257
Views
25
CrossRef citations to date
0
Altmetric
Review

Growth Factor Gene-Modified Mesenchymal Stem Cells in Tissue Regeneration

, &
Pages 1241-1256 | Published online: 26 Mar 2020

Abstract

There have been marked changes in the field of stem cell therapeutics in recent years, with many clinical trials having been conducted to date in an effort to treat myriad diseases. Mesenchymal stem cells (MSCs) are the cell type most frequently utilized in stem cell therapeutic and tissue regenerative strategies, and have been used with excellent safety to date. Unfortunately, these MSCs have limited ability to engraft and survive, reducing their clinical utility. MSCs are able to secrete growth factors that can support the regeneration of tissues, and engineering MSCs to express such growth factors can improve their survival, proliferation, differentiation, and tissue reconstructing abilities. As such, it is likely that such genetically modified MSCs may represent the next stage of regenerative therapy. Indeed, increasing volumes of preclinical research suggests that such modified MSCs expressing growth factors can effectively treat many forms of tissue damage. In the present review, we survey recent approaches to producing and utilizing growth factor gene-modified MSCs in the context of tissue repair and discuss its prospects for clinical application.

Background

In settings where the human body is unable to partially or fully heal a given tissue injury, the use of stem-cell based regenerative therapies offers great promise as a means of improving patient outcomes.Citation1 Indeed, such therapies can support heart or kidney transplants, bone reconstruction, or the repair of skin, cartilage, and neurons. In patients suffering from pathological conditions, such therapies can also potentially restore compromised tissue function.Citation2Citation4 Mesenchymal stem cells (MSCs) are a form of multipotent stem cell capable of differentiating into a subset of distinct cell types such as myocytes, adipocytes, chondrocytes, and osteoblasts. As they are capable of differentiating into several cell types, homing to target tissues, and secreting growth factors and immunomodulatory compounds, MSCs represent an ideal cell type to use for treating a range of disease types. Importantly, these cells can also be easily obtained and amplified in vitro without engendering substantial ethical concerns, allowing them to be safely and readily used in patients.

Most organs in human adults are limited in their ability to undergo tissue regeneration, instead undergoing scarring that can disrupt organ function. As such, the utilization of MSCs to facilitate true tissue reconstruction rather than scarring represents an ideal means of maintaining normal tissue function in the context of injury. Many studies to date have explored the ability of MSCs to support bone development, restoration of ventricular functional, and improved renal tubular function in vivo and in clinical settings.Citation4Citation6 Unfortunately, however, these cells are limited in their therapeutic efficacy, particularly in contexts where injuries or the associated ischemic damage are severe and irreversible. Indeed, preclinical animal models suggest that MSCs have a poor ability to engraft, and they are also hampered by limited homing and survival in vivo owing to factors including inflammation, ischemia, and anoikis.Citation7 One strategy proposed to overcome such limitations centers on the use of MSCs engineered to express specific genes.

Growth factors (GFs) are well known to be key mediators that can support MSC survival and proliferation, in addition to being key drivers of tissue regenerative processes. Many recent studies have utilized MSCs in order to deliver specific GFs to a target site of tissue regeneration either via utilizing cells naturally secreting these factors, or by engineering these cells to overexpress GFs of interest. Indeed, many recent studies have explored the therapeutic potential of MSCs engineered to express particular GFs in a therapeutic context. In the present review, we offer an overview of recent studies exploring the application of GF gene-modified MSCs in the field of tissue repair and reconstruction.

The Relationship Between MSC Biology and GF Secretion

MSCs are a readily isolated cell type that expand rapidly in culture without losing the ability to undergo self-renewal, permitting their use for reconstructing damaged tissues and organs via extensive amplification.Citation8 In addition to their multipotent ability to differentiate into a range of cell types, MSCs can orchestrate and enhance proximal or distal cell functionality via paracrine signaling and endocrine mechanisms. Studies have shown MSCs to be capable of promoting tissue regeneration via secreting exosomes and GFs including hepatocyte growth factor (HGF), fibroblast growth factor (FGF), and vascular endothelial growth factor (VEGF).Citation9 Additionally, these cells express high levels of factors known to regulate hematopoietic cell function such as CXCL12, vascular cell adhesion molecule 1, interleukin-7, angiopoietin-1 (Ang-1), and osteopontin.Citation10 Consistent with these findings, in vivo studies also support the fact that the paracrine secretion of GFs by MSCs is a key mechanism whereby they support target tissue healing, as while these cells can migrate to sites of injury, the cells derived therefrom contribute only to a limited degree to therapeutic efficacy. Many recent studies have suggested that the secretion of GFs and other bioactive molecules may be one of the primary mechanisms whereby MSCs mediate their therapeutic efficacy. These secreted compounds can inhibit a range of processes such as apoptotic cell death and fibrosis,Citation11 in addition to being able to drive angiogenesis,Citation12,Citation13 and to regulate the immune response.Citation14,Citation15

Without any exogenous manipulation, MSCs achieve limited therapeutic efficacy due to their poor survival and limited GF secretion upon transplantation. The therapeutic efficacy of MSCs ultimately depend upon the number of cells implanted, the function of these cells, when they are administered, and what condition they are being used to treat.Citation9,Citation16-18 Poor MSC engraftment can be attributable to limited cell survival as a consequence of ischemia, anoikis, loss of trophic factors, or localized inflammation.Citation19 It is thus vital that MSC survival and differentiation be improved following transplantation in order to enhance therapeutic outcomes in treated patients. To that end, studies have explored the use of MSCs modified to express certain exogenous genes that can enhance their ability to promote angiogenesis and target tissue homing.Citation13,Citation20 These genetically engineered MSCs can thereby both improve MSC engraftment and functionality, while also allowing for the targeted delivery of therapeutic gene products that can enhance local tissue healing.Citation21 Indeed, MSCs can secret a broad profile of active molecules including hematopoietic growth factors, angiogenic growth factors, trophic molecules, immunomodulatory cytokines, and chemokines. The best-characterized GFs and cytokines produced by these cells are compiled in . Based on these previous findings, it is clear that engineering MSCs to overexpress GFs may be an optimal means of improving the therapeutic efficacy of these cells.

Table 1 Secretome of Mesenchymal Stem Cells

Vectors Used for GF Overexpression in MSCs

Both non-viral vectors such as lipids or polymers, as well as viral vectors (including retroviruses, adenoviruses, lentiviruses and adeno-associated viruses) have been used to mediate GF overexpression in MSCs. The most common vectors used for such approaches are compiled in .Citation31Citation39 Using viral vectors to insert genes into MSCs is a high transduction efficiency approach that has the potential to induce off-target effects owing to insertional mutagenesis.Citation32,Citation35,Citation40,Citation41 Viral systems are also limited by relatively small transgene cargo capacity, high production cost, difficulties in production and scale-up, and adverse immune reactions. There are advantages and disadvantages to all known viral vectors, with the selection of an appropriate vector being dependent upon transduction rates and the desired duration of treatment and target gene expression. It is also essential that such modified MSCs be extensively screened for safety reasons, thus potentially reducing the cost-effectiveness of such approaches in a clinical context.

