Skip to main content
CNS Neuroscience & Therapeutics logoLink to CNS Neuroscience & Therapeutics
. 2020 Mar 10;26(6):616–627. doi: 10.1111/cns.13300

Potential of mesenchymal stem cells alone, or in combination, to treat traumatic brain injury

Alison E Willing 1,, Mahasweta Das 2,3, Mark Howell 2,3, Shyam S Mohapatra 3,4, Subhra Mohapatra 2,3
PMCID: PMC7248546  PMID: 32157822

Abstract

Traumatic brain injury (TBI) causes death and disability in the United States and around the world. The traumatic insult causes the mechanical injury of the brain and primary cellular death. While a comprehensive pathological mechanism of TBI is still lacking, the focus of the TBI research is concentrated on understanding the pathophysiology and developing suitable therapeutic approaches. Given the complexities in pathophysiology involving interconnected immunologic, inflammatory, and neurological cascades occurring after TBI, the therapies directed to a single mechanism fail in the clinical trials. This has led to the development of the paradigm of a combination therapeutic approach against TBI. While there are no drugs available for the treatment of TBI, stem cell therapy has shown promising results in preclinical studies. But, the success of the therapy depends on the survival of the stem cells, which are limited by several factors including route of administration, health of the administered cells, and inflammatory microenvironment of the injured brain. Reducing the inflammation prior to cell administration may provide a better outcome of cell therapy following TBI. This review is focused on different therapeutic approaches of TBI and the present status of the clinical trials.

Keywords: clinical trials, combination treatment, stem cells, traumatic brain injury

1. INTRODUCTION

The Centers for Disease Control and Prevention defines a traumatic brain injury (TBI) as a disruption in normal brain function as a result of any blow to the head.1 It is a major health concern in the United States and around the world. According to the Health United States Report 2016, 2.8 million people in this country sustain this injury annually, and it is estimated that of these, ~50 000 die, ~282 000 are hospitalized, and the remaining 2.5 million (or 89%) are treated and released from the emergency department.2 Long‐term disability depends on the severity of the TBI,3 the presence of diffuse axonal injury on imaging,4 and the intensity of neurorehabilitation.5 Further, recovery may take an extended period of time6 and the patient may be left with neurobehavioral deficits including mental health disorders such as depression, anxiety or psychotic disorders, cognitive disorders related to executive functioning, and aggression.7 In a prospective study that followed TBI patients for up to 1 year, the distribution of mild, moderate, and severe TBI was comparable to what is observed in the real‐world population with 49% having mild TBI, 34% having moderate TBI, and 17% having a severe injury. About half of the study population did not return to their previous work after 1 year, and ~28% never returned to work of any kind.8 Also, long‐term disability is seen occasionally even in those with mild TBI.9 Thorough reviews of TBI epidemiology have recently been published.10, 11

Thus, while TBI is a significant public health problem, unfortunately there is no single therapy that has proved efficacious in its treatment. Similar to the situation with other brain injuries (such as the failure of neuroprotective glutamate receptor antagonists and antioxidant treatments in clinical trials for stroke12, 13) and neurodegenerative diseases, there have been myriad‐positive preclinical studies in TBI models and all of these promising therapies have failed in clinical trials. Various reasons have been advanced for these failures, including, but not limited to, differences in brain anatomy and physiology between rodents and humans, inadequate animal models, failure to test the treatment in a clinically relevant way coupled with failure to remain faithful to the preclinical testing parameters in the clinical trials, underpowered studies, heterogeneity of TBI injury, and insensitive outcome measures in both preclinical and clinical studies. There is no dearth of discussions in the literature identifying these shortcomings in the therapeutic development and testing of potential new treatments for TBI.14, 15, 16, 17, 18

What we are left with for treatments is a general approach that is akin to crisis management. According to the current Brain Trauma Foundation Guidelines, based on the best available medical evidence for the management of severe TBI, it is imperative to provide adequate nutrition, support breathing by tracheostomy, and perform a large decompressive craniectomy.19 The underlying problems for developing an effective treatment for TBI are 2‐fold. First, the injury can be unique to the patient, depending on the type of TBI and the region of the brain affected. Second, once that injury occurs, a complicated neurodegenerative cascade is triggered; resolving any one of these pathological processes is not enough to prevent or terminate the others. In this review, we will discuss the pathophysiology of TBI with emphasis on immune and inflammatory function. We will also discuss the evidence for the development of a mesenchymal stem cell (MSC)‐based treatment that can suppress immune and inflammatory/degenerative cascades and provide neuroprotection.

2. PATHOPHYSIOLOGY OF TBI

The pathophysiologic mechanisms of TBI are poorly understood as the anatomy of the brain is uniquely complex with multiple cell types (neurons, astrocytes, oligodendrocytes, and microglia) and multiple subtypes of these cells. While we now know that neural stem cells (NSC) exist within the adult brain and some degree of axonal, dendritic, and synaptic plasticity occurs, we have yet to fully exploit the brain's regenerative capacity to repair an injury. Add to this the complicated neuronal networks throughout the brain and neural repair is a daunting task. Ramon Y Cajal, widely regarded as one of the fathers of modern neuroscience, stated in his treatise on neural development and regeneration “…once development was ended, the founts of growth and regeneration of the axons and dendrites dried up irrevocably. In the adult centers the nerve paths are something fixed, ended and immutable. Everything must die, nothing may be regenerated. It is for the science of the future to change, if possible, this harsh decree.20 When we consider additional factors such as the confined space within the skull that contributes to increased intracranial pressure as edema develops and the blood‐brain barrier (BBB), which can make therapeutic access to the brain difficult, the task of repairing the brain after TBI may seem insurmountable.

The pathophysiology of TBI occurs in two main phases—the primary insult and the secondary sequelae. The primary insult is the initial blow to the head. It can be a penetrating wound or a closed‐head injury. The nature of the injury can be focal, involving a very circumscribed area of the brain, or it can be more diffuse causing widespread axonal injury. Then, there is the special case of the coup‐countercoup injury as the brain impacts the skull in two sites on opposite sides of the brain; this is observed with trauma that involves deceleration of the head. Depending on the nature of the trauma, neurons, astrocytes, and oligodendrocytes may be destroyed, bleeding may occur, axons may be severed, and a contusion may form.21 The immediate pathological consequences of these injuries are similar to those observed with cerebral ischemia—excitotoxicity, changes in ion flux (Ca++, Na+, and K+) across the cell membrane, loss of ATP, lactate production, induction of cortical spreading depression, cytokine production, and loss of barrier function at the BBB.22, 23, 24

The severity of the initial blow and the immediate pathophysiologic changes that occur determines the severity of any subsequent secondary degenerative processes. In the acute period, neurons and axons continue to die, and there is damage to the vascular endothelium; this, in turn, allows blood components to leak into the brain parenchyma, including peripheral immune cells, which then contribute to the pro‐inflammatory environment. Astrocytes swell and tissue edema occurs. If edema is not controlled, intracranial pressure increases, which can lead to compression of arteries and decreased cerebral blood flow; cerebral ischemia commonly occurs under these conditions leading to a vicious cycle of increasing edema, increasing intracranial pressure, and increased ischemia that can lead to death.

3. INFLAMMATION IN TBI

The main architects of the local inflammation at the site of injury are the microglia. Once a TBI has occurred, it is the microglia that proliferate and migrate to the site of the damage. As these microglia work to remove the cellular debris at the lesion site, they produce cytokines and chemokines that activate pattern recognition receptors to bind damage‐associated molecular patterns, and attract and polarize peripheral immune cells. The first peripheral immune cells into the damaged tissue are neutrophils25 followed 24‐48 hours later by monocytes or macrophage, and T cells all of which are releasing cytokines and chemokines. Once the peripheral immune cells have established a strong pro‐inflammatory response in the brain in the acute to subacute stage of TBI, then tissue damage is likely to be exacerbated. Further, the T cells may become activated through antigen presentation by microglia and macrophage, mobilizing the adaptive immune system26 and potentially leading to autoimmunity.26, 27

In addition to local neuroinflammatory processes within the brain after TBI, systemic immune and inflammatory processes are also impacted and have recently been reviewed.28, 29 Peripheral immune cells are mobilized from the bone marrow,30 thymus,31 and spleen31 early after the TBI. While there is a brief recovery in thymocytes and classical monocytes in thymus and spleen within the first 2 weeks postinjury, over time these cell populations decline again and may be responsible for the post‐TBI immune suppression that is observed in patients.29, 31 Also implicated in the peripheral immune response and long‐term immune suppression is both the activation of the hypothalamus‐pituitary‐adrenal axis, through the release of glucocorticoids and the sympathetic nervous system, through release of catecholamines.29, 32

Ideally, inflammation will be a self‐limiting process. There are endogenously formed products of arachidonic acid metabolism that actively inhibit pro‐inflammatory responses.33 These include the lipoxins, resolvins, protectins, and maresins, which decrease pro‐inflammatory cytokine secretion,34 alter migratory signals for peripheral immune cells,35 and stimulate neuroprotective and tissue regeneration processes.36 There has been very little research on these resolving mediators in TBI, but administering the lipoxin, LXA4, into the lateral ventricles 10 minutes after a TBI induced by weight drop, reduced pro‐inflammation, BBB disruption, and lesion size.34 Further, its receptor, ALX/FPR2, is upregulated in astrocytes.37

Unfortunately, in many instances inflammation may not resolve and becomes chronic.38 In the aged brain, there is increased recruitment of peripheral macrophage into the TBI brain.39 In addition to cellular infiltration of pro‐inflammatory immune cells after TBI, sustained complement C3 activation leads to chronic inflammation by activating microglia and astrocytes in the region around the initial lesion and contributes to further neuronal loss 30 days post‐TBI.40 Using a combination of magnetic resonance imaging, magnetic resonance spectroscopy, and positron emission tomography in a rat lateral fluid percussion model, inflammation was shown to be present still 6 months post‐TBI.41 Even 12 months after a controlled cortical impact in mouse, there is increased immunolabeling for IBA1 (microglia) and glial fibrillary acidic protein (GFAP).42 What's more, this chronic inflammation is associated with continued behavioral deficits.

