Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Stem Cells. Apr 26, 2014; 6(2): 179-194
Published online Apr 26, 2014. doi: 10.4252/wjsc.v6.i2.179
Neurotrauma and mesenchymal stem cells treatment: From experimental studies to clinical trials
Ana Maria Blanco Martinez, Camila de Oliveira Goulart, Bruna dos Santos Ramalho, Júlia Teixeira Oliveira, Fernanda Martins Almeida, Laboratory of Neurodegeneration and Repair, Institute of Biomedical Sciences, Health Science Center, 21941-902, Rio de Janeiro, Brazil
Ana Maria Blanco Martinez, Camila de Oliveira Goulart, Bruna dos Santos Ramalho, Fernanda Martins Almeida, Pathology Department, Faculty of Medicine, Federal University of Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
Fernanda Martins Almeida, Federal University of Rio de Janeiro, Campus Macaé, 27930-560, Rio de Janeiro, Brazil
Author contributions: Martinez AMB and Almeida FM conceived and designed the manuscript; all authors contributed equally to the acquisition and analysis of data and the manuscript writing; Martinez AMB and Almeida FM revised and approved the final version of the manuscript.
Correspondence to: Ana Maria Blanco Martinez, MD, PhD, Laboratory of Neurodegeneration and Repair, Institute of Biomedical Sciences, Health Science Center, bloco F - sala 12, 21941-902, Rio de Janeiro, Brazil. martinez@histo.ufrj.br
Telephone: +55-21-25626431 Fax: +55-21-25626431
Received: October 29, 2013
Revised: February 26, 2014
Accepted: March 11, 2014
Published online: April 26, 2014

Abstract

Mesenchymal stem cell (MSC) therapy has attracted the attention of scientists and clinicians around the world. Basic and pre-clinical experimental studies have highlighted the positive effects of MSC treatment after spinal cord and peripheral nerve injury. These effects are believed to be due to their ability to differentiate into other cell lineages, modulate inflammatory and immunomodulatory responses, reduce cell apoptosis, secrete several neurotrophic factors and respond to tissue injury, among others. There are many pre-clinical studies on MSC treatment for spinal cord injury (SCI) and peripheral nerve injuries. However, the same is not true for clinical trials, particularly those concerned with nerve trauma, indicating the necessity of more well-constructed studies showing the benefits that cell therapy can provide for individuals suffering the consequences of nerve lesions. As for clinical trials for SCI treatment the results obtained so far are not as beneficial as those described in experimental studies. For these reasons basic and pre-clinical studies dealing with MSC therapy should emphasize the standardization of protocols that could be translated to the clinical set with consistent and positive outcomes. This review is based on pre-clinical studies and clinical trials available in the literature from 2010 until now. At the time of writing this article there were 43 and 36 pre-clinical and 19 and 1 clinical trials on injured spinal cord and peripheral nerves, respectively.

Key Words: Neurotrauma, Stem cell therapy, Mesenchymal stem cells, Pre-clinical studies, Clinical trials

Core tip: Basic and pre-clinical studies have highlighted the positive effects of mesenchymal stem cell (MSC) treatment after spinal cord injury (SCI) and nerve trauma. There are many pre-clinical studies on MSC treatment for SCI and nerve injuries. However, the same is not true for clinical trials, particularly those concerned with nerve trauma. As for clinical trials for SCI, the results obtained so far are not as beneficial as those described in experimental studies. For these reasons basic and pre-clinical studies dealing with MSC therapy should emphasize the standardization of protocols that could be translated to the clinical set with consistent and positive outcomes.



SPINAL CORD LESION: MECHANISMS OF DEGENERATION AND REGENERATION

Spinal cord injury (SCI) causes motor and sensory deficits that impair functional performance, and significantly impacts expectancy and quality of life of affected individuals. The estimated annual global incidence of SCI is 15-40 cases per million inhabitants[1]. In addition to the sensory and functional deficits, spinal cord injury also causes great economic impact on the whole society and it is estimated that this impact is greater than 4 billion dollars per year[2].

SCI results from primary and secondary injury mechanisms. Primary injury refers to the immediate physical injury to the spinal cord as a consequence of laceration, contusion, compression, and contraction of the neural tissue[3]. Pathological changes resulting from primary injury mechanisms include severed axons, direct mechanical damage to cells, and ruptured blood vessels. Secondary injury is responsible for the expansion of the injury site which, in turn, limits the restorative process[4,5]. Specific secondary sequel include alterations in local ionic concentrations, loss of regulation of local and systemic blood pressure, reduced spinal cord blood flow, breakdown of the blood-brain barrier, penetration of serum proteins into the spinal cord, inflammatory responses (alterations in chemokines and cytokines), apoptosis, excitotoxicity, calpain proteases activation, neurotransmitter accumulation, production of free radicals/lipid peroxidation, and imbalance of activated metalloproteinases. These changes lead to demyelination, ischemia, necrosis, and apoptosis of spinal cord parenchyma[5]. These intrinsic responses to tissue injury contribute to an environment that is inhibitory to axonal regrowth[6]. As a consequence of these negative influences when axons in the central nervous system (CNS) are damaged they mount a poor regenerative response.

An injury in the central nervous system generally leads to transection of some nerve fibers as well as damage to the surrounding tissues. The distal ends of the damaged axons form dystrophic growth cones that are exposed to a glial hostile microenvironment. During the initial phase of lesion, inhibitors associated with intact myelin oligodendrocyte and myelin debris, such as NOGO (no go), MAG (myelin associated glycoprotein) and OMGp (oligodendrocyte myelin glycoprotein) proteins can restrict axonal growth[7]. In addition, the recruitment of inflammatory cells and astrocytes, in an attempt to restore the blood-brain barrier, leads to the formation of glial scar, which is usually accompanied by cavities filled with astrocytes secreted substances, such as chondroitin sulfate proteoglycans, which also acts as axon growth inhibitory molecules[8,9]. Furthermore, there is also a lack of trophic factors in the lesion milieu due to intrinsic changes in neurons such as atrophy and death after axonal injury. Together, all these inhibitory molecules form a glial microenvironment which is hostile to axonal repair[2,4,10,11].

Although effective treatments for SCI remain limited, there have been many studies in recent years that have promised for the future from a clinical translational perspective. In general, basic science, preclinical, and clinical studies are aimed at overcoming the factors that are involved in impeding recovery from SCI. Current research is aimed at preventing secondary injury, promoting regeneration, and replacing destroyed spinal cord tissue. In particular, a variety of therapies have been addressed to alter neuro-inflammation[12-14], reduce free radical damage[15-17], reduce excitotoxic damage to neurons[18,19], improve blood flow[20,21], and counteract the effects of local ionic changes[20,22-25]. Current experimental studies and the knowledge of clinical situations provide us with a better understanding of the complex interaction of the pathophysiologic events after SCI. Future approaches should involve strategies aimed at blocking the multiple mechanisms of progressive pathogenesis in SCI and therefore promoting neuroregeneration.

Methylprednisolone (MP), a glucocorticoid, is the only current pharmacotherapy approved for SCI in the human clinic. Although therapy with methylprednisolone has been shown to be protective, its efficacy is limited and it only marginally improves outcomes[14]. Recent advances in SCI research have led to a variety of novel experimental therapeutic strategies. The approach based on cell therapy using various lineages of stem cells has been considered as the most potential for the treatment of spinal cord injuries[26]. Cell transplantation after spinal cord injury has several goals, among them, filling the cavity of the lesion to make a bridge that joins the edges of conserved areas, restore dead cells (neurons or myelinating cells) and make a favorable environment for axonal regeneration. Our laboratory employed in vivo experiments using predifferentiated embryonic stem cells[27], human dental pulp stem cells[28], and mesenchymal stem cell (MSC) (data not published) as a therapy for compressive spinal cord injury in mice, and our results show that these treatments lead to positive and similar functional and morphological responses. Among these lineages, mesenchymal stem cells have strengths such as easy extraction and cultivation, and do not involve ethical and moral issues, making them one of the favorite lineages for spinal cord injury treatment.

MSC THERAPY FOR SPINAL CORD LESION: FROM EXPERIMENTAL STUDIES TO CLINICAL TRIALS

MSC transplantation has been extensively investigated by several groups and these cells can be considered a feasible candidate for treatment of central nervous system diseases because they have characteristics that address the multifactorial events that occur after SCI. These cells have anti-inflammatory, immunomodulatory[29] and neuroprotective[30] effects. It has also been shown that MSC can secrete trophic factors thus exerting a paracrine effect that can stimulate axon regeneration contributing to functional recovery enhancement.

Concerning the paracrine effect, some groups have identified the ability of these cells in secreting pro-survival factor such as insulin-like growth factor (IGF) brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), granulocyte-macrophage colony stimulating factor (GMCSF), fibroblast growth factor-2 (FGF2) and transforming growth factor beta (TGF-β)[31-33]. In addition, MSC can be combined with gene therapy, by introducing genes to generate molecules with great therapeutic potential in promoting neuron survival and regeneration[34]. Table 1 is a summary of preclinical studies using MSC for spinal cord injury, from 2010 until now.

Table 1 Summary of pre-clinical studies using mesenchymal stem cell for spinal cord injury.
AnimalLesion typeCells sourceRoute of administrationEffects on CNS regenerationRef.
RatContusionHuman mesenchymal precursor cellsLesion siteImprovement in functional recovery and tissue sparing and reduction of cyst volume[35]
RatContusionHuman bone marrow-MSCLesion site , intracisternal, intravenousImprovement in functional recovery[36]
RatHemisectionBone marrow-MSC induced into Schwann CellsLesion siteImprovement in locomotor and sensory scores, axonal regeneration and remyelination[37]
RatContusionBone marrow-MSCLesion site , intravenousImprovement in locomotor scores and NGF expression[38-40]
RatTransection to the dorsal columns and tractsBone marrow-MSC, adipose derived-MSCLesion siteImprovement in locomotor scores, increased angiogenesis, preserved axons, decreased numbers of ED1-positive macrophages and reduced lesion cavity formation[41]
RatHemisectionHuman umbilical cord-derived MSCLesion siteSuppress mechanical allodynia, and this effect seems to be closely associated with the modulation of spinal cord microglia activity and NR1 phosphorylation[42,43]
RatHemisectionHuman bone marrow-MSCLesion siteImprovement in locomotor scores, shorter latency of somatosensory-evoked potentials and differentiation into various cells types[44]
RatHemisectionBone marrow-MSCLesion siteImprovement in locomotor scores and reduced lesion cavity formation[45]
MouseCompressionBone marrow-MSCLesion siteImprovement in locomotor and sensory scores and reduced lesion volume[46]
RatContusionHuman bone marrow-MSCLesion siteImprovement in functional recovery, tissue sparing and reduction in the volume of lesion cavity and in the white matter loss[35,47-49]
RatContusionHuman umbilical cord-MSCLesion siteImprovement in functional recovery, reduction of the extent of astrocytic activation and increased axonal preservation[50]
DogCompressionBone marrow, adipose, Wharton’s jelly, umbilical cord derived-MSCLesion siteImprovement in functional recovery, increased numbers of surviving neurons, smaller lesion sizes and fewer microglia and reactive astrocytes in the epicenter of lesion[51]
RatCompressionBone marrow-MSCIntravenousImprovement in functional recovery, increase of NGF expression, higher tissue sparing and density of blood vessels[52]
RatContusionHuman umbilical cord-MSCLesion siteImprovement in functional recovery, endogenous cell proliferation and oligogenesis, and smaller cavity volume[53,54]
RatTransectionHuman-MSCLesion siteImprovement in functional recovery, increased amplitude of motor-evoked potentials, differentiation into neural cells[55,56]
RatContusionBone marrow-MSCLesion siteImprovement in functional recovery, preservation of axons, less scar tissue formation and increase in myelin sparing; higher levels of IL-4 and IL-3 and higher numbers of M2 macrophages, and reduction in TNF-α and IL-6 levels, and in numbers of M1 macrophages[57-60]
DogCompressionNeural-induced adipose derived-MSCLesion siteImprovement in functional recovery and neuronal regeneration, and reduction of fibrosis[61]
MouseTransectionBone marrow-MSCLesion siteImprovement in functional recovery and neuronal survival, reduction of cavity volume and attenuation of inflammation, promotion of angiogenesis and reduction of cavity formation[62-64]
RatCompressionBone marrow-MSCLesion siteImprovement in functional recovery, up-regulation of VEGF mRNA expression, increase in angiogenesis and prevention of tissue atrophy[65-67]
RatCompressionHuman umbilical cord-MSCLesion siteImprovement in functional recovery, increase in the intensity of 5-HT fibers and in the volume of spared myelination; decrease in the area of the cystic cavity[68]
DogCompressionUmbilical cord-MSCLesion siteImprovement in functional recovery, promotion of neuronal regeneration and reduction of fibrosis[69]
DogCompressionHuman umbilical cord-MSCLesion siteImprovement in functional recovery and remyelination[70]
RatContusionBone marrow-MSCIntrathecalImprovement in functional recovery[71]
RatContusionHuman bone marrow-MSCLesion site , lumbar punctureImprovement in functional and sensory recovery[72]
RatContusionNeural differentiated and undifferentiated MSCLesion siteImprovement in functional recovery and reduction of cavitation[73]
Sources of MSC

MSC reside in a range of adult tissues that are easily accessible such as bone marrow, adipose tissue, skin, and even peripheral blood[34]. Most of the studies in SCI use MSC derived from bone marrow and adipose tissue, but it is also possible to get MSC from a perinatal source like umbilical cord blood, umbilical cord matrix[74], amniotic fluid and placenta[75-77]. MSC can be extracted from these tissues and plated to be used in autologous transplantation, minimizing the rejection risk.

Studies using MSC extracted from bone marrow in rodents have demonstrated a beneficial effect of cell transplantation after SCI. The beneficial effect of MSC is usually attributed to secretion of neurotrophic factors[78,79] and anti-inflammatory cytokines[71,80,81]. Studies performed with pigs[82] and monkeys[83] showed that MSC can promote axonal growth and sprouting, corroborating the previous results in rodents, thus supporting the clinical use of MSC.

MSC extracted from adipose tissue is considered an attractive source of cells due to easiness of isolation, obtention of a large amount of cells per donor, and also due to the fact that this tissue is usually discarded after liposuctions. In SCI models, treatment with these cells have resulted in cell survival, neuroprotection, attenuation of secondary damage, axonal regeneration, decrease of gliosis, angiogenesis and enhanced functional recovery[61,84-90]. A comparative study using MSC extracted from both bone marrow and adipose tissue after SCI found that both sources of MSC expressed similar surface protein markers, but animals that received adipose tissue cells presented higher levels of tissue BDNF, increased angiogenesis, higher number of preserved axons and a decrease in the number of macrophages, suggesting that the use of MSC extracted from adipose tissue is a better candidate for SCI treatment[41]. However, this is not a consensus and should be further investigated because in another comparative study published in 2012, the authors did not find any difference between animals that received MSC derived from bone marrow or adipose tissue, in terms of axonal regeneration, neuroprotection and functional recovery after a compression lesion in dogs[51].

Despite being less investigated in terms of SCI treatment, MSC extracted from perinatal tissues also present a therapeutic potential. Human umbilical cord blood cells (hUCBC) transplantation in rats submitted to an injury, resulted in differentiation of these cells into neural cells and downregulation of the fas/caspase-3 pathway in neurons and oligodendrocytes, and also increased levels of anti-apoptotic proteins[91,92].

The umbilical cord matrix, also known as Wharton’s jelly, possesses a stem cell population that present some advantages in comparison to other sources because they can proliferate more rapidly and extensively than adult MSC[76,93] and also because they are easily obtained after normal and cesarean births, with low risk of viral contamination[94,95]. Other advantage is the possibility of using them for allogenic transplantation because they act by suppressing immune response and are, therefore, considered non-immunogenic cells[96]. Some studies using umbilical cord matrix-derived MSC indicated that these cells can survive in the injury site and promote repair and recovery after SCI. This improvement is attributed to immunomodulatory and trophic effects through secretion of glial-derived neurotrophic factor (GDNF), BDNF and nerve growth factor (NGF) which are known as supporters of cell survival and regeneration[54,97].

The amniotic fluid cells constitute another source of MSC, which are obtained from discarded post-partum tissue, without any ethical objections about their use. They present similar proliferation and differentiation patterns in comparison to adult MSC[98,99]. According to few studies, these cells are able to enhance cell survival and axon myelination and improve hind limb function, after transplantation in SCI models[100]. Some studies have also demonstrated the immunomodulatory effect and trophic support provided by these cells after SCI[101,102].

Issues regarding the quantity and best via of administration of MSC for SCI

Two important questions that should be addressed when we discuss MSC and its efficacy in treating central nervous system disorders are: the ideal quantity of cells and the best administration via Concerning the cell quantity, the literature presents several studies using different amount of cells. In terms of cell administration, most transplantation is delivered directly into the injury site or adjacent to it, by injecting few microliters of cell suspension[103]. Attempts have been made to inject cells intravenously or intraperitoneally in order to decrease tissue damage and, thus, avoiding subjecting the individual to another surgical intervention.

There are several studies that injected different quantity of cells with similar results. Apart from the difference on the quantity of cells, there are other points that make the comparison among these studies difficult, such as the diversity of lesion models, animal types and route of cell administration. For example, Cizkova and colleagues[104] demonstrated cell survival and enhancement in locomotor performance after MSC transplantation delivered by intravenous injection (one million cells in a volume of 0.5 mL of DMEM) in a model of balloon compressive injury in rats, while Sheth et al[105] performed cell transplantation (600000 cells in a volume of 6 μL) directly into the injury site after contusive injury in rats, and also observed an enhancement in locomotor function and a decrease in the lesion volume, indicating a neuroprotective effect of these cells. Thus, it is still difficult to determine the ideal quantity of cells and the best via for stem cell transplantation after SCI. The questions that arise from these studies are: Is there a minimum number of transplanted cells that can be used and yet giving the best results in terms of functional recovery? Can we get similar results with cells injected systemically in comparison to local injection? Studies using the same type of lesion and different amount of cells and administration via should be further undertaken in order to better clarify this issue.

Time point for cell transplantation

Other crucial issue that should be further addressed here is the time point for cell transplantation after lesion. This is important because the environment created after SCI is hostile for regeneration and can negatively influence cell survival and differentiation. Thus, depending on the time that the treatment is performed the results can be completely different. Most studies have been performed in acute or sub-acute phases, which means immediately or 1-2 wk after injury, respectively[35,103]. There are fewer studies in the SCI chronic phase, when cells are delivered in later stages, when the glial scar is already present[38,41].

Clinical trials

The clinical trials conducted for SCI comprise three different phases with human participation in all phases. The phase 1 trial begins with the administration of the cell transplants to a human subject with the aim to investigate the presence of adverse or toxic effects and treatment safety. People who participate in these trials may experience some risks and have limited benefits. In phase 2, the objective is to determine the potential and variability of a therapy in comparison with a control group. The participants are usually recruited and randomly assigned to the groups (experimental or control) and both, participants and investigators, do not know to which study they have been assigned to. The phase 3 clinical trials are usually the definitive clinical trial. The aim is to confirm the preliminary results obtained at the phase 2, with a statistically significant clinical benefit of the therapeutic intervention. The number of subjects is also larger and multiple study centers are involved[106,107]. The majority of the studies using MSC transplantation after spinal cord injury are in phase 1 or 2.

At the time of writing this article there were twenty clinical trials being either completed, ongoing or in the recruitment stage, using either adult or perinatal sources of mesenchymal stem cells in different phases of the disease, and most of them use autologous transplantation to minimize the risk of rejection. Table 2 list the clinical trials listed on the clinical trials.gov.

Table 2 Summary of clinical trials studies using mesenchymal stem cell for spinal cord injury.
TitleLesion typeCells sourcePhase of the studyStatusEffects on CNS regeneration
Clinical study of treatment for acute SCI using cultured bone marrow stromal cellsCervical SCIAutologous Bone marrow-MSCTerminated1/2Rapid and remarkable recovery of ASIA B and C patients, but gradual or limited in ASIA A patients.
Autologous mesenchymal stem cell in SCI patientsComplete cervical or thoracic SCIAutologous bone marrow-MSCEnrolling by invitation2Not informed
Different efficacy between rehabilitation therapy and umbilical cord derived MSCs transplantation in patients with chronic SCI in ChinaTraumatic SCIUmbilical cord derived-MSCRecruiting3Not informed
A phase III/IV clinical trial to evaluate the safety and efficacy of bone marrow-derived MSC transplantation in patients with chronic SCICervical SCIAutologous bone marrow-MSCRecruiting1/2Not informed
Phase I/II trial of autologous bone marrow derived MSCs to patients with SCISafety of autologous adipose derived MSCs in patients with SCITraumatic thoracic or lumbar SCIClinical diagnosis of SCI (ASIA A to C)Autologous bone marrow-MSCAutologous Adipose derived-MSCRecruitingCompleted1/21Not informed
The effect of intrathecal transplantation of autologous adipose tissue derived MSCs in the patients with SCI, phase I clinical studyClinical diagnosis of SCIAutologous Adipose derived-MSCRecruiting1Not informed
Phase I, single center, trial to assess safety and tolerability of the intrathecal infusion of ex-vivo expanded bone-marrow derived MSCs for the treatment of SCIClinical diagnosis of SCI (ASIA A)Autologous bone marrow-MSCActive, not recruiting1Not informed
Study the safety and efficacy of bone marrow derived autologous cells for the treatment of SCIClinical diagnosis of SCIAutologous bone marrow-MSCRecruiting1/2Not informed
Surgical transplantation of autologous bone marrow stem cells with glial scar resection for patients of chronic SCI and intra-thecal injection for acute and subacute injury-a preliminary studyComplete spinal cord trans-sectionAutologous bone marrow-MSCCompleted1/2Not informed
To study the safety and efficacy of autologous bone marrow stem cells in patients with SCISCI below C5 (ASIA A to C)Autologous bone marrow-MSCRecruiting1/2Not informed
Safety of autologous stem cell treatment for SCI in childrenClinical diagnosis of SCI (ASIA A to D)Bone marrow-MSCRecruiting1Not informed
Autologous bone marrow derived cell transplant in SCI patientsTraumatic SCIAutologous bone marrow-MSCCompleted1/2Not informed
Phase 1 study of autologous bone marrow stem cell transplantation in patients with SCITraumatic thoracic or lumbar SCIAutologous bone marrow-MSCNot informed1Not informed
Phase I pilot study to evaluate the security of local Administration of autologous stem cells obtained from the bone marrow stroma, in traumatic injuries of the spinal cordTraumatic SCI between C3 and L1Autologous bone marrow-MSCRecruiting1Not informed
Feasibility and safety of umbilical cord blood cell transplant into injured spinal cord: an open-labeled, dose-escalating clinical trialChronic SCI between C5 and T11 (ASIA A)Umbilical cord blood mononuclear derived-MSCActive, not recruiting1/2Not informed
Efficacy difference between rehabilitation therapy and umbilical Cord derived transplantation in patients with acute or chronic SCI in ChinaClinical diagnosis of SCIUmbilical cord derived-MSCNot informed2Not informed
Safety and feasibility of umbilical cord blood cell Transplant Into Injured Spinal cord: an open-labeled, dose-escalating clinical trialChronic SCI between C5 and T11 (ASIA A)Umbilical cord derived-MSCActive, not recruiting1/2Not informed
Safety and effect of lithium, umbilical cord blood cells and the combination in the treatment of acute and sub-acute spinal cord injury : a randomized, double-blinded placebo-controlled clinical trialAcute or Subacute traumatic SCI between C5 and T11 (ASIA A)Umbilical cord derived-MSCActive, not recruiting1/2Not informed

