Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
16
Phil Chambers1, Helen O. McCarthy1, Nicholas J. Dunne1,2,3,4,5 1School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom; 2Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, Ireland; 3Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; 4Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; 5School of Mechanical and Manufacturing Engineering of Ireland, Dublin City University, Dublin, Ireland
16.1 Why nucleic acids for bone repair Within this section an overview of why nucleic acids could be the future of bone repair, the bone microenvironment is also considered. Many second-generation bone repair therapies (such as polymer and ceramic scaffolds) have exhibited successful bone formation and biomineralization, the newest generation of therapy options aim to further enhance the osteoinductive potential of these materials through strict biomimicry of the bone tissue microenvironment, resulting in a more defined regulation of osteoprogenitor cell function and homeostasis. Essential to this microenvironment are the inclusion of stimuli that promote favorable cellular response, or activation of specific genes, increasing the osteoinductivity of the material. A combination of biochemical stimuli (addition of biomolecules, surface functionalisation of the material, etc.) and mechanical stimuli (such as three-dimensional (3D) porous structures to promote cell infiltration, attachment, and proliferation) are usually implemented in third-generation therapies to fulfil these requirements. Historically, most research in tissue engineering has tended to focus primarily on the material properties of bone regeneration scaffolds, and this alone is not enough to mimic the in vivo microenvironment. More recently, the importance of cellular interplay and response has become apparent. Most studies now include biomaterials cultured with one type of cell, usually mesenchymal stem cells (MSCs) or osteoblasts, but in recent years focus has turned to coculture systems and/or the use of gene therapy approaches in an aim to better understand the in vivo process of bone formation. In an attempt to promote vascularization of newly formed bone, Buschmann et al. ([2007]) utilized a coculture system of human osteoblasts (hOB) and human endothelial cells (hEC) on a highly porous poly(ester urethane) foam. This coculture scaffold was compared to control groups containing (1) only hOB cells or (2) only hEC cells. Bone Repair Biomaterials. https://doi.org/10.1016/B978-0-08-102451-5.00016-0 Copyright © 2019 Elsevier Ltd. All rights reserved.
412
Bone Repair Biomaterials
After culture for 7 days in vitro and 15 days ex vivo the scaffold containing the coculture showed vastly improved cell proliferation, 3D scaffold invasion, extracellular matrix (ECM) formation, and blood perfusion exhibiting clearly the benefits of cocultured systems on bone regeneration [1]. Cocultured systems appear to benefit from cellular cross-talk and as previous investigations have shown that the paracrine effects of MSCs include stimulation of angiogenesis, apoptotic protection, the recruitment of host MSCs and other progenitor cells in addition, to stimulation of proliferation and differentiation [2,3]. Furthermore, MSCs transplanted into mice with a stabilized tibial fracture have been found to migrate to the fracture site, integrate within the callus and secrete BMP-2 to aid with the fracture healing [4]. Therefore, the signals and secretions of cells cultured on a bone regeneration scaffold are extremely important, and the cross-talk between cell types will affect the efficacy and quality of any therapeutic treatment. To this end, gene therapy strategies (such as the intracellular delivery of nucleic acids) are gaining momentum for the purposes of bone regeneration. In gene therapy delivery, cells seeded on the bone regeneration scaffolds are used as bioactive carriers of osteoinductive genes facilitating sustained local delivery, allowing these osteogenic factors to be expressed not only for sustained periods, but in more physiological forms and concentrations (rather than the supraphysiological concentrations required for recombinant growth factor delivery) [5]. As a consequence, this has been shown to direct increased natural bone formation [6] and the utilization of genetically altered proosteoinductive cells within a bone regeneration scaffold have exhibited superior performance in a range of animal models [7]. Most gene therapy approaches for bone formation have focused on the use of genes within the bone morphogenetic protein (BMP) family, in order to induce the subsequent osteogenic cascade [7–10]. However, a common issue exists, which is that a suitable and efficient delivery system for such gene therapies is required. There are numerous physical and biological barriers to be overcome for successful nucleic acid delivery for an intended therapeutic effect. Current methods employed to overcome this issue include viral (such as Adenovirus, Retrovirus, Lentivirus), nonviral (plasmids, cationic polymers, liposomes), and cell mediated gene transfer [6,11].
16.2 The biological barriers to nucleic acid delivery systems in the bone environment Such biological barriers include protection of the cargo from degradation in-situ, entry into the cells, disruption from the endosome, release of the nucleic acid payload into the cytoplasm and finally active transport into the nucleus for DNA. For the purposes of nucleic delivery, the treatment efficacy is highly dependent on the successful transport of a chosen therapy to the target-specific cells. A need exists for a suitable delivery vehicle to protect the cargo from degradation, facilitate cellular membrane penetration, accumulation at the required sites, and successful transfection
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
413
of the target cells. Historically, the safest and easiest method is the incorporation of naked DNA [12], however, in vivo gene delivery using this method has continually shown poor efficiency requiring large amounts of DNA, and any attempts to increase efficiency to a level that would allow clinical use have so far been impossible [13]. The introduction of physical methods to counteract the poor efficacy of delivering naked DNA such as ballistic gene delivery, electroporation, sonoporation, and microinjection [14] have increased the ex vivo applications, but in vivo efficacy still remains poor. Therefore, if the delivery of nucleic acids is to be a successful clinical platform technology in bone tissue regeneration, a suitable vehicle for the efficient delivery and promotion of osteogenesis must be found. The human body employs a host of sophisticated defense mechanisms to protect cells from the invasion of foreign material and therefore many hurdles must be overcome for the successful transfer of nucleic acids to the intracellular region for the intended effect. Some of the most relevant barriers to cellular uptake are described below.
16.2.1 Extracellular barriers 16.2.1.1 Skin The first barrier that any therapy must overcome is the skin, specifically the epidermis. The epidermis functions as a physical barrier to the external environment, protecting from infection and modulating water transfer between the external and internal environments [15]. Therefore, the only way to penetrate this barrier is with use of physical methods like those described previously (microneedles, ballistic gene delivery etc.), with the use of ex vivo scaffolds [16], or by utilizing systematic delivery of the osteogenic factors, usually via an implant for the specific case of bone regeneration.
16.2.1.2 Mononuclear phagocyte system The mononuclear phagocyte system (MPS), also known as the reticuloendothelial system, consists of phagocytic cells employed by the immune system for the identification and removal of foreign material. In the environment of bone repair, osteoclasts in the bone tissue, and monocytes in the bone marrow and blood, are the specific cell types that phagocytose any foreign biomolecules present [17]. The MPS binds serum proteins known as opsonins to the surface of foreign molecules, in a process known as opsonization [18]. Opsonization essentially “flags” the material for phagocytes to being the process of engulfment and subsequent digestion. Therefore, any new therapy for bone regeneration must exhibit the necessary physiochemical and morphological characteristics to avoid detection by the MPS. An increase in opsonization is found with nanoparticle (NP) and gene delivery systems with a cationic charge (increasing the likelihood of binding to the anionic serum proteins). This can be an issue as most cell-targeting delivery systems tend to be cationic in nature to facilitate increased binding to the proteoglycans on the cell membranes. For this reason, the surface charge of any therapeutic agents must be optimized for each application, to ensure that the cell-targeting nature of the molecules is not affected, whilst also ensuring that the
414
Bone Repair Biomaterials
increased cationic charge will not result in opsonization. The morphological characteristics are also important to evade detection by the MPS. It has been reported that particles <150 nm in size [19], and spherical in shape [20], experience a decrease in serum protein binding. The rationale for this effect is that NPs in this size range are seen to be similar in size to the serum proteins, and as a consequence, the potential contact area between the two is minimized; that is, neither the particle nor the serum protein has enough contact points to facilitate binding [21]. Many delivery systems utilize extra methods to avoid detection by the MPS including PEGylation and biomimetic surface coatings, such as using autologous erythrocytes/leukocytes or “selflike” peptides [22–24]. However, the addition of surface moieties has a detrimental effect on the efficient delivery of cargo (explained further in Cationic liposomes section), and must be carefully balanced for optimal therapeutic efficacy.
16.2.1.3 Extracellular matrix (ECM) and the interstitial space Another major barrier to the targeting of cells within the bone microenvironment is to cross the interstitium or ECM. The ECM is comprised of a cross-linked network of collagen and elastin fibers, proteoglycans and hyaluronic acid [25], and is responsible for the transport of oxygen and other supportive nutrients for the proliferation and maintenance of cells, in addition, to providing the structural integrity required for bone. A highly developed ECM may prove to exhibit significant resistance to the diffusion of therapeutic cargo, causing therapies to be delivered an unwanted distance away from the intended target. The spaces within the ECM are filled with interstitial fluid (ISF), forming a hydrophilic gel [26]. A pressure gradient within the ISF is present as a means for active diffusion and transport of molecules throughout the interstitial space. Therefore, any delivered therapeutic agent must remain stable in the presence of the ISF and the associated pressure gradient and fluid flow. Surface modifications or coatings to intended deliverables (such as the use of superparamagnetic NPs) [27], as well as adhering to strict morphological characteristics are methods currently utilized to aid the delivery of therapeutic agents through the interstitial space and ECM.
16.2.1.4 Phospholipid bilayer The last barrier for a therapeutic agent to overcome on its journey to the inside of a cell is the phospholipid bilayer. This semipermeable cell membrane consists of a structured dual layer of lipid molecules responsible for the mediation of molecule diffusion from the extracellular to intracellular regions (or vice versa). Cellular uptake from the extracellular region usually occurs via two main mechanisms: (1) endocytosis or (2) direct translocation [28].
Endocytosis Endocytosis is a process that occurs naturally in all cell types. The triggering of a cell to take up a molecule via endocytosis can occur via electrostatic interaction with cell surface receptors (proteoglycans), or via direct interaction with the plasma membrane [29]. Endocytosis is the umbrella term for several pathways that are responsible for the cellular
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
415
uptake of “large” particles (0.5–10 μm) via phagocytosis [30], or for smaller particles (<0.5 μm) via pinocytosis (including clathrin or caveolae-mediated internalization). Clathrin-mediated endocytosis refers to the uptake of material via the “packaging” of the cargo within a clathrin-coated vesicle, formation of which is assisted by the budding caused by the GTPase dynamin. The clathrin-coated vesicle is then sorted by early endosomes and either sent back to the surface, or moved to more mature endosomes prior to later compartmentalization in multivesicular bodies or fusion with lysosomes for subsequent acidic degradation [31]. Clathrin-mediated endocytosis only occurs for particles <200 nm in size, due to the observed upper limit on the size of a clathrin-coated vesicle [32], and the rate of uptake via this method also appears to be size-dependent, with particles in the range of 50–100 nm observed to be taken up at a higher rate [30]. As this method of entry involves encapsulation in endosomes, special care must be taken to ensure the required cargo to be delivered is not susceptible to acidic degradation in the early/late endosomal environment with intravascular pH levels ranging from pH 6.5–4.5 [33]. Caveolae-mediated endocytosis is another energy-dependent, but clathrin-independent, pathway of cellular uptake. Caveolae are a subdomain of the well-characterized glycolipid rafts, rich in cholesterol and sphingolipid, the function and maintenance of which is primarily due to caveolin-1 [34]. This method of entry is often exploited by pathogens for direct entry into a cell, or for the assistance of further pathogen entry that are larger than a single caveolae [35]. A single caveolae has dimensions of ∼70 nm [36] consequently is only utilized to take up particles in this region of size, or smaller. This process of entry is favorable to pathogens due to the nonacidic environment, protecting them from any potential lysosomal degradation. This process of cellular entry, however, is much slower than the clathrin-mediated process, due to the low mobility of caveolae [37]. Although clathrin and caveolae are the most utilized method of endocytosis by cells, any cells that are devoid of clathrin or caveolin-1 utilize a wide range of other methods for cellular uptake (collectively known as clathrin and caveolae-independent endocytosis). Currently many of the mechanisms are poorly understood or yet to be discovered, however, Lundmark et al. [38] were able to identify a highly prevalent method of endocytosis in HeLa cells involving GTPase Regulator Associated with Focal Adhesion Kinase-1 (GRAF1). GRAF1 was observed to regulate the clathrin/ caveolae-independent endocytic pathway known as the CLIC/GEEC pathway. Macropinocytosis is an actin-dependent process initiated by the rearrangement of the actin cytoskeleton by the activation of receptor tyrosine kinases, such as the epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor (PDGFR) [39]. This process of actin rearrangement leads to surface membrane ruffles that give rise to large endocytic vacuoles called macropinosomes, which encapsulate the extracellular fluid (ECF) and any solute present. Macropinocytosis is usually employed by cells for the uptake of particle clusters, or for larger macromolecules [40].
Direct translocation Direct translocation is the term given to energy-independent mechanisms employed by cells for the cellular uptake of extracellular molecules, without the use of internal cell internalization machinery, but by the perturbation of the cellular membrane for
416
Bone Repair Biomaterials
entry. Direct translocation is the main and most relevant method of cellular entry for cell-penetrating peptides (CPPs), amongst other direct intracellular delivery vehicles [28]. Direct translocation can describe multiple methods of entry, of which pore formation, inverted micelle formation and “carpet”-like model are among some of the main methods observed. Pore formation occurs due to interactions between the CPP and the phosphate groups on either side of the phospholipid bilayer once the CPP surface density reaches a certain threshold, leading to a formation of a pore and subsequent potential across the membrane, allowing influx of particles within the cell [41]. Inverted micelle formation occurs due to an initial electrostatic interaction between the CPP and the membrane, causing a disruption and the flip-flopping of phospholipids, leading to the formation of an inverted micellar structure. This structure permits the peptide to remain trapped within a hydrophilic environment, and the micelle is then capable of moving across the bilayer to the other side, where it then releases the CPP into the cytosol [42]. The “carpet” model of direct penetration describes the process whereby a cationic peptide binds to the negatively charged phospholipids, resulting in a “carpeting” of the cellular membrane. Above a threshold concentration, the CPP disrupts the lipid packing, facilitating the internalization of the CPP [42]. Although distinction has been made between the various methods of traversing the cellular membrane (clathrin/caveolae endocytosis, direct penetration, etc.), in reality a combination of entry methods are utilized simultaneously. For this reason, it is important to select a delivery vehicle that can be tailored to a predictable method of cellular uptake, as the fate of the cargo will vary depending on the method of internalization. As described previously, physical characteristics such as particle size, are also reported to play an important part in the internalization fate. Other characteristics that can have an effect include particle charge, aspect ratio and topography, as well as the specific internalization characteristics of the cell, which can vary between cell types.
16.2.2 Intracellular barriers 16.2.2.1 Endosomal escape The endocytic pathway is the most predominantly utilized uptake mechanism, and a membrane bound vesicle known as an endosome will engulf any cargo. If the cargo cannot escape during the early stages of endosomal maturation, it will be destined for lysosomal degradation and secretion by the cell. The cargo must be stable at pH 6.5–6.0 in the early endosome and at pH 5.5–4.5 in the mature late endosome [33]. Any cargo contained within the endosome must escape at this late stage, before the endosome fuses with the digestive enzyme-containing lysosomal vesicle. Failure to overcome this hurdle leads to a very low level of therapeutic delivery to intracellular targets. For these reasons, many therapeutic agents with promising in vitro promise fail to realize a potential in in vivo applications due to the lack of bioavailability. While many viral-based delivery systems have displayed efficient endosomal escape both in vitro and in vivo [43,44], endosomal escape is still a rate-limiting factor in
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
417
nonviral delivery systems. Many methods have been investigated for the combating of this issue, and one of the most promising methods involves the use of fusogenic peptides. A fusogenic peptide will undergo conversion from a β-sheet structure at the physiological pH 7.4 to conform to an α-helical structure at the acidic endosomal pH. The helical structure facilitates fusion to the endosomal membrane, allowing the escape of any endosomal inclusions [45]. Other methods of endosomal escape utilized by therapeutic delivery vehicles include pore formation by cationic amphiphilic peptides (AMPs) and membrane rupture due to the “proton sponge effect” caused by protonation [46]. Protonation can be exploited in the lower pH of the endosomal environment. Typically, these delivery systems have a high buffering capacity that swell under protonation, and examples of such delivery systems include cationic polymers and CPPs. The acidic endosomal pH causes protonation leading to the inflow of H+, Cl− and water into the endosomes, resulting in osmotic swelling and eventual rupture of the endosome.
16.2.2.2 Cytoplasm Following endosomal escape, therapeutic agents introduced within the cell are now located within the cytoplasm. The cytoplasm is a densely packed region full of cellular organelles (such as mitochondria, endoplasmic reticulum, Golgi apparatus) and cytosolic components (cytoskeletal filaments, soluble macromolecules such as proteins) [47], and as such is molecularly crowded. Therefore, transport of any delivered cargo within the cytoplasm that is intended for nuclear localization is severely hindered. For gene therapy, the cytoplasm is an inhospitable environment for DNA due to low transport mobility coupled with the presence of nucleases which will degrade free DNA. Lechardeur et al. [48] demonstrated that free DNA was degraded in the cytoplasm of HeLa and COS cells, exhibiting a half-life of only 50–90 min. This poses a significant barrier to the delivery of naked DNA, or DNA complexes that tend to disassociate in the cytoplasm before reaching the nucleus. Recently, studies have found the limiting factor in nucleic acid mobility through the cytoplasm is the size and spherical nature of the molecule, with circular plasmid DNA (pDNA) exhibiting faster transport than the equivalent linear structures [49]. There can only be two mechanisms by which cytoplasmic pDNA is transported to the nucleus; either by (1) diffusion or (2) active transport. However, due to the crowded nature of the cytoplasm, and the size of the DNA, it is highly unlikely that diffusion is the main transport mechanism. A study by Lukacs et al. [50] demonstrated that mobility of DNA larger than 250 bps inside the cytoplasm of cells was significantly slowed (>17-fold) compared to mobility in water, and that DNA larger than 2000 bp was simply not able to diffuse in a timeframe that is considered physiologically viable. It is thought that the high concentration of actin filaments, tubules and other microstructural cellular components are a major retardant to the diffusion of DNA in the cytoplasm. The highly cross-linked nature of the actin cytoskeleton was observed to be a major limiting factor, as the size-dependent diffusion rate of DNA appeared to no longer apply when the actin network was disrupted with the use of cytochalasin D [51]. This observation makes physical sense, as previous studies have reported that actin filaments tend to be approximately 100–500 nm
418
Bone Repair Biomaterials
in length, with the spacing between filaments observed to be ∼100 nm, and a 250 bp fragment of double-stranded DNA would exhibit a linear size of around 85 nm [52]. Therefore, it is easy to see why DNA may experience diffusional hindrance in the cytoplasmic environment. Consequently, since it is possible to show successful nuclear transfection with DNA it is assumed that the main mechanism of nuclear localization is by active transport along the cytoskeletal elements. Many viruses have been shown to utilize such mechanisms efficiently, such as the herpes simplex virus (HSV), which has been shown to shuttle along the microtubule elements in the cytoplasm of HeLa cells, moving from the periphery of the cell toward the nucleus with the molecular motor dynein implicated for such movement [53,54]. Further studies have shown that depolymerization of the microtubule network in HeLa cells using an actin depolymerization agent (nocodazole), showed a two-fold decrease in infection by HSV, further supporting the importance of cytoskeletal trafficking of complexes [55].
16.2.3 Suitable vehicles for the efficient delivery of therapeutic agents As described in Sections 16.1 and 16.2, many hurdles must be overcome for the successful intracellular delivery of any therapeutic agents in vivo. Therefore, the design of a suitable delivery vehicle is imperative for the successful delivery of the required cargo to the cytoplasm (in the case of RNAi) or the nucleus (pDNA), avoiding degradation or loss of function in the process. The delivery system must facilitate binding to the therapeutic agent, increase the extent and rate of cellular uptake, protect from degradation during circulation/endocytosis, facilitate endosomal escape, and disassociate from the cargo once cytoplasmic (or nuclear) delivery has occurred. Furthermore, if DNA is the chosen therapeutic agent, nuclear targeting and permeation of the nuclear membrane is one more limiting factor that needs to be overcome, usually achieved with the use of nuclear localization sequences (NLS) or other targeting ligands [56,57].
16.2.3.1 Nonviral versus viral delivery methods Having evolved to enable effective gene delivery into cells, viruses are currently one of the most effective methods of cell transfection, and in hard-to-transfect cells. Recombinant viruses can be generated by the insertion of exogenous DNA with the genome of the virus and is subsequently delivered inside the target cell via infection [58]. A wide variety of viral vectors have been developed to deliver therapeutic agents (most commonly genes), for the transient (adenovirus, vaccinia virus) or stable (retrovirus, lentivirus) transgene expression [59]. Viral delivery vehicles remain the “gold standard” in efficient in vitro and in vivo cellular transfection, and have been the subject of much research, with promising results being reported from various clinical trials [60]. However, due to potentially fatal adverse effects with the use of viral vectors, at this stage the FDA has yet to approve any virus-based therapies. The potential side effects of using viral vectors include: immunogenicity, off-target effects, toxicity, and insertional mutagenesis, including activation or proto-oncogenes and inactivation of tumor suppressor genes [61].
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
419
The inherently dangerous adverse side effects of using viral vectors for the transfection of cells, coupled with the associated expense, has led to extensive research into nonviral delivery methods that are simpler, more predictable, reproducible, and most importantly, provide a safer alternative. Historically, the major limitation on nonviral delivery systems was the observation of low levels of transfection and short-term gene expression observed in host cells in comparison with the viral alternatives. Fortunately, since only brief expression of osteogenic genes is enough for the stimulation of osteogenesis, nonviral delivery methods can be valuable to bone regeneration applications [62]. In addition, many recent studies have been able to exhibit levels of nonviral transfection on par with viral delivery systems [63,64], which points to the exciting potential of nonviral delivery systems. Additionally, such systems were also seen to exhibit much lower levels of toxicity, greater site-specificity, low immunogenicity and an ability to deliver large genes [65] making nonviral delivery systems an attractive and more clinically relevant option for the delivery of therapeutic agents. Some of the most commonly utilized nonviral systems for the delivery of therapeutic agents are described below and put into context for applicability for bone regeneration.
16.2.3.2 Nonviral: physical methods Physical methods used for the intracellular delivery of therapeutic agents rely on the application of external forces to the cellular membranes to increase transient permeability allowing entry of intended deliverables. Physical methods are often selected for use in hard-to-transfect cells, or to supplement another method of delivery. Usually employed in vitro, and rarely used in delivery of miRNAs, physical delivery of therapeutics can damage cell integrity and elicit a level of apoptotic response. Some of the physical methods employed include the use of mechanical, electrical, ultrasonic, hydrodynamic, and laser-based energy [66]. However, for bone regeneration scaffolds the use of physical methods do not present a viable option because of the need for a long term sustained release of therapeutic agents in vivo, which is simply not possible by way of physical intracellular delivery methods.
Microinjection of naked DNA The most simple and obvious method of introducing genes within a cell or tissue is by mechanical injection directly within the required location. Requiring no carrier system, this method of introduction is inherently safe, however, a major limitation of this method exists in the very low levels of efficiency observed [13], in some cases as low as 1% of cells experiencing transgene expression, due to rapid degradation by nucleases in the serum, or clearance by the MPS.
Biolistic delivery (the “gene gun”) Biolistic (a contraction of biological and ballistic) delivery involves the high-speed permeation of cell membrane with the use of a ballistic “gene gun.” Tungsten or gold particles (∼1–3 μm in diameter) are coated with DNA and accelerated using high voltage electric sparks, or a helium discharge [67,68]. This method aimed to overcome the difficulty of delivering naked pDNA by attachment to a high velocity particle carrier.
420
Bone Repair Biomaterials
Ballistic therapeutic delivery has shown to be effective in the delivery to exposed tissues such as skin (where it can penetrate into the epidermal and dermal layers) and any exposed surgical tissues, and has been particularly effective for use in DNA immunization and vaccination [69], and in the treatment of skin wounds and burns [70]. The efficiency of ballistic DNA delivery depends on the parameters selected, including the amount of DNA used, particle size, timing of the delivery, and the optimal acceleration conditions for the final localization of the DNA-coated particles [14]. Limitations to this method of therapeutic delivery include the slow preparation of particle carriers, often taking up to 2 days to prepare, and large variations in transfection efficiency due to shearing of DNA or particle carrier agglomeration [59]. Inflammation and irritation have also been reported at the introduction site of therapeutics delivered by ballistic means.
Electroporation Electroporation utilizes an electrical field to increase cell permeability, in a phenomenon known as electropermeabilization. First reported by Neuman et al. in 1982 [71], they found that by applying an electrical field greater than the capacitance of the membrane, charges of opposite polarity would arrange on opposing sides of the cell surface. This would create the formation of a pore due to the potential difference seen at this precise point, facilitating cellular entry through the pore. If the cells are to remain viable, the pore can then be closed if the field strength and pulse duration were correct. However, if the pore remains open, this leads to cell death, a method that is often utilized for the destruction of cancerous cells in patients [64]. Drawbacks to the use of electroporation exist in the difficulty in selecting the correct electrical field properties for optimal electroporation of the intended cell or tissue type, as incorrect properties may lead to irreversible cell damage. Furthermore, it has been reported that different conditions are required (field strength, pulse duration, etc.) for the delivery of differing molecules, with size and concentration affecting such properties. However, even with these described drawbacks, electroporation has been shown to be one of the most effective methods of nonviral delivery, and a reliable method of in vivo gene delivery in bone [72], and various other tissue types such as muscle [73], skin [74] and liver [75].
16.2.3.3 Nonviral: chemical methods Chemical carriers for the non-viral delivery can be broadly split into the categories of inorganic nanoparticles, lipid based, polymer based and peptide-based systems [76].
Inorganic nanoparticles Inorganic NPs (iNPs) used for the delivery of therapeutic agents include NPs prepared from ceramics (such as carbonate or phosphate salts of calcium, magnesium, or silicon) or metals (including iron, gold, and silver) [77]. Particles with a complex chemistry in the size range <100 nm can be easily prepared from these inorganic components, which facilitate surface coating for the binding of DNA, or for gene targeting. The nanoscale of iNPs offers various advantages to the delivery of therapeutic agents, including the ability to transfect cells successfully both in vitro and in vivo via
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
421
nonendocytic routes delivering NPs directly to the nucleus avoiding potential degradation in the endosome/lysosome [78]. Qureshi et al. [79] utilized silver nanoparticle complexes for the successful delivery of photoactivated miR-148b mimics, resulting in the osteogenic differentiation of MSCs. The transfection efficiency with the use of iNPs is not as good as other nonviral transfection options, however, iNPs are able to escape from the MPS, are inert to immune response due to the bio-inert chemistry, exhibit low toxicity and possess good storage stability. It must be noted that if DNA is to be delivered with the use of iNPs, by binding to the surface of the NP, further protection from intracellular degradation by nucleases is necessary. The use of superparamagnetic based iNPs (such as iron oxide NPs) are also of major interest for therapeutic delivery, as they are responsive to magnetic fields, facilitating the targeted delivery of cargo via magnetic field manipulation [80].
Cationic liposomes Cationic liposomes are synthetic lipid spheres that are composed by 1 or 2 fatty acids and alkyl functional groups, in addition, to a positively charged head group and hydrophobic tail group, connected via a linker structure. Cationic lipids possess the ability to form unilamellar vesicles, often with the help of a neutral or helper lipid, such as dioleoylphosphatidylethanolamine (DOPE) or dioleoylphosphatidylcholine (DOPC) [81,82], and these vesicles are capable of spontaneous interaction with DNA. The positively charged group on the cationic lipid interacts with the negatively charge phosphate groups on the DNA, resulting in self-assembly of lipid-DNA complexes known as a lipoplex. The delivery of genetic material within a cell with the use of liposomes (lipofection) is a process that was first pioneered in 1987 by Felgner et al. [83], but is now an intense area of interest for the nonviral delivery of therapeutic agents, and is currently one the most extensively investigated and commonly used delivery systems [84]. Liposomal delivery has been shown to be a highly effective method of gene transfer, and for bone regeneration in particular Park et al. [7] were able to report a comparable level of bone regeneration in critical-size defects of rat mandible treated with liposomal gene delivery to that by adenoviral vectors, negating the safety issues and constraints of requiring viral delivery. Cationic liposomes are more commonly used than anionic or neutral liposomes due to ease of production, high affinity for the negatively charged glycoproteins and proteoglycans of cell membranes facilitating cellular uptake, and a nonpathogenic and nonimmunogenic response. However, anionic and neutral liposomes are often utilized in the delivery of miRNAs, due to the lack of stability of cationic liposomes in circulation. Cationic liposomes suffer from unspecific binding to serum proteins, causing accumulation and subsequent removal by the MPS. For example, Huang et al. [85] were able to successfully increase miR29b expression in acute myeloid leukemia via miRNA delivery in anionic lipoplexes, and similarly, miR-34a transfer in lymphoma has been achieved through the use of neutral lipids [86]. The stability of cationic liposomes can be increased with the use of surface modifications, such as conjugation with polyethylene glycol (PEGylation), to create so-called “stealth liposomes” that are capable of avoiding detection by the MPS, consequently increasing circulation time [87,88]. It should be noted however, additions of PEG moieties to the liposome are associated with a decrease in interaction
422
Bone Repair Biomaterials
between the liposome and cellular membrane, leading to a decrease in levels of transfection [89]. Although the stability of cationic liposomes can be an issue, the main drawback with liposomal delivery is the acute cytotoxicity observed due to the strong cationic nature and lack of linker group degradation [90]. Optimal delivery of DNAlipoplexes has been reported when the positive/negative charge ratio is around 1, and liposomes are observed to become cytotoxic when the ratio of lipid:DNA surpasses 3:1 [64,83]. Liposomes continue to be an area of interest in nonviral delivery, and hundreds of combinations have been designed involving various lipids and surface modifications, including peptides and ligands, to combat stability and toxicity issues, and to offer more control over particle size, morphology, and degradation characteristics [91]. Many commercially available liposome transfection agents are commonly used as a benchmark for transfection efficacy such as Metafectene, Lipofectamine 2000, and Lipofectin [92], and as of August 2016 lipofection accounts for 4.7% (n = 115) of viral and nonviral vectors used in human clinical gene therapy trials worldwide [93].
Cationic polymers Cationic polymers were introduced around the same time as cationic liposomes by Wu et al. in 1988, who successfully utilized a poly-l-lysine (PLL) carrier to deliver pDNA to rats in vivo [94]. Like liposomes, cationic polymers are capable of interacting with DNA material to form a complex known as a polyplex. Further development lead to the second generation of polymeric carriers, the first of which was pioneered by Boussif et al. in 1995 who introduced the use of polyethylenimine (PEI) for the intracellular delivery of DNA [95]. PEI is a dense, highly cationic polymer that exists in either branched (BPEI) or linear (LPEI) forms, both of which are seen to exhibit high transfection activity [95–97] in vitro and moderate activity in vivo. Comparably, LPEI has been observed to exhibit lower toxicity and better gene transfer efficiency than the BPEI counterpart [98]. Although PLL is still the most commonly investigated polymer delivery vector due to the good biodegradability (resulting in lower toxicity) and has been used in a wide range of applications, PEI is considered by many to be the gold standard in polymeric carriers. PEI possesses high transfection efficacies [97], which are attributed to the high density of amine groups, the majority of which are unprotonated at a physiological pH. Upon entry into the cell via endocytosis, and compartmentalization in the endosome, the unprotonated amines exerts the “proton sponge effect,” as described in Section 16.2.1, effectively absorbing protons transported into the intracellular region by the ATPase proton pump, preventing acidification of the endosome [99]. What follows is an influx in chloride ions into the endo-lysosomal vesicle, resulting in osmotic swelling and destabilization of the membrane, preventing subsequent lysosomal degradation of the contained polyplexes. Further intracellular trafficking of DNA delivered by PEI-DNA polyplexes has been observed to result in high levels of transgene expression [100]. Disadvantages of PEI include low levels of biodegradability [101], with subsequent toxicity. The toxicity of PEI in particular has been observed to behave in a dose-dependent manner, and for this reason, the use of this polymer in vivo is severely limited due to the high concentrations necessary for sufficient transfection
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
423
efficacy [102]. Although many PEI-derivatives have exhibited lower toxicity (involving functionalization with other copolymers such as PEG [103]) and higher transfection efficacies (with the inclusion of hydrophobic substituents such as lipids or polycaprolactone, or targeting ligands such as transferrin [104,105]), PEI-based therapeutics are still a long way from use in clinical treatment. Based on a record of proven clinical safety [106], poly(lactic-co-glycolic acid) (PLGA) has been widely investigated as a nontoxic alternative to PEI/PLL, and a suitable carrier of therapeutic cargo. PLGA is a biodegradable polyester that undergoes bulk hydrolysis, facilitating a sustained release of cargo, with the degraded products easily removed by the citric acid (Krebs) cycle [107]. Due to the attractive biodegradability, biocompatibility and ease of processing, FDA-approved PLGA carriers are extensively used in drug delivery [107–110]. In particular, the ability of PLGA to bind to and protect DNA from in vivo degradation, by the rapid escape from the endo-lysosome, has led to the increased use of PLGA polyplexes for the delivery of nucleic acids in addition, to proteins and peptides [110–113]. PLGAcontaining drug delivery devices are routinely in clinical use, such as Lupron Depot, utilized in the treatment of prostate cancer [114]. Although safer than other cationic polymeric carriers, conventional PLGA contains negative charges which severely hinder the encapsulation of anionic cargo (such as DNA) and reduce overall transfection efficacy. It is therefore necessary to add other copolymers such as chitosan [115,116].
Chitosan Chitosan is a naturally occurring cationic polysaccharide that has proved to be an attractive option for use in the polymeric delivery of therapeutics due to the outstanding biodegradability, excellent biocompatibility, low immunogenicity, antimicrobial activity and ease of production [117,118]. The in vitro transfection efficiency of chitosan has been observed to correlate with such factors as the degree of deacetylation, molecular weight of the chitosan selected, and in the case of DNA-complexing, the ratio of nitrogen atoms in the chitosan to the phosphorus atoms in the DNA (known as the N:P ratio). Therefore, varying efficacy is seen in the intracellular delivery using chitosan depending on the chosen chitosan carrier. For example, high molecular weight (HMW) chitosan produces a more stable particle compared to low molecular weight (LMW) chitosan (including better DNA condensation) but disadvantages include increased aggregation of particles and a low solubility at a physiological pH [119]. LMW chitosan outperforms HMW with respect to intracellular release but the degree of complexation is severely hampered [120]. Chitosan does not have the pH buffering capacity required for endosomal disruption [121]. However, abundant amine and hydroxyl groups contained within chitosan enables simple modifications for increasing transfection efficiency or gene transfer. Typically, chitosan is cografted with other polymers such as PEI or PEG [122,123]. The naturally occurring biocompatibility and biodegradability properties of chitosan make it a highly investigated option for the targeted delivery of therapeutics, but low solubility, poor transfection efficiencies and a lack of targeting are limiting factors.
424
Bone Repair Biomaterials
Dendrimers Dendrimers are synthetic, homogeneous, monodisperse and spherically symmetrical polymers consisting of a well-defined tree-like branch structure [59]. Dendrimers contain positively charged terminal groups that are capable of functionality for the attachment of antibodies, therapeutic agents, or DNA at a physiological pH, forming dendriplexes, like lipoplexes or polyplexes [124]. Modification of dendrimer terminal groups results in a hydrophilic interior and hydrophobic exterior or vice versa. These characteristics confer flexibility of use [59]. The most common dendrimers (and commercially available) are poly(amidoamine) (PAMAM) and subsequent derivatives, which are observed to form spherical polymers with good aqueous solubility and the amount of positively charged exposed surface groups is an advantage facilitating endo-lysosomal escape [125]. The nanosize (5–20 nm) of PAMAM dendrimers in addition, to high molecular uniformity, ease of preparation, ease of large scale production, and the ability for precise functionalization due to numerous copies of surface groups present, has meant that they are an extensively used in the delivery of therapeutics [126–128]. Oliveira et al. [129] were able to induce osteogenic differentiation in rat bone marrow-derived MSCs (BMSCs) via scaffold-mediated delivery of dexamethasone loaded PAMAM dendrimers. While Santos et al. [130] used dendrimers to deliver a plasmid encoding BMP-2 inducing osteogenic differentiation in rat BMSCs. The structure of a dendrimer starts with a core molecule that is subsequently grown through a stepwise polymerization process. With each new layer (known as a generation G1, G2, G3, etc.), the number of surface amine groups is observed to increase, as is the MW and subsequent charge density [131]. The transfection efficiency and toxicity of dendrimers is generation-dependent. For example, PAMAM dendrimers G0–G3 have been reported to exhibit poor gene transfection efficiency and a low cytotoxicity, but higher generations such as G4–G8 exhibit a better gene transfection but with an associated increase in toxicity [132]. This is similar to the optimization trade-off between LMW and HMW chitosan polymer carriers, as described in Chitosan section. As the ideal transfection vector requires the highest possible efficiency and the lowest possible toxicity, many researchers are working to optimize the properties of PAMAM dendrimers for therapeutic delivery [133,134].
Cell-penetrating peptides Cationic peptide-based vectors are generally considered to hold many advantages over the various other options of nonviral delivery and have been shown to complex anionic cargoes, target specific intracellular or extracellular receptors, disrupt endosomal membranes, and protect the cargo in the cytoplasmic environment [135]. Additionally, peptide sequences have been conjugated to lipoplexes, polyplexes, or dendriplexes, to target or improve efficacy [136,137]. CPPs are a unique class of fusogenic peptide sequences designed to interact with the endosomal membrane, facilitating escape and intracellular delivery of therapeutic agents. CPPs rose to prominence in the late 1980s following multiple reports that the transactivator of transcription (Tat) protein of the human immunodeficiency virus
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
425
(HIV) was able to efficiently enter tissue-cultured cells and promote viral gene expression [138,139]. Generally no more than 30 amino acids in length, most known CPPs exhibit a net positive charge at physiological pH (ideal for the condensation of DNA) and normally contain a high content of basic residues, ranging from 17% in human calcitonin and its C-terminal fragment 9–32 (hCT(9–32)) [140] to 100% with polyarginines [141]. It is generally accepted that CPPs can be placed into one of three major classifications; protein-derived CPPs, chimeric CPPs, or synthetic CPPs (Table 16.1) [28]. Protein-derived peptides are normally derived from the minimal effective partial sequence of the parent translocation protein that interacts with cell membranes, known as protein transduction domains. Chimeric peptides usually involve the fusion of hydrophilic and hydrophobic domains from other natural sources. Synthetic peptides are rationally designed sequences that aim to create well-defined α-helical structures for amphipathicity, usually by involving motifs of other known CPPs. With the exception of polycationic homopolymers such as polyarginines or polylisines, most CPPs exhibit a distinct amphipathic structure (such as the chimeric CPP transportan [146]), or are seen to become amphipathic when conforming to an α-helical structure (such as the synthetic peptide MAP) [148]. CPPs are readily internalized by a wide range of cell lines [142] and primary mammalian cells (such as rat brain and rat spinal cord [142], calf aorta [151], and porcine and human umbilical vein endothelium [148]) for the delivery of a wide array of both large and small cargo such as nanoparticles, proteins, liposomes and DNA [152]. Suh et al. [153] were able to report the efficient intracellular delivery and subsequent upregulation of microRNA-29b into hMSCs with the use of an arginine-rich CPP, and reported a significant increase in osteogenic effect compared to the same miRNA delivered via lipoplexes. Furthermore, the successful in vivo efficacy of CPPs is routinely reported, and is widely used in the delivery of small interfering RNA (siRNA). Intravenous injection of MPG/cyclin-B1 siRNA particles was reported to efficiently block tumor growth [154,155], and the use of a small peptide derived from the rabies virus glycoprotein linked to polyarginine-9 was capable of siRNA delivery to the central nervous system. Although CPPs are successfully utilized for the intracellular delivery of therapeutic cargo, the method of internalization has many conflicting reports. It is thought that CPPs are taken up by cells via the energy-dependent endocytic routes (see Endocytosis section ), however, a large number of investigations report internalization of CPPs under conditions that should otherwise prevent translocation via endocytosis, such as low temperatures (e.g., 0–4°C) or in the presence of various endocytic inhibitors [142–144,146,156]. However, endocytosis does appear to play a role in the uptake of CPPs to a certain extent [157], therefore it is clear that CPPs are not limited by one method of internalization, but more likely use a combination of pathways. Differing CPP sequences have been observed to translocate by different methods, alluding to a dependence on CPP sequence as a potential contributor to the method of internalization. In Tatderived CPPs it has been postulated that endocytosis could be an exclusive method,
426
Table 16.1 Commonly utilized cell-penetrating peptides (CPPs) including sequences and origin Classification
Name
Sequence
Origin
Ref.
Protein-derived
Penetratin
RQIKIWFQNRRMKWKK
[142]
Tat pVEC
GRKKRRQRRRPPQC LLIILRRRIRKQAHAHSK
MPG
GALFLGFLGAAGSTMGAWSQPKSKRKV
Transportan
GWTLNSAGYLLGKINLKALAALAKISIL
Pep-1 MAP Polyarganines R6W3
KETWWETWWTEWSQPKKKRKV KLALKLALKALKAALKLA (R)n 6 < n < 12 RRWWRRWRR
Drosophila antennapedia homeodomain (amino acids 43–58) Protein from HIV-1 (amino acids 48–60) Derived from murine vascular endothelial cadherin (amino acids 615–632) Peptide derived from fusion sequence of HIV-1 gp41 protein coupled to peptide derived from the nuclear localization sequence of SV40 T-antigen minimal active part of galanin (amino acids 1–12) coupled to mastoparan via Lys13 HIV-reverse transcriptase/SV40 T-antigen de novo Based on Tat peptide Based on penetratin
Chimeric
Synthetic
[143] [144] [145]
[146]
Bone Repair Biomaterials
[147] [148] [149] [150]
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
427
as Tat 48–60 was observed to not enter live HeLa cells at 4°C, and has been recently thought to enter cells primarily via lipid raft-mediated macropinocytosis [158,159]. Internalization of CPPs as a result of direct penetration is reported as a primary method of entry (see Direct translocation section), with translocation occurring via such as mechanisms as the “carpet” model for pVEC and transportan-10 [160], or via the adoption of an α-helical structure of MAP when in contact with lipid membranes [161] leading to pore formation. It has been reported that Tat-derived CPPs (the most commonly utilized category of CPP) are limited in an ability to escape the endo- lysosome, and as such with functionalization of Tat via addition of a histidine tail, Lo et al. [162] were able to improve the gene efficiency more than 7000-fold compared to controls. Histidine is an amino acid that is often included in the peptide sequence of CPPs due to the promotion of the “proton sponge effect” for endo-lysosomal membrane disruption. Endosomal escape of therapeutic cargo delivered by CPP carriers is of the utmost importance to avoid subsequent lysosomal degradation. Many membrane destabilizing sequences can be found naturally, such as the influenza virus haemagglutinin 2 protein (HA-2) [163]. Upon a decrease in pH, the acidic residues contained within this peptide are protonated, facilitating the formation of an α-helical structure. PLL-DNA polyplexes conjugated with HA-2-derived peptide sequences have been reported to increase in vitro gene expression from as little as 10-fold, to as much as 10,000-fold increase over PLL-DNA complexes without the addition of the fusogenic peptide [135]. The pH dependent endosomal disruption behavior of this peptide sequence found in nature has subsequently lead to the creation of synthetic alternatives, such as GALA, KALA and H5WYG [164–166]. GALA is a sequence of glutamic acid (E), alanine (A), leucine (L), alanine (A) amino acid repeats (forming EALA sequence), which creates a fusogenic peptide capable of taking on the conformation of a strong α-helical structure at a range of pH levels (including low pH), exhibiting regions of both hydrophilicity and hydrophobicity [164]. At physiological pH, the glutamic acid residues cause a charge repulsion, however, as the pH is lowered (such as in the endosome) these charges are neutralized, allowing adoption of the α-helical shape. At the endosomal pH, GALA is capable of fusing to the bilayer membranes, inducing leakage of phosphatidylcholine vesicles [135]. Despite exhibiting good results in condensing cationic products [167], due to the anionic nature of GALA, it cannot be used as an efficient method of condensing anionic complexes (such as DNA). KALA is a modified version of GALA with the glutamic acid (E) repeats substituted for lysine (K) [165]. With the addition of lysine, it follows that KALA is cationic in nature and can therefore be used to condense anionic complexes. However, the presence of the lysine residues means that the α-helicity of KALA is disrupted as the pH level decreases, causing a decrease in the fusogenic ability, as well as inciting cytotoxic effects of the peptide [165]. With respect to these properties, KALA is therefore not desirable for use in situations where there may be a variation in pH, such as in the endosomal environment. As such, the ideal CPP should exhibit the properties of GALA (ability to hold α-helicity at a lower pH) but also be similar to KALA with respect to the condensation of anionic entities.
428
Bone Repair Biomaterials
Arginine Alanine Leucine Glutamic acid Tryptophan Histidine
Figure 16.1 Alpha-helical structure of RALA peptide. The arginine-rich region provides RALA with its hydrophilic nature and facilitates the binding of anionic complexes. The leucine-rich domain provides the hydrophobic region and allows interaction with lipid membranes. The hydrophobic and hydrophilic regions are separated by an alanine-rich region, subsequently giving RALA amphipathic properties. Tryptophan increases stability within the cell and glutamic acid increases solubility at physiological pH.
To this end, McCarthy et al. [168–171] have designed a fusogenic peptide that fits these desired characteristics of a peptide for the condensation and delivery of anionic complexes. Termed RALA, the lysine residues are substituted with arginine (Fig. 16.1). Arginine is found in naturally occurring condensation motifs and has been shown to exhibit superior DNA transfection compared to lysine [172]. RALA demonstrates strong nanoparticle formation of plasmid DNA, and maintain desirable α-helicity at reduced pH (∼pH 5.0) facilitating endosomal escape of NP cargo into the cytosol of cells. This is an important property of any CPP, as the pH in the endosome has been shown to be in the region of pH 5.0–5.5 [172]. In addition, RALA displays an in vitro transfection efficacy on par with commercially available transfection agents such as Lipofectamine, while beneficially exhibiting a superior lower cytotoxicity profile [168]. CPPs, and fusogenic CPPs in particular, are a highly attractive option for the intracellular delivery of oligonucleotides due to reduced cytotoxicity and immunogenicity compared to viral alternatives. CPPs can be designed to be multi-functional, are easily manufactured and scaled up and so it follows that this class of delivery systems are gaining momentum in the drug delivery field. With respect to bone tissue engineering and regeneration applications, there is the potential for the use of oligonucleotides directly delivered via CPPs such as RALA into the cytosol of cells, facilitating an increase in osteogenic differentiation and accelerating the rate of mineralization of osteoprogenitor/osteoblast cells (Fig. 16.2).
Thymine
Guanine
Adenine
Size < 100 nm Charge ~ +20 mV
Cytosine
4 Arginine
Amphipathic cationic peptide (RALA)
RALA/pDNA complex
1 Clathrin/caveolae mediated endocytosis
Nucle
us
pH<5 –
Osteogenic genes Osteogenic differentiation
Cell
3 Cytosolic release
2 Endosomal acidification
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
pDNA
Figure 16.2 Endosomal escape of RALA/pDNA NPs. Cationic peptide RALA interacts with anionic plasmid DNA to create net cationic RALA/ pDNA NPs that are internalized via clathrin/caveolae-mediated endocytosis (1). Upon the decrease in pH experienced within the endosomal environment (2), RALA conforms to the alpha-helical shape, facilitating endosomal escape (3), and delivery of the pDNA to the cytosol for an increase in osteogenic genes (4). 429
430
Bone Repair Biomaterials
16.3 MicroRNA: the future of nucleic acid therapies for bone regeneration MicroRNAs (miRNA) are small single strand, noncoding RNA molecules (approximately 22 nucleotides in length) that regulate posttranscriptional expression of messenger RNA (mRNA) and proteins [173]. First discovered in 1993 by Ambros et al. [174], at the time of writing there are more than 2000 mature human m iRNAs identified and cataloged [175], and undoubtedly more will be discovered. The power of miRNA therapeutics comes from the fact that a single molecule can target more than 100 genes, and each gene is normally a target of multiple miRNAs. Given that such a vast number of miRNAs have been identified it has been predicted that miRNA is responsible for the influence of anywhere from 30% to 80% of the human genome [176,177]. miRNAs have been shown to possess an important role in cellular processes such as differentiation, proliferation and apoptosis [178–180], but also in many biological processes such as the pathogenesis of human diseases such as diabetes [181], neurodegenerative diseases [182], cancer development [183] and tissue regeneration [184,185]. More recently, miRNAs have also been shown to participate greatly in osteogenesis, displaying involvement in osteoblast differentiation through the regulation of signaling pathways, either enhancing or inhibiting differentiation via binding to osteogenic transcription factors (TF) [186–188]. Additionally, miRNAs have been identified as regulators of osteoblast and osteoclast behavior and as a result play are involved in the process of bone remodeling and many bone metabolic diseases (such as osteoporosis) [189].
16.3.1 Biogenesis of microRNA The biogenesis of miRNA in mammalian cells is summarized in Fig. 16.3 and begins with RNA polymerase II transcription of a hairpin primary miRNA (pri-miRNA). The pri-miRNA is transcribed from a miRNA coding region which occurs either in the portions of noncoding regions (introns) of protein-coding genes, or as independent transcriptional units [190]. The pri-miRNA is then cropped by the nuclear RNaseIII type endonuclease Drosha, resulting in a double-stranded stem-loop intermediate, called precursor miRNA (pre-miRNA) that is 70 nucleotides in length [191]. Drosha is assisted by the nuclear protein DiGeorge syndrome critical region gene 8 (DGCR8). DGCR8 is responsible for stabilizing Drosha and recognizes the double-stranded RNA structure of the hairpin pri-RNA. It is also thought that DGCR8 is responsible for the correct orientation of Drosha on the pri-RNA [192]. The cleaving of the primiRNA is an important event that predetermines the mature sequence of the miRNA, as well as the substrate for further downstream events [193]. Cleaved pre-miRNA is subsequently exported from the nucleus into the cytoplasm via the transporter complex exportin-5, which not only facilitates nuclear exportation, but also protects pre-miRNA from nuclear digestion [194]. Once in the cytoplasm, premiRNA is once again cleaved by the RNase Type III endonuclease Dicer resulting in a ∼22 base pair (bp) double-stranded miRNA duplex [195]. Subsequently, via binding
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
Nucleus
RNA polymerase II
431
Cytoplasm
1 Transcription
(A)n
Hairpin pri-miRNA
Drosha DGCR8 (A)n
DNA
3’ UTR of target messenger RNA
2 Cleavage pre-miRNA
5 mRNA pairing
degraded strand Exportin-5 mature strand 3 Nuclear export
miRNA duplex
Risc
pre-miRNA Dicer 4 Cleavage
Figure 16.3 Schematic representation of the biogenesis of miRNA.
with Argonaute (Ago) proteins, one strand of this duplex is selected for degradation (passenger strand), while the other remains as a mature miRNA and is incorporated into the RNA-induced silencing complex (RISC) [196]. The inherent thermodynamic asymmetry of the miRNA duplex is the primary determinant of which strand will be selected to form the mature RNA, that is, the RNA with the 5′ strand that is the least stably bound with the opposite strand is selected [197]. Finally, mature miRNA complexes typically target the 3′ untranslated region (3′UTR) of the mRNA. The miRNA binds via what is known as the “seed” region, located between nucleotides 2–8 on the 5′- end of the miRNA [197], by either complementary or partially complementary base pairing. Once bound, the mature RISC complex controls specific gene expression at a posttranslational level by the recruitment of various protein factors, and degradation of the mRNA [198].
16.3.2 Prominent microRNAs involved in osteogenesis It is well established that MSCs are the source for osteoblast cells recruited for osteogenesis. MSCs are capable of differentiation into chondrocytes, adipocytes, and osteoblasts, determined by a complex array of osteogenic factors and signals received at a posttranscriptional level, of which miRNAs are among some of the most crucial. Since
432
Bone Repair Biomaterials
Table 16.2 Commonly reported miRNAs involved in osteogenesis
miRNA
Gene target(s)
miR-26a miR-29a miR-29b
SMAD 1, GSKβ, TOB1 RUNX2, COL1A, COL5A TGFβ3, HDAC4, DUSP2, AcvR2b FAK DLX5, Wnt signaling target CDC25 A HOXC8, HOXB7 TGFβ, VEGF, AcvR1b RUNX2, DKK1 RUNX2, FGFR2 GaINT7, CASP3, GSK3β Osterix HDAC5
miR-138 miR-141 miR-196a miR-210 miR-335 miR-338 miR-378 miR-637 miR-2861
Promotion (↑) or inhibition (↓) or miR required for osteogenesis
Ref.
↑/↓ ↑/↓ ↑
[187,199,200] [201–203] [188,203]
↓ ↓
[204,205] [206–208]
↑ ↑ ↑/↓ ↓ ↑ ↓ ↑
[209,210] [211,212] [213,214] [215–217] [218–220] [215,221] [222,223]
the miRNAs are capable of binding to the target sequences in the UTR of mRNAs, causing translational arrest or degradation by the RISC, it is clear miRNA can potentially have a considerable inhibitory or enhancing effect on osteogenesis by binding to osteogenic TFs. Vast numbers of miRNAs have been reported to be involved in the differentiation of BMSCs into mature osteoblasts, and a selection of the most commonly reported miRNAs involved have been summarized in Table 16.2. Some of the microRNAs studied are downregulated during normal osteogenesis, and as a result are considered as osteogenic inhibitors (such as miR-138, -141, -338), while others are upregulated and are considered osteogenic promoters (such as miR29b, -196a, 378). A selection of the miRNAs reported in literature show clear contradictory behavior (such as miR-26a, -29a, -335) with many researchers reporting either inhibitory or promoting behavior depending on their experimental conditions, which is testimony to the inherent complexity and cross-talk of the molecular mechanisms at play during the osteogenic differentiation of MSCs. Recently, many TFs have been identified to be involved in bone regeneration, however, two of the most prominent TFs in osteoblastogenesis specifically are RUNX2 and Osx (which functions downstream from RUNX2 [224]). RUNX2 functions by binding to the promotor region of osteoblast specific genes (ALP, COL1, OPN, OCN, and bone sialoprotein) and initiates transcription [225]. Furthermore, the expression of RUNX2 is significantly increased during the early stages of osteoblast differentiation at the preosteoblast stage. RUNX2 is therefore identified as an essential transcriptional marker for the detection of osteoblast differentiation. It has been shown that upregulation of RUNX2 by MSCs induces osteogenic differentiation, whilst suppression of RUNX2 induces the MSCs to undergo chondrogenic or adipogenic differentiation [226]. Although RUNX2 is essential during the preosteoblast stage, expression
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
433
of RUNX2 has been shown to not be necessary for the continued expression of the major osteogenic genes in mature osteoblasts [227]. Many miRNA are reported to affect the expression of RUNX2, either by direct targeting, or indirectly by targeting RUNX2 inhibitors/promotors. miR-23, miR-24, miR-106a, miR-133, miR-222, miR335, miR-338 and miR-433 among others inhibit osteogenic differentiation by the direct targeting of RUNX2 [215,216,223,228]. Conversely miR-764 promotes osteoblast differentiation by inhibiting RUNX2 degradation and miR-3960 promotes the osteoblast differentiation via targeting homeobox A2 (HOXA2), a known RUNX2 inhibitor [223]. Osx functions downstream from RUNX2 [224], and is a vital transcription factor involved in osteogenesis involved in may signaling pathways, including MAPK and protein kinase D [229]. Osx functions during MSC differentiation from preosteoblasts, and for mediation of function during the mature osteoblast phase. Furthermore, BMP-2 mediated induction of Osx was inhibited by the blocking of RUNX2, further indicating the regulatory role of RUNX2 in the expression of Osx [224]. Osx-null mice die at birth due to lack of mineralized skeletons [230] and Osx-deactivated mice have been reported to experience spinal deformities [231] indicating the important role of Osx in skeletal development. miR-637 has been reported to directly target Osx, and maintains the balance between adipocytes and osteoblasts. Expression of miR-637 during osteogenic differentiation is decreased, with miR-637 supressing COL4A1 and Osx, consequently inhibiting osteogenic differentiation [221]. Furthermore, Osx has been shown to be important in osteoblast mineralization, and overexpression of Osx-targeting miR-93 has been reported to inhibit mineralization [232]. Goettsch et al. [233] reported a decrease in Osxtargeting miR-125b during the transdifferentiation of vascular smooth muscle cells, and subsequently that the inhibition of endogenous miR-125b can in fact promote osteogenesis. In addition, miR-31, miR-142 and miR-138 have been identified as Osx inhibitors [234], and miR-322 was shown to be a positive modulator of Osx, by targeting Tob2, which regulates Osx degradation [235]. There are numerous other important TFs involved in osteogenesis that are targeted and finely tuned by miRNAs, and interestingly, many of which are known to be associated to RUNX2 as downstream, upstream, cofactors or binding partners with RUNX2 [234]. Furthermore, miRNAs also participate in the mediation of the osteogenic signaling pathways such as BMP/TGF-β, Wnt/β-catenin and MAPK pathways [236]. With the vast number of miRNAs identified (with more constantly being discovered), and with the numerous targets of each miRNA, the precise effect a particular miRNA has on the osteogenic differentiation of MSCs is not well known. Therefore, miRNAs are of great interest, and worthy of much more investigation for use in enhancing bone regeneration.
16.3.3 Bone repair and miRNAs Due to the infancy of this therapeutic alternative, there exists only a few reports of miRNAs successfully utilized in vivo for the repair of nonunion. However, in patients with fractures, miR-92a is downregulated and after 21 days the levels of
434
Bone Repair Biomaterials
miR-92a expression are seen to recover, indicating the inhibitory effect of miR-92a on bone regeneration. To this end, Murata et al. [237] performed systematic delivery of anti-miR-26a on young mice with femoral fractures and saw an enhanced effect in fracture healing with increased callus formation and vascularization. Most often, miRNAs are utilized in a scaffold based system and have been exhibiting promising results. Downregulation of miR-31 in BMSCs has been shown to promote osteogenesis, and Deng et al. [238] seeded anti-miR-31 expressing BMSCs onto a polyglycerol sebacate scaffold, and reported effective bone repair in rats with a critical-size calvarial defect. In a similar study, BMSCs transfected with anti-miR-31 were seeded on β-TCP based scaffolds, and implanted into the medial orbital bone defects in a canine model, and efficient repair with increased bone mineral density and volume was observed [239]. Angiogenesis, along with osteogenesis, is also a necessary factor in the regeneration of healthy bone tissue, and miR-26a is a known factor in both processes. Li et al. [187] were able to exhibit improved bone repair with both osteogenesis and angiogenesis by the seeding of miR-26a transfected BMSCs on a hydrogel and implanted in a rat critical-size cavarial defect. The scaffolds in this study exhibited complete bone repair with improved vascularization, hinting at the importance of osteogenic/angiogenic coupling. Therefore, with the increased exposure of miRNA as an enhanced therapeutic option for the treatment of bone regeneration, the use of bone scaffolds containing miRNA therapeutics is certainly a promising and highly exciting area of tissue engineering.
16.4 Final conclusions In summary, the most recent advances and breakthroughs in bone regeneration strategies are highlighting the importance of the bone microenvironment, with strong interrogation of the “cellular cross-talk” and paracrine effects of any cell components that are included in such technologies. Many promising results are being reported through the cellular delivery of nucleic acids for the purposes of genetic alteration. The use of plasmid DNAs, mRNAs, siRNAs, and most recently miRNAs to introduce the upregulation of osteogenic-promoting genes or the downregulation of osteogenic-inhibiting genes in MSCs or osteoprogenitor cells has led to the creation of third-generation bone regeneration strategies that are proving to be significantly more efficacious than their second generation (biomaterial only) counterparts. Additionally, such strategies may also remove some of the associated risks with cell-seeded regeneration scaffolds (such as immunogenic rejection) as the genetic engineering of cells can be done via an autologous cell transplant. However, to further drive the advancement of such technology, a more robust understanding of the cellular mechanisms and signaling pathways involved in osteogenesis must be uncovered, to better inform future research. Furthermore, a well-chosen nucleic acid strategy is only theoretical and is inherently limited by the percentage of cells that can be successfully transfected with the intended genetic
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
435
cargo. Therefore, selection of a suitable nonviral delivery vehicle for nucleic acid therapies is of the utmost importance to ensure the highest levels of efficacy, and the use of CPPs such as RALA are exhibiting some of the highest levels of transfection efficiency (higher than CaP or PEI delivery vectors) without the high levels of toxicity usually associated high transfection rates (such as that seen during lipofection). The combination effects of a well-chosen biomaterial in addition, to the use of a well understood nucleic acid therapy, to better mimic the native processes of bone regeneration on a molecular scale, may demonstrate some of the best results to-date for bone regeneration technologies.
References
[1] Buschmann J, Welti M, Hemmi S, et al. Three-dimensional co-cultures of osteoblasts and endothelial cells in DegraPol foam: histological and high-field magnetic resonance imaging analyses of pre-engineered capillary networks in bone grafts. Tissue Eng Part A 2011;17(3–4):291–9. [2] Meirelles Lda S, Fontes AM, Covas DT, Caplan AI. Mechanisms involved in the therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor Rev 2009;20:419–27. [3] Linero I, Chaparro O. Paracrine effect of mesenchymal stem cells derived from human adipose tissue in bone regeneration. PLoS One 2009;9. e107001. [4] Granero-Moltó F, Weis JA, Miga MI, et al. Regenerative effects of transplanted mesenchymal stem cells in fracture healing. Stem Cells 2009;27(8):1887–98. [5] Walker DH, Wright NM. Bone morphogenetic proteins and spinal fusion. Neurosurg Focus 2002;13(6):e3. 2002. [6] Lattanzi W, Parrilla C, Fetoni A, et al. Ex vivo-transduced autologous skin fibroblasts expressing human Lim mineralization protein-3 efficiently form new bone in animal models. Gene Ther 2008;19:1330–43. [7] Park J, Ries J, Gelse K, et al. Bone regeneration in critical size defects by cell-mediated BMP-2 gene transfer: a comparison of adenoviral vectors and liposomes. Gene Ther 2003;10:1089–98. [8] Zhu W, Rawlins BA, Boachie-Adjei O, et al. Combined bone morphogenetic protein-2 and -7 gene transfer enhances osteoblastic differentiation and spine fusion in a rodent model. J Bone Miner Res 2004;19:2021–32. [9] Partridge K, Yang X, Clarke NM, et al. Adenoviral BMP-2 gene transfer in mesenchymal stem cells: in vitro and in vivo bone formation on biodegradable polymer scaffolds. Biochem Biophys Res Commun 2002;292:144–52. [10] Tang Y, Tang W, Lin Y, et al. Combination of bone tissue engineering and BMP-2 gene transfection promotes bone healing in osteoporotic rats. Cell Biol Int 2008;32:1150–7. [11] Asahara T, Kalka C, Isner JM. Stem cell therapy and gene transfer for regeneration. Gene Ther 2000;7(6):451–7. [12] Fang J, Zhu YY, Smiley E, et al. Stimulation of new bone formation by direct transfer of osteogenic plasmid genes. Proc Natl Acad Sci USA 1996;93(12):5753–8. [13] Herweijer H, Wolff JA. Progress and prospects: naked DNA gene transfer and therapy. Gene Ther 2003;10(6):453–8. [14] Mehier-Humbert S, Guy RH. Physical methods for gene transfer: improving the kinetics of gene delivery into cells. Adv Drug Deliv Rev 2005;57:733–53.
436
Bone Repair Biomaterials
[15] Natsuga K. Epidermal barriers. Cold Spring Harbor Perspect Med 2014;4(4):a018218. [16] Bleich NK, Kallai I, Lieberman JR, et al. Gene therapy approaches to regenerating bone. Adv Drug Deliv Rev 2012;64(12):1320–30. [17] Wang W, Ferguson DJ, Quinn JM, et al. Biomaterial particle phagocytosis by bone-resorbing osteoclasts. J Bone Joint Surg Br 1997;79(5):849–56. [18] Demetzos C. Pharmaceutical nanotechnology: fundamentals and practical applications. Singapore: Springer; 2016. [19] Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol 2015;33(9):941–51. [20] Champion JA, Mitragotri S. Role of target geometry in phagocytosis. Proc Natl Acad Sci USA 2006;103(13):4930–4. [21] Nie S. Understanding and overcoming major barriers in cancer nanomedicine. Nanomedicine 2010;5(4):523–8. [22] Harris JM, Chess RB. Effect of pegylation on pharmaceuticals. Nat Rev Drug Discov 2003;2:214–21. [23] Parodi A, Quattrocchi N, van de Ven AL, et al. Biomimetic functionalization with leukocyte membranes imparts cell like functions to synthetic particles. Nature Nanotechnol 2013;8(1):61–68. [24] Rodriguez PL, Harada T, Christian DA. Minimal “self” peptides that inhibit phagocytic clearance and enhance delivery of nanoparticles. Science 2013;339(6122):971–5. [25] Sriraman SK, Aryasomayajula B, Torchilin VP. Barriers to drug delivery in solid tumors. Tissue Barriers 2014;2. e29528. [26] Barua S, Mitragotri S. Challenges associated with penetration of nanoparticles across cell and tissue barriers: a review of current status and future prospects. Nano Today 2014;9(2):223–43. [27] Kuhn SJ, Finch SK, Hallahan DE, Giorgio TD. Proteolytic surface functionalization enhances in vitro magnetic nanoparticle mobility through extracellular matrix. Nano Lett 2006;6:306–12. [28] Madani F, Lindberg S, Langel Ü, et al. Mechanisms of cellular uptake of cell-penetrating peptides. J Biophys 2011:414729. 2011. [29] Bechara C, Sagan S. Cell-penetrating peptides: 20 years later, where do we stand? FEBS Lett 2013;587:1693–702. [30] Rejman J, Oberle V, Zuhorn IS, Hoekstra D. Size-dependent internalization of particles via the pathways of clathrin- and caveolae-mediated endocytosis. Biochem J 2004;377(Pt 1):159–69. [31] McMahon HT, Boucrot E. Molecular mechanism and physiological functions of clathrin-mediated endocytosis. Nat Rev Mol Cell Biol 2011;12(8):517–33. [32] Ehrlich M, Boll W, van Oijen A, et al. Endocytosis by random initiation and stabilization of clathrin-coated pits. Cell 2004;118(5):591–605. [33] Sorkin A, von Zastrow M. Signal transduction and endocytosis: close encounters of many kinds. Nat Rev Mol Cell Biol 2002;3(8):600–14. [34] Nabi IR, Le PU. Caveolae/raft-dependent endocytosis. J Cell Biol 2003;161(4):673–7. [35] Parton RG, Simons K. The multiple faces of caveolae. Nat Rev Mol Cell Biol 2007;8(3):185–94. [36] Stan RV. Structure of caveolae. Biochim Biophys Acta 2005;1746:334–48. [37] Thomsen P, Roepstorff K, Stahlhut M, van Deurs B. Caveolae are highly immobile plasma membrane microdomains, which are not involved in constitutive endocytic trafficking. Mol Biol Cell 2002;13(1):238–50.
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
437
[38] Lundmark R, Doherty GJ, Howes MT, et al. The GTPase-activating protein GRAF1 regulates the CLIC/GEEC endocytic pathway. Curr Biol 2008;18(22–2):1802–8. [39] Kerr MC, Teasdale RD. Defining macropinocytosis. Traffic 2009;10(4):364–71. [40] Swanson JA. Shaping cups into phagosomes and macropinosomes. Nat Rev Mol Cell Biol 2008;9(8):639–49. [41] Di Pisa M, Chassaing G, Swiecicki JM. Translocation mechanism(s) of cell-penetrating peptides: biophysical studies using artificial membrane bilayers. Biochemistry 2015;54:194–207. [42] Magzoub M, Gräslund A. Cell-penetrating peptides: small from inception to application. Q Rev Biophys 2004;37:147–95. [43] Greber UF, Willetts M, Webster P, Helenius A. Stepwise dismantling of adenovirus 2 during entry into cells. Cell 1993;75:477–86. [44] Vorburger SA, Hunt KK. Adenoviral gene therapy. The Oncol 2002;7(1):46–59. [45] Lear JD, DeGrado WF. Membrane binding and conformational properties of peptides representing the NH2 terminus of influenza HA-2. J Biol Chem 1987;262(14):6500–5. [46] Varkouhi AK, Scholte M, Storm G, Haisma HJ. Endosomal escape pathways for delivery of biologicals. J Control Release 2011;151:220–8. [47] Alberts B, Johnson A, Lewis J, et al. Molecular biology of the cell: the compartmentalization of cells. 4th ed. New York: Garland Science; 2002. [48] Lechardeur D, Sohn KJ, Haardt M, et al. Metabolic instability of plasmid DNA in the cytosol: a potential barrier to gene transfer. Gene Ther 1999;6(4):482–97. [49] Ward CM, Read ML, Seymour LW. Systemic circulation of poly(l-lysine)/DNA vectors is influenced by polycation molecular weight and type of DNA: differential circulation in mice and rats and the implications for human gene therapy. Blood 2001;97(8):2221–9. [50] Lukacs G, Haggie P, Seksek O, et al. Size-dependent DNA mobility in cytoplasm and nucleus. J Biol Chem 2000;275(3):1625–9. [51] Dauty E, Verkman AS. Actin cytoskeleton as the principal determinant of size-dependent DNA mobility in cytoplasm. J Biol Chem 2005;280:7823–8. [52] Niederman R, Amrein PC, Hartwig J. Three-dimensional structure of actin filaments and of an actin gel made with actin-binding protein. J Cell Biol 1983;96(5):1400–13. [53] McDonald D, Vodicka MA, Lucero G, et al. Visualization of the intracellular behavior of HIV in living cells. J Cell Biol 2002;159(3):441–52. [54] Petit C, Giron ML, Tobaly-Tapiero J, et al. Targeting of incoming retroviral Gag to the centrosome involves a direct interaction with the dynein light chain 8. J Cell Sci 2003;116:3433–42. [55] Bukrinskaya A, Brichacek B, Mann A, Stevenson M. Establishment of a functional human immunodeficiency virus type 1 (HIV-1) reverse transcription complex involves the cytoskeleton. J Exp Med 1998;188(11):2113–25. [56] Lange A, Mills RE, Lange CJ, et al. Classical nuclear localization signals: definition, function, and interaction with importin a. J Biol Chem 2007;282:5101–5. [57] Lam A, Dean D. Progress and prospects: nuclear import of nonviral vectors. Gene Ther 2010;17(4):439–47. [58] Hamer DH, Smith KD, Boyer SH, Leder P. SV40 recombinants carrying rabbit beta- globin gene coding sequences. Cell 1979;17(3):725–35. [59] Prokop A, Davidson JM. Nanovehicular intracellular delivery systems. J Pharmaceut Sci 2008;97(9):3518–90. [60] Kotterman MA, Chalberg TW, Schaffer DV. Viral vectors for gene therapy: translational and clinical outlook. Annu Rev Biomed Eng 2015;17:63–89.
438
Bone Repair Biomaterials
[61] Yi Y, Hahm SH, Lee KH, et al. Retroviral gene therapy: safety issues and possible solutions. Curr Gene Ther 2005;5(1):25–35. [62] Noël D, Gazit D, Bouquet C, et al. Short-term BMP-2 expression is sufficient for in vivo osteochondral differentiation of mesenchymal stem cells. Stem Cell 2004;22:74–85. [63] McMahon JM, Signori E, Wells KE, et al. Optimisation of electrotransfer of plasmid into skeletal muscle by pretreatment with hyaluronidase - increased expression with reduced muscle damage. Gene Ther 2001;8:1264–70. [64] Ramamoorth M, Narvekar A. Non viral vectors in gene therapy- an overview. J Clin Diagn Res 2015;9(1):GE01–6. [65] Schatzlein AG. Non-viral vectors in cancer gene therapy: principles and progress. Anticancer Drugs 2001;12:275–304. [66] Nayerossadat N, Maedeh T, Ali PA. Viral and nonviral delivery systems for gene delivery. Adv Biomed Res 2012;1:27. [67] Klein T, Wolf E, Sanford J. High-velocity microprojectiles for delivering nucleic acids into living cells. Nature 1987;327:70–3. [68] Williams RS, Johnston SA, Riedy M, et al. Introduction of foreign genes into tissues of living mice by DNA-coated microprojectiles. Proc Natl Acad Sci USA 1991;88:2726–30. [69] Yang NS, Burkholder J, Roberts B, et al. In vivo and in vitro gene transfer to mammalian somatic cells by particle bombardment. Proc Natl Acad Sci USA 1990;87(24):9568–72. [70] Davidson JM, Krieg T, Eming SA. Particle-mediated gene therapy of wounds. Wound Repair Regen 2000;8:452–9. [71] Neumann E, Schaefer-Ridder M, Wang Y, Hofschneider PH. Gene transfer into mouse lyoma cells by electroporation in high electric fields. EMBO J 1982;1(7):841–5. [72] Kawai M, Bessho K, Maruyama H, et al. Simultaneous gene transfer of bone morphogenetic protein (BMP) -2 and BMP-7 by in vivo electroporation induces rapid bone formation and BMP-4 expression. BMC Muscoskel Disord 2006;7:62. [73] Aihara H, Miyazaki J. Gene transfer into muscle by electroporation in vivo. Nat Biotechnol 1998;16:867–70. [74] Vanbever R, Preat VV. In vivo efficacy and safety of skin electroporation. Adv Drug Deliv Rev 1999;35:77–88. [75] Suzuki T, Shin BC, Fujikura K, et al. Direct gene transfer into rat liver cells by in vivo electroporation. FEBS Lett 1998;425:436–40. [76] Jin L, Zeng X, Liu M, et al. Current progress in gene delivery technology based on chemical methods and nano-carriers. Theranostics 2014;4(3):240–55. [77] Sokolova V, Epple M. Inorganic nanoparticles as carriers of nucleic acids into cells. Angew Chem Int Ed Engl 2008;47(8):1382–95. [78] Davis PB, Cooper MJ. Vectors for airway gene delivery. AAPS J 2007;9:11–7. [79] Qureshi AT, Monroe WT, Dasa V, et al. miR-148b-nanoparticle conjugates for light mediated osteogenesis of human adipose stromal/stem cells. Biomaterials 2013;34:7799–810. [80] Mok H, Zhang M. Superparamagnetic iron oxide nanoparticle-based delivery systems for biotherapeutics. Expet Opin Drug Deliv 2013;10(1):73–87. [81] Mochizuki S, Kanegae N, Nishina K, et al. The role of the helper lipid dioleoylphosphatidylethanolamine (DOPE) for DNA transfection cooperating with a cationic lipid bearing ethylenediamine. Biochimica et Biophysica Acta 2013;1828:412–8. [82] Hui SW, Langner M, Zhao YL, et al. The role of helper lipids in cationic liposome-mediated gene transfer. Biophys J 1996;71(2):590–9. [83] Felgner PL, Gadek TR, Holm M, et al. Lipofection: a highly efficient, lipid-mediated DNA-transfection procedure. Proc Natl Acad Sci USA 1987;84:7413–7.
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
439
[84] Pezzoli D, Kajaste-Rudnitski A, Chiesa R, Candiani G. Lipid-based nanoparticles as nonviral gene delivery vectors. Methods Mol Biol 2013;1025:269–79. [85] Huang X, Schwind S, Yu B, et al. Targeted delivery of microRNA-29b by transferrin conjugated anionic lipopolyplex nanoparticles: a novel therapeutic strategy in acute myeloid leukemia. Clin Cancer Res 2013;19(9):2355–67. [86] Craig VJ, Tzankov A, Flori M, et al. Systemic microRNA-34a delivery induces apoptosis and abrogates growth of diffuse large B-cell lymphoma in vivo. Leukemia 2012;26:2421–4. [87] Bikram M, Lee M, Chang CW, et al. Long-circulating DNA-complexed biodegradable multiblock copolymers for gene delivery: degradation profiles and evidence of dysopsonization. J Control Release 2005;103:221–33. [88] Kim HK, Davaa E, Myung CS, Park JS. Enhanced siRNA delivery using cationic liposomes with new polyarginine-conjugated PEG-lipid. Int J Pharm 2010;392(1–2):141–7. [89] Xu L, Wempe MF, Anchordoquy TJ. The effect of cholesterol domains on PEGylated liposomal gene delivery in vitro. Ther Deliv 2011;2(4):451–60. [90] Lv H, Zhang S, Wang B, et al. Toxicity of cationic lipids and cationic polymers in gene delivery. J Control Release 2006;1(114):100–9. [91] Li WJ, Szoka FC. Lipid-based nanoparticles for nucleic acid delivery. Pharm Res 2007;3(24):438–49. [92] Yamano S, Dai J, Moursi AM. Comparison of transfection efficiency of nonviral gene transfer reagents. Mol Biotechnol 2010;46:287–300. [93] Vectors used in gene therapy clinical trials. J Gene Med 2016. http://wiley.co.uk/genmed/ clinical. [94] Wu GY, Wu CH. Receptor-mediated gene delivery and expression in vivo. J Biol Chem 1988;263:14621–4. [95] Boussif O, Lezoualc’h F, Zanta MA, et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc Natl Acad Sci USA 1995;92:7297–301. [96] Ulasov AV, Khramtsov YV, Trusov GA, et al. Properties of PEI-based polyplex nanoparticles that correlate with their transfection efficacy. Mol Ther 2011;19(1):103–12. [97] Sonawane ND, Szoka FC, Verkman AS. Chloride accumulation and swelling in endosomes enhances DNA transfer by polyamine-DNA polyplexes. J Biol Chem 2003;278(45):44826–31. [98] McKenzie DL, Collard WT, Rice KG. Comparative gene transfer efficiency of low molecular weight polylysine DNA-condensing peptides. J Pept Res 1999;54(4):311–8. [99] Yamashiro DJ, Fluss SR, Maxfield FR. Acidification of endocytic vesicles by an ATPdependent proton pump. J Cell Biol 1983;97:929–34. [100] Coll JL, Chollet P, Brambilla E, et al. In vivo delivery to tumors of DNA complexed with linear polyethylenimine. Hum Gene Ther 1999;10:1659–66. [101] Merkel OM, Urbanics R, Bedocs P, et al. In vitro and in vivo complement activation and related anaphylactic effects associated with polyethylenimine and polyethylenimine-graft-poly(ethylene glycol) block copolymers. Biomaterials 2011;21(32): 4936–42. [102] Thomas M, Klibanov AM. Enhancing polyethylenimine’s delivery of plasmid DNA into mammalian cells. Proc Natl Acad Sci USA 2002;99(23):14640–5. [103] Tian H, Deng C, Lin H, et al. Biodegradable cationic PEG-PEI-PBLG hyperbranched block copolymer: synthesis and micelle characterization. Biomaterials 2005;26:4209–17. [104] Liu ZH, Zhang ZY, Zhou CR, Jiao YP. Hydrophobic modifications of cationic polymers for gene delivery. Prog Polym Sci 2010;9(35):1144–62.
440
Bone Repair Biomaterials
[105] Ogris M, Brunner S, Schüller S, et al. PEGylated DNA/transferrin-PEI complexes: reduced interaction with blood components, extended circulation in blood and potential for systemic gene delivery. Gene Thery 1999;6:595–605. [106] Langer R. Drug delivery and targeting. Nature 1998;392(6679 Suppl.):5–10. [107] Shive MS, Anderson JM. Biodegradation and biocompatibility of PLA and PLGA microspheres. Adv Drug Deliv Rev 1997;28(1):5–24. [108] Makadia HK, Siegel SJ. Poly lactic-co-glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers 2011;3(3):1377–97. [109] Panyam J, Labhasetwar V. Biodegradable nanoparticles for drug and gene delivery to cells and tissue. Adv Drug Deliv Rev 2003;55(3):329–47. [110] Shah B, Kona S, Gilbertson TA, Nguyen KT. Effects of poly-(lactide-co-glycolide) nanoparticles on electrophysiological properties of enteroendocrine cells. J Nanosci Nanotechnol 2011;11(4):3533–42. [111] Babar IA, Cheng CJ, Booth CJ, et al. Nanoparticle-based therapy in an in vivo microRNA-155 (miR-155)-dependent mouse model of lymphoma. Proc Natl Acad Sci USA 2012;109(26):E1695–704. [112] Blum JS, Saltzman WM. High loading efficiency and tunable release of plasmid DNA encapsulated in submicron particles fabricated from PLGA conjugated with poly-L-lysine. J Control Release 2008;129(1):66–72. [113] Panyam J, Zhou WZ, Prabha S, et al. Rapid endo-lysosomal escape of poly(-lactide-co-glycolide) nanoparticles: implications for drug and gene delivery. FASEB J 2002;16(10):1217–26. [114] Lü JM, Wang X, Marin-Muller C, et al. Current advances in research and clinical applications of PLGA-based nanotechnology. Expert Rev Mol Diagn 2009;9(4):325–41. [115] Zeng P, Xu Y, Zeng CH, et al. Chitosan-modified poly(d,l-lactide-co-glycolide) nanospheres for plasmid DNA delivery and HBV gene-silencing. Int J Pharm 2011;415(1–2):259–66. [116] Ravi Kumar MNV, Bakowsky U, Lehr CM. Preparation and characterization of cationic PLGA nanospheres as DNA carriers. Biomaterials 2004;25:1771–7. [117] Chandy T, Sharma CP. Chitosan - as a biomaterial. Biomater Artif Cells Artif Organs 1990;1(18):1–24. [118] Prabaharan M, Mano JF. Chitosan-based particles as controlled drug delivery systems. Drug Deliv 2005;1(12):41–57. [119] Mansouri S, Lavigne P, Corsi K, et al. Chitosan-DNA nanoparticles as non-viral vectors in gene therapy: strategies to improve transfection efficacy. Eur J Pharm Biopharm 2004;1(57):1–8. [120] Lavertu M, Methot S, Tran-Khanh N, Buschmann MD. High efficiency gene transfer using chitosan/DNA nanoparticles with specific combinations of molecular weight and degree of deacetylation. Biomaterials 2006;27(27):4815–24. [121] Liang W, Lam JKW. Endosomal escape pathways for non-viral nucleic acid delivery systems. In: Brian Ceresa D, editor. Molecular regulation of endocytosis. InTech; 2012. [122] Park SC, Nam JP, Kim YM, et al. Branched polyethylenimine-grafted-carboxymethyl chitosan copolymer enhances the delivery of pDNA or siRNA in vitro and in vivo. Int J Nanomed 2013;8:3663–77. [123] He XK, Yuan ZX, Wu XJ, et al. Low molecular weight hydroxyethyl chitosan-prednisolone conjugate for renal targeting therapy: synthesis, characterization and in vivo studies. Theranostics 2012;2(11):1054–63. [124] Shcharbin D, Pedziwiatr E, Bryszewska M. How to study dendriplexes I: Characterization. J Control Release 2009;135(3):186–97.
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
441
[125] Majoros IJ, Thomas TP, Mehta CB, Baker Jr JR. Poly(amidoamine) dendrimer-based multi-functional engineered nanodevice for cancer therapy. Journal of Medicinal Chemistry 2005;48:5892–9. [126] Morgan JR, Cloninger MJ. Heterogeneously functionalized dendrimers. Curr Opin Drug Discov Dev 2002;5:966–73. [127] Daneshvar N, Abdullah R, Shamsabadi FT, et al. PAMAM dendrimer roles in gene delivery methods and stem cell research. Cell Biol Int 2013;5(37):415–9. [128] Kukowska-Latallo JF, Bielinska AU, Johnson J, et al. Efficient transfer of genetic material into mammalian cells using starburst polyamidoamine dendrimers. Proc Natl Acad Sci USA 1996;10(93):4897–902. [129] Oliveira JM, Sousa RA, Kotobuki N, et al. The osteogenic differentiation of rat bone marrow stromal cells cultured with dexamethasone loaded carboxymethylchitosan/poly(amidoamine) dendrimer nanoparticles. Biomaterials 2009;30(5):804–13. [130] Santos JL, Oramas E, Pego AP, et al. Osteogenic differentiation of mesenchymal stem cells using PAMAM dendrimers as gene delivery vectors. J Control Release 2009;134(2):141–8. [131] Maiti PK, Cagin T, Wang GF, Goddard WA. Structure of PAMAM dendrimers: generations 1 through 11. Macromolecules 2004;37:6236–54. [132] Santander-Ortega MJ, Uchegbu IF, Schätzlein AG. Dendrimer-based gene delivery systems: administration routes and in vivo evaluation. In: Dendrimer-based drug delivery systems: from theory to practice. Hoboken: John Wiley & Sons; 2012. [133] Kim JB, Choi JS, Nam K, et al. Enhanced transfection of primary cortical cultures using arginine-grafted PAMAM dendrimer, PAMAM-arg. J Control Release 2006;1(114):110–7. [134] Santos JL, Oliveira H, Pandita D, et al. Functionalization of poly(amidoamine) dendrimers with hydrophobic chains for improved gene delivery in mesenchymal stem cells. J Control Release 2010;1(144):55–64. [135] Martin ME, Rice KG. Peptide-guided gene delivery. AAPS J 2007;9(1):E18–29. [136] Saccardo P, Villaverde A, González-Montalbán N. Peptide-mediated DNA condensation for non-viral gene therapy. Biotechnol Adv 2009;27(4):432–8. [137] Levine RM, Scott CM, Kokkoli E. Peptide functionalized nanoparticles for nonviral gene delivery. Soft Matter 2013;9(4):985–1004. [138] Frankel AD, Pabo CO. Cellular uptake of the tat protein from human immunodeficiency virus. Cell 1988;55(6):1189–93. [139] Green M, Loewenstein PM. Autonomous functional domains of chemically synthesized human immunodeficiency virus tat trans-activator protein. Cell 1988;55:1179–88. [140] Rennert R, Wespe C, Beck-Sickinger AG, Neundorf I. Developing novel hCT derived cell-penetrating peptides with improved metabolic stability. Biochim Biophys Acta 2006;1758:347–54. [141] Schmidt N, Mishra A, Lai GH, Wong GC. Arginine-rich cell-penetrating peptides. FEBS Lett 2010;584:1806–13. [142] Derossi D, Joliot AH, Chassaing G, Prochiantz A. The 3rd helix of the Antennapedia homeodomain translocates through biological-membranes. J Biol Chem 1994;269: 10444–50. [143] Vivés E, Brodin P, Lebleu B. A truncated HIV-1 Tat protein basic domain rapidly translocates through the plasma membrane and accumulates in the cell nucleus. J Biol Chem 1997;272:16010–7. [144] Elmquist A, Lindgren M, Bartfai T, Langel Ü. VE-cadherin-derived cell-penetrating peptide, pVEC, with carrier functions. Exp Cell Res 2001;269:237–44.
442
Bone Repair Biomaterials
[145] Morris MC, Vidal P, Chaloin L, et al. A new peptide vector for efficient delivery of oligonucleotides into mammalian cells. Nucleic Acids Res 1997;25:2730–6. [146] Pooga M, Hällbrink M, Zorko M, Langel Ü. Cell penetration by transportan. FASEB J 1998;12:67–77. [147] Morris MC, Depollier J, Mery J, et al. A peptide Carrier for the delivery of biologically active proteins into mammalian cells. Nat Biotechnol 2001;19:1173–6. [148] Oehlke J, Scheller A, Wiesner B, et al. Cellular uptake of an α-helical amphipathic model peptide with the potential to deliver polar compounds into cell interior non-endocytotically. Biochim Biophys Acta 1998;1414:127–39. [149] Rothbard JB, Garlington S, Lin Q, et al. Conjugation of arginine oligomers to cyclosporin A facilitates topical delivery and inhibition of inflammation. Nat Med 2000;6:1253–7. [150] Delaroche D, Aussedat B, Aubry S, et al. Tracking a new cell-penetrating (W/R) nonapeptide, through an enzyme-stable mass spectrometry reporter tag. Anal Chem 2007;79:1932–8. [151] Scheller A, Oehlke J, Wiesneret B, et al. Structural requirements for cellular uptake of alpha-helical amphipathic peptides. J Pept Sci 1999;5(4):185–94. [152] Gupta B, Levchenko TS, Torchilin VP. Intracellular delivery of large molecules and small particles by cell-penetrating proteins and peptides. Adv Drug Deliv Rev 2005;57(4):637–51. [153] Suh JS, Lee JY, Choi YS, et al. Peptide-mediated intracellular delivery of miRNA-29b for osteogenic stem cell differentiation. Biomaterials 2013;34:4347–59. [154] Crombez L, Morris MC, Dufort S, et al. Targeting cyclin B1 through peptide-based delivery of siRNA prevents tumour growth. Nucleic Acids Research 2009;37(14):4559–69. [155] Kumar P, Wu H, McBride JL, et al. Transvascular delivery of small interfering RNA to the central nervous system. Nature 2007;448:39–43. [156] Suzuki T, Futaki S, Niwa M, et al. Possible existence of common internalization mechanisms among arginine-rich peptides. J Biol Chem 2002;277(4):2437–43. [157] Vives E, Richard JP, Rispal C, Lebleu B. TAT peptide internalization: seeking the mechanism of entry. Curr Protein Pept Sci 2003;4:125–32. [158] Richard JP, Melikov K, Vives E, et al. Cell-penetrating peptides. A re-evaluation of the mechanism of cellular uptake. J Biol Chem 2003;278:585–90. [159] Wadia JS, Stan RV, Dowdy SF. Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nature Med 2004;10:310–5. [160] Nekhotiaeva N, Elmquist A, Rajarao GK, et al. Cell entry and antimicrobial properties of eukaryotic cell-penetrating peptides. FASEB J 2004;18:394–6. [161] Zemel A, Fattal DR, Ben-Shaul A. Energetics and self-assembly of amphipathic peptide pores in lipid membranes. Biophys J 2003;84(4):2242–55. [162] Lo SL, Wang S. An endosomolytic Tat peptide produced by incorporation of histidine and cysteine residues as a nonviral vector for DNA transfection. Biomaterials 2008;29:2408–14. [163] Wagner E, Plank C, Zatloukal K, et al. Influenza virus hemagglutinin HA-2 N-terminal fusogenic peptides augment gene transfer by transferrin-polylysine-DNA complexes: toward a synthetic virus-like gene-transfer vehicle. Proc Natl Acad Sci USA 1992;89:7934–8. [164] Li W, Nicol F, Szoka FC. GALA: a designed synthetic pH-responsive amphipathic peptide with applications in drug and gene delivery. Adv Drug Deliv Rev 2004;56:967–85. [165] Wyman TB, Nicol F, Zelphati O, et al. Design, synthesis, and characterization of a cationic peptide that binds to nucleic acids and permeabilizes bilayers. Biochemistry 1997;36:3008–17.
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
443
[166] Midoux P, Kichler A, Boutin V, et al. Membrane permeabilization and efficient gene transfer by a peptide containing several histidines. Bioconjug Chem. 1998;9:260–7. [167] Simoes S, Slepushkin V, Gaspar R, et al. Gene delivery by negatively charged ternary complexes of DNA, cationic liposomes and transferrin or fusogenic peptides. Gene Ther 1998;5(7):955–64. [168] McCarthy HO, McCaffery J, McCrudden CM, et al. Development and characterization of self-assembling nanoparticles using a bio-inspired amphipathic peptide for gene delivery. J Control Release 2014;189:141–9. [169] Bennett R, Yakkundi A, McKeen HD, et al. RALA-mediated delivery of FKBPL nucleic acid therapeutics. Nanomedicine 2015;10:2989–3001. [170] McCaffrey J, McCrudden CM, Ali AA, et al. Transcending epithelial and intracellular biological barriers; a prototype DNA delivery device. J Control Release 2016;226:238–47. [171] Massey AS, Pentlavalli S, Cunningham R, et al. Potentiating the anticancer properties of bisphosphonates by nanocomplexation with the cationic amphipathic peptide. RALA. Mol Pharm 2016. [Epub ahead of print]. [172] Geisow MJ, Evans WH. pH in the endosome. Measurements during pinocytosis and receptor-mediated endocytosis. Exp Cell Res 1984;150:36–46. [173] MacFarlane L-A, Murphy PR. MicroRNA: biogenesis, function and role in cancer. Curr Genom 2010;11(7):537–61. [174] Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993;75(5):843–54. [175] Griffiths-Jones S. miRBase: the microRNA sequence database. Methods Mol Biol 2006;342:129–38. [176] Lu J, Clark AG. Impact of microRNA regulation on variation in human gene expression. Genome Res 2012;22(7):1243–54. [177] Davis BN, Hata A. Regulation of MicroRNA biogenesis: a miRiad of mechanisms. Cell Commun Signal: CCS 2009;7:18. [178] Schober A, Nazari-Jahantigh M, Wei Y, et al. MicroRNA-126-5p promotes endothelial proliferation and limits atherosclerosis by suppressing Dlk1. Nat Med 2014;20(4): 368–76. [179] Ivey KN, Srivastava D. MicroRNAs as regulators of differentiation and cell fate decisions. Cell Stem Cell 2010;7(1):36–41. [180] Jovanovic M, Hengartner MO. miRNAs and apoptosis: RNAs to die for. Oncogene 2006;25:6176–87. [181] Pandey AK, Agarwal P, Kaur K, Datta M. MicroRNAs in diabetes: tiny players in big disease. Cell Physiol Biochem 2009;23:221–32. [182] Nelson PT, Wang W-X, Rajeev BW. MicroRNAs (miRNAs) in neurodegenerative diseases. Brain Pathol 2008;18(1):130–8. [183] Lee YS, Dutta A. MicroRNAs in cancer. Annu Rev Pathol 2009;4:199–227. [184] Sessa R, Hata A. Role of microRNAs in lung development and pulmonary diseases. Pulm Circ 2013;3(2):315–28. [185] Sen CK, Ghatak S. miRNA control of tissue repair and regeneration. Am J Pathol 2015;185(10):2629–40. [186] Titorencu I, Pruna V, Jinga VV, et al. Osteoblast ontogeny and implications for bone pathology: an overview. Cell Tissue Res 2014;355(1):23–33. [187] Li Y, Fan L, Liu S, et al. The promotion of bone regeneration through positive regulation of angiogenic-osteogenic coupling using microRNA-26a. Biomaterials 2013;34(21):5048–58.
444
Bone Repair Biomaterials
[188] Li Z, Hassan MQ, Jafferji M, et al. Biological functions of miR-29b contribute to positive regulation of osteoblast differentiation. J Biol Chem 2009;284(23):15676–84. [189] Van Wijnen AJ, van de Peppel J, van Leeuwen JP, et al. MicroRNA functions in osteogenesis and dysfunctions in osteoporosis. Curr Osteoporos Rep 2013;11(2):72–82. [190] Lee Y, Kim M, Han J, et al. MicroRNA genes are transcribed by RNA polymerase II. EMBO J 2004;23(20):4051–60. [191] Han J, Lee Y, Yeom K-H, et al. The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev 2004;18(24):3016–27. [192] Landthaler M, Yalcin A, Tuschl T. The human DiGeorge syndrome critical region gene 8 and its D. melanogaster homolog are required for miRNA biogenesis. Curr Biol 2004;14:2162–7. [193] Kim VN. MicroRNA precursors in motion: exportin-5 mediates their nuclear export. Trends Cell Biol 2004;14:156–9. [194] Zeng Y, Cullen BR. Structural requirements for pre-microRNA binding and nuclear export by Exportin 5. Nucleic Acids Res 2004;32(16):4776–85. [195] Gregory RI, Chendrimada TP, Cooch N, Shiekhattar R. Human RISC couples microRNA biogenesis and posttranscriptional gene silencing. Cell 2005;123:631–40. [196] Huang C, Wang X, Liu X, et al. RNAi pathway participates in chromosome segregation in mammalian cells. Cell Discov 2015;1:15029. [197] Liu X, Fortin K, Mourelatos Z. MicroRNAs: biogenesis and molecular functions. Brain Pathol 2008;18:113–21. [198] Van den Berg A, Mols J, Han J. RISC-target interaction: cleavage and translational suppression. Biochimica et biophysica acta 2008;1779(11):668–77. [199] Luzi E, Marini F, Sala SC, et al. Osteogenic differentiation of human adipose tissue-derived stem cells is modulated by the miR-26a targeting of the SMAD1 transcription factor. J Bone Miner Res 2008;23:287–95. [200] Wang Z, Xie Q, Yu Z, et al. A regulatory loop containing miR-26a, GSK3β and C/EBPα regulates the osteogenesis of human adipose-derived mesenchymal stem cells. Sci Rep 2015;5:15280. [201] van Rooij E, Sutherland LB, Thatcher JE, et al. Dysregulation of microRNAs after myocardial infarction reveals a role of miR-29 in cardiac fibrosis. Proc Natl Acad Sci USA 2008;105(35):13027–32. [202] Ko J-Y, Chuang P-C, Ke H-J, et al. MicroRNA-29a mitigates glucocorticoid induction of bone loss and fatty marrow by rescuing RUNX2 acetylation. Bone 2015;81:80–8. [203] Trompeter HI, Dreesen J, Hermann E, et al. MicroRNAs miR-26a, miR-26b, and miR29b accelerate osteogenic differentiation of unrestricted somatic stem cells from human cord blood. BMC Genom 2013;14:111. [204] Eskildsen T, Taipaleenmäki H, Stenvang J, et al. MicroRNA-138 regulates osteogenic differentiation of human stromal (mesenchymal) stem cells in vivo. Proc Natl Acad Sci USA 2011;108:6139–44. [205] Yan J, Zhang C, Zhao Y, et al. Non-viral oligonucleotide antimiR-138 delivery to mesenchymal stem cell sheets and the effect on osteogenesis. Biomaterials 2014;35:7734–49. [206] Itoh T, Nozawa Y, Akao Y. MicroRNA-141 and -200a are involved in bone morphogenetic protein-2-induced mouse pre-osteoblast differentiation by targeting distal-less homeobox 5. J Biol Chem 2009;284:19272–9. [207] Qiu W, Kassem M. MiR-141-3p inhibits human stromal (mesenchymal) stem cell proliferation and differentiation. Biochim Biophys Acta Mol Cell Res 2014;1843(9):2114–21. [208] Li Z, Hassan MQ, Volinia S, et al. A microRNA signature for a BMP2-induced osteoblast lineage commitment program. Proc Natl Acad Sci USA 2008;105:13906–11.
Emerging areas of bone repair materials: nucleic acid therapy and drug delivery
445
[209] Kim YJ, Bae SW, Yu SS, et al. miR-196a regulates proliferation and osteogenic differentiation in mesenchymal stem cells derived from human adipose tissue. J Bone Miner Res 2009;24:816–25. [210] Candini O, Spano C, Murgia A, et al. Mesenchymal progenitors aging highlights a miR196 switch targeting HOXB7 as master regulator of proliferation and osteogenesis. Stem Cell 2015;33:939–50. [211] Liu XD, Cai F, Liu L, et al. MicroRNA-210 is involved in the regulation of postmenopausal osteoporosis through promotion of VEGF expression and osteoblast differentiation. Biol Chem 2015;396(4):339–47. [212] Mizuno Y, Tokuzawa Y, Ninomiya Y, et al. miR-210 promotes osteoblastic differentiation through inhibition of AcvR1b. FEBS Lett 2009;583:2263–8. [213] Zhang J, Tu Q, Bonewald LF, et al. Effects of miR-335-5p in modulating osteogenic differentiation by specifically downregulating Wnt antagonist DKK1. J Bone Miner Res 2011;26:1953–63. [214] Tome M, López-Romero P, Albo C, et al. MiR-335 orchestrates cell proliferation, migration and differentiation in human mesenchymal stem cells. Cell Death Differ 2011;18:985–95. [215] Zhang Y, Xie RL, Croce CM, et al. A program of microRNAs controls osteogenic lineage progression by targeting transcription factor RUNX2. Proc Natl Acad Sci USA 2011;108(24):9863–8. [216] Liu H, Sun Q, Wan C, et al. MicroRNA-338-3p regulates osteogenic differentiation of mouse bone marrow stromal stem cells by targeting RUNX2 and Fgfr2. J Cell Physiol 2014;229(10):1494–502. [217] Guo DW, Han YX, Cong L, et al. Resveratrol prevents osteoporosis in ovariectomized rats by regulating microRNA-338-3p. Mol Med Rep 2015;12:2098–106. [218] Hupkes M, Sotoca AM, Hendriks JM, et al. MicroRNA miR-378 promotes BMP2induced osteogenic differentiation of mesenchymal progenitor cells. BMC Mol Biol 2014;15(1). [219] You L, Gu W, Chen L, et al. MiR-378 overexpression attenuates high glucose-suppressed osteogenic differentiation through targeting CASP3 and activating PI3k/Akt signaling pathway. Int J Clin Exp Pathol 2014;7(10):7249–61. [220] Kahai S, Lee S-C, Lee DY, et al. MicroRNA miR-378 regulates nephronectin expression modulating osteoblast differentiation by targeting GalNT-7. PLoS One 2009;4(10):e7535. [221] Zhang JF, Fu WM, He ML, et al. MiR-637 maintains the balance between adipocytes and osteoblasts by directly targeting Osterix. Mol Biol Cell 2011;22(21):3955–61. [222] Li H, Xie H, Liu W, et al. A novel microRNA targeting HDAC5 regulates osteoblast differentiation in mice and contributes to primary osteoporosis in humans. J Clin Invest 2009;119:3666–77. [223] Hu R, Liu W, Li H, et al. A RUNX2/miR-3960/miR-2861 regulatory feedback loop during mouse osteoblast differentiation. J Biol Chem 2011;286(14):12328–39. [224] Matsubara T, Kida K, Yamaguchi A, et al. BMP2 regulates osterix through Msx2 and Runx2 during osteoblast differentiation. J Biol Chem 2008;283(43):29119–25. [225] Jang WG, Kim EJ, Kim DK, et al. BMP2 protein regulates oteocalcin expression via Runx2-mediated Atf6 gene transcription. J Biol Chem 2012;287(2):905–15. [226] Lian JB, Javed A, Zaidi SK, et al. Regulatory controls for osteoblast growth and differentiation: role of Runx/Cbfa/AML factors. Crit Rev Eukaryot Gene Exp 2004;14:1–41.
446
Bone Repair Biomaterials
[227] Maruyama Z, Yoshida CA, Furuichi T, et al. Runx2 determines bone maturity and turnover rate in postnatal bone development and is involved in bone loss in estrogen deficiency. Dev Dyn 2007;236:1876–90. [228] Hassan MQ, Gordon JAR, Belot MM, et al. A network connecting Runx2, SATB2, and the miR-23a 27a 24–2 cluster regulates the osteoblast differentiation program. Proc Natl Acad Sci USA 2010;107:19879–84. [229] Nishio Y, Dong Y, Paris M, et al. Runx2-mediated regulation of the zinc finger Osterix/ Sp7 gene. Gene 2006;372:62–70. [230] Nakashima K, Zhou X, Kunkel G, et al. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell 2002;108(1):17–29. [231] Chen S, Feng J, Zhang H, et al. Key role for the transcriptional factor, osterix, in spine development. Spine J 2014;14(4):683–94. [232] Yang L, Cheng P, Chen C, et al. miR-93/Sp7 function loop mediates osteoblast mineralization. J Bone Miner Res 2012;27(7):1598–606. [233] Goettsch C, Rauner M, Pacyna N, et al. miR-125b regulates calcification of vascular smooth muscle cells. Am J Pathol 2011;179:1594–600. [234] Fang S, Deng Y, Gu P, Fan X. MicroRNAs regulate bone development and regeneration. Int J Mol Sci 2015;16(4):8227–53. [235] Gámez B, Rodríguez-Carballo E, Bartrons R, et al. MicroRNA-322 (miR-322) and its target protein Tob2 modulate Osterix (Osx) mRNA stability. J Biol Chem 2013;288(20):14264–75. [236] Zhao X, Xu D, Li Y, et al. MicroRNAs regulate bone metabolism. J Bone Miner Metab 2014;32:221–31. [237] Murata K, Ito H, Yoshitomi H, et al. Inhibition of miR-92a enhances fracture healing via promoting angiogenesis in a model of stabilized fracture in young mice. J Bone Miner Res 2014;29(2):316–26. [238] Deng Y, Bi X, Zhou H, et al. Repair of critical-sized bone defects with anti-miR-31- expressing bone marrow stromal stem cells and poly(glycerol sebacate) scaffolds. Eur Cell Mater 2014;27:13–24. [239] Deng Y, Zhou H, Gu P, Fan X. Repair of canine medial orbital bone defects with miR-31-modified bone marrow mesenchymal stem cells. Invest Ophthalmol Vis Sci 2014;55(9):6016–23.
Further reading [1] Shah N, Steptoe RJ, Parekh HS. Low-generation asymmetric dendrimers exhibit minimal toxicity and effectively complex DNA. J Pept Sci. 6(17):470–478. [2] Zorko M, Langel U. Cell-penetrating peptides: mechanism and kinetics of cargo delivery. Adv Drug Deliv Rev 2005;57:529–45. [3] Maurer B, Stanczyk J, Jüngel A, et al. MicroRNA-29, a key regulator of collagen expression in systemic sclerosis. Arthritis Rheum 2010;62:1733–43.