4. EXPERIMENTAL PHARMACEUTIC TREATMENTS FOR TBI

4.1. Pharmaceuticals

The focus of current pharmacological interventions after TBI is to manage level of consciousness, neuropsychiatric, neurocognitive, and neurobehavioral symptoms that may arise.43 With the occurrence of so many interconnected neuro‐immune‐inflammatory pathologic cascades engaged after TBI, it is not surprising that therapies targeting one specific degenerative pathway have failed to demonstrate efficacy in clinical trials. Drug interventions that have been studied can generally be categorized by their therapeutic target. One class of drugs are those that prevent calcium ion flux. An example of such an approach is the calcium channel blocker, Nimodipine, which showed promising effects in rodents but exhibited only a small effect on TBI patients.44, 45 Disruption of intracellular calcium signaling may also improve outcome after TBI. Cyclosporin is a T‐cell immunosuppressant that acts by binding to cyclophilin; the cyclosporine‐cyclophilin complex binds to calcineurin preventing dephosphorylation of NFAT, translocation to the nucleus, and increased transcription of interleukin (IL) 2.46 In the absence of cyclosporine, calcineurin is regulated by calcium and calmodulin. Cyclosporin prevents calcium ion transport into the mitochondria in animal models but not in TBI patients.47

There are also a number of studies that have targeted excitotoxicity, specifically glutamate release and overstimulation of the NMDA receptor. For example, Selfotel is a NMDA antagonist and the first glutamate antagonist to enter into Phase III clinical trial. This trial was discontinued because of high mortality and a failure to improve Glasgow Outcome Score.48 The results from clinical studies of other NMDA receptor antagonists also failed to demonstrate any efficacy of treatment.49 While not a true antagonist, magnesium blocks the NMDA receptor calcium channel. Increasing available magnesium also had no effect on TBI outcome.50

Another approach has been to target oxidative stress produced by oxygen radical formation and lipid peroxidation. The lipid peroxidation inhibitor, Tirilazad mesylate, which is an approved drug in Europe to treat aneurismal subarachnoid hemorrhage, showed promising neuro‐ and vaso‐protective responses in animal models of moderate‐to‐severe TBI but failed to show improvement over placebo control in Phase III clinical trials involving human TBI patients.51, 52 Pegylated superoxide dismutase, a free radical scavenger, was found to be effective in preventing secondary injury in preclinical and Phase I clinical studies but failed to show reduction in mortality or improve neurologic outcome in Phase III trials.53, 54 Another antioxidant that may be promising is N‐acetylcysteine. When administered to patients within 24 hours of mild TBI, symptoms were significantly better compared to a placebo‐controlled group.55

Another target that has been examined is the treatment of inflammation with corticosteroids, statins,56 cannabinoids, and bradykinin B2 receptor antagonists.50 In addition, the gonadal hormones, estrogen and progesterone, both showed promising results in preclinical studies but failed to show beneficial effects in clinical trials.57, 58, 59

The more recent approach has been to search for potential therapies that target more than one pathway. One such strategy is to use a pharmacologic that interacts with multiple receptor types, which, thereby, produces more than one effect. For example, sigma receptor agonists selective for either sigma 1 (σ1) or σ2 receptors (or both) have both neuroprotective and anti‐inflammatory effects in rodent models of stroke.60 More recently, σ1‐selective agonists have been shown to decrease neuroinflammation,61 while σ2‐selective agonists are neuroprotective after TBI.62

4.2. Combination drug treatment regimens for TBI

In an effort to increase treatment efficacy, multiple drug combinations have been administered together to target multiple neurodegenerative pathways. Based on reported success in the treatment of HIV/AIDS with HAART,63 recently a combination drug therapy was designed for the treatment of TBI64 combining vitamin D3, progesterone, omega 3 fatty acids, and glutamine administration for the first 72 hours for TBI patients with a poor prognosis; all patients improved beyond original expectations. However, these case studies included only three patients, there were no controls, and a larger study has not been performed to validate these observations. Another example of a combination of drug therapy is the progesterone and 1,25‐dihydroxyvitamin D3 combination, which was effective in reducing neuroinflammation as compared to treatment with the drugs separately.65, 66

5. CELL THERAPY IN TBI

A number of different cell types have been examined as potential therapeutics for TBI. The first studies in the field focused on replacing neurons in order to rebuild the neural circuitry. The earliest cells examined were already postmitotic neurons,67, 68, 69 and there was variable therapeutic success. Gradually, these studies were replaced by studies using stem cells. In a mouse model, NSC survived, differentiated, migrated to the lesion site, and improved motor and cognitive function after TBI.70 Also, embryonic stem cells were shown to improve functional outcome after TBI in rodents, but tumors also formed.71 Later studies involved predifferentiating stem cells into more lineage‐restricted precursors in order to reduce the likelihood of tumorigenesis.72, 73 In an effort to reduce reliance on embryonic or fetal tissue, there have been a number of studies more recently focused on induced pluripotent stem cells derived from adult somatic cells.74, 75, 76

In addition to these embryonic and NSC for which there are direct developmental pathways to produce neurons, astrocytes, and oligodendrocytes, another source of stem cells that has received a great deal of attention is MSCs. Originally, MSCs were isolated from bone marrow where they support hematopoiesis. However, it has become clear that MSCs reside in many tissues in the body,77 which may explain why they appear to be efficacious for treating so many different injuries and diseases. Because of their pleiotropic characteristics, these cells have significant therapeutic potential for various diseases including TBI. Following administration, MSCs have shown to penetrate the BBB, migrate to the site of injury, and secrete several growth factors including brain‐derived neurotrophic factor (BDNF), glial‐derived neurotrophic factor (GDNF), vascular endothelial growth factor (VEGF), nerve growth factor (NGF), and regenerate BBB and neuronal and glial tissues.78, 79, 80, 81 MSCs also modulated inflammation by inhibiting interleukin six (IL‐6) and IL1‐β and enhancing IL‐10.82 The anti‐inflammatory effect of MSC was reported in a study involving combined administration of MSC and NSC that led to increased recovery from stroke‐induced cerebral damage in rats as compared to MSC or NSC alone.83 Early studies demonstrated that intravenous,84 intraarterial,85 and intracerebral 86 administration of these cells improved motor and neurological outcome after TBI. Other methods of MSC delivery that have been used since include intranasal,87, 88, 89 intrathecal,90 and intracisternal.91 The postulated mechanism of repair in these early studies was transdifferentiation of the cells into neural cells. A number of studies were able to demonstrate that some of the transplanted MSCs expressed neuronal and astrocytic markers in vivo, but few cells survived.84, 85, 92, 93, 94 The results of in vitro studies were more positive95; however, the transformation from MSC to mature neuron occurred within hours,96 was reversible when the initiating stimuli were removed, and occurred in the presence of protein synthesis inhibition.97 It has been suggested that the “transdifferentiation” of MSCs into neurons requires MSCs to be in a toxic, stressful environment.96, 97 However, the ultimate proof that the cells transdifferentiate into neurons is still lacking; these “neurons” have never been shown to produce an action potential.98

In the ensuing years, several other putative mechanisms of repair for these cells have been studied. Thus, it has been suggested that MSCs may induce brain repair through the production of trophic factors or stimulating release of trophic factors from endogenous cells.99, 100, 101 These paracrine mechanisms of repair have recently been reviewed in some detail.102, 103, 104 More recently, MSC‐derived exosomes have been examined as the paracrine source of neuroprotection and anti‐inflammation. These exosomes improved cognitive function and reduced inflammation as determined by decreased IL‐1 and increased IL‐10 in the brain after TBI.105 Later studies are consistent with these results.106, 107 MSC‐derived exosomes have also been tested in porcine model of TBI coupled with hemorrhagic shock; consistent with the rodent data, the pigs had fewer cognitive deficits as determined with a neurological severity score and recovered faster than nontreated pigs.108 There is also evidence for MSC inducing immune suppression109, 110 and anti‐inflammation106, 111, 112, 113; stimulating neurogenesis88, 114, 115 and angiogenesis107, 116, 117; activating survival pathways118 and inhibiting apoptotic pathways101, 119; and enhancing neuroplasticity through neurite outgrowth120, 121 and synaptogenesis.78, 107

5.1. Clinical translation of cell therapy for TBI

As described in the previous section, many different approaches have shown promise for treating TBI‐induced pathologies and stimulating tissue regeneration in animal models. However, none of these have thus far translated into therapeutic benefit in human patients and the early clinical trials have not used pure MSC. Table 1 shows the current comprehensive status of clinical trials of stem cell therapy for TBI. The earliest clinical studies reported in the literature used bone marrow‐derived mononuclear cells (BMMNCs), which is a heterogeneous mixture of immune cells and stem cells including MSCs. The first major cell therapy trials for TBI using BMMNCs were conducted on children by Cox and colleagues in 2011.122 In the preclinical studies and Phase 1 clinical trial, treatment reduced BBB permeability and neuroinflammation after TBI. In a second pediatric study from the same research group, the TBI‐induced increase in intracranial pressure was reduced in the cell treatment group.123

Table 1.

Current status of cell therapy clinical trials of traumatic brain injury (TBI)

NCT number Title Status Interventions Phases Enrollment
NCT04063215 A clinical trial to determine the safety and efficacy of hope biosciences autologous mesenchymal stem cell therapy for the treatment of traumatic brain injury and hypoxic‐ischemic encephalopathy Not yet recruiting Drug: HB‐adMSC Phase 1 Phase 2 24
NCT02525432 Autologous stem cell study for adult TBI (Phase 2b) Enrolling by invitation Biological: Placebo Infusion Biological: Autologous BMMNC Infusion Phase 2 55
NCT01575470 Treatment of severe adult traumatic brain injury using bone marrow mononuclear cells Completed Biological: autologous bone marrow mononuclear cells Phase 1|Phase 2 25
NCT02416492 A study of modified stem cells in TBI Completed Biological: SB623 cells Procedure: Sham Control Phase 2 61
NCT01851083 pediatric autologous bone marrow mononuclear cells for severe traumatic brain injury Active, not recruiting Biological: autologous bone marrow mononuclear cells|Other: Placebo Infusion Phase 1|Phase 2 47
NCT02959294 Use of adipose‐derived stem/stromal cells in concussion and traumatic brain injuries Enrolling by invitation Procedure: Microcannula Harvest Adipose|Device: Centricyte 1000|Procedure: Sterile Normal Saline IV deployment AD‐cSVF Phase 1|Phase 2 200

There are also clinical trials of BMMNCs in the adults with severe TBI. In this population, treatment with BMMNCs resulted in structural preservation of the corpus callosum and corticospinal tract and these changes were correlated to neurocognitive outcomes; in addition, there was a reduction in the pro‐inflammatory cytokine response to injury (NCT01575470).124 A Phase 2 (NCT02525432) and Phase IIb (NCT02416492) follow‐up studies are currently underway.

Of the currently registered clinical trials specifically investigating MSCs, there are two. Hope Bioscience has a safety and efficacy clinical trial of its adipose‐derived MSCs. In another clinical trial (NCT02416492), the safety and efficacy of San Bio's proprietary adult bone marrow‐derived MSCs genetically modified to express the intracellular domain of human Notch‐1 to treat chronic TBI. Clinical trial of these cells in stroke patients demonstrated that the cells were safe and induced significant motor function improvement in adults according to European Stroke Scale, the NIH Stroke Scale, and the Fugl‐Meyer scale.125

5.2. Potential of MSC and anti‐inflammatory combination treatments for TBI

Because of inflammation and other ongoing neurodegenerative cascades, the brain environment post‐TBI is a hostile environment for transplanted cells. Without some adjunctive treatment, cell survival is limited. For NSC and neurons, where the cells are needed to rebuild neural circuitry, survival is critical. For MSCs, whether survival is necessary depends on where and how the cells are having their effects. The immune‐suppressive effects of the cells are systemic, so delivery to the brain is not critical.113, 126, 127 Their anti‐inflammatory effects are both systemic and local within the brain, so some cells need to enter the brain.111, 112 These functions have led to studies of these cells as cotransplants to enhance the survival of other cells; an example from the spinal cord injury literature demonstrated that MSCs cotransplanted with NSC in injured spinal cord resulted in increased survival of the NSC.128 While MSCs do have anti‐inflammatory properties, the noxious environment may decrease their survival as well,129 which may limit their neuroprotective effects. To deal with the problem of a toxic, degenerative environment in the brain post‐TBI, investigators have adopted multiple approaches to enhance cell survival.

One approach has been to delay transplantation so that the pathophysiology can stabilize; for example, in a study comparing MSCs with MSCs in a scaffold,130 MSCs were administered two months post‐TBI so that regenerative and repair physiologic processes would dominant in the parenchyma around the TBI lesion. Another approach has been to administer a drug treatment along with the cells or prior to cell transplantation. For example, Mahmood and associates administered MSCs intravenously 24 hours after TBI.131, 132 At the same time, statin treatment was started and continued for 14 days. Combination treatment with either atorvastatin or simvastatin improved recovery on the modified Neurological Severity Score (mNSS). In another study, investigators combined an early (1 hour post‐TBI) injection of the β adrenergic antagonist, propranolol, with an intravenous administration of MSCs at 72 hours post‐TBI.133 The underlying premise of the study was that propranolol decreases the TBI‐induced Sympathetic Nervous System hyperactivity; this decreased activity then helps to maintain cerebral perfusion, thereby decreasing post‐TBI ischemia and cell death. The MSCs were administered to manage the secondary inflammatory state. Unfortunately, the combined effects of propranolol and MSCs were not synergistic or additive; favorable outcomes (decreased serum norepinephrine, BBB permeability, microglial activation, cognitive function) could be achieved solely by the MSCs. In another study, investigators administered a calpain inhibitor 30 minutes after TBI and then transplanted the MSCs at 24 hours post‐TBI.134 There were significant decreases in pro‐inflammatory cytokines around the lesion, increased survival of the MSCs, and improvements on the mNSS.

We have taken a similar approach, but instead of modifying sympathetic activity in conjunction with targeting inflammation, we combined two inflammation‐modulating treatments. Based on earlier studies in which we identified chemokine (C‐C motif) ligand 20 (CCL20) as being significantly elevated after TBI,135, 136 we combined treatment with pioglitazone, a peroxisome proliferator‐activated receptor gamma agonist that inhibits CCL20, with treatment with MSCs.88 The pioglitazone was administered once a day for 5 days after TBI. On day 5, MSCs were administered intranasally. The combination of pioglitazone and MSCs was significantly better than either treatment alone on measures of anxiety, inflammation in the brain, and endogenous NSC proliferation. A similar approach was taken by Peruzzaro and associates when they engineered MSC's to overexpress IL‐10137 before transplantation in a TBI model.

Growth or trophic factor delivery in conjunction with cell administration is another approach that has been studied. These growth/trophic factors may favorably condition the environment in the TBI brain, or they may protect the transplanted cells from cell death. GDNF, epidermal growth factor, and VEGF have been shown to protect the brain from neuronal injury and increase regeneration of different cell types.138, 139, 140 However, growth factor delivery can be problematic. Growth factors have very short half‐lives, necessitating continual local delivery.141 Further, systemic delivery is often associated with side effects.142 As a result, it is imperative to deliver the growth factors into the brain near the injury. Liu et al143 showed that intracerebroventricular administration of fibroblast growth factor 2 (FGF2) for 7 days beginning at the time of MSC transplantation resulted in faster improvement in the forelimb placing and balance beam tests compared to the no treatment TBI group and the cell‐only group. Insulin‐like growth factor‐1 known to have a crucial role in MSC proliferation and putative differentiation to neuronal cells.144 This approach has shown to improve cell injury and motor activity of injured rats and improved metabolic and nutritional outcomes following TBI.145, 146 However, even growth factor delivery directly into the central nervous system (CNS) can lead to adverse effects. In a recent review of NGF trials for CNS diseases, the authors point out significant adverse effects such as pain and weight loss led to the discontinuation of the studies.142, 147

The most common procedure for combining a growth factor treatment with the delivery of MSCs has been to transfect the cells with specific growth factors. For example, overexpressing FGF21 in MSC resulted in improved performance on the Morris water maze, increased hippocampal neurogenesis and dendritic outgrowth.148 Other investigators have focused on the neurotrophins. Wu and associates overexpressed NT3 in MSCs and observed decreased glial activation, a smaller brain lesion, and decreased edema in the brain,149 while a number of investigators have focused on increasing BDNF expression in the cells, essentially improving functional outcome.150, 151

More recently, transplanting MSCs with a bioactive scaffold or biomaterial that secretes growth factors has been used to enhance survival, migration, and differentiation of transplanted cells.152 In one version of a scaffold, Bhang and associates suspended MSCs in a fibrin gel laced with FGF2.153 The MSCs in the fibrin glue with FGF2 decreased lesion size and apoptotic cell death more than MSCs in glue alone. When a functional peptide derivative of BDNF was incorporated in a self‐assembling hydrogel scaffold prior to seeding with MSCs and activated astrocytes, the resulting structure was able to reduce TBI lesion size.154

6. FUTURE PERSPECTIVES

Over the last two decades, there is a wealth of preclinical data, suggesting that MSCs may be an effective treatment, either alone or in combination, to improve outcome after TBI. Clinical trials are still in the early stages. Even with an abundance of data, there are still questions that should be addressed before these cells are routinely used to treat TBI or other CNS injuries or disease. Perhaps, most important is the issue of dosing. All studies in TBI to date have used a single dose of MSCs and doses used in the clinical studies have ranged from a total of 2.5 × 106 cells up to 12 × 106 cells per kilogram.124, 125 Not only do different research groups use different cell doses, in those studies using combination treatments, there is no indication whether dosing of the cell and/or the pharmacologic changes when the two are used in combination. To complicate issues further, patients may be taking a plethora of medications either prophylactically (such as baby aspirin or statins) or to treat common chronic diseases (beta blockers). For example, aspirin altered the immune and inflammatory profile of both endogenous monocytes harvested from stroke patients or healthy controls and MSCs in culture.155 When rats were treated with aspirin and cord blood‐derived MSCs singly or in combination, outcome tended to be somewhat worse with the combination treatment.156 These considerations are especially important as the population ages, since the elderly are at risk for TBI and polypharmacy is an issue in this population. In another example, when type I diabetic rats underwent a middle cerebral artery occlusion followed by MSC transplantation 24 hours later, the cells induced hemorrhagic transformation of the stroke and increased BBB leakage.157 The addition of niacin to the MSC therapy prevented these adverse effects.158 It is, therefore, essential that MSCs alone or in combination with other drugs must be tested for efficacy against a background of commonly prescribed drugs or medical conditions.

Another consideration with dosing is whether or not a single injection of cells is enough to maintain long‐term improvements in the functional outcome. As mentioned previously, TBI is accompanied by chronic inflammation.38, 40, 41, 42 As of yet, there are no studies in rodent models of TBI that have examined repeated administration of MSCs. There are indications from small clinical studies in other neurologic diseases or injuries that may hint at the potential efficacy of this dosing approach in TBI. For example, in patients with incomplete spinal cord injury, 30 × 106 cells were administered into the subarachnoid space four times at three‐month intervals 159; while improvements in function were noted, the improvements were different for each individual patient. Patients with amyotrophic lateral sclerosis (ALS) were injected twice intrathecally with autologous MSCs (1 × 106 cells/kg) 28 days apart and followed for 1 year.160 These patients had no severe adverse events and function as measured with the ALS Functional Rating Scale‐Revised stabilized.

Another issue that needs to be addressed in this field is the reliance on rodents in the early testing of potential new therapeutics. While using mice and rats is cost‐effective and allows manipulation of the genome, there are significant differences in the structure of the rodent brain and the human brain, not only in size, but also in, cerebrovascular volume, oxygen and glucose requirements, lissencephalic vs gyrencephalic architecture, and the amount of white matter present.161 It is imperative that larger animal models are used for the testing of new therapies including cell therapies so that we can distinguish good candidate therapies that are likely to succeed in clinical trials from those that will not. There has been some work in the development and characterization of porcine and ovine models of TBI, but there are currently no studies of MSC therapies and only a handful of MSC‐derive exosome studies108 in these larger animal models TBI models.161, 162

7. SUMMARY

The preclinical data on MSCs both alone and in conjunction with other treatment strategies are promising. These cells have entered at least Phase I (safety) clinical trials for multiple nervous system diseases and injuries, most notably cerebral ischemia,163, 164, 165, 166 multiple sclerosis,167, 168, 169, 170, 171, 172, 173, 174 Alzheimer's disease,175 and TBI.133, 150, 176 While we have a better understanding of the pathologic cascades triggered after TBI and the mechanisms by which MSCs repair the brain and improve functional outcomes, we are still years away from realizing an effective regenerative medicine therapy for TBI that is widely available to patients.

CONFLICT OF INTEREST

The authors declare no conflict of interest.

ACKNOWLEDGMENTS

This work is partly supported by Veterans Affairs Merit Review grant (BX002668) to Dr Subhra Mohapatra, and Research Career Scientist Awards to Dr Subhra Mohapatra (IK6BX004212) and Dr Shyam Mohapatra (IK6 BX003778). Though this report is based upon work supported, in part, by the Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development, the contents of this report do not represent the views of the Department of Veterans Affairs or the United States Government.

Willing AE, Das M, Howell M, Mohapatra SS, Mohapatra S. Potential of mesenchymal stem cells alone, or in combination, to treat traumatic brain injury. CNS Neurosci Ther. 2020;26:616–627. 10.1111/cns.13300

REFERENCES

  • 1. Centers for Disease Control and Prevention . Surveillance Report of Traumatic Brain Injury‐related Emergency Department Visits, Hospitalizations, and Deaths—United States, 2014. Centers for Disease Control and Prevention, U.S. Department of Health and Human Services.
  • 2. National Center for Health Statistics. Health, United States , 2016: With Chartbook on Long‐term Trends in Health. Hyattsville, MD. 2017. [PubMed]
  • 3. Walker WC, Stromberg KA, Marwitz JH, et al. Predicting long‐term global outcome after traumatic brain injury: development of a practical prognostic tool using the traumatic brain injury model systems national database. J Neurotrauma. 2018;35(14):1587‐1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. van Eijck MM, Schoonman GG, van der Naalt J, de Vries J, Roks G. Diffuse axonal injury after traumatic brain injury is a prognostic factor for functional outcome: a systematic review and meta‐analysis. Brain Inj. 2018;32(4):395‐402. [DOI] [PubMed] [Google Scholar]
  • 5. Konigs M, Beurskens EA, Snoep L, Scherder EJ, Oosterlaan J. Effects of timing and intensity of neurorehabilitation on functional outcome after traumatic brain injury: a systematic review and meta‐analysis. Arch Phys Med Rehabil. 2018;99(6):1149‐1159.e1. [DOI] [PubMed] [Google Scholar]
  • 6. Schmidt RH, Scholten KJ, Maughan PH. Time course for recovery of water maze performance and central cholinergic innervation after fluid percussion injury. J Neurotrauma. 1999;16(12):1139‐1147. [DOI] [PubMed] [Google Scholar]
  • 7. Warden DL, Gordon B, McAllister TW, et al. Guidelines for the pharmacologic treatment of neurobehavioral sequelae of traumatic brain injury. J Neurotrauma. 2006;23(10):1468‐1501. [DOI] [PubMed] [Google Scholar]
  • 8. Singh R, Choudhri K, Sinha S, Mason S, Lecky F, Dawson J. Global outcome after traumatic brain injury in a prospective cohort. Clin Neurol Neurosurg. 2019;186:105526. [DOI] [PubMed] [Google Scholar]
  • 9. Lloyd J, Wilson ML, Tenovuo O, Saarijarvi S. Outcomes from mild and moderate traumatic brain injuries among children and adolescents: a systematic review of studies from 2008–2013. Brain Inj. 2015;29(5):539‐549. [DOI] [PubMed] [Google Scholar]
  • 10. Faul M, Coronado V. Epidemiology of traumatic brain injury. Handb Clin Neurol. 2015;127:3‐13. [DOI] [PubMed] [Google Scholar]
  • 11. Iaccarino C, Carretta A, Nicolosi F, Morselli C. Epidemiology of severe traumatic brain injury. J Neurosurg Sci. 2018;62(5):535‐541. [DOI] [PubMed] [Google Scholar]
  • 12. Roth S, Liesz A. Stroke research at the crossroads ‐ where are we heading? Swiss Med Wkly. 2016;146:w14329. [DOI] [PubMed] [Google Scholar]
  • 13. Moretti A, Ferrari F, Villa RF. Neuroprotection for ischaemic stroke: current status and challenges. Pharmacol Ther. 2015;146:23‐34. [DOI] [PubMed] [Google Scholar]
  • 14. Carbonara M, Fossi F, Zoerle T, et al. Neuroprotection in traumatic brain injury: mesenchymal stromal cells can potentially overcome some limitations of previous clinical trials. Front Neurol. 2018;9:885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Chakraborty S, Skolnick B, Narayan RK. Neuroprotection trials in traumatic brain injury. Curr Neurol Neurosci Rep. 2016;16(4):29. [DOI] [PubMed] [Google Scholar]
  • 16. Cox CS Jr, Juranek J, Bedi S. Clinical trials in traumatic brain injury: cellular therapy and outcome measures. Transfusion. 2019;59(S1):858‐868. [DOI] [PubMed] [Google Scholar]
  • 17. (STAIR) STAIR . Recommendations for standards regarding preclinical neuroprotective and restorative drug development. Stroke. 1999;30:2752‐2758. [DOI] [PubMed] [Google Scholar]
  • 18. Fisher M, Feuerstein G, Howells DW, et al. Update of the stroke therapy academic industry roundtable preclinical recommendations. Stroke. 2009;40(6):2244‐2250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Volovici V, Steyerberg EW, Cnossen MC, et al. Evolution of evidence and guideline recommendations for the medical management of severe traumatic brain injury. J Neurotrauma. 2019;36(22):3183‐3189. [DOI] [PubMed] [Google Scholar]
  • 20. Ramon y Cajal S. Cajal's Degeneration and Regeneration of the Nervous System (History of Neuroscience), 1st edn New York, NY: Oxford University Press, Inc.; 1991. [Google Scholar]
  • 21. Bramlett HM, Dietrich WD. Pathophysiology of cerebral ischemia and brain trauma: similarities and differences. J Cereb Blood Flow Metab. 2004;24(2):133‐150. [DOI] [PubMed] [Google Scholar]
  • 22. Ladak AA, Enam SA, Ibrahim MT. A Review of the Molecular Mechanisms of Traumatic Brain Injury. World Neurosurg. 2019;131:126‐132. [DOI] [PubMed] [Google Scholar]
  • 23. Ng SY, Lee AYW. Traumatic brain injuries: pathophysiology and potential therapeutic targets. Front Cell Neurosci. 2019;13:528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Romeu‐Mejia R, Giza CC, Goldman JT. Concussion pathophysiology and injury biomechanics. Curr Rev Musculoskelet Med. 2019;12(2):105‐116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Liu Y‐W, Li S, Dai S‐S. Neutrophils in traumatic brain injury (TBI): friend or foe? J Neuroinflammation. 2018;15(1):146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Jassam YN, Izzy S, Whalen M, McGavern DB, El Khoury J. Neuroimmunology of traumatic brain injury: time for a paradigm shift. Neuron. 2017;95(6):1246‐1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Goverman J. Autoimmune T cell responses in the central nervous system. Nat Rev Immunol. 2009;9(6):393‐407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Nizamutdinov D, Shapiro LA. Overview of traumatic brain injury: an immunological context. Brain Sci. 2017;7(1):11 10.3390/brainsci7010011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Hazeldine J, Lord JM, Belli A. Traumatic brain injury and peripheral immune suppression: primer and prospectus. Front Neurol. 2015;6:235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Livingston DH, Anjaria D, Wu J, et al. Bone marrow failure following severe injury in humans. Ann Surg. 2003;238(5):748‐753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Schwulst SJ, Trahanas DM, Saber R, Perlman H. Traumatic brain injury‐induced alterations in peripheral immunity. J Trauma Acute Care Surg. 2013;75(5):780‐788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Hoover DB. Cholinergic modulation of the immune system presents new approaches for treating inflammation. Pharmacol Ther. 2017;179:1‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Martini AC, Forner S, Bento AF, Rae GA. Neuroprotective effects of lipoxin A4 in central nervous system pathologies. Biomed Res Int. 2014;2014:316204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Luo C‐L, Li Q‐Q, Chen X‐P, et al. Lipoxin A4 attenuates brain damage and downregulates the production of pro‐inflammatory cytokines and phosphorylated mitogen‐activated protein kinases in a mouse model of traumatic brain injury. Brain Res. 2013;1502:1‐10. [DOI] [PubMed] [Google Scholar]
  • 35. Hawkins KE, DeMars KM, Singh J, et al. Neurovascular protection by post‐ischemic intravenous injections of the lipoxin A4 receptor agonist, BML‐111, in a rat model of ischemic stroke. J Neurochem. 2014;129(1):130‐142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Serhan CN. Pro‐resolving lipid mediators are leads for resolution physiology. Nature. 2014;510(7503):92‐101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Bisicchia E, Sasso V, Catanzaro G, et al. Resolvin D1 halts remote neuroinflammation and improves functional recovery after focal brain damage via ALX/FPR2 receptor‐regulated microRNAs. Mol Neurobiol. 2018;55(8):6894‐6905. [DOI] [PubMed] [Google Scholar]
  • 38. Acosta SA, Tajiri N, Shinozuka K, et al. Long‐term upregulation of inflammation and suppression of cell proliferation in the brain of adult rats exposed to traumatic brain injury using the controlled cortical impact model. PLoS One. 2013;8(1):e53376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Morganti JM, Riparip L‐K, Chou A, Liu S, Gupta N, Rosi S. Age exacerbates the CCR2/5‐mediated neuroinflammatory response to traumatic brain injury. J Neuroinflammation. 2016;13(1):80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Alawieh A, Langley EF, Weber S, Adkins D, Tomlinson S. Identifying the role of complement in triggering neuroinflammation after traumatic brain injury. J Neurosci. 2018;38(10):2519‐2532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Yasmin A, Pitkanen A, Jokivarsi K, Poutiainen P, Grohn O, Immonen R. MRS reveals chronic inflammation in T2w MRI‐negative perilesional cortex ‐ a 6‐months multimodal imaging follow‐up study. Front Neurosci. 2019;13:863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Pischiutta F, Micotti E, Hay JR, et al. Single severe traumatic brain injury produces progressive pathology with ongoing contralateral white matter damage one year after injury. Exp Neurol. 2018;300:167‐178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Talsky A, Pacione LR, Shaw T, et al. Pharmacological interventions for traumatic brain injury. BC Med J. 2011;53:1. [Google Scholar]
  • 44. Gurkoff G, Shahlaie K, Lyeth B, Berman R. Voltage‐gated calcium channel antagonists and traumatic brain injury. Pharmaceuticals (Basel). 2013;6(7):788‐812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Narayan RK, Michel ME, Ansell B, et al. Clinical trials in head injury. J Neurotrauma. 2002;19(5):503‐557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Matsuda S, Koyasu S. Mechanisms of action of cyclosporine. Immunopharmacology. 2000;47(2–3):119‐125. [DOI] [PubMed] [Google Scholar]
  • 47. Aminmansour B, Fard SA, Habibabadi MR, Moein P, Norouzi R, Naderan M. The efficacy of cyclosporine‐A on diffuse axonal injury after traumatic brain injury. Adv Biomed Res. 2014;3:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Morris GF, Bullock R, Marshall SB, et al. Failure of the competitive N‐methyl‐D‐aspartate antagonist Selfotel (CGS 19755) in the treatment of severe head injury: results of two phase III clinical trials. The Selfotel Investigators. J Neurosurg. 1999;91(5):737‐743. [DOI] [PubMed] [Google Scholar]
  • 49. Muir KW. Glutamate‐based therapeutic approaches: clinical trials with NMDA antagonists. Curr Opin Pharmacol. 2006;6(1):53‐60. [DOI] [PubMed] [Google Scholar]
  • 50. Beauchamp K, Mutlak H, Smith WR, Shohami E, Stahel PF. Pharmacology of traumatic brain injury: where is the "golden bullet"? Mol Med. 2008;14(11–12):731‐740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Del Zoppo GJ, Moskowitz MA. Translating interventions from ischemic stroke models to patients: the view in 2009. Front Neurol Neurosci. 2009;25:34‐38. [DOI] [PubMed] [Google Scholar]
  • 52. Hall ED, Vaishnav RA, Mustafa AG. Antioxidant therapies for traumatic brain injury. Neurotherapeutics. 2010;7(1):51‐61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Dekmak AmiraSan, Mantash S, Shaito A, et al. Stem cells and combination therapy for the treatment of traumatic brain injury. Behav Brain Res. 2018;340:49‐62. [DOI] [PubMed] [Google Scholar]
  • 54. Muizelaar JP, Kupiec JW, Rapp LA. PEG‐SOD after head injury. J Neurosurg. 1995;83(5):942. [DOI] [PubMed] [Google Scholar]
  • 55. Hoffer ME, Balaban C, Slade MD, Tsao JW, Hoffer B. Amelioration of acute sequelae of blast induced mild traumatic brain injury by N‐acetyl cysteine: a double‐blind, placebo controlled study. PLoS One. 2013;8(1):e54163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Chen S‐F, Hung T‐H, Chen C‐C, et al. Lovastatin improves histological and functional outcomes and reduces inflammation after experimental traumatic brain injury. Life Sci. 2007;81(4):288‐298. [DOI] [PubMed] [Google Scholar]
  • 57. He J, Hoffman SW, Stein DG. Allopregnanolone, a progesterone metabolite, enhances behavioral recovery and decreases neuronal loss after traumatic brain injury. Restor Neurol Neurosci. 2004;22(1):19‐31. [PubMed] [Google Scholar]
  • 58. Skolnick BE, Maas AI, Narayan RK, et al. A clinical trial of progesterone for severe traumatic brain injury. N Engl J Med. 2014;371(26):2467‐2476. [DOI] [PubMed] [Google Scholar]
  • 59. VanLandingham JW, Cekic M, Cutler S, Hoffman SW, Stein DG. Neurosteroids reduce inflammation after TBI through CD55 induction. Neurosci Lett. 2007;425(2):94‐98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Ajmo CT Jr, Vernon DO, Collier L, Pennypacker KR, Cuevas J. Sigma receptor activation reduces infarct size at 24 hours after permanent middle cerebral artery occlusion in rats. Curr Neurovasc Res. 2006;3(2):89‐98. [DOI] [PubMed] [Google Scholar]
  • 61. Dong H, Ma Y, Ren Z, et al. Sigma‐1 receptor modulates neuroinflammation after traumatic brain injury. Cell Mol Neurobiol. 2016;36(5):639‐645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Vázquez‐Rosa E, Watson MR, Sahn JJ, et al. Neuroprotective efficacy of a sigma 2 receptor/TMEM97 modulator (DKR‐1677) after traumatic brain injury. ACS Chem Neurosci. 2019;10(3):1595‐1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Palella FJ, Delaney KM, Moorman AC, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med. 1998;338(13):853‐860. [DOI] [PubMed] [Google Scholar]
  • 64. Matthews LR, Danner OK, Ahmed YA, et al. Combination therapy with vitamin D3, progesterone, omega‐3 fatty acids, and glutamine reverses coma and improves clinical outcomes inpatients with severe traumatic brain injuries: a case series. Int J Case Rep Images. 2013;4(4):143‐149. [Google Scholar]
  • 65. Tang H, Hua F, Wang J, et al. Progesterone and vitamin D combination therapy modulates inflammatory response after traumatic brain injury. Brain Inj. 2015;29(10):1165‐1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Cekic M, Cutler SM, VanLandingham JW, Stein DG. Vitamin D deficiency reduces the benefits of progesterone treatment after brain injury in aged rats. Neurobiol Aging. 2011;32(5):864‐874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Muir JK, Raghupathi R, Saatman KE, et al. Terminally differentiated human neurons survive and integrate following transplantation into the traumatically injured rat brain. J Neurotrauma. 1999;16(5):403‐414. [DOI] [PubMed] [Google Scholar]
  • 68. Soares H, McIntosh TK. Fetal cortical transplants in adult rats subjected to experimental brain injury. J Neural Transplant Plast. 1991;2(3–4):207‐220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Soares HD, Sinson GP, McIntosh TK. Fetal hippocampal transplants attenuate CA3 pyramidal cell death resulting from fluid percussion brain injury in the rat. J Neurotrauma. 1995;12(6):1059‐1067. [DOI] [PubMed] [Google Scholar]
  • 70. Riess P, Zhang C, Saatman KE, et al. Transplanted neural stem cells survive, differentiate, and improve neurological motor function after experimental traumatic brain injury. Neurosurgery. 2002;51(4):1043‐1054; discussion 52–54. [DOI] [PubMed] [Google Scholar]
  • 71. Riess P, Molcanyi M, Bentz K, et al. Embryonic stem cell transplantation after experimental traumatic brain injury dramatically improves neurological outcome, but may cause tumors. J Neurotrauma. 2007;24(1):216‐225. [DOI] [PubMed] [Google Scholar]
  • 72. Le Belle JE, Caldwell MA, Svendsen CN. Improving the survival of human CNS precursor‐derived neurons after transplantation. J Neurosci Res. 2004;76(2):174‐183. [DOI] [PubMed] [Google Scholar]
  • 73. Lepore AC, Han SS, Tyler‐Polsz CJ, Cai J, Rao MS, Fischer I. Differential fate of multipotent and lineage‐restricted neural precursors following transplantation into the adult CNS. Neuron Glia Biol. 2004;1(2):113‐126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Dunkerson J, Moritz KE, Young J, et al. Combining enriched environment and induced pluripotent stem cell therapy results in improved cognitive and motor function following traumatic brain injury. Restor Neurol Neurosci. 2014;32(5):675‐687. [DOI] [PubMed] [Google Scholar]
  • 75. Furmanski O, Nieves MD, Doughty ML. Controlled cortical impact model of mouse brain injury with therapeutic transplantation of human induced pluripotent stem cell‐derived neural cells. J Vis Exp. 2019;149:e59561 10.3791/59561 [DOI] [PubMed] [Google Scholar]
  • 76. Tang H, Sha H, Sun H, et al. Tracking induced pluripotent stem cells‐derived neural stem cells in the central nervous system of rats and monkeys. Cell Reprogram. 2013;15(5):435‐442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. da Silva ML, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post‐natal organs and tissues. J Cell Sci. 2006;119(Pt 11):2204‐2213. [DOI] [PubMed] [Google Scholar]
  • 78. Gutiérrez‐Fernández M, Rodríguez‐Frutos B, Ramos‐Cejudo J, et al. Effects of intravenous administration of allogenic bone marrow‐ and adipose tissue‐derived mesenchymal stem cells on functional recovery and brain repair markers in experimental ischemic stroke. Stem Cell Res Ther. 2013;4(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Walker PA, Shah SK, Harting MT, Cox CS Jr. Progenitor cell therapies for traumatic brain injury: barriers and opportunities in translation. Dis Model Mech. 2009;2(1–2):23‐38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Mahmood A, Lu D, Chopp M. Intravenous administration of marrow stromal cells (MSCs) increases the expression of growth factors in rat brain after traumatic brain injury. J Neurotrauma. 2004;21(1):33‐39. [DOI] [PubMed] [Google Scholar]
  • 81. Mahmood A, Lu D, Qu C, Goussev A, Chopp M. Long‐term recovery after bone marrow stromal cell treatment of traumatic brain injury in rats. J Neurosurg. 2006;104(2):272‐277. [DOI] [PubMed] [Google Scholar]
  • 82. Ma H, Lam PK, Tong CSW, Lo KKY, Wong GKC, Poon WS. The neuroprotection of hypoxic adipose tissue‐derived mesenchymal stem cells in experimental traumatic brain injury. Cell Transplant. 2019;28(7):874‐884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Hosseini SM, Farahmandnia M, Razi Z, et al. Combination cell therapy with mesenchymal stem cells and neural stem cells for brain stroke in rats. Int J Stem Cells. 2015;8(1):99‐105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Lu D, Mahmood A, Wang L, Li Y, Lu M, Chopp M. Adult bone marrow stromal cells administered intravenously to rats after traumatic brain injury migrate into brain and improve neurological outcome. Neuroreport. 2001;12(3):559‐563. [DOI] [PubMed] [Google Scholar]
  • 85. Lu D, Li Y, Wang L, Chen J, Mahmood A, Chopp M. Intraarterial administration of marrow stromal cells in a rat model of traumatic brain injury. J Neurotrauma. 2001;18(8):813‐819. [DOI] [PubMed] [Google Scholar]
  • 86. Mahmood A, Lu D, Wang L, Chopp M. Intracerebral transplantation of marrow stromal cells cultured with neurotrophic factors promotes functional recovery in adult rats subjected to traumatic brain injury. J Neurotrauma. 2002;19(12):1609‐1617. [DOI] [PubMed] [Google Scholar]
  • 87. Chau MJ, Deveau TC, Gu X, et al. Delayed and repeated intranasal delivery of bone marrow stromal cells increases regeneration and functional recovery after ischemic stroke in mice. BMC Neurosci. 2018;19(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Das M, Mayilsamy K, Tang X, et al. Pioglitazone treatment prior to transplantation improves the efficacy of human mesenchymal stem cells after traumatic brain injury in rats. Sci Rep. 2019;9(1):13646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Donega V, Nijboer CH, van Velthoven CTJ, et al. Assessment of long‐term safety and efficacy of intranasal mesenchymal stem cell treatment for neonatal brain injury in the mouse. Pediatr Res. 2015;78(5):520‐526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Vaquero J, Zurita M, Bonilla C, et al. Progressive increase in brain glucose metabolism after intrathecal administration of autologous mesenchymal stromal cells in patients with diffuse axonal injury. Cytotherapy. 2017;19(1):88‐94. [DOI] [PubMed] [Google Scholar]
  • 91. Shin DA, Kim J‐M, Kim H‐I, et al. 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(10):1943‐1950. [DOI] [PubMed] [Google Scholar]
  • 92. Chen X, Katakowski M, Li YI, et al. Human bone marrow stromal cell cultures conditioned by traumatic brain tissue extracts: growth factor production. J Neurosci Res. 2002;69(5):687‐691. [DOI] [PubMed] [Google Scholar]
  • 93. Hong S‐Q, Zhang H‐T, You J, et al. Comparison of transdifferentiated and untransdifferentiated human umbilical mesenchymal stem cells in rats after traumatic brain injury. Neurochem Res. 2011;36(12):2391‐2400. [DOI] [PubMed] [Google Scholar]
  • 94. Mahmood A, Lu D, Wang L, Li Y, Lu M, Chopp M. Treatment of traumatic brain injury in female rats with intravenous administration of bone marrow stromal cells. Neurosurgery. 2001;49(5):1196‐1204; discussion 203–204. [PubMed] [Google Scholar]
  • 95. Woodbury D, Reynolds K, Black IB. Adult bone marrow stromal stem cells express germline, ectodermal, endodermal, and mesodermal genes prior to neurogenesis. J Neurosci Res. 2002;69(6):908‐917. [DOI] [PubMed] [Google Scholar]
  • 96. Croft AP, Przyborski SA. Formation of neurons by non‐neural adult stem cells: potential mechanism implicates an artifact of growth in culture. Stem Cells. 2006;24(8):1841‐1851. [DOI] [PubMed] [Google Scholar]
  • 97. Lu P, Blesch A, Tuszynski MH. Induction of bone marrow stromal cells to neurons: differentiation, transdifferentiation, or artifact? J Neurosci Res. 2004;77(2):174‐191. [DOI] [PubMed] [Google Scholar]
  • 98. Barnabé GF, Schwindt TT, Calcagnotto ME, et al. Chemically‐induced RAT mesenchymal stem cells adopt molecular properties of neuronal‐like cells but do not have basic neuronal functional properties. PLoS One. 2009;4(4):e5222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Chen Q, Long Y, Yuan X, et al. Protective effects of bone marrow stromal cell transplantation in injured rodent brain: synthesis of neurotrophic factors. J Neurosci Res. 2005;80(5):611‐619. [DOI] [PubMed] [Google Scholar]
  • 100. Feng Y, Ju Y, Cui J, Wang L. Bone marrow stromal cells promote neuromotor functional recovery, via upregulation of neurotrophic factors and synapse proteins following traumatic brain injury in rats. Mol Med Rep. 2017;16(1):654‐660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Kim HJ, Lee JH, Kim SH. Therapeutic effects of human mesenchymal stem cells on traumatic brain injury in rats: secretion of neurotrophic factors and inhibition of apoptosis. J Neurotrauma. 2010;27(1):131‐138. [DOI] [PubMed] [Google Scholar]
  • 102. Yang Y, Ye Y, Su X, He J, Bai W, He X. MSCs‐derived exosomes and neuroinflammation, neurogenesis and therapy of traumatic brain injury. Front Cell Neurosci. 2017;11:55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Matthay MA, Pati S, Lee JW. Concise review: mesenchymal stem (stromal) cells: biology and preclinical evidence for therapeutic potential for organ dysfunction following trauma or sepsis. Stem Cells. 2017;35(2):316‐324. [DOI] [PubMed] [Google Scholar]
  • 104. Xiong Y, Mahmood A, Chopp M. Emerging potential of exosomes for treatment of traumatic brain injury. Neural Regen Res. 2017;12(1):19‐22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Kim DK, Nishida H, An SY, Shetty AK, Bartosh TJ, Prockop DJ. Chromatographically isolated CD63+CD81+ extracellular vesicles from mesenchymal stromal cells rescue cognitive impairments after TBI. Proc Natl Acad Sci USA. 2016;113(1):170‐175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Ni H, Yang SU, Siaw‐Debrah F, et al. Exosomes derived from bone mesenchymal stem cells ameliorate early inflammatory responses following traumatic brain injury. Front Neurosci. 2019;13:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Zhang Y, Chopp M, Zhang ZG, et al. Systemic administration of cell‐free exosomes generated by human bone marrow derived mesenchymal stem cells cultured under 2D and 3D conditions improves functional recovery in rats after traumatic brain injury. Neurochem Int. 2017;111:69‐81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Williams AM, Dennahy IS, Bhatti UF, et al. mesenchymal stem cell‐derived exosomes provide neuroprotection and improve long‐term neurologic outcomes in a swine model of traumatic brain injury and hemorrhagic shock. J Neurotrauma. 2019;36(1):54‐60. [DOI] [PubMed] [Google Scholar]
  • 109. Le Blanc K, Tammik L, Sundberg B, Haynesworth SE, Ringdén O. Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompatibility complex. Scand J Immunol. 2003;57(1):11‐20. [DOI] [PubMed] [Google Scholar]
  • 110. Maitra B, Szekely E, Gjini K, et al. Human mesenchymal stem cells support unrelated donor hematopoietic stem cells and suppress T‐cell activation. Bone Marrow Transplant. 2004;33(6):597‐604. [DOI] [PubMed] [Google Scholar]
  • 111. Lin C‐H, Lin W, Su Y‐C, et al. Modulation of parietal cytokine and chemokine gene profiles by mesenchymal stem cell as a basis for neurotrauma recovery. J Formos Med Assoc. 2019;118(12):1661–1673. [DOI] [PubMed] [Google Scholar]
  • 112. Liu YI, Zhang R, Yan KE, et al. Mesenchymal stem cells inhibit lipopolysaccharide‐induced inflammatory responses of BV2 microglial cells through TSG‐6. J Neuroinflammation. 2014;11:135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Zhang R, Liu YI, Yan KE, et al. Anti‐inflammatory and immunomodulatory mechanisms of mesenchymal stem cell transplantation in experimental traumatic brain injury. J Neuroinflammation. 2013;10:106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Liu XY, Wei MG, Liang J, et al. Injury‐preconditioning secretome of UCMSCs amplified the neurogenesis and cognitive recovery after severe traumatic brain injury in rats. J Neurochem. 2020;153:230‐251. [DOI] [PubMed] [Google Scholar]
  • 115. Shin MS, Park HK, Kim TW, et al. Neuroprotective effects of bone marrow stromal cell transplantation in combination with treadmill exercise following traumatic brain injury. Int Neurourol J. 2016;20(Suppl 1):S49‐S56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Guo S, Zhen Y, Wang A. Transplantation of bone mesenchymal stem cells promotes angiogenesis and improves neurological function after traumatic brain injury in mouse. Neuropsychiatr Dis Treat. 2017;13:2757‐2765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Li L, Chopp M, Ding GL, et al. MRI measurement of angiogenesis and the therapeutic effect of acute marrow stromal cell administration on traumatic brain injury. J Cereb Blood Flow Metab. 2012;32(11):2023‐2032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Isele NB, Lee H‐S, Landshamer S, et al. Bone marrow stromal cells mediate protection through stimulation of PI3‐K/Akt and MAPK signaling in neurons. Neurochem Int. 2007;50(1):243‐250. [DOI] [PubMed] [Google Scholar]
  • 119. Xu L, Xing QU, Huang T, et al. HDAC1 silence promotes neuroprotective effects of human umbilical cord‐derived mesenchymal stem cells in a mouse model of traumatic brain injury via PI3K/AKT pathway. Front Cell Neurosci. 2018;12:498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Thakor DK, Wang L, Benedict D, Kabatas S, Zafonte RD, Teng YD. Establishing an organotypic system for investigating multimodal neural repair effects of human mesenchymal stromal stem cells. Curr Protoc Stem Cell Biol. 2018;47(1):e58. [DOI] [PubMed] [Google Scholar]
  • 121. Zeng X, Ma YH, Chen YF, et al. Autocrine fibronectin from differentiating mesenchymal stem cells induces the neurite elongation in vitro and promotes nerve fiber regeneration in transected spinal cord injury. J Biomed Mater Res A. 2016;104(8):1902‐1911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Cox CS, Baumgartner JE, Harting MT, et al. Autologous bone marrow mononuclear cell therapy for severe traumatic brain injury in children. Neurosurgery. 2011;68(3):588‐600. [DOI] [PubMed] [Google Scholar]
  • 123. Liao GP, Harting MT, Hetz RA, et al. Autologous bone marrow mononuclear cells reduce therapeutic intensity for severe traumatic brain injury in children. Pediatr Crit Care Med. 2015;16(3):245‐255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Cox CS, Hetz RA, Liao GP, et al. Treatment of severe adult traumatic brain injury using bone marrow mononuclear cells. Stem Cells. 2017;35(4):1065‐1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Steinberg GK, Kondziolka D, Wechsler LR, et al. Clinical outcomes of transplanted modified bone marrow‐derived mesenchymal stem cells in stroke: a phase 1/2a study. Stroke. 2016;47(7):1817‐1824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Tatebayashi K, Takagi T, Fujita M, et al. Adipose‐derived stem cell therapy inhibits the deterioration of cerebral infarction by altering macrophage kinetics. Brain Res. 2019;1712:139‐150. [DOI] [PubMed] [Google Scholar]
  • 127. Xu K, Lee J‐Y, Kaneko Y, et al. Human stem cells transplanted into the rat stroke brain migrate to the spleen via lymphatic and inflammation pathways. Haematologica. 2019;104(5):1062‐1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Oh JS, Ha Y, An SS, et al. 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(3):215‐219. [DOI] [PubMed] [Google Scholar]
  • 129. Garcia‐Olmo D, Garcia‐Arranz M, Herreros D, Pascual I, Peiro C, Rodriguez‐Montes JA. A phase I clinical trial of the treatment of Crohn's fistula by adipose mesenchymal stem cell transplantation. Dis Colon Rectum. 2005;48(7):1416‐1423. [DOI] [PubMed] [Google Scholar]
  • 130. Bonilla Horcajo C, Zurita Castillo M, Vaquero CJ. Platelet‐rich plasma‐derived scaffolds increase the benefit of delayed mesenchymal stromal cell therapy after severe traumatic brain injury. Cytotherapy. 2018;20(3):314‐321. [DOI] [PubMed] [Google Scholar]
  • 131. Mahmood A, Goussev A, Lu D, et al. Long‐lasting benefits after treatment of traumatic brain injury (TBI) in rats with combination therapy of marrow stromal cells (MSCs) and simvastatin. J Neurotrauma. 2008;25(12):1441‐1447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Mahmood A, Lu D, Qu C, Goussev A, Chopp M. Treatment of traumatic brain injury with a combination therapy of marrow stromal cells and atorvastatin in rats. Neurosurgery. 2007;60(3):546‐554; discussion 53–54. [DOI] [PubMed] [Google Scholar]
  • 133. Kota DJ, Prabhakara KS, van Brummen AJ, et al. Propranolol and mesenchymal stromal cells combine to treat traumatic brain injury. Stem Cells Transl Med. 2016;5(1):33‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Hu J, Chen L, Huang X, et al. Calpain inhibitor MDL28170 improves the transplantation‐mediated therapeutic effect of bone marrow‐derived mesenchymal stem cells following traumatic brain injury. Stem Cell Res Ther. 2019;10(1):96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Das M, Leonardo CC, Rangooni S, Pennypacker KR, Mohapatra S, Mohapatra SS. Lateral fluid percussion injury of the brain induces CCL20 inflammatory chemokine expression in rats. J Neuroinflammation. 2011;8:148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Leonardo CC, Musso J, Das M, et al. CCL20 is associated with neurodegeneration following experimental traumatic brain injury and promotes cellular toxicity in vitro. Transl Stroke Res. 2012;3(3):357‐363. [DOI] [PubMed] [Google Scholar]
  • 137. Peruzzaro ST, Andrews MMM, Al‐Gharaibeh A, et al. Transplantation of mesenchymal stem cells genetically engineered to overexpress interleukin‐10 promotes alternative inflammatory response in rat model of traumatic brain injury. J Neuroinflammation. 2019;16(1):2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Minnich JE, Mann SL, Stock M, et al. Glial cell line‐derived neurotrophic factor (GDNF) gene delivery protects cortical neurons from dying following a traumatic brain injury. Restor Neurol Neurosci. 2010;28(3):293‐309. [DOI] [PubMed] [Google Scholar]
  • 139. Scafidi J, Hammond TR, Scafidi S, et al. Intranasal epidermal growth factor treatment rescues neonatal brain injury. Nature. 2014;506(7487):230‐234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Zhang A, Liang L, Niu H, Xu P, Hao Y. Protective effects of VEGF treatment on focal cerebral ischemia in rats. Mol Med Rep. 2012;6(6):1315‐1318. [DOI] [PubMed] [Google Scholar]
  • 141. Wang Z, Wang Z, Lu WW, Zhen W, Yang D, Peng S. Novel biomaterial strategies for controlled growth factor delivery for biomedical applications. NPG Asia Mater. 2017;9(10):e435‐e435. [Google Scholar]
  • 142. Aloe L, Rocco ML, Bianchi P, Manni L. Nerve growth factor: from the early discoveries to the potential clinical use. J Transl Med. 2012;10(1):239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Liu Y, Yi X‐C, Guo G, et al. Basic fibroblast growth factor increases the transplantation‐mediated therapeutic effect of bone mesenchymal stem cells following traumatic brain injury. Mol Med Rep. 2014;9(1):333‐339. [DOI] [PubMed] [Google Scholar]
  • 144. Huat TJ, Khan AA, Pati S, Mustafa Z, Abdullah JM, Jaafar H. IGF‐1 enhances cell proliferation and survival during early differentiation of mesenchymal stem cells to neural progenitor‐like cells. BMC Neurosci. 2014;15:91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Hatton J, Kryscio R, Ryan M, Ott L, Young B. Systemic metabolic effects of combined insulin‐like growth factor‐I and growth hormone therapy in patients who have sustained acute traumatic brain injury. J Neurosurg. 2006;105(6):843‐852. [DOI] [PubMed] [Google Scholar]
  • 146. Li XS, Williams M, Bartlett WP. Induction of IGF‐1 mRNA expression following traumatic injury to the postnatal brain. Brain Res Mol Brain Res. 1998;57(1):92‐96. [DOI] [PubMed] [Google Scholar]
  • 147. Eriksdotter Jönhagen M, Nordberg A, Amberla K, et al. Intracerebroventricular Infusion of nerve growth factor in three patients with Alzheimer's disease. Dement Geriatr Cogn Disord. 1998;9(5):246‐257. [DOI] [PubMed] [Google Scholar]
  • 148. Shahror RA, Linares GR, Wang Y, et al. Transplantation of mesenchymal stem cells overexpressing fibroblast growth factor 21 facilitates cognitive recovery and enhances neurogenesis in a mouse model of traumatic brain injury. J Neurotrauma. 2020;37(1):14‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Wu KE, Huang D, Zhu C, et al. NT3(P75–2) gene‐modified bone mesenchymal stem cells improve neurological function recovery in mouse TBI model. Stem Cell Res Ther. 2019;10(1):311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Wang S, Cheng H, Dai G, et al. Umbilical cord mesenchymal stem cell transplantation significantly improves neurological function in patients with sequelae of traumatic brain injury. Brain Res. 2013;1532:76‐84. [DOI] [PubMed] [Google Scholar]
  • 151. Wang Z, Wang YU, Wang Z, et al. Engineered mesenchymal stem cells with enhanced tropism and paracrine secretion of cytokines and growth factors to treat traumatic brain injury. Stem Cells. 2015;33(2):456‐467. [DOI] [PubMed] [Google Scholar]
  • 152. Huang C, Zhao L, Gu J, et al. The migration and differentiation of hUC‐MSCs(CXCR4/GFP) encapsulated in BDNF/chitosan scaffolds for brain tissue engineering. Biomed Mater. 2016;11(3):035004. [DOI] [PubMed] [Google Scholar]
  • 153. Bhang SH, Lee YE, Cho S‐W, et al. Basic fibroblast growth factor promotes bone marrow stromal cell transplantation‐mediated neural regeneration in traumatic brain injury. Biochem Biophys Res Commun. 2007;359(1):40‐45. [DOI] [PubMed] [Google Scholar]
  • 154. Shi W, Huang CJ, Xu XD, et al. Transplantation of RADA16‐BDNF peptide scaffold with human umbilical cord mesenchymal stem cells forced with CXCR4 and activated astrocytes for repair of traumatic brain injury. Acta Biomater. 2016;45:247‐261. [DOI] [PubMed] [Google Scholar]
  • 155. Satani N, Giridhar K, Cai C, et al. Aspirin in stroke patients modifies the immunomodulatory interactions of marrow stromal cells and monocytes. Brain Res. 2019;1720:146298. [DOI] [PubMed] [Google Scholar]
  • 156. Shamsara A, Sheibani V, Asadi‐Shekaari M, Nematollahi‐Mahani SN. Neural like cells and acetyl‐salicylic acid alter rat brain structure and function following transient middle cerebral artery occlusion. Biomol Concepts. 2018;9(1):155‐168. [DOI] [PubMed] [Google Scholar]
  • 157. Chen J, Ye X, Yan T, et al. Adverse effects of bone marrow stromal cell treatment of stroke in diabetic rats. Stroke. 2011;42(12):3551‐3558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Yan T, Ye X, Chopp M, et al. Niaspan attenuates the adverse effects of bone marrow stromal cell treatment of stroke in type one diabetic rats. PLoS One. 2013;8(11):e81199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Vaquero J, Zurita M, Rico MA, et al. Repeated subarachnoid administrations of autologous mesenchymal stromal cells supported in autologous plasma improve quality of life in patients suffering incomplete spinal cord injury. Cytotherapy. 2017;19(3):349‐359. [DOI] [PubMed] [Google Scholar]
  • 160. Oh K‐W, Moon C, Kim HY, et al. Phase I trial of repeated intrathecal autologous bone marrow‐derived mesenchymal stromal cells in amyotrophic lateral sclerosis. Stem Cells Transl Med. 2015;4(6):590‐597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Sorby‐Adams AJ, Vink R, Turner RJ. Large animal models of stroke and traumatic brain injury as translational tools. Am J Physiol Regul Integr Comp Physiol. 2018;315(2):R165‐R190. [DOI] [PubMed] [Google Scholar]
  • 162. Kinder HA, Baker EW, West FD. The pig as a preclinical traumatic brain injury model: current models, functional outcome measures, and translational detection strategies. Neural Regen Res. 2019;14(3):413‐424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Díez‐Tejedor E, Gutiérrez‐Fernández M, Martínez‐Sánchez P, et al. Reparative therapy for acute ischemic stroke with allogeneic mesenchymal stem cells from adipose tissue: a safety assessment: a phase II randomized, double‐blind, placebo‐controlled, single‐center, pilot clinical trial. J Stroke Cerebrovasc Dis. 2014;23(10):2694‐2700. [DOI] [PubMed] [Google Scholar]
  • 164. Hess DC, Sila CA, Furlan AJ, Wechsler LR, Switzer JA, Mays RW. A double‐blind placebo‐controlled clinical evaluation of MultiStem for the treatment of ischemic stroke. Int J Stroke. 2014;9(3):381‐386. [DOI] [PubMed] [Google Scholar]
  • 165. Lee JS, Hong JM, Moon GJ, Lee PH, Ahn YH, Bang OY. A long‐term follow‐up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem Cells. 2010;28(6):1099‐1106. [DOI] [PubMed] [Google Scholar]
  • 166. Bang OY, Lee JS, Lee PH, Lee G. Autologous mesenchymal stem cell transplantation in stroke patients. Ann Neurol. 2005;57(6):874‐882. [DOI] [PubMed] [Google Scholar]
  • 167. Mohyeddin Bonab M, Ali Sahraian M, Aghsaie A, et al. Autologous mesenchymal stem cell therapy in progressive multiple sclerosis: an open label study. Curr Stem Cell Res Ther. 2012;7(6):407‐414. [DOI] [PubMed] [Google Scholar]
  • 168. Connick P, Kolappan M, Crawley C, et al. Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open‐label phase 2a proof‐of‐concept study. Lancet Neurol. 2012;11(2):150‐156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Dahbour S, Jamali F, Alhattab D, et al. Mesenchymal stem cells and conditioned media in the treatment of multiple sclerosis patients: clinical, ophthalmological and radiological assessments of safety and efficacy. CNS Neurosci Ther. 2017;23(11):866‐874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Fernández O, Izquierdo G, Fernández V, et al. Adipose‐derived mesenchymal stem cells (AdMSC) for the treatment of secondary‐progressive multiple sclerosis: A triple blinded, placebo controlled, randomized phase I/II safety and feasibility study. PLoS One. 2018;13(5):e0195891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Karussis D, Karageorgiou C, Vaknin‐Dembinsky A, et al. Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Arch Neurol. 2010;67(10):1187‐1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Li J‐F, Zhang D‐J, Geng T, et al. The potential of human umbilical cord‐derived mesenchymal stem cells as a novel cellular therapy for multiple sclerosis. Cell Transplant. 2014;23(Suppl 1):S113‐S122. [DOI] [PubMed] [Google Scholar]
  • 173. Llufriu S, Sepúlveda M, Blanco Y, et al. Randomized placebo‐controlled phase II trial of autologous mesenchymal stem cells in multiple sclerosis. PLoS One. 2014;9(12):e113936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Riordan NH, Morales I, Fernández G, et al. Clinical feasibility of umbilical cord tissue‐derived mesenchymal stem cells in the treatment of multiple sclerosis. J Transl Med. 2018;16(1):57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Kim HJ, Seo SW, Chang JW, et al. Stereotactic brain injection of human umbilical cord blood mesenchymal stem cells in patients with Alzheimer's disease dementia: a phase 1 clinical trial. Alzheimers Dement (N Y). 2015;1(2):95‐102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Tian C, Wang X, Wang X, et al. Autologous bone marrow mesenchymal stem cell therapy in the subacute stage of traumatic brain injury by lumbar puncture. Exp Clin Transplant. 2013;11(2):176‐181. [DOI] [PubMed] [Google Scholar]

Articles from CNS Neuroscience & Therapeutics are provided here courtesy of Wiley

RESOURCES