Progr Neurobiology Con Portada

download Progr Neurobiology Con Portada

of 10

Transcript of Progr Neurobiology Con Portada

  • 8/3/2019 Progr Neurobiology Con Portada

    1/10

    This article appeared in a journal published by Elsevier. The attached

    copy is furnished to the author for internal non-commercial research

    and education use, including for instruction at the authors institution

    and sharing with colleagues.

    Other uses, including reproduction and distribution, or selling or

    licensing copies, or posting to personal, institutional or third partywebsites are prohibited.

    In most cases authors are permitted to post their version of the

    article (e.g. in Word or Tex form) to their personal website or

    institutional repository. Authors requiring further information

    regarding Elseviers archiving and manuscript policies are

    encouraged to visit:

    http://www.elsevier.com/copyright

    http://www.elsevier.com/copyrighthttp://www.elsevier.com/copyright
  • 8/3/2019 Progr Neurobiology Con Portada

    2/10

    Author's personal copy

    Functional recovery after severe CNS trauma: Current perspectives for celltherapy with bone marrow stromal cells

    Jesus Vaquero *, Mercedes Zurita

    Surgical Research Service, Neurosciences Unit, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain

    Contents

    1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341

    2. Cell therapy strategies for TBI. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342

    3. Current perspectives for BMSC in the treatment of TBI. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343

    4. Cell therapy strategies for SCI. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 344

    5. Current perspectives for BMSC in the treatment of SCI. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 345

    6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 347

    Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 347

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 347

    Between these two faults, boldness is preferable to timidity. Dare

    wins or is defeated, but excessive prudence leads us to a shameful

    inactivity.

    Santiago Ramon y Cajal, 1923

    1. Introduction

    In recent years, numerous research groups have studied on

    experimental models of brain or spinal cord trauma the possible

    therapeutic effect of neurotrophic factors, able to create in the

    central nervous system (CNS) an environment conducive to

    regeneration. Among these neurotrophic factors, nerve growth

    factor (NGF) (Dixon et al., 1997; Zhou et al., 2003), fibroblastgrowth factor-2 (FGF-2) (Yoshimura et al., 2003), and insulin-

    like growth factor-1 (IGF-1) (Saatman et al., 1997) have been

    considered. Simultaneously, gene therapy protocols, which are

    able to bring these factors to damaged areas, have been studied

    (Longhi et al., 2004). Although these new strategies offer

    interesting perspectives, the truth is that human clinical

    applications still require many studies regarding the routes

    and patterns of administration and the use of viral vectors. A

    second line of research with an increasing importance in recent

    years is the use of cell therapy with stem cells. These cells would

    be able firstly, to release neurotrophic factors, and secondly, to

    regenerate damaged nerve tissue through differentiation or

    transdifferentiation into mature neural cells. The first experi-

    mental studies used fetal tissue or embryonic stem cells, but it is

    Progress in Neurobiology 93 (2011) 341349

    A R T I C L E I N F O

    Article history:

    Received 4 September 2010

    Received in revised form 30 November 2010

    Accepted 7 December 2010

    Available online 14 December 2010

    Keywords:

    Bone marrow stromal cells

    Cell transplantation

    Traumatic brain injury

    Traumatic spinal cord injury

    Stem cells

    A B S T R A C T

    The aim of this paper is to identify current perspectives for cell therapy applied to traumatic injuries of

    the central nervous system (CNS). After using diverse types of cell therapy, at present there is a growing

    experimentalevidence thattransplantation of bone marrow stromalcells (BMSC) can be useful to reverse

    the sequels of trauma affecting the brain and spinal cord. Although we still do not know many details

    about how these cells achieve their beneficial effects, the application of BMSC in humans, for brain or

    spinal cord repair, is beginning. An exquisite caution and strict methodological controls are needed to

    determine with certainty whether we can open a door of hope for many patients who currently suffer

    severe neurological deficits that are now supposedly irreversible.

    2010 Elsevier Ltd. All rights reserved.

    Abbreviations: BMSC, bone marrow stromal cells; CNS, central nervous system;

    FGF-2, fibroblast growth factor-2; IGF-1, insulin like growth factor; iPS, induced

    pluripotent stem cells;NSC, neural stemcells; SCI,spinal cord injury; TBI,traumatic

    brain injury.

    * Corresponding author.

    E-mail address: [email protected] (J. Vaquero).

    Contents lists available at ScienceDirect

    Progress in Neurobiology

    j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / p n e u r o b i o

    0301-0082/$ see front matter 2010 Elsevier Ltd. All rights reserved.

    doi:10.1016/j.pneurobio.2010.12.002

  • 8/3/2019 Progr Neurobiology Con Portada

    3/10

    Author's personal copy

    difficult for these studies to lead to clinical applications in

    patients because of logistical, immunological, and ethical

    reasons (Widner and Brundin, 1988; Hoffer and Olson, 1991).

    Furthermore, the potential usefulness of embryonic stem cells

    for the treatment of human diseases has been hampered by the

    risk of tumorigenesis (Wu et al., 2007).

    Multipotent neural progenitors and neural stem cells (NSC) canbe isolated from either embryonic or adult brain tissue and,

    following engraftmentintothe adultbrain, theydifferentiate mainly

    into glial cells, suggesting possible efficacy in cell therapy strategies

    applied to traumatic CNS injuries (Park et al., 1999). At present, the

    functional effect achieved for these cells in the treatment of

    experimental CNS injuries has been very limited, and their clinical

    usealso raises many problems becauseof the difficulty of obtaining

    NSC (Cao et al., 2002). The use of immortalized neural cell lines

    represents an alternative strategy (Whittemore and Onifer, 2000)

    but raises the possibility of risks inherent to genetic manipulation,

    which in practice means that many issues must be resolved before

    considering its clinical application. The same considerations can be

    applied to recent studies with the so-called induced pluripotent

    stem cells (iPS), which can achieve remyelinization, axonal

    regrowth, and functional improvement in spinal cord injury (SCI)

    models (Tsuji et al., 2010). Because of these limitations, researchers

    have paidattention to adult stemcells generally obtained frombone

    marrow stroma (Opydo-Chanek, 2007).

    The growing interest in cell therapy has resulted in much

    knowledge about the many of the details of bone marrow stromal

    cells (BMSC). These cells are isolated from the mononuclear

    fraction of bone marrow and represent a population of undiffer-

    entiated cells that do not express markers of hematopoietic stem

    cells, such as CD14, CD19, CD34, CD45 and CD79, but they are

    positive for CD73, CD105, and other adhesion molecules, such as

    CD90 and CD106 (Horwitz et al., 2005; Dominici et al., 2006). We

    also know that BMSC show a low expression of the major

    histocompatibility complex antigens (Class II). Therefore, these

    cells have low antigenicity, which is one of the main advantages in

    the consideration of cell therapy protocols (Le Blanc and Ringden,

    2005). Furthermore, these cells show a high expression of growth

    factors, cytokines, and extracellular matrix molecules that, under

    normal conditions, can contribute to the formation and function of

    the bone marrow stromal microenvironment, inducing regulatory

    signals not only for stromal stem cells themselves but also for

    hematopoietic stem cells (Garca et al., 2004; Chen et al., 2005; Ye

    et al., 2005).

    Stromal cell cultures are morphologically heterogeneous and

    show different cell types including a population of elongated cells

    with low proliferation and differentiation, and a second population

    of cells with a high capacity of proliferation and differentiation.

    This latter cell population, in turn, can be of two types: (1) some

    very small and rounded cells, or (2) cells that are larger withsomewhatstarry elements that appear to be progenitors of stromal

    cells in culture. We accept that within the set of stromal stem cells

    there is a subpopulation of cells known as multipotent adult

    progenitor (MAP) cells, which proliferate indefinitely in vitro. It is

    assumed that they are probably a rare form of stem cells that are

    maintained with this potential from the embryonic period until

    adulthood, and that, under certain circumstances, they can

    differentiate into the cells of all three embryonic layers (Jiang

    et al., 2002; Zeng et al., 2006; Raedt et al., 2007).

    Earlier this decade, a diverse set of studies suggested that BMSC

    can undergo in vitro transdifferentiation phenomenon when the

    medium is treated with different chemical agents, resulting in cells

    withadult neuronal-likemorphology (Woodburyet al.,2000;Mezey

    and Chandross, 2000; Sanchez-Ramos et al.,2000; Hung et al.,2002;Dezawa et al., 2004; Hermann et al., 2004; Bossolasco et al., 2005).

    Several authors have questioned whether this is true neuronal

    transdifferentiation of BMSC, because the transformation into

    neuronal-like morphology could be due to nonspecific changes of

    the cell cytoskeleton. The truth is that there is increasing evidence,

    both in vitro and in vivo, that it is possible to transform BMSC to

    neurons and glial cells, thus creating the potential to regenerate

    injured CNS tissue (Zurita and Vaquero, 2004, 2006; Zurita et al.,

    2005, 2008; Vaquero et al., 2006; Bonilla et al., 2009).Studies done in our lab using BMSC for CNS trauma were

    conducted in experimental models of paraplegia and have

    reported that BMSC can regenerate the injured spinal cord and

    produce newly formed nervoustissuethrough which axons cango

    upward and downward in the year after transplantation (Zurita

    andVaquero, 2006). In these studies,the labelingof BMSC through

    gene transfection showed that neurons and glial cells present in

    the regenerated tissue came from the transplanted BMSC. An

    important detail was to test how BMSC also differentiate to cells

    that form new vessels and promote angiogenesis (Zurita and

    Vaquero, 2006; Zuritaet al.,2008). Inthe caseof traumatic injuries

    of the spinal cord, it is relatively easy to quantify the functional

    results obtained with these techniques and to study the

    morphological changes that can occur in previously injured

    areas. However, the evaluation of neurological deficits and the

    possibility of recovery are more difficult in the case of

    experimental traumatic brain injury (TBI). Nevertheless, numer-

    ous studies are underway to determine whether the therapeutic

    effects that seem to make BMSC useful in the treatment of

    experimental spinal cord lesions are also applicable to the

    treatment of severe TBI.

    2. Cell therapy strategies for TBI

    Our understanding of cell therapy for TBI has accrued over the

    last decades and has shown promise in the management of this

    devastating disease. Sinson et al. (1996) grafted fetal rodent

    cortical tissue into the injured brains of adult rats resulting in

    significant improvement in motor and cognitive function. The

    potential effectiveness of neural stem cells engineered to secrete

    neurotrophic factors has been studied by some authors on

    experimental models of TBI and usually during the acute period

    after trauma. So, Philips et al. (2001) described positive results

    after intracerebral transplantation of immortalized neural stem

    cells that were retrovirally transduced to produce NGF and Bakshi

    et al. (2006b) reported cognitive function improvement after

    transplantation into the perilesional brain tissue of neural

    progenitor cells engineered to secrete glial cell line-derived

    neurotrophic factor (GDNF). However, other studies using neural

    stem cells have shown no functional improvement (McMahon et

    al., 2010). In any case, when analyzing the literature, we can see

    that in contrast to what has happened in SCI research, experimen-

    tal studies using cell therapy in TBI are relatively few, and theresults have been contradictory (Harting et al., 2008). Because

    severe experimental brain trauma produces significant loss of

    brain tissue in rodents andthe difficulty in achievingan acceptable

    survival of transplanted cells in cell therapy protocols, the results

    are difficult to evaluate.

    Nevertheless in recent years, Mahmood et al. reported

    numerous experimental studies using BMSC for the treatment of

    TBI (Mahmood et al., 2001a,b, 2002, 2003, 2004a,b, 2005, 2006,

    2007; Lu et al., 2001). In these studies, cell therapy was used in the

    early phases after trauma and a significant reduction of

    neurological deficits was observed. Because of these and other

    observations, BMSC appear to be highly promising for the

    treatment of severe TBI to obtain neuroprotection or functional

    recovery. In parallel, clinical trials in children using an intravenousadministration of autologous bone marrow-derived mononuclear

    cells have been initiated (Harting et al., 2008).

    J. Vaquero, M. Zurita / Progress in Neurobiology 93 (2011) 341349342

  • 8/3/2019 Progr Neurobiology Con Portada

    4/10

    Author's personal copy

    3. Current perspectives for BMSC in the treatment of TBI

    At present, the experimental studies by Mahmood et al. have

    provided further evidence about the possibility of reversing

    functional deficits in adult rats subjected to traumatic brain injury

    through the administration of adult mesenchymal stem cells

    obtained from bone marrow stroma. This group has reported inrecent years that BMSC reverse functional deficits when cells were

    administered locally, intravenously, or intra-arterially (Mahmood

    et al., 2001a,b, 2002, 2003, 2004a,b, 2005, 2006, 2007; Lu et al.,

    2001). This effect is accompanied by identification of these cells in

    areas of brain damage, irrespective of the route of administration,

    and morphological characteristics suggesting their differentiation

    into neural cells (Lu et al., 2001, 2006). However, these studies

    generally show a discrepancy between the positive results

    obtained and the low rate of cell transdifferentiation, suggesting

    that the effect of stromal cells, at least in part, may be due to the

    release of neurotrophic factors that induce regeneration in the host

    tissue.

    Conclusion from these experiences is that beneficial effect can

    be achieved when human stromal cells are administered

    intravenously to rat TBI models, and without signs of immune

    rejection (Mahmood et al., 2003). This finding supports the low

    antigenicity of BMSC, which is an important argument when

    clinical applications are considered. Perhaps the biggest criticism

    of these experiences lies in the fact that in almost all of the studies,

    BMSC are administeredat an early stage after TBI, whichis far from

    the actual conditions in which these new therapies could be

    applied in humans; it seems reasonable that patients would only

    use this technique during a chronic phase after having exhausted

    the possibilities of rehabilitation treatment. Moreover, the

    variability of brain lesions after an experimental trauma, and

    the variable functional recovery of animals subjected to similar

    parameters of traumatic injury, makes it difficult to evaluate theactual effectiveness of cell therapy.

    We are currently developing experimental studies trying to

    confirm the therapeutic efficacy of the intracerebral administra-

    tion of BMSC during the chronic phase of severe TBI where there is

    an established neurological deficit. Our results in rodents have

    shownthat thelocal administration of 10 106 BMSCin the areaof

    brain injury two months after trauma achieved a clear recovery

    according to a functional assessment using a modified sensory-

    motor neurological deficit (mNSS) scale (see Fig. 1 and Supple-

    mental video 1). This improvement was morphologically correlat-

    ed with the presence of transplanted BMSC in the injured area,

    some of which showed transdifferentiation to neurons and glial

    cells together with an increase of endogenous neurogenesis

    (Bonilla et al., 2009).

    Although these initial findings appear promising, further

    studies are required. The size of the brain lesions and their

    variability appear to be determinants of the need to find biological

    matrices allowing cell survival and differentiation of the trans-

    planted stem cells. Similarly, the critical number of BMSC that are

    necessary to restore the functional deficits after experimental

    Fig. 1. BMSC can improve established neurological sequels after severe TBI in rodents (see Bonilla et al., 2009). (A) Macroscopical view of the brain two months after

    experimental TBI by weight-drop impact in Wistar rat. (B and C) Immunohistochemical studies in injured brain, two months after intracerebral administration of BMSC.

    Donor cells were obtained from male-donor rats, and they were transplanted into female rats subjected to TBI. Transplanted BMSC cells showing the SrY gene of Y-

    chromosome can be seen into the injury zone by means of in situ hybridization. In (B), double immunostain permits the identification of neurons showing SrY gene (arrows,

    SrY gene immunostained in red), and suggesting a neuronal transdifferentiation of the transplanted BMSC (green: neuron nuclei, showing expression of neuron-specific

    nuclear antigen (NeuN). Original magnification: 1000. In (C), cells with nuclear presence of SrY gene (arrows) and cytoplasmic expression of gliofibrillary acidic protein

    (GFAP, in green) can be seen,suggesting astroglial transdifferentiationof the transplanted BMSC.Original magnification:400. (DF)Histologicalfields in BMSC-treated rats,

    four months after severe TBI, and two months after local BMSC transplantation. (D) A stream of cells, identified as migratory neuroblasts due to doublecortin-positivity,

    extends from the subventricular zone (SVZ). Original magnification: 40. (E) Injury zone, showing doublecortin-positive cells at the edges (top). Original magnification:

    200. (F) Doublecortin-positive cells filling the injury zone can be found. Original magnification: 200. The associated graph shows the temporal profile of functional

    recovery in a seriesof brain-injured ratsthat received intracerebral administrationof saline(n: 1 0 )or 5 106 BMSC (n: 10)in saline, twomonths after severeTBI.UsingmNSS

    test, significant differences were detected starting 6 weeks after intracerebral BMSC transplantation; p < 0.05). See Supplemental video 1.

    J. Vaquero, M. Zurita / Progress in Neurobiology 93 (2011) 341349 343

  • 8/3/2019 Progr Neurobiology Con Portada

    5/10

    Author's personal copy

    brain trauma represents one of the main issues to be resolved. In

    one of the experiments reported by Mahmood et al. (2005),

    different doses of BMSC were intravenously administered in adult

    rats one week after TBI. Three months later, the rats that had

    received more than 4 106 BMSC showed some functional

    recovery, but rats that received only 2 106 BMSC did not,

    suggesting that the number of administered stem cells is adetermining factor for achievement of the therapeutic effect. If this

    hypothesis is correct, strategies should aim at making large

    quantities of adult stem cells that can reach areas of brain injury.

    Nevertheless at this point, one consideration must be born in mind.

    Although the number of administered stem cells is important, we

    believe that the future research in this field should aim to ensure

    that the cells survive a long-term in the brain. Regardless of the

    number of cells administered various factors are present in the

    host tissue, most of which are related to the pathophysiology of

    traumatic injury, that decreases the survival of the transplanted

    cells.

    However, the experiments conducted in our lab using BMSC

    labeled with Indium-111 suggest that, after the systemic

    administration of these cells, they preferentially colonize in

    infusion-rich organs, such as the liver and spleen, at least when

    the BMSC administration is performed during the chronic phase of

    CNS trauma (De Haro et al., 2005; Vaquero et al., 2006). Therefore,

    it is necessary to know the best route for the administration of cell

    therapy to achieve a better survival of the transplanted cells after

    administration and the best time for the application of cell therapy

    after trauma. Even though there are unresolved issues, it is obvious

    that in recentyears newtechniques of cell therapy with adult stem

    cells, together with the new concepts about the possibility of

    regeneration of the adult nervous system, have opened an avenue

    of hope for the treatment of the sequels of TBI. Any advance in this

    field, as in many other areas of neurobiology, will require close

    collaboration between basic and clinical researchers.

    4. Cell therapy strategies for SCI

    Although it has been classically considered that traumatic

    paraplegia is irreversible, over the last century, numerous

    experimental models have been designed to investigate if, in

    some cases, the limited regenerative activity following SCI as

    described by Ramon y Cajal (1914) can be increased. Around 1940,

    transplants were performed into injured spinal cord tissue with

    variable results (Brown and Mc Cough, 1947; Barnard and

    Carpenter, 1950). Subsequently, Kao (1974) tried to interpose

    various neural tissues, such as peripheral nerve, ganglion

    nodosum, or cultured cerebellar brain tissue, between the ends

    of a transected spinal cord in an attempt to restore its continuity.

    As a result of these studies, new evidence showed that the

    peripheral nerve, implanted between the ends of a transectedspinal cord, achieved a bridge while reducingthe local mesodermic

    scar. By 1980, the group of Aguayo, discussed both the anatomical

    and functional possibilities, of the bridges of peripheral nerve

    placed between the ends of a transected spinal cord ( David and

    Aguayo, 1981). Through axonal labeling techniques using peroxi-

    dase, these authors showed that spinal neurons projected their

    axons along these transplants and also provided evidence that, at

    least in part, the axons that invaded the transplants represented

    the regeneration of the previously damaged axons. These

    experiences, and those of Wrathall et al. (1982) or Fernandez et

    al. (1986), showed that peripheral nerve grafts may constitute a

    valid support for axonal growth perhaps due to trophic factors

    produced by Schwann cells. Despite this evidence, the literature

    contains few attempts at transplanting nervous tissue in theinjured spinal cord. The explanation appears to lie in the technical

    difficulty in performing these transplants and from the poor

    functional results obtained in the few experiences (Nygren et al.,

    1977; Das, 1983a,b, 1987).

    In association with these procedures, the placement of tubes

    of biological or synthetic materials surrounding the area of SCI has

    been studied. These tubes might act as a guide for regenerative

    axons especially when various neurotrophic substances are

    injected in the interior. The most commonly used materialsinclude biodegradable polymers of a polyester type that carry

    Schwann cells or brain derived neurotrophic factor (BDNF). The

    demonstration that olfactory bulb ensheathing cells are capable of

    achieving the myelination of CNS axons has also led to the use of

    these cells,togetherwithSchwann cells and the tube systems, with

    promising results in rats subjected to transection (Ramon-Cueto et

    al., 1998, 2000).

    However, the first clinical studies in human paraplegia using

    olfactory ensheathing cell transplantation have not demonstrated

    functional improvement (Mackay-Sim et al., 2008).

    Fetal neural cells have been used by a diverse set of researchers

    in an attempt to regain function after spinal cord trauma with

    experimental results that suggest that age may be a determining

    factor for functional recovery (Bregman et al., 1993; Bregman,

    1994; Diener and Bregman, 1998; Reier, 2004). These studies have

    been generally performed on incomplete SCI models during acute

    stages after injury. However, when looking for donor nervous

    tissue capable of reconstructing the injured spinal cord, it seems

    logical to consider the use of spinal cord tissue and specifically,

    fetal spinal cord tissue. Although the first experimental transplants

    of fetal spinal cord to a sectioned adult spinal cord showed low

    survival (Das, 1983a) compared with other types of transplants

    such as fetal brain tissue, the fetal spinal cord is seen as the type of

    donor tissue most suitable for these experimental studies. The

    experience using fetal spinal cord transplantation showed that the

    survival of the graft within the previously injured spinal cord

    depended on the age of the donor, and it is best when the fetus

    corresponded to a period between 13 and 15 days of gestation in

    the rat. We also know that such transplants can survive long time

    and experience changes that include the formation of unmyelin-

    ated areas that resemble the gelatinous substance of dorsal horn.

    Immunofluorescence studies indicate that neurons in the trans-

    plants may project axons to the host tissue (Reier, 1985). Pallini et

    al. (1989) reported their experience grafting fetal spinal cord (13

    14 days of gestation) into the spinal cord of adult rats. The

    transplant was done immediately after transection and achieved a

    survival rate of 55% with good morphological integration, but the

    clinical and electrophysiological assessment of the animals did not

    show any kind of functional recovery. However, there are other

    publications that show functional recovery of animals after

    transection and subsequent reconstruction with fetal spinal cord

    transplants (Zompa et al., 1997). In studies using fetal brain tissue

    to reconstruct the injured spinal cord, the first experimentsindicated a low viability of the grafts, perhaps as a result of a

    deficient surgical technique. At least theoretically, the use of fetal

    brain tissue may have the advantage of a high proliferative

    capacity, which helps to restore the injured tissue (Das, 1987).

    Although at present limited experience exists about the useof fetal

    cortex transplants in the intact or injured spinal cord (Bernstein et

    al., 1984; Das, 1987), the grafted tissue can survive in the adult

    spinal cord using an appropriate surgical technique, with easier

    integration at the level of the gray matter. However, most

    experimental models of SCI in which neural transplants have

    been performed have used spinal cord transection, which may

    differ significantly from what occurs in humans, where traumatic

    paraplegia is usually the result of a contusion injury. In addition,

    the neural transplants in these experimental models wereperformed almost always immediately after transection in models

    of acute paraplegia, such as the studies by Ramon-Cueto et al.

    J. Vaquero, M. Zurita / Progress in Neurobiology 93 (2011) 341349344

  • 8/3/2019 Progr Neurobiology Con Portada

    6/10

    Author's personal copy

    (2000) using olfactory bulb ensheathingcells. It must be taken into

    account that performing the transplant in an acute phase may be

    unfavorable to the survival of the transplanted tissue. Further-

    more, with the logical objective of a possible clinical application of

    these techniques, it is obvious that their use in humans should be

    done in phases of chronic paraplegia, where the chances of

    spontaneous functional recovery have already been dismissed.Bearing in mind all these considerations, by 1990 we aimed to

    study the functional and morphological results of delayed

    transplantation of fetal neocortical tissue in a model of spinal

    cord contusion causing chronic paraplegia. The use of neocortical

    tissue was based on the finding that this tissue shows a high

    mitotic capacity, which theoretically may lead to fill the area of

    posttraumatic intramedullary necrosis. As a result of these pilot

    studies (Vaquero et al., 1992a,b), we obtained evidence that

    transplantationof fetal brain tissuein the injured spinal cord in the

    adult rat achieved a survival rate greater than 80% if donor tissue

    was obtained from fetuses at 18 days of gestation and transplan-

    tation was performed at least one week after the traumatic injury.

    These results were similar if the transplantation was performed at

    one week or at 34 months after SCI, in a situation of chronically

    established paraplegia. Furthermore, grafted tissue showed

    integration with the host tissue and neurons within the

    transplanted tissue underwent maturational changes, showing

    the morphological appearance of cerebral neurons. At this point,

    the discrepancy between the good histological features (e.g., the

    integration of transplanted fetal brain tissue into the spinal cord)

    andthe absence of significant motor recovery led us to believe that

    the transplanted animals were followed for a sufficiently long

    evolutionary time, especially given the fact that experience of

    literature is limited to a few months of follow-up. A proper

    microsurgical technique, the prevention of infections, bladder

    emptying in the early stages after SCI, healing of pressure ulcers,

    and the presence of a single caregiver for our animals, were the

    factors that allowed us to maintain paraplegic animals for more

    than one year after transplantation. This achievement led us to the

    observation that six months after transplantation, the animals

    showedsigns of motorand sensory recoverythat was initiatedby a

    series of spontaneous movements of thetail and hind legs, together

    with a response to pain stimulus applied at these loci. However,

    animals that survive long-term after transplantation often have

    significant rigidities that prevent any joint movement. Thus by

    1995, we started a new experimental phase, incorporating

    intensive rehabilitation in the paraplegic animals that allowed

    us to keep them alive and free of joint stiffness and avoided, as in

    humans, most of the complications of chronically established

    paraplegia. At the same time, diverse donor neural tissues were

    studied, and the best tissue regeneration rates wereobtainedwhen

    the centromedullary posttraumatic cavities were filled with fetal

    brain tissue and peripheral nerve, possibly because of the knownneurotrophic effect of Schwann cells (Zurita et al., 2000, 2001).

    With this experimental model, we kept paraplegic rats more than

    two years after intralesional transplantation of fetal cerebral

    cortex with peripheral nerve fragments. A larger follow-up was

    limited by the age of the animal itself, which hardly reached the 2-

    year survival in Wistar rats. This technique allowed us to confirm

    that transplanted rats showed a progressive recovery of sensory

    and motor function that became very evident 68 months after

    transplantation. Furthermore, the transplanted rats showed

    recovery of muscle atrophy, which was clearly significant one

    year of transplantation (Zurita et al., 2001). Histological examina-

    tion of animals after more than one year of evolution showed a

    seamless integration of the transplanted tissue with identifiable

    adult brain tissue at the level of what was once an intramedullarycavity. Parallel to these experimental studies, some authors (Falci

    et al., 1997; Wirth et al., 2001) marked the starting point for the

    implementation of these new techniques of cell therapy in patients

    with established paraplegia. In these early clinical trials, fetal

    spinal cord tissue was used and, although some patients described

    recovery of sensation in the lower limbs, there was no clear motor

    recovery. In our opinion, these poor results could be due to the use

    of fetal spinal cord tissue rather than brain tissue because in our

    experience, fetal cerebral cortex tissue has a higher proliferativecapacity than fetal spinal cord tissue and the transplanted spinal

    cord tissue failed to completely fill the posttraumatic cavities

    (Wirth et al., 2001). According to our experimental studies, delayed

    transplantation of fetal spinal cord in the spinal cord of paraplegic

    adult rats has never achieved motor recovery, at least in the 10

    months following the transplantation procedure. However, based

    on similar parameters of neurological injury, the use of fetal brain

    tissue in co-transplantation with peripheral nerve provided

    acceptable functional results (Zurita et al., 2001). Regardless of

    these observations, it is obvious that the experimental animal and

    man represent different biological systems, making it difficult to

    predict that the results obtained in rodents can be successfully

    applied to the treatment of paraplegic patients. We must consider

    that studies with higher numbers of animals are problematic

    because of the difficulty in the long-term maintenance of the

    paraplegic animals and the difficulty of their rehabilitation.

    By 2000, the use of fetal brain tissue for transplantation into

    the posttraumatic spinal cord cavities in paraplegic patients

    appeared to be a reasonable option. However, this approach was

    faced with enormous technical difficulties because it was

    extremely difficult to obtain human brain tissue with good cell

    viability from either spontaneous or therapeutic abortions. In

    addition, the need for immunosuppression added a new difficulty

    to the technique, especially considering that paraplegic patients

    often have urinary tract infections. Given these difficulties, we

    considered alternatives in paraplegicpatients and fundamentally,

    we explored the possibilities for CNS regeneration provided from

    so-called adult stemcells. Among thedifferent types of adult stem

    cells, BMSC were soon regarded as potentially useful, in the same

    way that these cells had been considered for the treatment of TBI

    because of their easy availability and the possibility of autologous

    transplantation.

    5. Current perspectives for BMSC in the treatment of SCI

    After the first descriptions that BMSC may be transformed in

    vitro into neuronal-like cells with the addition of specific chemical

    agents to the culture medium (Woodbury et al., 2000; Mezey and

    Chandross, 2000; Sanchez-Ramos et al., 2000; Hung et al., 2002;

    Dezawa et al., 2004; Hermann et al., 2004; Bossolasco et al., 2005),

    several research groups studied the possibility of applying this to

    the clinic, specifically to the treatment of traumaticparaplegia. As a

    result, comments on the usefulness of BMSC to restore functionaldeficits in experimental models of acute or incomplete SCI were

    soon published (Chopp et al., 2000; Chopp and Li, 2002; Hofstetter

    et al., 2002; Lee et al., 2003; Ankeny et al., 2004 ). In 2004, we

    demonstrated in our laboratory that this form of cell therapy may

    also be usefulif applied to complete SCI in a situationof chronically

    established paraplegia (Zurita and Vaquero, 2004). In these

    experimental conditions, intralesional transplantation of BMSC

    was followed by clear signs of neurological recovery. At one year,

    an almost complete motor recovery was observed in over 60% of

    cases that was associated with an apparent regeneration of nerve

    tissue at the previously injured area (Vaquero et al., 2006; Zurita

    and Vaquero, 2006).

    Once we confirmed the efficacy of BMSC transplantation in the

    regeneration of damaged spinal tissue to achieve the motorrecovery of paraplegic animals, and keeping in mind that some

    publications indicated that intravenous administration of BMSC

    J. Vaquero, M. Zurita / Progress in Neurobiology 93 (2011) 341349 345

  • 8/3/2019 Progr Neurobiology Con Portada

    7/10

    Author's personal copy

    reversed functional deficits in rats subjected to TBI (Mahmood et

    al., 2001a) we asked whether the systemic administration of these

    cells could also be effective. As a first step, we labeled BMSC with

    bisbenzimide or with Indium-111 and verified the distribution of

    labeled cells following intravenous administration. The results

    showed that after intravenous administration, labeled BMSC

    preferentially colonized the spleen, liver, and kidneys but didnot significantly reach the area of the SCI (De Haro et al., 2005).

    These studies were the first to report the usefulness of Indium-111

    to the in vivo study of the distribution of BMSC after their

    administration in cell therapy procedures. However, it is obvious

    that systemic administration of BMSC prevents these cells from

    arriving in significant number to areas of traumatic SCI, at least

    during the chronic phase after SCI. In another study in chronic

    paraplegic rats, we compared the usefulness of systemic adminis-

    tration of BMSC with local administration, and a clear difference in

    favor of the local administration was found (Vaquero et al., 2006).

    These good results, after more than 150 transplants in rodents,

    have been reproduced in adult pigs with chronically established

    paraplegia, which suggests that this technique may also be helpful

    in humans (Zurita et al., 2008) (see Fig. 2 and Supplemental videos

    2 and 3).

    As a result of these and other experimental studies, the

    implementation of this type of cell therapy in patients is beginning.

    Early clinical trials have used stem cells that include both stromal

    cells andhematopoieticstem cells and have confirmed theabsence

    of sideeffects (Park et al., 2005; Sykova et al., 2006; Chernykhet al.,

    2007; Yoon et al., 2007; Saito et al., 2008; Deda et al., 2008; Pal et

    al., 2009). Recently, Geffner et al. (2008) reported a preliminary

    study in a series of paraplegic patients treated with the local

    transplantation of stem cells derived from bone marrow. The

    patients have been evaluated for one year after transplantation and

    show recovery in sensitivity, motility, and bowel control. These

    early results seem to justify the initiation of clinical trials in

    patients to evaluate a sufficient number of cases to obtain reliable

    conclusions.

    In the present state of our knowledge, it is difficult to argue

    about the advantages of using only BMSC for these transplants, as

    diverse groups have done in preclinical or clinical studies (Chopp

    et al., 2000; Hofstetter et al., 2002; Ohta et al., 2004; Zurita and

    Vaquero, 2004, 2006; Bakshi et al., 2006a; Vaquero et al., 2006;Himes et al., 2006; Parr et al., 2007; Saito et al., 2008; Pal et al.,

    2009) or either use, as has been done in the first human clinical

    experiences, a mixture of bone marrow mononuclear cells (Park

    et al., 2005; Sykova et al., 2006; Chernykh et al., 2007; Yoon et al.,

    2007; Deda et al., 2008; Geffner et al., 2008). Given that stromal

    cells represent less than 0.1% of bone marrowcells, their use in cell

    therapy protocol requires manipulation of the cells in a high

    biosecurity environment to expandthe cells andobtain a sufficient

    number before transplantation. It involves technical complexity

    and this is possibly the main argument for the use of bone marrow

    stem cells without excluding the fraction of hematopoietic stem

    cells. However, stromal cells have the advantage of their great

    specificity to achieve neural transdifferentiation within the host

    tissue, ease of expansion, taking into account that the effectiveness

    of transplantation appears to be dose-dependent, and finally, low

    antigenicity, which could eventually lead to the use of allogeneic

    stromal cells in humans.

    It is obvious that we have yet to gain a better understanding of

    the mechanisms by which such cell therapy achieves neurological

    recovery. In experimental studies, it is noteworthy that the

    functional recovery of animals begins before the establishment of a

    tissue bridge suitable for thepassage of axons (Zurita and Vaquero,

    2004, 2006; Vaquero et al., 2006). Therefore, it is obvious that after

    transplantation, at least at an early stage, different regenerative

    processes, including release of neurotrophic factors by the

    transplanted cells or activation of endogenous mechanisms, can

    work together to restore neurological functions.

    Fig. 2. Experimental studies show the effectiveness of cell therapy with BMSC in adult pigs (AC) and rodents (D and E) suffering established paraplegia. (A and B)

    Somatosensory-evoked potentials (SSEP) in our experimental model of SCI in minipig (for technical details, see Zurita et al., 2008). (A) Absence of SSEP, three months after

    severe SCI.(B) Recovery of SSEP,three months afterlocal BMSCtransplantation(70 106 autologousBMSC). (C)Two months afterlocal BMSCtransplantation, motorrecovery

    canbe seen in previouslyparaplegic pigs. At this time, some treatedanimalsget up spontaneously. See Supplemental videos2 and3. (Dand E)SCI model inadult Wistarrats.

    (D) Postraumatic cavities were filled, eight months after intralesional administration of 5 106 BMSC. A clear difference, compared with controls, can be seen. (E) One yearafterlocal administration of 5 106 BMSC,a newnervoustissue, fillingthe previous traumaticcentral spinalcordcavity, canbe seen.H.E.,original magnification:40.At this

    time, a clear functional recovery is observed in transplanted animals. See Zurita and Vaquero (2006).

    J. Vaquero, M. Zurita / Progress in Neurobiology 93 (2011) 341349346

  • 8/3/2019 Progr Neurobiology Con Portada

    8/10

    Author's personal copy

    Based on experimental studies, cell therapy strategies for

    repair of SCI should be addressed to bring the greatest possible

    number of stem cells to the injured area. This strategy increases

    cell survival and facilitates neural differentiation. The adminis-

    tration of stem cells in the subarachnoid space can be a

    complementary approach to local administration (Ohta et al.,

    2004; Sateke et al., 2004; Bakshi et al., 2004, 2006a; Himes et al.,2006), and it is necessary to optimize transplantation procedures

    using biological scaffolds. Therecentfinding in ourlaboratorythat

    plasma-derived scaffolds are extremely useful in achieving this

    aim (Zurita et al., 2010) offers new perspectives in this exciting

    field of research.

    6. Conclusions

    In recent decades, the development of new techniques for cell

    therapy allowed to consider its possible application to the

    treatment of traumatic injuries of the CNS. After several years of

    experimental studies, there is growing evidence in favor that

    intralesional transplantation of adult stem cells obtained from

    bone marrow stroma can be useful to reverse the sequels oftraumatic injuries affecting the brain and spinal cord. The

    application of these techniques to patients is beginning, but it is

    obvious that we still do not know many details about how these

    cells operate and how we canoptimize the seemingly good results.

    Prudence and submission to ethical and methodological standards

    are basic premises in bringing these studies to patients.

    Conflict of interest

    The authors have no relevant affiliations or financial involve-

    ment with any organization or entity with a financial interest in or

    financial conflict with the subject matter or material discussed in

    the manuscript. This includes employment, consultancies, hono-

    raria, stock ownership or options, expert testimony, grants or

    patents received or pending, or royalties.

    Acknowledgements

    Most of the experimental work by the authors cited in this

    review has been possible by grants received from the Mapfre

    Foundation, Mutua Madrilena Foundation, and the Institute of

    Health Carlos III (Fondo de Investigacion Sanitaria, FIS 07/0621 and

    PS09/01105).

    Appendix A. Supplementary data

    Supplementary data associated with this article can be found, in

    the online version, at doi:10.1016/j.pneurobio.2010.12.002.

    References

    Ankeny, D.P., Mc Tigue,D.M.,Jakeman, L.B., 2004. Bone marrowtransplantsprovidetissue protection and directional guidance for axons after contusive spinal cordinjury in rats. Exp. Neurol. 190, 1731.