Table 2 Summary of Common Vectors Used for GF Expression in MSCs

To avoid the limitations of viral vectors, non-viral vectors such as nanoparticles (NPs) or cationic liposomes have been utilized to deliver vectors into MSCs. These alternative delivery strategies are more scalable and flexible, easier to synthesize and target to tissues, less likely to drive immune stimulation, and more amenable to scale-up manufacturing.Citation37 However, the disadvantages of non-viral vectors can include their transient expression with low efficiencies, and their potential for associated toxicity. He et alCitation42 utilized the cationic polymer pullulan-spermine to overexpress HGF encoded in the pMEX vector in MSCs, resulting in high in vitro HGF expression.Citation43 In contrast to such success, however, Tan et alCitation44 found that such plasmid-containing liposomes were only able to mediate FGF expression in BMSCs at a relatively low transfection efficiency, although they were able to achieve expression at levels sufficient to support periodontal regeneration. Still other authors have utilized lipid-based NPs to achieve target gene expression in MSCs for a sustained period of time.Citation45,Citation46 These vectors, however, have recently been suggested to have the potential to induce genotoxicity, thus potentially mediating oncogenesis.Citation47,Citation48 It is thus important to weight the relative costs and benefits of these different strategies to MSC engineering in order to produce a safe, effective, and sustainable approach for clinical use.

The Impact of GF Overexpression on MSCs in Tissue Regeneration

The Primary Impact of GF Gene-Modified MSCs in Tissue Regeneration

Many previous studies have explored the ability of GFs to regulate MSC growth in vitro via adding these GFs to cell culture media and/or by inhibiting their cognate receptors, allowing for the study of concentration-dependent effects. When MSCs are treated with GFs including FGF-2, PDGF-B, TGF-β1, and VEGF-A, this has been shown to result in enhanced production and secretion of GFs by MSCs.Citation49 As such, overexpressing target GFs in MSCs may be able to yield similar therapeutic effects to those observed upon adding recombinant GFs to MSC cultures, although they can also affect the biology of cells in a therapeutically uncertain manner. Indeed, the secretion of exosomes and GFs such as HGF, FGF-B, and VEGF is potentially key to the regenerative abilities of MSCs.Citation23 When these GFs are overexpressed, this is associated with significant enhancement of MSC-mediated regeneration of tissues, making such GF overexpression strategies a focus of key therapeutic interest. A general overview of the therapeutic utilization of GF gene-modified MSCs is shown in . First, MSCs are extracted from humans or animals, identified, and amplified. Second, the GF gene of interest is integrated into the vector and jointly introduced into the MSCs. Third, GF modified MSCs are delivered to the target tissues of the recipient organism wherein they can play a therapeutic role via secreting GFs, promoting angiogenesis, and enhancing homing functions.

Figure 1 An overview of the therapeutic utilization of GF gene-modified MSCs.

Abbreviation: GF, growth factor; MSC, Mesenchymal stem cell.
Figure 1 An overview of the therapeutic utilization of GF gene-modified MSCs.

The Selection of GFs in MSC Modification

Initially selection of GFs used to treat MSCs was based on prior understanding of the role of these GFs in cellular differentiation and morphogenesis, with experiments being aimed at exploring the ability of these GFs to drive MSC differentiation towards particular lineages. For example, HGF is a multifunctional factor produced by MSCs which can bind to its cognate receptor c-Met on cells of the vascular endothelium.Citation50 Studies using mice lacking expression of HGF in specific tissues highlighted the ability of this GF to support tissue repair and regeneration,Citation51 and the implantation of MSCs overexpressing HGF led to enhanced left ventricular remodeling,Citation52 reductions in neurological deficits,Citation53 and enhanced liver function.Citation43 Similarly, MSCs engineered to overexpress VEGF have been shown to enhance the viability of cells in the context of in vitro hypoxia and can also improve capillary formation in animal models of myocardial infarction,Citation54 hind limb ischemia,Citation49 and skin defects.Citation55 Certain GFs exhibit similar repair effects in MSCs for many tissue types. For instance, angiopoietin-1 (Ang-1) is a growth factor that specifically acts on endothelial cells and can drive angiogenesis.Citation56 MSCs overexpressing Ang-1 have been shown to inhibit cardiac remodeling and to drive improved myocardial angiogenesis and arteriogenesis relative to control MSCs.Citation57 Such cells were also able to markedly reduce pulmonary inflammationCitation58 and to facilitate tissue repair.Citation59 MSCs overexpressing Ang-1 have also been shown to improve wound healing in a rat model system, enhancing angiogenesis in addition to dermal and epidermal tissue regeneration.Citation60 Tissue-specific repair factor modifications enhance the repair capabilities of MSCs in specific tissues. For example, BDNF promotes the survival and differentiation of neuronal tissue by acting on receptor kinases,Citation61 and BDNF-MSCs have primarily been used to promote the survival of neurons in the context of brain injury.Citation62,Citation63 Similarly, TGF family proteins are closely linked to MSC survival and differentiation.Citation64,Citation65 In particular, TGF-β superfamily genes are often used to drive MSC chondrogenic differentiation.Citation66 Therefore, TGF-β1 has been chosen to engineer rat MSCs to support enhanced regeneration of cartilage.Citation42 Hence, the selection of a particular GF for use in the modification of MSCs depends upon the effect of growth factors on MSCs and also on the response of the damaged tissue itself.

MSCs Overexpressing Multiple GF Genes Exhibit Therapeutic Utility

The primary mechanism whereby such gene-modified MSCs contribute to tissue repair is via the secretion of these multifactorial GFs rather than via their ability to differentiate into particular cell types, with these cells serving key roles in inhibiting fibrosis and inflammation while promoting angiogenesis.Citation49 Some studies have modified MSCs to express multiple synergistic genes in an effort to enhance their therapeutic utility. For example, IGF-1 is a GF that promotes cell survival,Citation67 whereas HGF promotes angiogenesis while suppressing fibrosis and inflammation.18 In a rat model of myocardial infarction, human adipose-derived stem cells that continuously produced IGF-1 and HGF were able to achieve a 1.3-fold increase in medium-sized blood vessel density at the infarct border zone relative to control cells.Citation68 In another study using a porcine model of myocardial infarction, such MSCs overexpressing HGF and IGF-1 were able to drive angiogenesis and suppress inflammation more effectively than other cells, although these cells also exhibited enhanced fibrosis suggesting that combined IGF-1 and HGF exposure over extended periods of time can induce both beneficial and counterproductive effects.Citation69 This suggests that the preparation of MSCs secreting both IGF-1 and HGF may not be an effective means of synergistically effective cardiac repair, with the elevated levels of either factor in the local environment potentially contributing to this effect.

The Role of Exosomes Derived from GF-Modified MSCs in Tissue Regeneration

Multiple studies have indicated a role for MSCs in regenerative medicine through their paracrine effects and ability to produce exosomes.Citation17,Citation70 Encapsulated with a lipid bilayer, exosomes can protect their contents from degradation and can transport a variety of small biomolecules including mRNAs, miRNAs, and proteins to surrounding cells. Moreover, MSC sources and culture conditions have been shown to influence the regenerative responses induced by exosomes, as a number of GFs can be detected in MSC-derived exosomes, including HGF, IGF family members, FGF2, and platelet-derived growth factor-AA (PDGF-AA).Citation69 As natural vesicles suitable for gene delivery, MSC-derived exosomes exhibit a broad range of therapeutic effects, and can mediate tissue repair, immunological regulation, and inflammatory control.Citation70,Citation72 Moreover, recent studies have revealed that MSC-derived exosomes can mediate therapeutic benefits in animal disease models, with previous studies of bone fracture, cutaneous wound, myocardial infarction, and acute hepatic injury all having demonstrated the clinical utility of such exosomes.Citation16,Citation17,Citation70,Citation73 Exosomes can modulate the differentiation and migration of MSCs in a targeted manner, offering an opportunity to promote tissue regeneration in a cell-free manner. Genetic manipulation can also be used to control the levels of such GFs in these exosomes, as in studies in which huc-MSCs were engineered to secrete GFs in a controlled fashion over an extended period.Citation71 Such genetically modified MSC-derived exosomes may thereby be able to mediate tissue regenerative benefits, making them ideal for future therapeutic regenerative regimens.

Preclinical Use of GF-Modified MSCs in Tissue Regeneration

The therapeutic value of MSCs stems largely from their ability to mediate angiogenesis and tissue regeneration,Citation75 secreting GFs and exosomes to achieve therapeutic efficacy and homing to target tissue sites.Citation9,Citation16,Citation17 A number of preclinical studies to date have sought to use genetically-modified MSCs that secrete GFs in order to treat a wide range of conditions associated with tissue injuries. A detailed overview of the uses of GF-modified MSCs in preclinical tissue repair studies is given in .

Table 3 Preclinical Studies of the Use of Genetically Engineered MSCs in Tissue Repair

Central Nervous System (CNS) Lesions

Occlusive cerebrovascular diseases can result in cerebral ischemia and significant neuropathology, leading to the exploration of many modes of treating such diseases including the application of MSC-based therapies. One of the keys to treating CNS lesions is to maintain the integrity of the blood-brain barrier and to reduce edema in the context of ischemia, thus reducing the severity of injury. Importantly, MSCs can home to the CNS in vivo, allowing them to improve functional recovery following stroke owing to their ability to drive angiogenesis and neurogenesis while suppressing local inflammation via GF and chemokine secretion.Citation88 MSCs overexpressing specific GFs can also help to facilitate efficient CNS tissue regeneration. For example, MSCs overexpressing HGF have shown superior efficacy in reducing neurological deficits in the rat middle cerebral artery occlusion (MCAO) model relative to unmodified MSCs.Citation53 Su et alCitation89 found that BMSCs engineered to overexpress GDNF using a lentivirus were able to protect against injury in PC12 cells, highlighting their potential therapeutic value in the context of Parkinson’s disease. However, there are still many obstacles to the widespread use of this technique in CNS lesions. Intracerebral injection remains particularly difficult if the lesions are widespread and numerous. In addition, intra-arterial injection will increase risk of embolic events,Citation90 and intravenous injections typically result in few cells reaching the target sites.Citation91,Citation92

Ischemic Heart Disease (IHD)

In many nations, the primary cause of morbidity and mortality is myocardial infarction (MI),Citation93 and as such it is one of the most common targets of therapeutic efforts to engineer MSCs to facilitate tissue repair. Indeed, a number of genes have been proposed as targets for MSC-mediated delivery in the context of MI including HO-1,Citation94 IGF-1,Citation69 Ang-1,Citation57,Citation81 SVV,Citation95 Bcl-2Citation96 and Akt1.Citation97,Citation98 Over 30 clinical studies to date have been registered using MSCs for the treatment of MI, but these studies have suggested the need for improved therapeutic efficacy of these MSCs. Angiogenesis mediates clinical benefits via the formation, remodeling, and maturation of blood vessels in injured tissues, making GF engineering an ideal means of achieved such angiogenic efficacy in a therapeutic setting. Moreover, among angiogenic growth factors, the HGF/Met pathway is a key mediator of cardiovascular remodeling following tissue injury,Citation99 with HGF mediating the migration and expression of cardiac-specific markers in MSCs.Citation100 Many studies have utilized murine, rat, and porcine models of MI to confirm the ability of such HGF-expressing MSCs to enhance cardiac function, drive angiogenesis, and decrease myocardial fibrosis.Citation79,Citation101-103 In addition, human BMSCs expressing HGF have been shown to have enhanced anti-arrhythmic properties.Citation104 Following the delivery of these modified cells to the infarcted region, low local nutrient and oxygen levels can result in poor survival and engraftment efficiency. VEGF is known to enhance the survival of these and other cell types upon transplantation in damaged tissues.Citation105 Normally, angiogenesis in the infarcted tissue is not sufficient to meet the needs of the remaining viable myocardial tissue, thereby compromising contractile compensation.Citation80 Moon et alCitation54 found that MSCs overexpressing VEGF were able to induce a 1.4-fold increase in VEGF expression upon hypoxic exposure relative to cells grown under normoxic conditions, and these modified MSCs were able to facilitate the enhanced microvascularization of infarcted myocardial tissues.

Musculoskeletal Defects and Skin Injuries

Bone, muscle, and skin are all highly metabolized tissues with a relatively high vascular supply, based on the homeostasis of biomaterial structures that need to be studied for growth and remodeling.Citation106 Kumar et alCitation87 found that mice transplanted with MSCs engineered to overexpress bone morphogenetic protein 2 (BMP2) exhibited increased bone mineral density and content and improved BMSC proliferation relative to control animals, with a corresponding improvement in bone formation. Dental pulp stem cells overexpressing HGF have also been shown to prevent bone loss in the early phase of ovariectomy-induced osteoporosis.Citation107 MSCs engineered to overexpress Ang-1 are also able to facilitate wound healing as well as dermal and epidermal regeneration and angiogenesis.Citation60 In addition, tissue engineering is usually achieved via inserting stem cells into three-dimensional scaffolds that are induced to generate new cells.Citation6,Citation108 GF-modified MSCs have been widely used in this innovative treatment for musculoskeletal defects and skin wounds, with many studies having explored optimal tissue engineering approaches to improving the efficiency of cells, scaffolds, and bioactive factors.Citation33 The most commonly studied technique is to add supplemental growth factors that locally provide signals that mimic the process of bone regeneration.Citation109 It is therefore important to design systems that provide this biological cue in a time-controlled manner so as to mimic the normal bone healing process. Brunger et al attempted to develop a system using poly-L-lysine to immobilize a lentivirus encoding TGF-β3 in a 3D woven poly scaffold to induce robust and sustained cartilaginous extracellular matrix formation by hMSCs.Citation110

Radiation Injury

Certain tissues including the lungs, intestines, and bone marrow are highly radiation sensitive. While hematopoietic stem cells can regenerate the bone marrow, strategies to mediate similar regeneration of lung and intestinal tissue are limited. GF-overexpressing MSCs may therefore represent an ideal approach to regenerating tissues following radiation injury and associated damage. For example, in a model of radiation-induced lung fibrosis, MSCs overexpressing HGF were shown to home to damaged lung tissue wherein they could promote epithelial cell proliferation and survival, thereby decreasing local inflammation and fibrosis.Citation104 Similarly, MSCs engineered to overexpress TGF-β2 using an adenoviral vector were able to reduce lung injury and protect alveolar type II cells from radiation-induced apoptosis and DNA damage while reducing local inflammation, highlighting the benefits of GF production by MSCs in a paracrine manner.Citation85 BMSCs engineered to express VEGF were similarly able to improve radiation-induced tissue injury repair owing to their ability to drive angiogenesis and regeneration of muscle fibers.Citation111 BMSCs modified to express both BD2 and PDGF-A using an adenoviral vector were also able to improve wound healing in a model of radiation-induced wounding.Citation84 MSCs overexpressing HGF suppress local inflammation and enhance small intestinal recovery in a murine model of radiation induced intestinal injury.Citation83 Irradiation of cardiac tissue can result in late cardiovascular complications, and HGF can reduce such radiation-induced cardiac injury in a model of irradiation-induced heart disease.Citation112 Adenoviral-mediated overexpression of HGF can also prevent radiation-induced hematopoietic damageCitation113 and can reduce radiation induced hepatic damage in a rat model system.Citation114

Other Tissue Injuries and Diseases

In addition to the diseases mentioned above, MSCs modified to overexpress GFs have been employed to treat a wide range of tissue injuries and diseases in preclinical studies. Studies have shown that MSCs overexpressing HGF and Ang-1, respectively, can improve therapeutic outcomes in ischemia/reperfusion injury in the lungCitation115 and in a Phosgene-induced model of lung injury owing to their ability to decrease pulmonary inflammation and endothelial permeability.Citation116 Furthermore, MSCs modified to overexpress HGF have been shown to improve such AKI in a rat model of ischemia/reperfusion injury via reducing kidney inflammation and apoptotic cell death, thus making these cells of value to human therapeutic implementation.Citation50 Moreover, MSCs expressing HGF can also enhance liver regeneration, making them viable for the treatment of those patients suffering from liver fibrosis or cirrhosis.Citation43

Clinical Trials Utilizing Genetically Modified MSCs

Given the number of preclinical studies demonstrating the potential utility of genetically modified MSCs, it is perhaps unsurprising that a number of clinical trials have been or are currently being conducted exploring the clinical value of such therapeutic approaches. To date over one thousand MSC-based trials have been conducted globally as reported in the US National Institute of Health database (ClinicalTrial.gov) in order to evaluate the safety and efficacy of either autologous or allogeneic MSCs. These trials are primarily focused on treating human diseases such as cancer,Citation117 metabolic and inflammatory diseases such as chronic obstructive pulmonary disease,Citation118 or adult respiratory distress syndrome.Citation119 These studies are primarily reliant upon the use of unmodified MSCs for these clinical efforts, with very few studies to date utilizing genetically modified MSCs. In 2006, Ripa et al published the results of a trial initiated in 2003 piloting the combination of VEGF gene therapy and stem cell mobilization in patients with severe chronic ischemic heart disease, finding this approach to be safe in humansCitation120 (NCT00135850). Another relevant study aims to explore the use of MSCs overexpressing BDNF for the treatment of Huntington’s disease (HD) patients in a pre-cellular therapy observational studyCitation121 (NCT01937923). At present, however, this study has only enrolled a cohort of individuals early-stage HD in order to characterize their clinical and biomarker findings at baseline for comparisons to a planned future Phase 1 trial safety and tolerability trial applying these BDNF-modified MSCs. This trial has been submitted as an Investigational New Drug application to the Food and Drug Administration.Citation122

A number of challenges still face the clinical implementation of GF gene-modified MSC-based therapies. Of particular difficulty is the production of clinical grade therapeutic products, as such cellular and gene therapies differ from traditional pharmaceutical compounds, instead representing a form of heterogenous biological product that can vary in response to a wide range of culture conditions. Modified MSCs also have the potential to become malignant upon transplant, and the use of recombinant viral vectors to manipulate these cells poses a significant safety concern.Citation109 Minimizing variability in sample preparation while still remaining cost-effective thus represents a significant challenge. Therefore, the production of modified MSCs for clinical applications must comply with the Good Manufacturing Practice (GMP) standards for medicinal products. These recommended approaches include product safety, cell characterization, and manufacturing process control.Citation123 Cell donors must be screened carefully and the stem cells expanded in the GMP production facilities should be tested using standardized procedures to assess their viability, sterility, genetic stability, tumorigenicity, adventitious agents, pyrogenicity, and mycoplasma infection status. Modified MSCs also have to be verified with respect to their identities, purity, stability and potency. In addition, the biological activity and toxicity of stem cell products must be tested in an applicable animal model under Good Laboratory Practice (GLP) conditions prior to administration into humans.Citation124 To ensure product efficacy, however, it is essential that these standardized production procedures do not compromise therapeutic efficacy. The fate of modified MSCs upon intravenous injection is also uncertain, as previous trials of unmodified MSCs have achieved limited efficacy owing to their quick elimination from circulation.Citation125 Therefore, to achieve significant functional benefits, this strategy requires a defined selection of the number and type of stem cells to be delivered, an explicit vector application method, and fixed transduction efficiency and time of administration. In addition, the design of novel bioactive materials such as three-dimensional spheroidsCitation126 and nano-active scaffoldsCitation109 to bolster stem cell survival, signaling, and function at the target site can also help to increase the cost-effectiveness of the applications of modified MSCs for tissue repair.

At present it remains unclear as to whether it will be legally permissible to utilize genetically modified MSCs for clinical treatment. The potential consequences of utilizing such cells in humans are not well understood, and as such the safety of these approaches needs to be more thoroughly examined in animal model systems in order to identify means overcoming any potential safety issues. In addition, many of the ethical issues associated with genetically-modified MSC research are similar to those arising in other MSC-based interventions. Efforts to address these issues typically focus upon minimizing the risk of harm, emphasizing the importance of informed consent and information disclosure, reducing the potential for overpromising, limiting excessive expectations and therapeutic misconceptions, and avoiding pressure from commercial entities and disease constituencies to move quickly into the clinic.Citation125,Citation127 In addition, justice is a necessary consideration given that stem cell interventions can be extraordinary costly and labor-intensive,Citation123 as can many other novel biotechnologies. Justice necessitates that additional attention be paid to the cost of genetically modified MSC interventions in an effort to make them available, effective, and safe, with the goal of reducing unfair disparities in treatment accessibility. These ethical considerations continue to provide crucial guidance for the clinical application of these approaches not only for the trials specifically considered, but also for investigators exploring new translational medicine pathways.

Current Challenges and Future Prospects

The therapeutic utility of GF gene-modified MSCs has been a focus of increasing research interest in recent years owing to their enhanced ability to suppress inflammation, home to target tissues, regulate immune responses, and facilitate tissue repair. Several preclinical and clinical studies have utilized MSC-based therapeutic strategies for treating a range of disorders and injuries. While efforts to modify MSCs to overexpress defined GFs are still in their early stages and are far from clinical application, although they offer a potentially ideal means of directed tissue regeneration. MSCs alone are limited in their ability to home to and survive in injured tissues, making the modification of MSCs to express such GF genes vital in order to facilitate more robust regenerative medicine approaches. While the outcomes of many of the studies reported in this review are promising, there remain many challenges which must be overcome. These include the need to optimize delivery strategies in human patients while simultaneously preventing immunogenicity or tumor formation. Preclinical findings highlight the safety and therapeutic efficacy of these GF-modified MSCs for the treatment of tissue damage.

In addition, large-scale, multi-center clinical trials are needed to conclusively demonstrate the long-term beneficial effects of such therapies.

Further ongoing clinical studies and efforts to demonstrate the long-term beneficial effects will help to ensure that these promising therapeutic tools soon become available to patients as a novel and efficacious form of regenerative medicine.

Data Sharing Statement

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

Disclosure

The authors report no conflicts of interest in this work.

Acknowledgments

The authors would like to thank the Jilin Scientific and Technological Development Program and the Interdisciplinary Research Funding Program for PhD Students of Jilin University, for their support in developing this paper.

References

  • RajabzadehN, FathiE, FarahzadiR. Stem cell-based regenerative medicine. Stem Cell Investig. 2019;6:19. doi:10.21037/sci
  • CoreyS, BonsackB, BorlonganCV. Stem cell-based regenerative medicine for neurological disorders: a special tribute to Dr. Teng Ma. Brain Circ. 2019;5(3):97–100. doi:10.4103/bc.bc_39_1931620654
  • LeeWS, KimHJ, KimKI, KimGB, JinW. Intra-articular injection of autologous adipose tissue-derived mesenchymal stem cells for the treatment of knee osteoarthritis: a phase IIb, randomized, placebo-controlled clinical trial. Stem Cells Transl Med. 2019;8(6):504–511. doi:10.1002/sctm.18-012230835956
  • KhanS, MafiP, MafiR, KhanW. A systematic review of mesenchymal stem cells in spinal cord injury, intervertebral disc repair and spinal fusion. Curr Stem Cell Res Ther. 2018;13(4):316–323. doi:10.2174/1574888X1166617090712003028891440
  • JayaramP, IkpeamaU, RothenbergJB, MalangaGA. Bone marrow-derived and adipose-derived mesenchymal stem cell therapy in primary knee osteoarthritis: a narrative review. PM R. 2019;11(2):177–191. doi:10.1016/j.pmrj.2018.06.01930010050
  • QaziTH, DudaGN, OrtMJ, PerkaC, GeisslerS, WinklerT. Cell therapy to improve regeneration of skeletal muscle injuries. J Cachexia Sarcopenia Muscle. 2019;10(3):501–516. doi:10.1002/jcsm.1241630843380
  • LemckeH, VoroninaN, SteinhoffG, DavidR. Recent progress in stem cell modification for cardiac regeneration. Stem Cells Int. 2018;2018:1909346.29535769
  • GrimmD, EgliM, KrugerM, et al. Tissue engineering under microgravity conditions-use of stem cells and specialized cells. Stem Cells Dev. 2018;27(12):787–804.29596037
  • MadrigalM, RaoKS, RiordanNH. A review of therapeutic effects of mesenchymal stem cell secretions and induction of secretory modification by different culture methods. J Transl Med. 2014;12:260.25304688
  • Mendez-FerrerS, MichurinaTV, FerraroF, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466(7308):829–834. doi:10.1038/nature0926220703299
  • RodriguesM, GriffithLG, WellsA. Growth factor regulation of proliferation and survival of multipotential stromal cells. Stem Cell Res Ther. 2010;1(4):32. doi:10.1186/scrt3220977782
  • HuMS, BorrelliMR, LorenzHP, LongakerMT, WanDC. Mesenchymal stromal cells and cutaneous wound healing: a comprehensive review of the background, role, and therapeutic potential. Stem Cells Int. 2018;2018:6901983. doi:10.1155/2018/690198329887893
  • ShafeiAE, AliMA, GhanemHG, et al. Mesenchymal stem cell therapy: a promising cell-based therapy for treatment of myocardial infarction. J Gene Med. 2017;19:12. doi:10.1002/jgm.2995
  • LiuF, QiuH, XueM, et al. MSC-secreted TGF-beta regulates lipopolysaccharide-stimulated macrophage M2-like polarization via the Akt/FoxO1 pathway. Stem Cell Res Ther. 2019;10(1):345. doi:10.1186/s13287-019-1447-y31771622
  • WuR, LiuC, DengX, ChenL, HaoS, MaL. Enhanced alleviation of aGVHD by TGF-beta1-modified mesenchymal stem cells in mice through shifting MPhi into M2 phenotype and promoting the differentiation of Treg cells. J Cell Mol Med. 2019.
  • ZhangB, WangM, GongA, et al. HucMSC-exosome med-Wnt4 signaling is required for cutaneous wound healing. Stem Cells. 2015;33(7):2158–2168. doi:10.1002/stem.177124964196
  • FurutaT, MiyakiS, IshitobiH, et al. Mesenchymal stem cell-derived exosomes promote fracture healing in a mouse model. Stem Cells Transl Med. 2016;5(12):1620–1630. doi:10.5966/sctm.2015-028527460850
  • WangLS, WangH, ZhangQL, YangZJ, KongFX, WuCT. Hepatocyte growth factor gene therapy for ischemic diseases. Hum Gene Ther. 2018;29(4):413–423. doi:10.1089/hum.2017.21729409352
  • SongH, SongBW, ChaMJ, ChoiIG, HwangKC. Modification of mesenchymal stem cells for cardiac regeneration. Expert Opin Biol Ther. 2010;10(3):309–319. doi:10.1517/1471259090345599720132054
  • LiX, WangQ, DingL, et al. Intercellular adhesion molecule-1 enhances the therapeutic effects of MSCs in a dextran sulfate sodium-induced colitis models by promoting MSCs homing to murine colons and spleens. Stem Cell Res Ther. 2019;10(1):267. doi:10.1186/s13287-019-1384-931443680
  • LiL, ZhangD, LiP, DamaserM, ZhangY. Virus integration and genome influence in approaches to stem cell based therapy for andro-urology. Adv Drug Deliv Rev. 2015;82-83:12–21. doi:10.1016/j.addr.2014.10.01225453258
  • SzeSK, de KleijnDP, LaiRC, et al. Elucidating the secretion proteome of human embryonic stem cell-derived mesenchymal stem cells. Mol Cell Proteomics. 2007;6(10):1680–1689. doi:10.1074/mcp.M600393-MCP20017565974
  • TsujiK, KitamuraS, WadaJ. Secretomes from mesenchymal stem cells against acute kidney injury: possible heterogeneity. Stem Cells Int. 2018;2018:8693137. doi:10.1155/2018/869313730651737
  • LiuCH, HwangSM. Cytokine interactions in mesenchymal stem cells from cord blood. Cytokine. 2005;32(6):270–279. doi:10.1016/j.cyto.2005.11.00316377203
  • HochAI, BinderBY, GenetosDC, LeachJK. Differentiation-dependent secretion of proangiogenic factors by mesenchymal stem cells. PLoS One. 2012;7(4):e35579. doi:10.1371/journal.pone.003557922536411
  • OskowitzA, McFerrinH, GutschowM, CarterML, PochampallyR. Serum-deprived human multipotent mesenchymal stromal cells (MSCs) are highly angiogenic. Stem Cell Res. 2011;6(3):215–225. doi:10.1016/j.scr.2011.01.00421421339
  • CaplanAI, DennisJE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98(5):1076–1084. doi:10.1002/(ISSN)1097-464416619257
  • AggarwalS, PittengerMF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105(4):1815–1822. doi:10.1182/blood-2004-04-155915494428
  • ZhangH, YangR, WangZ, LinG, LueTF, LinCS. Adipose tissue-derived stem cells secrete CXCL5 cytokine with neurotrophic effects on cavernous nerve regeneration. J Sex Med. 2011;8(2):437–446. doi:10.1111/j.1743-6109.2010.02128.x21114767
  • ArthurA, CakourosD, CooperL, et al. Twist-1 enhances bone marrow mesenchymal stromal cell support of hematopoiesis by modulating CXCL12 expression. Stem Cells. 2016;34(2):504–509. doi:10.1002/stem.226526718114
  • ChenX, NomaniA, PatelN, NouriFS, HatefiA. Bioengineering a non-genotoxic vector for genetic modification of mesenchymal stem cells. Biomaterials. 2018;152:1–14. doi:10.1016/j.biomaterials.2017.10.02829078136
  • ChanJ, O’DonoghueK, DeL, et al. Human fetal mesenchymal stem cells as vehicles for gene delivery. Stem Cells. 2010;23(1):93–102. doi:10.1634/stemcells.2004-0138
  • ParkJS, SuryaprakashS, LaoYH, LeongKW. Engineering mesenchymal stem cells for regenerative medicine and drug delivery. Methods. 2015;84:3–16. doi:10.1016/j.ymeth.2015.03.00225770356
  • AslanH, ZilbermanY, ArbeliV, et al. Nucleofection-based ex vivo nonviral gene delivery to human stem cells as a platform for tissue regeneration. Tissue Eng. 2006;12(4):877–889. doi:10.1089/ten.2006.12.87716674300
  • Santiago-TorresJE, LovaszR, BertoneAL. Fetal vs adult mesenchymal stem cells achieve greater gene expression, but less osteoinduction. World J Stem Cells. 2015;7(1):223–234. doi:10.4252/wjsc.v7.i1.22325621122
  • StenderS, MurphyM, O’BrienT, et al. Adeno-associated viral vector transduction of human mesenchymal stem cells. Eur Cell Mater. 2007;13:93–99; discussion 99. doi:10.22203/eCM.v013a10
  • HamannA, NguyenA, PannierAK. Nucleic acid delivery to mesenchymal stem cells: a review of nonviral methods and applications. J Biol Eng. 2019;13(1):7. doi:10.1186/s13036-019-0140-030675180
  • SantosJL, PanditaD, RodriguesJ, PegoAP, GranjaPL, TomasH. Non-viral gene delivery to mesenchymal stem cells: methods, strategies and application in bone tissue engineering and regeneration. Curr Gene Ther. 2011;11(1):46–57. doi:10.2174/15665231179452010221182464
  • GheisariY, SoleimaniM, AzadmaneshK, ZeinaliS. Multipotent mesenchymal stromal cells: optimization and comparison of five cationic polymer-based gene delivery methods. Cytotherapy. 2008;10(8):815–823. doi:10.1080/1465324080247430719058061
  • LuCH, LinKJ, ChiuHY, et al. Improved chondrogenesis and engineered cartilage formation from TGF-beta3-expressing adipose-derived stem cells cultured in the rotating-shaft bioreactor. Tissue Eng Part A. 2012;18(19–20):2114–2124. doi:10.1089/ten.tea.2012.001022712565
  • LoWH, HwangSM, ChuangCK, ChenCY, HuYC. Development of a hybrid baculoviral vector for sustained transgene expression. Mol Ther. 2009;17(4):658–666. doi:10.1038/mt.2009.1319240695
  • HeCX, ZhangTY, MiaoPH, et al. TGF-beta1 gene-engineered mesenchymal stem cells induce rat cartilage regeneration using nonviral gene vector. Biotechnol Appl Biochem. 2012;59(3):163–169. doi:10.1002/bab.100123586825
  • SeoKW, SohnSY, BhangDH, NamMJ, LeeHW, YounHY. Therapeutic effects of hepatocyte growth factor-overexpressing human umbilical cord blood-derived mesenchymal stem cells on liver fibrosis in rats. Cell Biol Int. 2014;38(1):106–116. doi:10.1002/cbin.1018624115681
  • TanZ, ZhaoQ, GongP, et al. Research on promoting periodontal regeneration with human basic fibroblast growth factor-modified bone marrow mesenchymal stromal cell gene therapy. Cytotherapy. 2009;11(3):317–325. doi:10.1080/1465324090282475719308772
  • GandraN, WangDD, ZhuY, MaoC. Virus-mimetic cytoplasm-cleavable magnetic/silica nanoclusters for enhanced gene delivery to mesenchymal stem cells. Angew Chem Int Ed Engl. 2013;52(43):11278–11281. doi:10.1002/anie.20130111324038718
  • MunJY, ShinKK, KwonO, LimYT, OhDB. Minicircle microporation-based non-viral gene delivery improved the targeting of mesenchymal stem cells to an injury site. Biomaterials. 2016;101:310–320. doi:10.1016/j.biomaterials.2016.05.05727315214
  • HubbsAF, MercerRR, BenkovicSA, et al. Nanotoxicology - a pathologist’s perspective. Toxicol Pathol. 2011;39(2):301–324. doi:10.1177/019262331039070521422259
  • NorppaH, CatalánJ, FalckG, HannukainenK, SiivolaK, SavolainenK. Nano-specific genotoxic effects. J Biomed Nanotechnol. 2011;7(1):19. doi:10.1166/jbn.2011.117921485781
  • FierroFA, KalomoirisS, SondergaardCS, NoltaJA. Effects on proliferation and differentiation of multipotent bone marrow stromal cells engineered to express growth factors for combined cell and gene therapy. Stem Cells. 2011;29(11):1727–1737. doi:10.1002/stem.72021898687
  • ChenYA, QianH, ZhuW, et al. Hepatocyte growth factor modification promotes the amelioration effects of human umbilical cord mesenchymal stem cells on rat acute kidney injury. Stem Cells Dev. 2011;20(1):103–113. doi:10.1089/scd.2009.049520446811
  • GonzálezMN, de MelloW, Butler-BrowneGS, et al. HGF potentiates extracellular matrix-driven migration of human myoblasts: involvement of matrix metalloproteinases and MAPK/ERK pathway. Skelet Muscle. 2017;7(1):20. doi:10.1186/s13395-017-0138-629017538
  • WangSX, QinX, SunDD, et al. Effects of hepatocyte growth factor overexpressed bone marrow-derived mesenchymal stem cells on prevention from left ventricular remodelling and functional improvement in infarcted rat hearts. Cell Biochem Funct. 2012;30(7):574–581. doi:10.1002/cbf.v30.722592978
  • ZhaoMZ, NonoguchiN, IkedaN, et al. Novel therapeutic strategy for stroke in rats by bone marrow stromal cells and ex vivo HGF gene transfer with HSV-1 vector. J Cereb Blood Flow Metab. 2006;26(9):1176–1188. doi:10.1038/sj.jcbfm.960027316421510
  • MoonHH, JooMK, MokH, et al. MSC-based VEGF gene therapy in rat myocardial infarction model using facial amphipathic bile acid-conjugated polyethyleneimine. Biomaterials. 2014;35(5):1744–1754. doi:10.1016/j.biomaterials.2013.11.01924280192
  • YanfuH, RanT, YanqingH, et al. Microencapsulated VEGF gene-modified umbilical cord mesenchymal stromal cells promote the vascularization of tissue-engineered dermis: an experimental study. Cytotherapy. 2014;16(2):160–169. doi:10.1016/j.jcyt.2013.10.01424438897
  • TeeJK, SetyawatiMI, PengF, LeongDT, HoHK. Angiopoietin-1 accelerates restoration of endothelial cell barrier integrity from nanoparticle-induced leakiness. Nanotoxicology. 2019;13(5):682–700. doi:10.1080/17435390.2019.157164630776942
  • SunL, CuiM, WangZ, et al. Mesenchymal stem cells modified with angiopoietin-1 improve remodeling in a rat model of acute myocardial infarction. Biochem Biophys Res Commun. 2007;357(3):779–784. doi:10.1016/j.bbrc.2007.04.01017445769
  • MeiSH, McCarterSD, DengY, ParkerCH, LilesWC, StewartDJ. Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin 1. PLoS Med. 2007;4(9):e269. doi:10.1371/journal.pmed.004026917803352
  • XuJ, QuJ, CaoL, et al. Mesenchymal stem cell-based angiopoietin-1 gene therapy for acute lung injury induced by lipopolysaccharide in mice. J Pathol. 2008;214(4):472–481. doi:10.1002/path.230218213733
  • LiY, ZhengL, XuX, et al. Mesenchymal stem cells modified with angiopoietin-1 gene promote wound healing. Stem Cell Res Ther. 2013;4(5):113. doi:10.1186/scrt32424477049
  • SchabitzWR, SommerC, ZoderW, KiesslingM, SchwaningerM, SchwabS. Intravenous brain-derived neurotrophic factor reduces infarct size and counterregulates Bax and Bcl-2 expression after temporary focal cerebral ischemia. Stroke. 2000;31(9):2212–2217. doi:10.1161/01.STR.31.9.221210978054
  • WangZ, YaoW, DengQ, ZhangX, ZhangJ. Protective effects of BDNF overexpression bone marrow stromal cell transplantation in rat models of traumatic brain injury. J Mol Neurosci. 2013;49(2):409–416. doi:10.1007/s12031-012-9908-023143881
  • KurozumiK, NakamuraK, TamiyaT, et al. BDNF gene-modified mesenchymal stem cells promote functional recovery and reduce infarct size in the rat middle cerebral artery occlusion model. Mol Ther. 2004;9(2):189–197.14759803
  • LiWJ, TuliR, OkaforC, et al. A three-dimensional nanofibrous scaffold for cartilage tissue engineering using human mesenchymal stem cells. Biomaterials. 2005;26(6):599–609.15282138
  • HerrmannJL, WangY, AbarbanellAM, WeilBR, TanJ, MeldrumDR. Preconditioning mesenchymal stem cells with transforming growth factor-alpha improves mesenchymal stem cell-mediated cardioprotection. Shock. 2010;33(1):24–30.19996917
  • HuangAH, MotlekarNA, SteinA, DiamondSL, ShoreEM, MauckRL. High-throughput screening for modulators of mesenchymal stem cell chondrogenesis. Ann Biomed Eng. 2008;36(11):1909–1921.18791827
  • ScioliMG, CervelliV, ArcuriG, et al. High insulin-induced down-regulation of Erk-1/IGF-1R/FGFR-1 signaling is required for oxidative stress-mediated apoptosis of adipose-derived stem cells. J Cell Physiol. 2014;229(12):2077–2087.24818995
  • SaviM, BocchiL, FiumanaE, et al. Enhanced engraftment and repairing ability of human adipose-derived stem cells, conveyed by pharmacologically active microcarriers continuously releasing HGF and IGF-1, in healing myocardial infarction in rats. J Biomed Mater Res A. 2015;103(9):3012–3025.25727843
  • Gomez-MauricioG, MoscosoI, Martin-CanchoMF, et al. Combined administration of mesenchymal stem cells overexpressing IGF-1 and HGF enhances neovascularization but moderately improves cardiac regeneration in a porcine model. Stem Cell Res Ther. 2016;7(1):94.27423905
  • ChenB, LiQ, ZhaoB, WangY. Stem cell-derived extracellular vesicles as a novel potential therapeutic tool for tissue repair. Stem Cells Transl Med. 2017;6(9):1753‐1758.
  • ChoiJS, YoonHI, LeeKS, et al. Exosomes from differentiating human skeletal muscle cells trigger myogenesis of stem cells and provide biochemical cues for skeletal muscle regeneration. J Control Release. 2016;222:107–115.26699421
  • TsujiK, KitamuraS, WadaJ. Immunomodulatory and regenerative effects of mesenchymal stem cell-derived extracellular vesicles in renal diseases. Int J Mol Sci. 2020;21:3.
  • TanCY, LaiRC, WongW, DanYY, LimS-K, HoHK. Mesenchymal stem cell-derived exosomes promote hepatic regeneration in drug-induced liver injury models. Stem Cell Res Ther. 2014;5(3):76.24915963
  • ChoiJS, YoonHI, LeeKS, et al. Exosomes from differentiating human skeletal muscle cells trigger myogenesis of stem cells and provide biochemical cues for skeletal muscle regeneration. J Control Release. 2016;222:107–115.26699421
  • ShafeiAE, AliMA, GhanemHG, et al. Mesenchymal stem cells therapy: a promising cell based therapy for treatment of myocardial infraction. J Gene Med. 2017;19:12. doi:10.1002/jgm.2995
  • IkedaN, NonoguchiN, ZhaoMZ, et al. Bone marrow stromal cells that enhanced fibroblast growth factor-2 secretion by herpes simplex virus vector improve neurological outcome after transient focal cerebral ischemia in rats. Stroke. 2005;36(12):2725–2730.16282547
  • PollockK, DahlenburgH, NelsonH, et al. Human mesenchymal stem cells genetically engineered to overexpress brain-derived neurotrophic factor improve outcomes in Huntington’s disease mouse models. Mol Ther. 2016;24(5):965–977.26765769
  • ZhaoGY, CuiL, GaoJ, DaiRT, ZhangP. Study on the levels of DA and metabolite in striatum in rats with Parkinson’s disease treated by BDNF gene modified bone mesenchymal stem cells. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 2013;29(1):82–85.23662416
  • ChenH, XiaR, LiZ, et al. Mesenchymal stem cells combined with hepatocyte growth factor therapy for attenuating ischaemic myocardial fibrosis: assessment using multimodal molecular imaging. Sci Rep. 2016;6:33700.27804974
  • GaoF, HeT, WangH, et al. A promising strategy for the treatment of ischemic heart disease: mesenchymal stem cell-mediated vascular endothelial growth factor gene transfer in rats. Can J Cardiol. 2007;23(11):891–898.17876381
  • LiZ, MeiJ, ZhangB. Cell transplantation of 5-aza cytidine induced bone marrow stromal cells transfected by angiogenin gene ex vivo into infarcted myocardium, an experimental study. Zhonghua Yi Xue Za Zhi. 2002;82(19):1319–1323.12509935
  • FanL, LinC, ZhuoS, et al. Transplantation with survivin-engineered mesenchymal stem cells results in better prognosis in a rat model of myocardial infarction. Eur J Heart Fail. 2009;11(11):1023–1030.19875403
  • WangH, SunRT, LiY, et al. HGF Gene Modification in mesenchymal stem cells reduces radiation-induced intestinal injury by modulating immunity. PLoS One. 2015;10(5):e0124420.25933295
  • HaoL, WangJ, ZouZ, et al. Transplantation of BMSCs expressing hPDGF-A/hBD2 promotes wound healing in rats with combined radiation-wound injury. Gene Ther. 2009;16(1):34–42.18701914
  • XueJ, LiX, LuY, et al. Gene-modified mesenchymal stem cells protect against radiation-induced lung injury. Mol Ther. 2013;21(2):456–465.23299797
  • HuangJH, SurgeryBDO, HospitalD, et al. Intravenous injection of nerve growth factor gene-modified bone marrow stromal stem cells on the apoptosis of the mouse hepatic cells induced by radiation. J Clin Rehab Tissue Eng Res. 2008;12(38):7503–7506.
  • KumarS, NagyTR, PonnazhaganS. Therapeutic potential of genetically modified adult stem cells for osteopenia. Gene Ther. 2010;17(1):105–116.19741731
  • DulameaAO. The potential use of mesenchymal stem cells in stroke therapy–From bench to bedside. J Neurol Sci. 2015;352(1–2):1–11.25818674
  • SuYR, WangJ, WuJJ, ChenY, JiangYP. Overexpression of lentivirus-mediated glial cell line-derived neurotrophic factor in bone marrow stromal cells and its neuroprotection for the PC12 cells damaged by lactacystin. Neurosci Bull. 2007;23(2):67–74.17592528
  • LundbergJ, JussingE, LiuZ, et al. Safety of intra-arterial injection with tumor-activated T cells to the rabbit brain evaluated by MRI and SPECT/CT. Cell Transplant. 2017;26(2):283–292.27725029
  • BhatiaV, GuptaV, KhuranaD, SharmaRR, KhandelwalN. Randomized assessment of the safety and efficacy of intra-arterial infusion of autologous stem cells in subacute ischemic stroke. AJNR. 2018;39(5):899–904.29545253
  • FengCJ, PerngCK, LinCH, TsaiCH, HuangPH, MaH. Intra-arterial injection of human adipose-derived stem cells improves viability of the random component of axial skin flaps in nude mice. J Plast Reconstr Aesthet Surg. 2020;73(3):598–607.
  • Di SpignaG, IannoneM, LadoganaP, et al. Human cardiac multipotent adult stem cells in 3D matrix: new approach of tissue engineering in cardiac regeneration post-infarction. J Biol Regul Homeost Agents. 2017;31(4):911–921.29254293
  • TangYL, TangY, ZhangYC, QianK, ShenL, PhillipsMI. Improved graft mesenchymal stem cell survival in ischemic heart with a hypoxia-regulated heme oxygenase-1 vector. J Am Coll Cardiol. 2005;46(7):1339–1350.16198853
  • LiuN, ZhangY, FanL, et al. Effects of transplantation with bone marrow-derived mesenchymal stem cells modified by Survivin on experimental stroke in rats. J Transl Med. 2011;9:105.21733181
  • LiW, MaN, OngLL, et al. Bcl-2 engineered MSCs inhibited apoptosis and improved heart function. Stem Cells. 2007;25(8):2118–2127.17478584
  • MangiAA, NoiseuxN, KongDL, et al. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat Med. 2003;9(9):1195–1201.12910262
  • GnecchiM, HeH, NoiseuxN, et al. Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J. 2006;20(6):661–669.16581974
  • GalloS, SalaV, GattiS, CrepaldiT. Cellular and molecular mechanisms of HGF/Met in the cardiovascular system. Clin Sci (Lond). 2015;129(12):1173–1193.26561593
  • ForteG, MinieriM, CossaP, et al. Hepatocyte growth factor effects on mesenchymal stem cells: proliferation, migration, and differentiation. Stem Cells. 2006;24(1):23–33. doi:10.1634/stemcells.2004-017616100005
  • LuF, ZhaoX, WuJ, et al. MSCs transfected with hepatocyte growth factor or vascular endothelial growth factor improve cardiac function in the infarcted porcine heart by increasing angiogenesis and reducing fibrosis. Int J Cardiol. 2013;167(6):2524–2532. doi:10.1016/j.ijcard.2012.06.05222981278
  • Gomez-MauricioG, MoscosoI, Martin-CanchoMF, et al. Combined administration of mesenchymal stem cells overexpressing IGF-1 and HGF enhances neovascularization but moderately improves cardiac regeneration in a porcine model. Stem Cell Res Ther. 2016;7(1):94. doi:10.1186/s13287-016-0350-z27423905
  • YangZJ, MaDC, WangW, et al. Experimental study of bone marrow-derived mesenchymal stem cells combined with hepatocyte growth factor transplantation via noninfarct-relative artery in acute myocardial infarction. Gene Ther. 2006;13(22):1564–1568. doi:10.1038/sj.gt.330282016810195
  • ZhangJ, WangLL, DuW, et al. Hepatocyte growth factor modification enhances the anti-arrhythmic properties of human bone marrow-derived mesenchymal stem cells. PLoS One. 2014;9(10):e111246. doi:10.1371/journal.pone.011124625360679
  • LaddhaAP, KulkarniYA. VEGF and FGF-2: promising targets for the treatment of respiratory disorders. Respir Med. 2019;156:33–46. doi:10.1016/j.rmed.2019.08.00331421589
  • GiriTK, AlexanderA, AgrawalM, SarafS, SarafS. Ajazuddin. Current status of stem cell therapies in tissue repair and regeneration. Curr Stem Cell Res Ther. 2019;14(2):117–126. doi:10.2174/1574888X1366618050210383129732992
  • KongF, ShiX, XiaoF, et al. Transplantation of HGF modified dental pulp stem cells prevents bone loss in the early phase of ovariectomy-induced osteoporosis. Hum Gene Ther. 2018;29(2):271–282. doi:10.1089/hum.2017.09128950723
  • ZhangJ, ChenJ. Bone tissue regeneration - application of mesenchymal stem cells and cellular and molecular mechanisms. Curr Stem Cell Res Ther. 2017;12(5):357–364. doi:10.2174/1574888X1166616092112155527658728
  • PacelliS, BasuS, WhitlowJ, et al. Strategies to develop endogenous stem cell-recruiting bioactive materials for tissue repair and regeneration. Adv Drug Deliv Rev. 2017;120:50–70. doi:10.1016/j.addr.2017.07.01128734899
  • BrungerJM, HuynhNP, GuentherCM, et al. Scaffold-mediated lentiviral transduction for functional tissue engineering of cartilage. Proc Natl Acad Sci U S A. 2014;111(9):E798–E806. doi:10.1073/pnas.132174411124550481
  • WangT, LiaoT, WangH, DengW, YuD. Transplantation of bone marrow stromal cells overexpressing human vascular endothelial growth factor 165 enhances tissue repair in a rat model of radiation-induced injury. Chin Med J (Engl). 2014;127(6):1093–1099.24622441
  • HuS, ChenY, LiL, et al. Effects of adenovirus-mediated delivery of the human hepatocyte growth factor gene in experimental radiation-induced heart disease. Int J Radiat Oncol Biol Phys. 2009;75(5):1537–1544. doi:10.1016/j.ijrobp.2009.07.169719931736
  • LiQ, SunH, XiaoF, et al. Protection against radiation-induced hematopoietic damage in bone marrow by hepatocyte growth factor gene transfer. Int J Radiat Biol. 2014;90(1):36–44. doi:10.3109/09553002.2014.84729424059647
  • ZhangJ, ZhouS, ZhouY, et al. Hepatocyte growth factor gene-modified adipose-derived mesenchymal stem cells ameliorate radiation induced liver damage in a rat model. PLoS One. 2014;9(12):e114670. doi:10.1371/journal.pone.011467025501583
  • ChenS, ChenX, WuX, et al. Hepatocyte growth factor-modified mesenchymal stem cells improve ischemia/reperfusion-induced acute lung injury in rats. Gene Ther. 2017;24(1):3–11. doi:10.1038/gt.2016.6427556817
  • ShaoY, ShenJ, ZhouF, HeD. Mesenchymal stem cells overexpressing Ang1 attenuates phosgene-induced acute lung injury in rats. Inhal Toxicol. 2018;30(7–8):313–320. doi:10.1080/08958378.2018.152148330395743
  • SageEK, ThakrarRM, JanesSM. Genetically modified mesenchymal stromal cells in cancer therapy. Cytotherapy. 2016;18(11):1435–1445. doi:10.1016/j.jcyt.2016.09.00327745603
  • WeissDJ, CasaburiR, FlanneryR, LeRoux-WilliamsM, TashkinDP. A placebo-controlled, randomized trial of mesenchymal stem cells in COPD. Chest. 2013;143(6):1590–1598. doi:10.1378/chest.12-209423172272
  • WilsonJG, LiuKD, ZhuoH, et al. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Respir Med. 2015;3(1):24–32. doi:10.1016/S2213-2600(14)70291-725529339
  • RipaRS, WangY, JørgensenE, JohnsenHE, HesseB, KastrupJ. Intramyocardial injection of vascular endothelial growth factor-A165 plasmid followed by granulocyte-colony stimulating factor to induce angiogenesis in patients with severe chronic ischaemic heart disease. European Heart Journal. 2006;27(15):1785–1792.16825290
  • WheelockV, TempkinT, DuffyA, et al. PRE-CELL: clinical and novel biomarker measures of disease progression in a lead-in-observational study for a planned phase 1 trial of genetically modified mesenchymal stem cells over-expressing BDNF in patients with Huntington’s disease (S25.004) Neurology. 2016;86(16 Supplement):S25.004.
  • DengP, TorrestA, PollockK, et al. Clinical trial perspective for adult and juvenile Huntington’s disease using genetically-engineered mesenchymal stem cells. Neural Regen Res. 2016;11(05):702–705. doi:10.4103/1673-5374.18268227335539
  • DaleyGQ, HyunI, ApperleyJF, et al. Setting global standards for stem cell research and clinical translation: the 2016 ISSCR guidelines. Stem Cell Reports. 2016;6(6):787–797. doi:10.1016/j.stemcr.2016.05.00127185282
  • Munoz RuizM, RegueiroJR. New tools in regenerative medicine: gene therapy. Adv Exp Med Biol. 2012;741:254–275.22457115
  • PoulosJ. The limited application of stem cells in medicine: a review. Stem Cell Res Ther. 2018;9(1):1. doi:10.1186/s13287-017-0735-729291747
  • ImamuraA, KajiyaH, FujisakiS, et al. Three-dimensional spheroids of mesenchymal stem/stromal cells promote osteogenesis by activating stemness and Wnt/beta-catenin. Biochem Biophys Res Commun. 2020;523(2):458–464. doi:10.1016/j.bbrc.2019.12.06631882121
  • KingNM, PerrinJ. Ethical issues in stem cell research and therapy. Stem Cell Res Ther. 2014;5(4):85. doi:10.1186/scrt47425157428