The number of clinical trials using MSCs for treatment of SCI is increasing, indicating that despite several questions that still need to be addressed at basic and pre-clinical levels, the MSC are considered potentially beneficial for translational studies.

According to PubMed database, in the last three years only three studies were published in “clinical trials” category, using MSC transplantation after SCI. One of them transplanted autologous bone marrow-derived MSC into the cerebrospinal fluid of patients with complete SCI. The authors described that 45% of the patients showed a recovery, but there was no difference between these patients and those from control groups; they emphasized that despite the fact that results were not positive, the transplantation was a feasible and safe technique, since patients did not present any adverse reaction[108]. On the other hand, Park et al[109] using the same cell source, and repeated cells injections directly into the spinal cord, demonstrated that three of ten patients presented a motor improvement, and significant magnetic resonance changes and electrophysiological results. These results are similar to those obtained by Dai et al[110] who also demonstrated a clinical improvement in patients that received autologous MSC transplantation. The results of these studies are not conclusive, and, unfortunately, not as good as those obtained in pre-clinical experiments. In spite of that, all of them emphasize mesenchymal stem cell clinical potential.

WALLERIAN DEGENERATION AND NERVE REGENERATION IN THE PERIPHERAL NERVOUS SYSTEM

Traumatic injury to nerves in the peripheral nervous system (PNS) is a large-scale problem annually affecting more than one million people worldwide. These injuries often result in pain and disabilities, owing to reduction in motor function and sensory perception. Moreover, the trauma can cause emotional, social and work-related disorders, and the affected individuals undergo a reduction in their quality of life[111,112].

While it is widely accepted that the PNS has an inherent potential for regeneration, functional recovery after a lengthy peripheral nerve injury (PNI) remains unsatisfactory[113]. After an extensive traumatic nerve injury with a large gap between the proximal and distal nerve stumps, a long period of time is required for regenerating axons to cross that gap. During that time, the ability of axotomized neurons to regenerate declines and Schwann Cells (SC) can no longer support regenerating neurons and their axons. As a result, regenerating axons fail to reach their target organs and the injury cannot be successfully repaired. In order to accelerate the rate of axonal growth many therapeutic strategies are being developed and investigated. The identification of crucial elements responsible for successful regeneration in injured peripheral nerves will be quintessential in improving regenerative outcomes after peripheral and central nerve injuries.

Nerve trauma elicits a cascade of molecular, cellular, and ultrastructural responses which are necessary for degeneration and posterior regeneration, including: disruption of axonal conduction; increase in cell body metabolism and protein synthesis; degeneration of the distal stump of injured axons; dedifferentiation and proliferation of SC; degradation of the myelin sheath; recruitment of macrophages to the site of injury[114], as well as the release of cytokines, neurotrophins and growth factors[115-117]. These events will allow rapid and efficient removal of the growth inhibitory cellular debris present in the injured peripheral nerve milieu, producing a favorable microenvironment for axonal growth[118].

After an injury the axon is divided into two segments: a proximal stump that remains in contact with the cell body, and a distal stump which is separated from the rest of the neuron. The distal nerve stump undergoes a cascade of events called “Wallerian degeneration”[119,120], which is initiated within 24 to 48 h by the entry of calcium in the axoplasm. Calcium influx activates proteases, such as calpains that promote cytoskeletal degradation and disintegration of axoplasm, myelin and axolemma[121,122]. The rupture of the blood-nerve barrier allows the entry of macrophages into the site of injury and, together with SC, these cells initiate intense phagocytosis and removal of degenerating axon and myelin debris. The barrier permeability decreases two weeks after the injury and then, in the fourth week, increases again in order to regain homeostasis after Wallerian degeneration[118].

Immediately after injury, the SC in the distal stump of the nerve begin the process of dedifferentiation. Even before axonal degeneration occurs, SC can modify its gene expression[123] and 48 h after injury, they decrease myelin protein expression, acquire a non-myelinating phenotype and begin to express genes related to regeneration, like growth associated protein 43 (GAP-43), neurotrophic factors and their receptors, neuroregulins and their receptors, and assume an intense proliferative activity[124,125]. About four days after injury SC reach their proliferation peak. These proliferative cells are confined within the tube formed by its own basal lamina and align forming the so called bands of Büngner. These bands columns will form a supportive substrate, providing clues that will guide axon growth toward the target organ, through the release of trophic factors. When SC contact the regenerating axons, the process of re-myelination is started[126].

The injury also causes a rapid arrival of signals from the damaged axons to the neuronal body resulting in an extraordinary change from a transmitting to a growth promoting phenotype. Cell body suffers a process called chromatolysis, which is characterized by swelling of the neuronal body and by dispersion of Nissl corpuscles[127,128]; These changes reflects variations in the metabolic activity of neurons which, as a result, fail to synthesize proteins required for neurotransmission, and start producing substances that are important for axonal sprouting and growth[129]. The regeneration that follows occurs via different mechanisms: the elongation of the distal end of injured axons and the growth of collateral axons from nodes of Ranvier in the proximal stump. However, the success of regeneration and target organ reinnervation depends mostly on the enhancement of the number of regenerating axons, the velocity of axon growth and on the ability of affected neurons to survive and acquire a regenerative phenotype.

In the clinical settings, reconstruction of transected peripheral nerve requires accurate microsurgical repair that connects the proximal and distal stumps of the nerve in a tension-free manner. In cases of injury with tissue loss, autologous peripheral nerve grafts, i.e., autografts, is considered by neurosurgeons the gold standard technique, but unfortunately, even in these cases, the clinical results remain disappointing and, therefore, the search for better strategies is an urgent necessity. In cases of digital nerve lesions, biodegradable artificial nerve conduits are being used in the clinical settings, but their use is still limited to these thin nerves. An advantage of the use of these conduits is that they can be combined with other pro-regenerative strategies, such as the local injection of neurotrophic factors and cells.

New therapeutic approaches should have as a goal an increase of the intrinsic regenerative capacity of transected nerve fibers and a decrease of the extrinsic factors that limit regeneration of severed nerve fibers, thus creating an appropriate environment in which, axon elongation, remyelination and proper reinnervation of target organ may occur. A stem cell-based therapy represents an important new strategy to manage peripheral nerve injury. In the next part of this review we will discuss the potential use of mesenchymal stem cells, in promoting nerve regeneration.

MSC THERAPY IN PNS: FROM EXPERIMENTAL STUDIES TO CLINICAL TRIALS

A number of experimental studies have shown the potential of MSC to improve peripheral nerve regeneration following traumatic injuries[130-135]. These cells may act on nerve regeneration mainly by paracrine, neuro/axonoprotective, or immunomodulatory effects; by transdifferentiation into SCs; by cell-to-cell contact; or even by a combination of the above mechanisms[134]. However, most of the beneficial effects exerted by the MSC are strongly correlated with the production of neutrophic substances, such as FGF, NGF, ciliary neurotrophic facto, BDNF, GDNF among others[132,133,136,137].

Our group showed the presence of high levels of NGF-b in the in MSC in vitro suggesting that they are also able to express this potent neurotrophic factor in vivo; this result could represent one mean by which these cells acted on the enhancement of axon regeneration and remyelination, consequently contributing to the observed return of motor function[133]. In agreement with these findings, bone marrow-MSC locally injected in the mouse ischiatic nerve resulted in improvement of regeneration of sensory and motor axons[134]. Because these authors also observed that these cells were capable of increasing neurite outgrowth in vitro through NGF releasing, and that they presented low potential to differentiate into SC in vivo, they suggested that the beneficial effects exerted by the implanted cells were mainly dependent on their trophic activity rather than their stemness potential[134]. In another work, our group also observed the benefits of bone marrow-MSC locally injected in the mouse median nerve following transection and conduit repair. This cell system was capable of increasing the number of both myelinated and unmyelinated fibers, preventing the muscle atrophy and, most importantly, improving functional performance[130].

It is also possible that MSC can act indirectly on nerve regeneration by modulating cellular behaviors such as inducing SC to survive, proliferate, produce neurotrophic factors and promote remyelination. A coculture system with rat bone marrow-MSC conditioned media and SC demonstrated cell-cell interactions despite no direct contact between the two population of cells. MSC not only favored survival and proliferation of SC but also induced them to express NGF, BDNF and NGF receptors[138]. This is an important MSC feature as it might indicates that MSC can relay and magnify neurotrophic function from stem cells to glia cells, thus improving peripheral nerve regeneration.

Besides rodents, larger animal models have also been used to investigate the effects of MSC-based therapy on more challenging nerve gaps. Few authors have shown the successful bridging of a 30 mm-long ischiatic nerve defect by means of a biodegradable conduit in dogs[139]. After six months of MSC implantation, they observed the reconstruction of ischiatic nerve trunk with restoration of nerve continuity, functional recovery for conducing electrical impulses and transporting materials, and muscle re-innervation, which lead to improvement of locomotion activities. Even more challenging, using a two-fold nerve gap in a similar experimental model but with addition of autologous MSC, the same group[140] demonstrated that the cellular treatment improved nerve regeneration and functional recovery in a manner comparable to the autograft-treated animals, which is considered by neurosurgeons the current gold standard for peripheral nerve repair.

As aforementioned, the great majority of the experimental studies of mesenchymal stem cell-based therapy on the peripheral nerve regeneration use rodents (mainly mice and rats) as animal models[130,133,134,138], perhaps because they are small size mammals and, consequently, easy to handle; also, they have been extensively used in the field of genetic engineering for a diversity of experimental trials of gain and loss of function as well as reporter assays. However, there are few studies using non-human primates such as cynomolgus and rhesus monkeys, which share high level of sequence homology with human genome, that have confirmed the feasibility of this cell system for improving nerve regeneration after severe nerve lesions. MSC transplantation into either allogeneic nerve grafts[141] or artificial conduits[142] for bridging severe upper extremity nerve defects in higher primates yielded structurally and functionally regenerated nerves; these studies proved to be safe and effective, thus giving great insight into the use of MSC in human clinics.

MSC obtained from human subjects have also been used in pre-clinical studies for promoting nerve regeneration, yielding promising results[143-145]. These studies are of great relevance because they address human MSC properties, clarifying their mechanisms of action, and also provide insight into their effects on peripheral nervous tissue recovery. Interestingly, the authors of these studies demonstrated that human MSC-based therapy improved peripheral nerve regeneration as well as functional recovery. However, McGrath et al[145] showed that MSC survived in the conduit and enhanced axonal regeneration only when transplantation was combined with the immunosuppressive treatment, cyclosporine A. As these results provide evidence of the nerve regeneration potential of human MSC, and taking into account that one of the great advantages of MSC is the possibility of auto transplantation without donor-site morbidity, they might encourage the use of this cell system for treating human peripheral nerve trauma.

Thus, the results of pre-clinical studies highlighting the improved outcomes yielded by using MSC with the aim to repair a large nerve gap may increase the feasibility of translation of MSC-based therapy to clinical trials for peripheral nerve applications.

Table 3 summarizes the studies using MSC for nerve injuries, either in pre-clinical or clinical trials, since 2010 until now. To date, only one clinical trial has used autologous bone marrow mononuclear cells within silicone tubes to repair human median or ulnar nerves[146]. In this study scores for motor function, sensation and the effect of pain on function were better than those obtained from individuals that had the tubular nerve repair only; However, a possible limitation in this study is the fact that there was a difference between groups regarding the age of individuals and the length of follow-up after treatment, which could represent biases in this study. So, the interval between injury and treatment was always longer than 75 d, which could possibly limit the positive effects exerted by the cells on the nerve regeneration process. Another possible disadvantage of this work is that nerve conduits were made of silicone, a non-biodegradable material, thus requiring a second surgery to remove the conduit. In spite of these limitations cells-treated patients presented a better recovery compared to the untreated. The results of this study will, hopefully, encourage subsequent clinical studies to be conducted safely, with fewer biases, and with the association of the cellular treatment with suitable biodegradable conduits, thus preventing discomfort and complications generated from the use of silicone material.

Table 3 Summary of pre-clinical and clinical studies using mesenchymal stem cell for peripheral nerve injury.
AnimalNerveLesion typeCells sourceRoute of administrationRef.
MouseMedianTransectionBone marrow-MSCLocal[147]
MouseIschiaticTransectionBone marrow-MSCLocal[148]
MouseIschiaticTransectionEmbryonic stem cell derived-MSCLocal[144]
MouseIschiaticCrushAdipose derived-MSCIntravenous[149]
MouseIschiaticTransectionBone marrow-MSCLocal[131]
RatIschiaticCrushAmniotic fluid derived-MSCLocal[150]
RatIschiaticCrushAmniotic fluid derived-MSCIntravenous[151-153]
RatFacialTransectionBone marrow-MSCLocal[154,155]
RatIschiaticCrushBone marrow-MSCLocal[156]
RatIschiaticTransectionAdipose derived-MSCLocal[157-159]
RatIschiaticTransectionUmbilical cord derived-MSCLocal[160,161]
RatIschiaticTransectionBone marrow-MSCLocal[145,158,162-169]
RatCavernousTractionBone marrow-MSCIntracavernousal injection[170]
RabbitFacialTransectionBone marrow-MSCLocal[171]
RabbitIschiaticTractionBone marrow-MSCLocal[172,173]
PigIschiaticTransectionBone marrow-MSCLocal[174]
DogIschiaticTransectionBone marrow-MSCLocal[140,175]
DogIschiaticTransectionAdipose derived-MSCLocal[176]
MonkeyMedianTransectionBone marrow-MSCLocal[142]
HumanMedian or ulnarTransectionBone marrow mononuclear cell fraction-MSCLocal[146]

Although important advances have been achieved in the use of stem cells for improving nerve regeneration, they are still limited to basic and pre-clinical trials. In addition, there are several variables among these studies, such as tissue source; methods of cell isolation, expansion and characterization; route of cell delivery; number of transplanted cells; therapeutic time window; animal and nerve models; type of injury; number of transplanted cells; and immunogenicity. These variables represent an important obstacle for comparing and contrasting study outcomes from different groups, thus hindering progress in the field.

In 2006, The International Society for Cellular Therapy proposed the development of a set of minimal criteria (adherence to plastic in standard culture conditions, expression of a number of markers and multipotent differentiation potential into osteoblasts, adipocytes and chondroblasts) for defining the MSC for research purposes[177]. Although this action represented a great attempt to allow for comparison of scientific studies among different groups, the criteria for mesenchymal cells from different species should be further considered and well-defined, in particular the non-human and human primate MSC.

CONCLUSION

Pre-clinical studies have shown the beneficial effects of MSC therapy in the neurotrauma field. Unfortunately, these effects are not usually seen in the clinical trials, and the results are far from being as good as those described in experimental studies. Therefore, there is an urgent need to seek for standardization of protocols in terms of source of cells, culture conditions, time of treatment after injury, number and via of administration of cells, plasticity and capability of human MSC after extraction and expansion in culture, among other concerns. Basic and pre-clinical studies focusing on these important points will, hopefully, be of great help in terms of their successful implementation in clinical trials.

Footnotes

P- Reviewers: Kita K, Zocchi E S- Editor: Song XX L- Editor: A E- Editor: Zhang DN

References
1.  Vawda R, Fehlings MG. Mesenchymal cells in the treatment of spinal cord injury: current & amp; future perspectives. Curr Stem Cell Res Ther. 2013;8:25-38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 61]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
2.  Varma AK, Das A, Wallace G, Barry J, Vertegel AA, Ray SK, Banik NL. Spinal cord injury: a review of current therapy, future treatments, and basic science frontiers. Neurochem Res. 2013;38:895-905.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 164]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
3.  Jellinger KA. Neurochemical aspects of neurotraumatic and neurodegenerative diseases. Eur J Neurol. 2011;18:e104-e104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
4.  Yiu G, He Z. Glial inhibition of CNS axon regeneration. Nat Rev Neurosci. 2006;7:617-627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1032]  [Cited by in F6Publishing: 1102]  [Article Influence: 61.2]  [Reference Citation Analysis (0)]
5.  Dumont RJ, Okonkwo DO, Verma S, Hurlbert RJ, Boulos PT, Ellegala DB, Dumont AS. Acute spinal cord injury, part I: pathophysiologic mechanisms. Clin Neuropharmacol. 2001;24:254-264.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Ramer MS, Harper GP, Bradbury EJ. Progress in spinal cord research - a refined strategy for the International Spinal Research Trust. Spinal Cord. 2000;38:449-472.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  McKerracher L, David S, Jackson DL, Kottis V, Dunn RJ, Braun PE. Identification of myelin-associated glycoprotein as a major myelin-derived inhibitor of neurite growth. Neuron. 1994;13:805-811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 835]  [Cited by in F6Publishing: 857]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
8.  Zuo J, Neubauer D, Dyess K, Ferguson TA, Muir D. Degradation of chondroitin sulfate proteoglycan enhances the neurite-promoting potential of spinal cord tissue. Exp Neurol. 1998;154:654-662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 235]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
9.  Bradbury EJ, Moon LD, Popat RJ, King VR, Bennett GS, Patel PN, Fawcett JW, McMahon SB. Chondroitinase ABC promotes functional recovery after spinal cord injury. Nature. 2002;416:636-640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1798]  [Cited by in F6Publishing: 1728]  [Article Influence: 78.5]  [Reference Citation Analysis (0)]
10.  Kamada T, Koda M, Dezawa M, Yoshinaga K, Hashimoto M, Koshizuka S, Nishio Y, Moriya H, Yamazaki M. Transplantation of bone marrow stromal cell-derived Schwann cells promotes axonal regeneration and functional recovery after complete transection of adult rat spinal cord. J Neuropathol Exp Neurol. 2005;64:37-45.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Wright KT, El Masri W, Osman A, Chowdhury J, Johnson WE. Concise review: Bone marrow for the treatment of spinal cord injury: mechanisms and clinical applications. Stem Cells. 2011;29:169-178.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Wingrave JM, Schaecher KE, Sribnick EA, Wilford GG, Ray SK, Hazen-Martin DJ, Hogan EL, Banik NL. Early induction of secondary injury factors causing activation of calpain and mitochondria-mediated neuronal apoptosis following spinal cord injury in rats. J Neurosci Res. 2003;73:95-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 70]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
13.  Das A, Smith JA, Gibson C, Varma AK, Ray SK, Banik NL. Estrogen receptor agonists and estrogen attenuate TNF-α-induced apoptosis in VSC4.1 motoneurons. J Endocrinol. 2011;208:171-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 53]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
14.  Bracken MB. Steroids for acute spinal cord injury. Cochrane Database Syst Rev. 2012;1:CD001046.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 137]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
15.  Samantaray S, Sribnick EA, Das A, Knaryan VH, Matzelle DD, Yallapragada AV, Reiter RJ, Ray SK, Banik NL. Melatonin attenuates calpain upregulation, axonal damage and neuronal death in spinal cord injury in rats. J Pineal Res. 2008;44:348-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 88]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
16.  Bains M, Hall ED. Antioxidant therapies in traumatic brain and spinal cord injury. Biochim Biophys Acta. 2012;1822:675-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 300]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
17.  Robert AA, Zamzami M, Sam AE, Al Jadid M, Al Mubarak S. The efficacy of antioxidants in functional recovery of spinal cord injured rats: an experimental study. Neurol Sci. 2012;33:785-791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
18.  Mazzone GL, Nistri A. Delayed neuroprotection by riluzole against excitotoxic damage evoked by kainate on rat organotypic spinal cord cultures. Neuroscience. 2011;190:318-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
19.  Rong W, Wang J, Liu X, Jiang L, Wei F, Zhou H, Han X, Liu Z. 17β-estradiol attenuates neural cell apoptosis through inhibition of JNK phosphorylation in SCI rats and excitotoxicity induced by glutamate in vitro. Int J Neurosci. 2012;122:381-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
20.  Ritz MF, Graumann U, Gutierrez B, Hausmann O. Traumatic spinal cord injury alters angiogenic factors and TGF-beta1 that may affect vascular recovery. Curr Neurovasc Res. 2010;7:301-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
21.  Lutton C, Young YW, Williams R, Meedeniya AC, Mackay-Sim A, Goss B. Combined VEGF and PDGF treatment reduces secondary degeneration after spinal cord injury. J Neurotrauma. 2012;29:957-970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
22.  Guha A, Tator CH, Piper I. Effect of a calcium channel blocker on posttraumatic spinal cord blood flow. J Neurosurg. 1987;66:423-430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 59]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
23.  Ray SK, Matzelle DD, Sribnick EA, Guyton MK, Wingrave JM, Banik NL. Calpain inhibitor prevented apoptosis and maintained transcription of proteolipid protein and myelin basic protein genes in rat spinal cord injury. J Chem Neuroanat. 2003;26:119-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 40]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
24.  Sribnick EA, Matzelle DD, Banik NL, Ray SK. Direct evidence for calpain involvement in apoptotic death of neurons in spinal cord injury in rats and neuroprotection with calpain inhibitor. Neurochem Res. 2007;32:2210-2216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 25]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
25.  Ray SK, Samantaray S, Smith JA, Matzelle DD, Das A, Banik NL. Inhibition of cysteine proteases in acute and chronic spinal cord injury. Neurotherapeutics. 2011;8:180-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
26.  Reier PJ. Cellular transplantation strategies for spinal cord injury and translational neurobiology. NeuroRx. 2004;1:424-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 156]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
27.  Marques SA, Almeida FM, Fernandes AM, dos Santos Souza C, Cadilhe DV, Rehen SK, Martinez AM. Predifferentiated embryonic stem cells promote functional recovery after spinal cord compressive injury. Brain Res. 2010;1349:115-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
28.  de Almeida FM, Marques SA, Ramalho Bdos S, Rodrigues RF, Cadilhe DV, Furtado D, Kerkis I, Pereira LV, Rehen SK, Martinez AM. Human dental pulp cells: a new source of cell therapy in a mouse model of compressive spinal cord injury. J Neurotrauma. 2011;28:1939-1949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 56]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
29.  Bai L, Lennon DP, Eaton V, Maier K, Caplan AI, Miller SD, Miller RH. Human bone marrow-derived mesenchymal stem cells induce Th2-polarized immune response and promote endogenous repair in animal models of multiple sclerosis. Glia. 2009;57:1192-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 384]  [Cited by in F6Publishing: 388]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
30.  Torres-Espín A, Corona-Quintanilla DL, Forés J, Allodi I, González F, Udina E, Navarro X. Neuroprotection and axonal regeneration after lumbar ventral root avulsion by re-implantation and mesenchymal stem cells transplant combined therapy. Neurotherapeutics. 2013;10:354-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
31.  Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, Pell CL, Johnstone BH, Considine RV, March KL. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation. 2004;109:1292-1298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1649]  [Cited by in F6Publishing: 1637]  [Article Influence: 81.9]  [Reference Citation Analysis (0)]
32.  Nakagami H, Maeda K, Morishita R, Iguchi S, Nishikawa T, Takami Y, Kikuchi Y, Saito Y, Tamai K, Ogihara T. Novel autologous cell therapy in ischemic limb disease through growth factor secretion by cultured adipose tissue-derived stromal cells. Arterioscler Thromb Vasc Biol. 2005;25:2542-2547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 448]  [Cited by in F6Publishing: 448]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
33.  Wei X, Du Z, Zhao L, Feng D, Wei G, He Y, Tan J, Lee WH, Hampel H, Dodel R. IFATS collection: The conditioned media of adipose stromal cells protect against hypoxia-ischemia-induced brain damage in neonatal rats. Stem Cells. 2009;27:478-488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 185]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
34.  Azari MF, Mathias L, Ozturk E, Cram DS, Boyd RL, Petratos S. Mesenchymal stem cells for treatment of CNS injury. Curr Neuropharmacol. 2010;8:316-323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
35.  Hodgetts SI, Simmons PJ, Plant GW. A comparison of the behavioral and anatomical outcomes in sub-acute and chronic spinal cord injury models following treatment with human mesenchymal precursor cell transplantation and recombinant decorin. Exp Neurol. 2013;248:343-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
36.  Shin DA, Kim JM, Kim HI, Yi S, Ha Y, Yoon do H, Kim KN. Comparison of functional and histological outcomes after intralesional, intracisternal, and intravenous transplantation of human bone marrow-derived mesenchymal stromal cells in a rat model of spinal cord injury. Acta Neurochir (Wien). 2013;155:1943-1950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
37.  Zaminy A, Shokrgozar MA, Sadeghi Y, Noroozian M, Heidari MH, Piryaei A. Mesenchymal stem cells as an alternative for Schwann cells in rat spinal cord injury. Iran Biomed J. 2013;17:113-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 17]  [Reference Citation Analysis (0)]
38.  Kim JW, Ha KY, Molon JN, Kim YH. Bone marrow-derived mesenchymal stem cell transplantation for chronic spinal cord injury in rats: comparative study between intralesional and intravenous transplantation. Spine (Phila Pa 1976). 2013;38:E1065-E1074.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 62]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
39.  Kang ES, Ha KY, Kim YH. Fate of transplanted bone marrow derived mesenchymal stem cells following spinal cord injury in rats by transplantation routes. J Korean Med Sci. 2012;27:586-593.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
40.  Osaka M, Honmou O, Murakami T, Nonaka T, Houkin K, Hamada H, Kocsis JD. Intravenous administration of mesenchymal stem cells derived from bone marrow after contusive spinal cord injury improves functional outcome. Brain Res. 2010;1343:226-235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 170]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
41.  Zhou Z, Chen Y, Zhang H, Min S, Yu B, He B, Jin A. Comparison of mesenchymal stromal cells from human bone marrow and adipose tissue for the treatment of spinal cord injury. Cytotherapy. 2013;15:434-448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 92]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
42.  Schira J, Gasis M, Estrada V, Hendricks M, Schmitz C, Trapp T, Kruse F, Kögler G, Wernet P, Hartung HP. Significant clinical, neuropathological and behavioural recovery from acute spinal cord trauma by transplantation of a well-defined somatic stem cell from human umbilical cord blood. Brain. 2012;135:431-446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 79]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
43.  Roh DH, Seo MS, Choi HS, Park SB, Han HJ, Beitz AJ, Kang KS, Lee JH. Transplantation of human umbilical cord blood or amniotic epithelial stem cells alleviates mechanical allodynia after spinal cord injury in rats. Cell Transplant. 2013;22:1577-1590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
44.  Choi JS, Leem JW, Lee KH, Kim SS, Suh-Kim H, Jung SJ, Kim UJ, Lee BH. Effects of human mesenchymal stem cell transplantation combined with polymer on functional recovery following spinal cord hemisection in rats. Korean J Physiol Pharmacol. 2012;16:405-411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
45.  Wei X, Wen Y, Zhang T, Li H. [Effects of bone marrow mesenchymal stem cells with acellular muscle bioscaffolds on repair of acute hemi-transection spinal cord injury in rats]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi. 2012;26:1362-1368.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Boido M, Garbossa D, Fontanella M, Ducati A, Vercelli A. Mesenchymal stem cell transplantation reduces glial cyst and improves functional outcome after spinal cord compression. World Neurosurg. 2014;81:183-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 48]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
47.  Fang KM, Chen JK, Hung SC, Chen MC, Wu YT, Wu TJ, Lin HI, Chen CH, Cheng H, Yang CS. Effects of combinatorial treatment with pituitary adenylate cyclase activating peptide and human mesenchymal stem cells on spinal cord tissue repair. PLoS One. 2010;5:e15299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
48.  Park WB, Kim SY, Lee SH, Kim HW, Park JS, Hyun JK. The effect of mesenchymal stem cell transplantation on the recovery of bladder and hindlimb function after spinal cord contusion in rats. BMC Neurosci. 2010;11:119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
49.  Alexanian AR, Fehlings MG, Zhang Z, Maiman DJ. Transplanted neurally modified bone marrow-derived mesenchymal stem cells promote tissue protection and locomotor recovery in spinal cord injured rats. Neurorehabil Neural Repair. 2011;25:873-880.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 44]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
50.  Zhilai Z, Hui Z, Anmin J, Shaoxiong M, Bo Y, Yinhai C. A combination of taxol infusion and human umbilical cord mesenchymal stem cells transplantation for the treatment of rat spinal cord injury. Brain Res. 2012;1481:79-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
51.  Ryu HH, Lim JH, Byeon YE, Park JR, Seo MS, Lee YW, Kim WH, Kang KS, Kweon OK. Functional recovery and neural differentiation after transplantation of allogenic adipose-derived stem cells in a canine model of acute spinal cord injury. J Vet Sci. 2009;10:273-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 124]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
52.  Quertainmont R, Cantinieaux D, Botman O, Sid S, Schoenen J, Franzen R. Mesenchymal stem cell graft improves recovery after spinal cord injury in adult rats through neurotrophic and pro-angiogenic actions. PLoS One. 2012;7:e39500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 153]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
53.  Park SI, Lim JY, Jeong CH, Kim SM, Jun JA, Jeun SS, Oh WI. Human umbilical cord blood-derived mesenchymal stem cell therapy promotes functional recovery of contused rat spinal cord through enhancement of endogenous cell proliferation and oligogenesis. J Biomed Biotechnol. 2012;2012:362473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
54.  Hu SL, Luo HS, Li JT, Xia YZ, Li L, Zhang LJ, Meng H, Cui GY, Chen Z, Wu N. Functional recovery in acute traumatic spinal cord injury after transplantation of human umbilical cord mesenchymal stem cells. Crit Care Med. 2010;38:2181-2189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 68]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
55.  Kang KN, Kim da Y, Yoon SM, Lee JY, Lee BN, Kwon JS, Seo HW, Lee IW, Shin HC, Kim YM. Tissue engineered regeneration of completely transected spinal cord using human mesenchymal stem cells. Biomaterials. 2012;33:4828-4835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
56.  Min SH, Lee SH, Shim H, Park JS, Lee YI, Kim HW, Hyun JK. Development of complete thoracic spinal cord transection model in rats for delayed transplantation of stem cells. Spine (Phila Pa 1976). 2011;36:E155-E163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
57.  Hara Y, Nishiura Y, Ochiai N, Sharula Y, Kubota S, Saijilafu H. New treatment for peripheral nerve defects: reconstruction of a 2 cm, monkey median nerve gap by direct lengthening of both nerve stumps. J Orthop Res. 2012;30:153-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
58.  Karaoz E, Kabatas S, Duruksu G, Okcu A, Subasi C, Ay B, Musluman M, Civelek E. Reduction of lesion in injured rat spinal cord and partial functional recovery of motility after bone marrow derived mesenchymal stem cell transplantation. Turk Neurosurg. 2012;22:207-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 18]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
59.  Alexanian AR, Kwok WM, Pravdic D, Maiman DJ, Fehlings MG. Survival of neurally induced mesenchymal cells may determine degree of motor recovery in injured spinal cord rats. Restor Neurol Neurosci. 2010;28:761-767.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
60.  Gu W, Zhang F, Xue Q, Ma Z, Lu P, Yu B. Transplantation of bone marrow mesenchymal stem cells reduces lesion volume and induces axonal regrowth of injured spinal cord. Neuropathology. 2010;30:205-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 74]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
61.  Park SS, Lee YJ, Lee SH, Lee D, Choi K, Kim WH, Kweon OK, Han HJ. Functional recovery after spinal cord injury in dogs treated with a combination of Matrigel and neural-induced adipose-derived mesenchymal Stem cells. Cytotherapy. 2012;14:584-597.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 85]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
62.  Zhang W, Yan Q, Zeng YS, Zhang XB, Xiong Y, Wang JM, Chen SJ, Li Y, Bruce IC, Wu W. Implantation of adult bone marrow-derived mesenchymal stem cells transfected with the neurotrophin-3 gene and pretreated with retinoic acid in completely transected spinal cord. Brain Res. 2010;1359:256-271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 50]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
63.  Zeng X, Zeng YS, Ma YH, Lu LY, Du BL, Zhang W, Li Y, Chan WY. Bone marrow mesenchymal stem cells in a three-dimensional gelatin sponge scaffold attenuate inflammation, promote angiogenesis, and reduce cavity formation in experimental spinal cord injury. Cell Transplant. 2011;20:1881-1899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 108]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
64.  Shi CY, Ruan LQ, Feng YH, Fang JL, Song CJ, Yuan ZG, Ding YM. [Marrow mesenchymal stem cell transplantation with sodium alginate gel for repair of spinal cord injury in mice]. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2011;40:354-359.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Liu WG, Wang ZY, Huang ZS. Bone marrow-derived mesenchymal stem cells expressing the bFGF transgene promote axon regeneration and functional recovery after spinal cord injury in rats. Neurol Res. 2011;33:686-693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
66.  Hejcl A, Sedý J, Kapcalová M, Toro DA, Amemori T, Lesný P, Likavcanová-Mašínová K, Krumbholcová E, Prádný M, Michálek J. HPMA-RGD hydrogels seeded with mesenchymal stem cells improve functional outcome in chronic spinal cord injury. Stem Cells Dev. 2010;19:1535-1546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 113]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
67.  Yu D, Lü G, Cao Y, Li G, Zhi X, Fan Z. [Effects of bone marrow mesenchymal stem cells transplantation on expression of vascular endothelial growth factor gene and angiogenesis after spinal cord injury in rats]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi. 2011;25:837-841.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Shang AJ, Hong SQ, Xu Q, Wang HY, Yang Y, Wang ZF, Xu BN, Jiang XD, Xu RX. NT-3-secreting human umbilical cord mesenchymal stromal cell transplantation for the treatment of acute spinal cord injury in rats. Brain Res. 2011;1391:102-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
69.  Park SS, Byeon YE, Ryu HH, Kang BJ, Kim Y, Kim WH, Kang KS, Han HJ, Kweon OK. Comparison of canine umbilical cord blood-derived mesenchymal stem cell transplantation times: involvement of astrogliosis, inflammation, intracellular actin cytoskeleton pathways, and neurotrophin-3. Cell Transplant. 2011;20:1867-1880.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
70.  Lee JH, Chung WH, Kang EH, Chung DJ, Choi CB, Chang HS, Lee JH, Hwang SH, Han H, Choe BY. Schwann cell-like remyelination following transplantation of human umbilical cord blood (hUCB)-derived mesenchymal stem cells in dogs with acute spinal cord injury. J Neurol Sci. 2011;300:86-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 50]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
71.  Cizkova D, Novotna I, Slovinska L, Vanicky I, Jergova S, Rosocha J, Radonak J. Repetitive intrathecal catheter delivery of bone marrow mesenchymal stromal cells improves functional recovery in a rat model of contusive spinal cord injury. J Neurotrauma. 2011;28:1951-1961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 78]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
72.  Pal R, Gopinath C, Rao NM, Banerjee P, Krishnamoorthy V, Venkataramana NK, Totey S. Functional recovery after transplantation of bone marrow-derived human mesenchymal stromal cells in a rat model of spinal cord injury. Cytotherapy. 2010;12:792-806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 53]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
73.  Pedram MS, Dehghan MM, Soleimani M, Sharifi D, Marjanmehr SH, Nasiri Z. Transplantation of a combination of autologous neural differentiated and undifferentiated mesenchymal stem cells into injured spinal cord of rats. Spinal Cord. 2010;48:457-463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
74.  Karahuseyinoglu S, Cinar O, Kilic E, Kara F, Akay GG, Demiralp DO, Tukun A, Uckan D, Can A. Biology of stem cells in human umbilical cord stroma: in situ and in vitro surveys. Stem Cells. 2007;25:319-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 359]  [Cited by in F6Publishing: 335]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
75.  Wang HS, Hung SC, Peng ST, Huang CC, Wei HM, Guo YJ, Fu YS, Lai MC, Chen CC. Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord. Stem Cells. 2004;22:1330-1337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 946]  [Cited by in F6Publishing: 889]  [Article Influence: 46.8]  [Reference Citation Analysis (0)]
76.  Weiss ML, Medicetty S, Bledsoe AR, Rachakatla RS, Choi M, Merchav S, Luo Y, Rao MS, Velagaleti G, Troyer D. Human umbilical cord matrix stem cells: preliminary characterization and effect of transplantation in a rodent model of Parkinson’s disease. Stem Cells. 2006;24:781-792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 476]  [Cited by in F6Publishing: 467]  [Article Influence: 24.6]  [Reference Citation Analysis (0)]
77.  De Coppi P, Bartsch G, Siddiqui MM, Xu T, Santos CC, Perin L, Mostoslavsky G, Serre AC, Snyder EY, Yoo JJ. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol. 2007;25:100-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1406]  [Cited by in F6Publishing: 1205]  [Article Influence: 70.9]  [Reference Citation Analysis (0)]
78.  Lu P, Jones LL, Tuszynski MH. Axon regeneration through scars and into sites of chronic spinal cord injury. Exp Neurol. 2007;203:8-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 122]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
79.  Novikova LN, Brohlin M, Kingham PJ, Novikov LN, Wiberg M. Neuroprotective and growth-promoting effects of bone marrow stromal cells after cervical spinal cord injury in adult rats. Cytotherapy. 2011;13:873-887.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
80.  Ohta M, Suzuki Y, Noda T, Ejiri Y, Dezawa M, Kataoka K, Chou H, Ishikawa N, Matsumoto N, Iwashita Y. Bone marrow stromal cells infused into the cerebrospinal fluid promote functional recovery of the injured rat spinal cord with reduced cavity formation. Exp Neurol. 2004;187:266-278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 193]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
81.  Urdzíková L, Jendelová P, Glogarová K, Burian M, Hájek M, Syková E. Transplantation of bone marrow stem cells as well as mobilization by granulocyte-colony stimulating factor promotes recovery after spinal cord injury in rats. J Neurotrauma. 2006;23:1379-1391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 154]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
82.  Zurita M, Vaquero J, Bonilla C, Santos M, De Haro J, Oya S, Aguayo C. Functional recovery of chronic paraplegic pigs after autologous transplantation of bone marrow stromal cells. Transplantation. 2008;86:845-853.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 71]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
83.  Deng YB, Liu XG, Liu ZG, Liu XL, Liu Y, Zhou GQ. Implantation of BM mesenchymal stem cells into injured spinal cord elicits de novo neurogenesis and functional recovery: evidence from a study in rhesus monkeys. Cytotherapy. 2006;8:210-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 76]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
84.  Kang SK, Shin MJ, Jung JS, Kim YG, Kim CH. Autologous adipose tissue-derived stromal cells for treatment of spinal cord injury. Stem Cells Dev. 2006;15:583-594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 116]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
85.  Kang SK, Yeo JE, Kang KS, Phinney DG. Cytoplasmic extracts from adipose tissue stromal cells alleviates secondary damage by modulating apoptosis and promotes functional recovery following spinal cord injury. Brain Pathol. 2007;17:263-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
86.  Zhang HT, Luo J, Sui LS, Ma X, Yan ZJ, Lin JH, Wang YS, Chen YZ, Jiang XD, Xu RX. Effects of differentiated versus undifferentiated adipose tissue-derived stromal cell grafts on functional recovery after spinal cord contusion. Cell Mol Neurobiol. 2009;29:1283-1292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 20]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
87.  Oh JS, Ha Y, An SS, Khan M, Pennant WA, Kim HJ, Yoon DH, Lee M, Kim KN. Hypoxia-preconditioned adipose tissue-derived mesenchymal stem cell increase the survival and gene expression of engineered neural stem cells in a spinal cord injury model. Neurosci Lett. 2010;472:215-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 65]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
88.  Oh JS, Park IS, Kim KN, Yoon DH, Kim SH, Ha Y. Transplantation of an adipose stem cell cluster in a spinal cord injury. Neuroreport. 2012;23:277-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
89.  Arboleda D, Forostyak S, Jendelova P, Marekova D, Amemori T, Pivonkova H, Masinova K, Sykova E. Transplantation of predifferentiated adipose-derived stromal cells for the treatment of spinal cord injury. Cell Mol Neurobiol. 2011;31:1113-1122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 61]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
90.  Chung JY, Kim W, Im W, Yoo DY, Choi JH, Hwang IK, Won MH, Chang IB, Cho BM, Hwang HS. Neuroprotective effects of adipose-derived stem cells against ischemic neuronal damage in the rabbit spinal cord. J Neurol Sci. 2012;317:40-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
91.  Dasari VR, Spomar DG, Gondi CS, Sloffer CA, Saving KL, Gujrati M, Rao JS, Dinh DH. Axonal remyelination by cord blood stem cells after spinal cord injury. J Neurotrauma. 2007;24:391-410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 83]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
92.  Dasari VR, Spomar DG, Li L, Gujrati M, Rao JS, Dinh DH. Umbilical cord blood stem cell mediated downregulation of fas improves functional recovery of rats after spinal cord injury. Neurochem Res. 2008;33:134-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 60]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
93.  Troyer DL, Weiss ML. Wharton’s jelly-derived cells are a primitive stromal cell population. Stem Cells. 2008;26:591-599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 542]  [Cited by in F6Publishing: 576]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
94.  Can A, Karahuseyinoglu S. Concise review: human umbilical cord stroma with regard to the source of fetus-derived stem cells. Stem Cells. 2007;25:2886-2895.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 297]  [Cited by in F6Publishing: 282]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
95.  Manca MF, Zwart I, Beo J, Palasingham R, Jen LS, Navarrete R, Girdlestone J, Navarrete CV. Characterization of mesenchymal stromal cells derived from full-term umbilical cord blood. Cytotherapy. 2008;10:54-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 40]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
96.  Weiss ML, Anderson C, Medicetty S, Seshareddy KB, Weiss RJ, VanderWerff I, Troyer D, McIntosh KR. Immune properties of human umbilical cord Wharton’s jelly-derived cells. Stem Cells. 2008;26:2865-2874.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 420]  [Cited by in F6Publishing: 400]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
97.  Yang CC, Shih YH, Ko MH, Hsu SY, Cheng H, Fu YS. Transplantation of human umbilical mesenchymal stem cells from Wharton’s jelly after complete transection of the rat spinal cord. PLoS One. 2008;3:e3336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 176]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
98.  Alviano F, Fossati V, Marchionni C, Arpinati M, Bonsi L, Franchina M, Lanzoni G, Cantoni S, Cavallini C, Bianchi F. Term Amniotic membrane is a high throughput source for multipotent Mesenchymal Stem Cells with the ability to differentiate into endothelial cells in vitro. BMC Dev Biol. 2007;7:11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 164]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
99.  Tsai MS, Hwang SM, Tsai YL, Cheng FC, Lee JL, Chang YJ. Clonal amniotic fluid-derived stem cells express characteristics of both mesenchymal and neural stem cells. Biol Reprod. 2006;74:545-551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 191]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
100.  Wu ZY, Hui GZ, Lu Y, Wu X, Guo LH. Transplantation of human amniotic epithelial cells improves hindlimb function in rats with spinal cord injury. Chin Med J (Engl). 2006;119:2101-2107.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Sankar V, Muthusamy R. Role of human amniotic epithelial cell transplantation in spinal cord injury repair research. Neuroscience. 2003;118:11-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 140]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
102.  Meng XT, Li C, Dong ZY, Liu JM, Li W, Liu Y, Xue H, Chen D. Co-transplantation of bFGF-expressing amniotic epithelial cells and neural stem cells promotes functional recovery in spinal cord-injured rats. Cell Biol Int. 2008;32:1546-1558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
103.  Tetzlaff W, Okon EB, Karimi-Abdolrezaee S, Hill CE, Sparling JS, Plemel JR, Plunet WT, Tsai EC, Baptiste D, Smithson LJ. A systematic review of cellular transplantation therapies for spinal cord injury. J Neurotrauma. 2011;28:1611-1682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 394]  [Cited by in F6Publishing: 427]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
104.  Cízková D, Rosocha J, Vanický I, Jergová S, Cízek M. Transplants of human mesenchymal stem cells improve functional recovery after spinal cord injury in the rat. Cell Mol Neurobiol. 2006;26:1167-1180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 153]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
105.  Sheth RN, Manzano G, Li X, Levi AD. Transplantation of human bone marrow-derived stromal cells into the contused spinal cord of nude rats. J Neurosurg Spine. 2008;8:153-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
106.  Lammertse D, Tuszynski MH, Steeves JD, Curt A, Fawcett JW, Rask C, Ditunno JF, Fehlings MG, Guest JD, Ellaway PH. Guidelines for the conduct of clinical trials for spinal cord injury as developed by the ICCP panel: clinical trial design. Spinal Cord. 2007;45:232-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 145]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
107.  Steeves JD, Lammertse D, Curt A, Fawcett JW, Tuszynski MH, Ditunno JF, Ellaway PH, Fehlings MG, Guest JD, Kleitman N. Guidelines for the conduct of clinical trials for spinal cord injury (SCI) as developed by the ICCP panel: clinical trial outcome measures. Spinal Cord. 2007;45:206-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 340]  [Cited by in F6Publishing: 331]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
108.  Karamouzian S, Nematollahi-Mahani SN, Nakhaee N, Eskandary H. Clinical safety and primary efficacy of bone marrow mesenchymal cell transplantation in subacute spinal cord injured patients. Clin Neurol Neurosurg. 2012;114:935-939.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 104]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
109.  Park JH, Kim DY, Sung IY, Choi GH, Jeon MH, Kim KK, Jeon SR. Long-term results of spinal cord injury therapy using mesenchymal stem cells derived from bone marrow in humans. Neurosurgery. 2012;70:1238-147; discussion 1247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 132]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
110.  Dai G, Liu X, Zhang Z, Yang Z, Dai Y, Xu R. Transplantation of autologous bone marrow mesenchymal stem cells in the treatment of complete and chronic cervical spinal cord injury. Brain Res. 2013;1533:73-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 84]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
111.  Noble J, Munro CA, Prasad VS, Midha R. Analysis of upper and lower extremity peripheral nerve injuries in a population of patients with multiple injuries. J Trauma. 1998;45:116-122.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Robinson LR. Traumatic injury to peripheral nerves. Muscle Nerve. 2000;23:863-873.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
113.  Huang JH, Cullen DK, Browne KD, Groff R, Zhang J, Pfister BJ, Zager EL, Smith DH. Long-term survival and integration of transplanted engineered nervous tissue constructs promotes peripheral nerve regeneration. Tissue Eng Part A. 2009;15:1677-1685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 55]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
114.  Lee HK, Shin YK, Jung J, Seo SY, Baek SY, Park HT. Proteasome inhibition suppresses Schwann cell dedifferentiation in vitro and in vivo. Glia. 2009;57:1825-1834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
115.  Gordon T. The role of neurotrophic factors in nerve regeneration. Neurosurg Focus. 2009;26:E3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 248]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
116.  Ruohonen S, Khademi M, Jagodic M, Taskinen HS, Olsson T, Röyttä M. Cytokine responses during chronic denervation. J Neuroinflammation. 2005;2:26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 46]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
117.  Shamash S, Reichert F, Rotshenker S. The cytokine network of Wallerian degeneration: tumor necrosis factor-alpha, interleukin-1alpha, and interleukin-1beta. J Neurosci. 2002;22:3052-3060.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 641]  [Cited by in F6Publishing: 639]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
118.  Gaudet AD, Popovich PG, Ramer MS. Wallerian degeneration: gaining perspective on inflammatory events after peripheral nerve injury. J Neuroinflammation. 2011;8:110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 569]  [Cited by in F6Publishing: 546]  [Article Influence: 42.0]  [Reference Citation Analysis (0)]
119.  Waller A. Experiments on the section of the glossopharyngeal and hypoglossal nerves of the frog, and observations of the alterations produced thereby in the structure of their primitive fibres. Phil Transact Royal Soc London. 1850;423:429.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Dubový P. Wallerian degeneration and peripheral nerve conditions for both axonal regeneration and neuropathic pain induction. Ann Anat. 2011;193:267-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 126]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
121.  Martinez AM, Ribeiro LC. Ultrastructural localization of calcium in peripheral nerve fibres undergoing Wallerian degeneration: an oxalate-pyroantimonate and X-ray microanalysis study. J Submicrosc Cytol Pathol. 1998;30:451-458.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Zhai Q, Wang J, Kim A, Liu Q, Watts R, Hoopfer E, Mitchison T, Luo L, He Z. Involvement of the ubiquitin-proteasome system in the early stages of wallerian degeneration. Neuron. 2003;39:217-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 261]  [Cited by in F6Publishing: 269]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
123.  Guertin AD, Zhang DP, Mak KS, Alberta JA, Kim HA. Microanatomy of axon/glial signaling during Wallerian degeneration. J Neurosci. 2005;25:3478-3487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 140]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
124.  White FV, Toews AD, Goodrum JF, Novicki DL, Bouldin TW, Morell P. Lipid metabolism during early stages of Wallerian degeneration in the rat sciatic nerve. J Neurochem. 1989;52:1085-1092.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
125.  Murinson BB, Archer DR, Li Y, Griffin JW. Degeneration of myelinated efferent fibers prompts mitosis in Remak Schwann cells of uninjured C-fiber afferents. J Neurosci. 2005;25:1179-1187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 51]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
126.  Griffin JW, Pan B, Polley MA, Hoffman PN, Farah MH. Measuring nerve regeneration in the mouse. Exp Neurol. 2010;223:60-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 84]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
127.  Gersh I, Bodian D. Some chemical mechanisms in chromatolysis. J Cell Comp Physiol. 1943;21:253-279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 89]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
128.  Lieberman AR. The axon reaction: a review of the principal features of perikaryal responses to axon injury. Int Rev Neurobiol. 1971;14:49-124.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  Deumens R, Bozkurt A, Meek MF, Marcus MA, Joosten EA, Weis J, Brook GA. Repairing injured peripheral nerves: Bridging the gap. Prog Neurobiol. 2010;92:245-276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 357]  [Cited by in F6Publishing: 347]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
130.  Oliveira JT, Almeida FM, Biancalana A, Baptista AF, Tomaz MA, Melo PA, Martinez AM. Mesenchymal stem cells in a polycaprolactone conduit enhance median-nerve regeneration, prevent decrease of creatine phosphokinase levels in muscle, and improve functional recovery in mice. Neuroscience. 2010;170:1295-1303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 62]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
131.  Frattini F, Lopes FR, Almeida FM, Rodrigues RF, Boldrini LC, Tomaz MA, Baptista AF, Melo PA, Martinez AM. Mesenchymal stem cells in a polycaprolactone conduit promote sciatic nerve regeneration and sensory neuron survival after nerve injury. Tissue Eng Part A. 2012;18:2030-2039.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
132.  Dezawa M, Takahashi I, Esaki M, Takano M, Sawada H. Sciatic nerve regeneration in rats induced by transplantation of in vitro differentiated bone-marrow stromal cells. Eur J Neurosci. 2001;14:1771-1776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 397]  [Cited by in F6Publishing: 422]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
133.  Pereira Lopes FR, Camargo de Moura Campos L, Dias Corrêa J, Balduino A, Lora S, Langone F, Borojevic R, Blanco Martinez AM. Bone marrow stromal cells and resorbable collagen guidance tubes enhance sciatic nerve regeneration in mice. Exp Neurol. 2006;198:457-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 74]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
134.  Ribeiro-Resende VT, Pimentel-Coelho PM, Mesentier-Louro LA, Mendez RM, Mello-Silva JP, Cabral-da-Silva MC, de Mello FG, de Melo Reis RA, Mendez-Otero R. Trophic activity derived from bone marrow mononuclear cells increases peripheral nerve regeneration by acting on both neuronal and glial cell populations. Neuroscience. 2009;159:540-549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 59]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
135.  Oliveira JT, Mostacada K, de Lima S, Martinez AM. Bone marrow mesenchymal stem cell transplantation for improving nerve regeneration. Int Rev Neurobiol. 2013;108:59-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
136.  Gu Y, Wang J, Ding F, Hu N, Wang Y, Gu X. Neurotrophic actions of bone marrow stromal cells on primary culture of dorsal root ganglion tissues and neurons. J Mol Neurosci. 2010;40:332-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 47]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
137.  Chen CJ, Ou YC, Liao SL, Chen WY, Chen SY, Wu CW, Wang CC, Wang WY, Huang YS, Hsu SH. Transplantation of bone marrow stromal cells for peripheral nerve repair. Exp Neurol. 2007;204:443-453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 222]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
138.  Wang J, Ding F, Gu Y, Liu J, Gu X. Bone marrow mesenchymal stem cells promote cell proliferation and neurotrophic function of Schwann cells in vitro and in vivo. Brain Res. 2009;1262:7-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 103]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
139.  Wang X, Hu W, Cao Y, Yao J, Wu J, Gu X. Dog sciatic nerve regeneration across a 30-mm defect bridged by a chitosan/PGA artificial nerve graft. Brain. 2005;128:1897-1910.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Xue C, Hu N, Gu Y, Yang Y, Liu Y, Liu J, Ding F, Gu X. Joint use of a chitosan/PLGA scaffold and MSCs to bridge an extra large gap in dog sciatic nerve. Neurorehabil Neural Repair. 2012;26:96-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 62]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
141.  Hu J, Zhu QT, Liu XL, Xu YB, Zhu JK. Repair of extended peripheral nerve lesions in rhesus monkeys using acellular allogenic nerve grafts implanted with autologous mesenchymal stem cells. Exp Neurol. 2007;204:658-666.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 121]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
142.  Hu N, Wu H, Xue C, Gong Y, Wu J, Xiao Z, Yang Y, Ding F, Gu X. Long-term outcome of the repair of 50 mm long median nerve defects in rhesus monkeys with marrow mesenchymal stem cells-containing, chitosan-based tissue engineered nerve grafts. Biomaterials. 2013;34:100-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 99]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
143.  Pan HC, Yang DY, Chiu YT, Lai SZ, Wang YC, Chang MH, Cheng FC. Enhanced regeneration in injured sciatic nerve by human amniotic mesenchymal stem cell. J Clin Neurosci. 2006;13:570-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 78]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
144.  Lee EJ, Xu L, Kim GH, Kang SK, Lee SW, Park SH, Kim S, Choi TH, Kim HS. Regeneration of peripheral nerves by transplanted sphere of human mesenchymal stem cells derived from embryonic stem cells. Biomaterials. 2012;33:7039-7046.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
145.  McGrath AM, Brohlin M, Kingham PJ, Novikov LN, Wiberg M, Novikova LN. Fibrin conduit supplemented with human mesenchymal stem cells and immunosuppressive treatment enhances regeneration after peripheral nerve injury. Neurosci Lett. 2012;516:171-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
146.  Braga-Silva J, Gehlen D, Padoin AV, Machado DC, Garicochea B, Costa da Costa J. Can local supply of bone marrow mononuclear cells improve the outcome from late tubular repair of human median and ulnar nerves? J Hand Surg Eur Vol. 2008;33:488-493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
147.  Oliveira JT, Almeida FM, Biancalana A, Baptista AF, Tomaz MA, Melo PA, Martinez AM. Mesenchymal stem cells in a polycaprolactone conduit enhance median-nerve regeneration, prevent decrease of creatine phosphokinase levels in muscle, and improve functional recovery in mice. Neuroscience. 2010;170:1295-1303.  [PubMed]  [DOI]  [Cited in This Article: ]
148.  Zhao Z, Wang Y, Peng J, Ren Z, Zhan S, Liu Y, Zhao B, Zhao Q, Zhang L, Guo Q. Repair of nerve defect with acellular nerve graft supplemented by bone marrow stromal cells in mice. Microsurgery. 2011;31:388-394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
149.  Marconi S, Castiglione G, Turano E, Bissolotti G, Angiari S, Farinazzo A, Constantin G, Bedogni G, Bedogni A, Bonetti B. Human adipose-derived mesenchymal stem cells systemically injected promote peripheral nerve regeneration in the mouse model of sciatic crush. Tissue Eng Part A. 2012;18:1264-1272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 137]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
150.  Cheng FC, Tai MH, Sheu ML, Chen CJ, Yang DY, Su HL, Ho SP, Lai SZ, Pan HC. Enhancement of regeneration with glia cell line-derived neurotrophic factor-transduced human amniotic fluid mesenchymal stem cells after sciatic nerve crush injury. J Neurosurg. 2010;112:868-879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 59]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
151.  Yang LJ, Chang KW, Chung KC. A systematic review of nerve transfer and nerve repair for the treatment of adult upper brachial plexus injury. Neurosurgery. 2012;71:417-29; discussion 429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 85]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
152.  Jia H, Wang Y, Tong XJ, Liu GB, Li Q, Zhang LX, Sun XH. Sciatic nerve repair by acellular nerve xenografts implanted with BMSCs in rats xenograft combined with BMSCs. Synapse. 2012;66:256-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
153.  Wang Y, Jia H, Li WY, Tong XJ, Liu GB, Kang SW. Synergistic effects of bone mesenchymal stem cells and chondroitinase ABC on nerve regeneration after acellular nerve allograft in rats. Cell Mol Neurobiol. 2012;32:361-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
154.  Satar B, Hidir Y, Serdar MA, Kucuktag Z, Ural AU, Avcu F, Safali M, Oguztuzun S. Protein profiling of anastomosed facial nerve treated with mesenchymal stromal cells. Cytotherapy. 2012;14:522-528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
155.  Costa HJ, Ferreira Bento R, Salomone R, Azzi-Nogueira D, Zanatta DB, Paulino Costa M, da Silva CF, Strauss BE, Haddad LA. Mesenchymal bone marrow stem cells within polyglycolic acid tube observed in vivo after six weeks enhance facial nerve regeneration. Brain Res. 2013;1510:10-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 34]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
156.  Dadon-Nachum M, Sadan O, Srugo I, Melamed E, Offen D. Differentiated mesenchymal stem cells for sciatic nerve injury. Stem Cell Rev. 2011;7:664-671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 58]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
157.  Zhang Y, Luo H, Zhang Z, Lu Y, Huang X, Yang L, Xu J, Yang W, Fan X, Du B. A nerve graft constructed with xenogeneic acellular nerve matrix and autologous adipose-derived mesenchymal stem cells. Biomaterials. 2010;31:5312-5324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 118]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
158.  Wang Y, Zhao Z, Ren Z, Zhao B, Zhang L, Chen J, Xu W, Lu S, Zhao Q, Peng J. Recellularized nerve allografts with differentiated mesenchymal stem cells promote peripheral nerve regeneration. Neurosci Lett. 2012;514:96-101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 79]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
159.  Carriel V, Garrido-Gómez J, Hernández-Cortés P, Garzón I, García-García S, Sáez-Moreno JA, Del Carmen Sánchez-Quevedo M, Campos A, Alaminos M. Combination of fibrin-agarose hydrogels and adipose-derived mesenchymal stem cells for peripheral nerve regeneration. J Neural Eng. 2013;10:026022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 95]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
160.  Matsuse D, Kitada M, Kohama M, Nishikawa K, Makinoshima H, Wakao S, Fujiyoshi Y, Heike T, Nakahata T, Akutsu H. Human umbilical cord-derived mesenchymal stromal cells differentiate into functional Schwann cells that sustain peripheral nerve regeneration. J Neuropathol Exp Neurol. 2010;69:973-985.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 88]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
161.  Gärtner A, Pereira T, Alves MG, Armada-da-Silva PA, Amorim I, Gomes R, Ribeiro J, França ML, Lopes C, Carvalho RA. Use of poly(DL-lactide-epsilon-caprolactone) membranes and mesenchymal stem cells from the Wharton’s jelly of the umbilical cord for promoting nerve regeneration in axonotmesis: in vitro and in vivo analysis. Differentiation. 2012;84:355-365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 54]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
162.  Zheng L, Cui HF. Use of chitosan conduit combined with bone marrow mesenchymal stem cells for promoting peripheral nerve regeneration. J Mater Sci Mater Med. 2010;21:1713-1720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 29]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
163.  Ladak A, Olson J, Tredget EE, Gordon T. Differentiation of mesenchymal stem cells to support peripheral nerve regeneration in a rat model. Exp Neurol. 2011;228:242-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 138]  [Article Influence: 10.6]  [Reference Citation Analysis (1)]
164.  Ao Q, Fung CK, Tsui AY, Cai S, Zuo HC, Chan YS, Shum DK. The regeneration of transected sciatic nerves of adult rats using chitosan nerve conduits seeded with bone marrow stromal cell-derived Schwann cells. Biomaterials. 2011;32:787-796.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 118]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
165.  Yang Y, Yuan X, Ding F, Yao D, Gu Y, Liu J, Gu X. Repair of rat sciatic nerve gap by a silk fibroin-based scaffold added with bone marrow mesenchymal stem cells. Tissue Eng Part A. 2011;17:2231-2244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 92]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
166.  Liao IC, Wan H, Qi S, Cui C, Patel P, Sun W, Xu H. Preclinical evaluations of acellular biological conduits for peripheral nerve regeneration. J Tissue Eng. 2013;4:2041731413481036.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
167.  Zheng L, Cui HF. Enhancement of nerve regeneration along a chitosan conduit combined with bone marrow mesenchymal stem cells. J Mater Sci Mater Med. 2012;23:2291-2302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 21]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
168.  Nijhuis TH, Bodar CW, van Neck JW, Walbeehm ET, Siemionow M, Madajka M, Cwykiel J, Blok JH, Hovius SE. Natural conduits for bridging a 15-mm nerve defect: comparison of the vein supported by muscle and bone marrow stromal cells with a nerve autograft. J Plast Reconstr Aesthet Surg. 2013;66:251-259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
169.  Zhao Z, Wang Y, Peng J, Ren Z, Zhang L, Guo Q, Xu W, Lu S. Improvement in nerve regeneration through a decellularized nerve graft by supplementation with bone marrow stromal cells in fibrin. Cell Transplant. 2014;23:97-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
170.  You D, Jang MJ, Lee J, Jeong IG, Kim HS, Moon KH, Suh N, Kim CS. Periprostatic implantation of human bone marrow-derived mesenchymal stem cells potentiates recovery of erectile function by intracavernosal injection in a rat model of cavernous nerve injury. Urology. 2013;81:104-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
171.  Wang X, Luo E, Li Y, Hu J. Schwann-like mesenchymal stem cells within vein graft facilitate facial nerve regeneration and remyelination. Brain Res. 2011;1383:71-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
172.  Shen J, Duan XH, Cheng LN, Zhong XM, Guo RM, Zhang F, Zhou CP, Liang BL. In vivo MR imaging tracking of transplanted mesenchymal stem cells in a rabbit model of acute peripheral nerve traction injury. J Magn Reson Imaging. 2010;32:1076-1085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
173.  Duan XH, Cheng LN, Zhang F, Liu J, Guo RM, Zhong XM, Wen XH, Shen J. In vivo MRI monitoring nerve regeneration of acute peripheral nerve traction injury following mesenchymal stem cell transplantation. Eur J Radiol. 2012;81:2154-2160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
174.  Cho HH, Jang S, Lee SC, Jeong HS, Park JS, Han JY, Lee KH, Cho YB. Effect of neural-induced mesenchymal stem cells and platelet-rich plasma on facial nerve regeneration in an acute nerve injury model. Laryngoscope. 2010;120:907-913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 99]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
175.  Ding F, Wu J, Yang Y, Hu W, Zhu Q, Tang X, Liu J, Gu X. Use of tissue-engineered nerve grafts consisting of a chitosan/poly(lactic-co-glycolic acid)-based scaffold included with bone marrow mesenchymal cells for bridging 50-mm dog sciatic nerve gaps. Tissue Eng Part A. 2010;16:3779-3790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 63]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
176.  Ghoreishian M, Rezaei M, Beni BH, Javanmard SH, Attar BM, Zalzali H. Facial nerve repair with Gore-Tex tube and adipose-derived stem cells: an animal study in dogs. J Oral Maxillofac Surg. 2013;71:577-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
177.  Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj, Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315-317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11055]  [Cited by in F6Publishing: 11776]  [Article Influence: 692.7]  [Reference Citation Analysis (1)]