    Bakshi, A., Barshinger, A.L., Swanger, S.A., Madhavani, V., Shumsky, J.S., Neuhuber,B., Fischer, I., 2006a. Lumbar puncture delivery of bone marrow stromal cells inspinal cord contusion: a novel method for minimally invasive cell transplanta-tion. J. Neurotrauma 23, 5565.

    Bakshi, A., Hunter, C., Swanger, S., Lepore, A., Fischer, I., 2004. Minimally invasivedelivery of stem cells for spinal cord injury: advantages of the lumbar puncturetechnique. J. Neurosurg. Spine 1, 330337.

    Bakshi, A.,Shimizu, S.,Keck,C.A., Cho, S.,LeBold,D.G.,Morales,D., Arenas, E.,Snyder,E.Y., Watson, D.J., Mcintosh, T.K., 2006b. Neural progenitor cells engineered tosecrete GDNF show enhanced survival, neuronal differentiation and improvecognitive function following traumatic brain injury. Eur. J. Neurosci. 23, 2119

    2134.Barnard, J.W., Carpenter, W., 1950. Lack of regeneration in spinal cord of rat. J.

    Neurophysiol. 13, 223228.

    Bernstein, J.J., Patel, U., Keleman, M., Jefferson, M., Turtil, S., 1984. Ultrastructure offetal spinal cord and cortex implants into adult rat spinal cord. J. Neurosci. Res.11, 359372.

    Bonilla, C., Zurita, M., Otero, L., Aguayo, C., Vaquero, J., 2009. Delayed intralesionaltransplantation of bone marrow stromal cells increases endogenous neurogen-esisand promotes functional recovery aftersevere traumatic braininjury. BrainInjury 23, 760769.

    Bossolasco, P., Cova, L., Calzarossa, C., Rimoldi, S.G., Borsotti, C., Deliliers, G.L., Silani,V., Soligo, D., Polli, E., 2005. Neuro-glial differentiation of human bone marrowstem cells in vitro. Exp. Neurol. 193, 312325.

    Bregman, B.S., Kunkel-Bagden, E., Reier, P.J., Dai, H.N., Mc Atee, M., Gao, D., 1993.Recovery of function after spinal cord injury: mechanisms underlying trans-plant-mediated recovery of funcyion differ after spinal cord injury in newbornand adult rats. Exp. Neurol. 123, 316.

    Bregman, B.S., 1994. Recovery of function after spinal cord injury: transplantationstrategies. In: Dunnett, S.B., Bjorkland, A. (Eds.), Functional Neural Transplan-tation. Raven Press, New York, pp. 489529.

    Brown, J.O., Mc Cough, G.P., 1947. Abortive regeneration of the transected spinalcord. J. Comp. Neurol. 87, 131137.

    Cao, Q., Benton, R.L., Whittemore, S.R., 2002. Stem cell repair of central nervoussystem injury. J. Neurosci. Res. 68, 501510.

    Chen, Q., Long, Y., Yuan, X., Zou, L., Sun, J., Chen, S., Perez-Polo, J.M., Yang, K., 2005.Protective effects of bone marrow stromal cells transplantation in injuredrodent brain: synthesis of neurotrophic factors. J. Neurosci. Res. 80, 611619.

    Chernykh, E.R.,Stupak, V.V.,Muradov,G.M., Sizikov, M.Y.,Shevela, E.Y.,Leplina, O.Y.,Tikhonova, M.A., Kulagin, A.D., Lisukov, I.A., Ostanin, A.A., Kozlov, V.A., 2007.

    Application of autologous bone marrow stem cells in the therapy of spinal cordinjury patients. Bull. Exp. Biol. Med. 143, 543547.

    Chopp,M., Li,Y., 2002. Treatmentof neuralinjury with marrowstromal cells.LancetNeurol. 1, 92100.

    Chopp, M., Zhang, X.H., Li, Y., Wang, L., Chen, J., Lu, D., Lu, M., Rosemblum, M., 2000.Spinal cord injury in rat: treatment with bone marrow stromal cell transplan-tation. Neuroreport 11, 30013005.

    Das, G.D., 1987. Neural transplantation in normal and traumatized spinal cord. In:Azmitia, E.C., Bjorklund, A. (Eds.), Cell and Tissue Transplantation into theAdult Brain, vol. 495. The New York Academy of Sciences, New York, pp. 5370.

    Das, G.D., 1983a. Neural transplantation in the spinal cord of adult rats. Conditions,survival, cytology and connectivity of the transplants. J. Neurol. Sci. 62, 191210.

    Das, G.D., 1983b. Neural transplantation in the spinal cord of the adult mammals.In: Kao, C.C., Bunge, R.P., Reier, P.J. (Eds.), Spinal Cord Reconstruction. RavenPress, New York, pp. 367396.

    David, S., Aguayo, A.J., 1981. Axonal elongation into peripheral nervous systembridges after central nervous system injury in adult rats. Science 214, 931933.

    De Haro, J.,Zurita, M., Ayllon, L., Vaquero, J., 2005. Detection of 111In-oxine-labeled

    bone marrow stromal cells after intravenous or intralesional administration inchronic paraplegic rats. Neurosci. Lett. 377, 711.

    Deda, H., Inci, M.C., Kurekci, A.E., Kayihan, K., Ozgun, E., Ustunsoy, G.E., Kocabay, S.,2008. Treatment of chronic spinal cord injured patients with autologous bonemarrow-derived hematopoietic stem cell transplantation: 1-year follow-up.Cytotherapy 10, 565574.

    Dezawa, M., Kanno, H., Hoshino, M., Cho, H., Matsumoto, N., Itokazu, Y., