble particles. Curr Mol Med. 2012;12(6):732–745. doi: 10.2174/156652412800792624. [DOI] [PubMed] [Google Scholar]
  • 163.Shaw BJ, Handy RD. Physiological effects of nanoparticles on fish: a comparison of nanometals versus metal ions. Environ Int. 2011;37(6):1083–1097. doi: 10.1016/j.envint.2011.03.009. [DOI] [PubMed] [Google Scholar]
  • 164.Braydich-Stolle L, Hussain S, Schlager JJ, Hofmann MC. In vitro cytotoxicity of nanoparticles in mammalian germline stem cells. Toxicol Sci. 2005;88(2):412–419. doi: 10.1093/toxsci/kfi256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Deng X, Luan Q, Chen W, Wang Y, Wu M, Zhang H, Jiao Z. Nanosized zinc oxide particles induce neural stem cell apoptosis. Nanotechnology. 2009;20(11):115101. doi: 10.1088/0957-4484/20/11/115101. [DOI] [PubMed] [Google Scholar]
  • 166.Heng BC, Zhao X, Xiong S, Ng KW, Boey FY, Loo JS. Cytotoxicity of zinc oxide (ZnO) nanoparticles is influenced by cell density and culture format. Arch Toxicol. 2011;85(6):695–704. doi: 10.1007/s00204-010-0608-7. [DOI] [PubMed] [Google Scholar]
  • 167.Taccola L, Raffa V, Riggio C, Vittorio O, Iorio MC, Vanacore R, Pietrabissa A, Cuschieri A. Zinc oxide nanoparticles as selective killers of proliferating cells. Int J Nanomedicine. 2011;6:1129–1140. doi: 10.2147/IJN.S16581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Rasmussen JW, Martinez E, Louka P, Wingett DG. Zinc oxide nanoparticles for selective destruction of tumor cells and potential for drug delivery applications. Expert Opin Drug Deliv. 2010;7(9):1063–1077. doi: 10.1517/17425247.2010.502560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Choi SJ, Oh JM, Choy JH. Toxicological effects of inorganic nanoparticles on human lung cancer A549 cells. J Inorg Biochem. 2009;103(3):463–471. doi: 10.1016/j.jinorgbio.2008.12.017. [DOI] [PubMed] [Google Scholar]
  • 170.Kim IS, Baek M, Choi SJ. Comparative cytotoxicity of Al2O3, CeO2, TiO2 and ZnO nanoparticles to human lung cells. J Nanosci Nanotechnol. 2010;10(5):3453–3458. doi: 10.1166/jnn.2010.2340. [DOI] [PubMed] [Google Scholar]
  • 171.Zhang QL, Li MQ, Ji JW, Gao FP, Bai R, Chen CY, Wang ZW, Zhang C, Niu Q. In vivo toxicity of nano-alumina on mice neurobehavioral profiles and the potential mechanisms. Int J Immunopathol Pharmacol. 2011;24(1 Suppl):23S–29S. [PubMed] [Google Scholar]
  • 172.Alshatwi AA, Vaiyapuri Subbarayan P, Ramesh E, Al-Hazzani AA, Alsaif MA, Alwarthan AA. Al(2)O(3) nanoparticles induce mitochondria-mediated cell death and upregulate the expression of signaling genes in human mesenchymal stem cells. J Biochem Mol Toxicol. 2012;26(11):469–476. doi: 10.1002/jbt.21448. [DOI] [PubMed] [Google Scholar]
  • 173.Atiyeh BS, Costagliola M, Hayek SN, Dibo SA. Effect of silver on burn wound infection control and healing: review of the literature. Burns. 2007;33(2):139–148. doi: 10.1016/j.burns.2006.06.010. [DOI] [PubMed] [Google Scholar]
  • 174.Lansdown AB. Silver in health care: antimicrobial effects and safety in use. Curr Probl Dermatol. 2006;33:17–34. doi: 10.1159/000093928. [DOI] [PubMed] [Google Scholar]
  • 175.Carlson C, Hussain SM, Schrand AM, Braydich-Stolle LK, Hess KL, Jones RL, Schlager JJ. Unique cellular interaction of silver nanoparticles: size-dependent generation of reactive oxygen species. J Phys Chem B. 2008;112(43):13608–13619. doi: 10.1021/jp712087m. [DOI] [PubMed] [Google Scholar]
  • 176.Braydich-Stolle LK, Lucas B, Schrand A, Murdock RC, Lee T, Schlager JJ, Hussain SM, Hofmann MC. Silver nanoparticles disrupt GDNF/Fyn kinase signaling in spermatogonial stem cells. Toxicol Sci. 2010;116(2):577–589. doi: 10.1093/toxsci/kfq148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Hussain SM, Hess KL, Gearhart JM, Geiss KT, Schlager JJ. In vitro toxicity of nanoparticles in BRL 3A rat liver cells. Toxicol In Vitro. 2005;19(7):975–983. doi: 10.1016/j.tiv.2005.06.034. [DOI] [PubMed] [Google Scholar]
  • 178.Ahamed M, Karns M, Goodson M, Rowe J, Hussain SM, Schlager JJ, Hong Y. DNA damage response to different surface chemistry of silver nanoparticles in mammalian cells. Toxicol Appl Pharmacol. 2008;233(3):404–410. doi: 10.1016/j.taap.2008.09.015. [DOI] [PubMed] [Google Scholar]
  • 179.Hackenberg S, Scherzed A, Kessler M, Hummel S, Technau A, Froelich K, Ginzkey C, Koehler C, Hagen R, Kleinsasser N. Silver nanoparticles: evaluation of DNA damage, toxicity and functional impairment in human mesenchymal stem cells. Toxicol Lett. 2011;201(1):27–33. doi: 10.1016/j.toxlet.2010.12.001. [DOI] [PubMed] [Google Scholar]
  • 180.Samberg ME, Loboa EG, Oldenburg SJ, Monteiro-Riviere NA. Silver nanoparticles do not influence stem cell differentiation but cause minimal toxicity. Nanomedicine (Lond) 2012;7(8):1197–1209. doi: 10.2217/nnm.12.18. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES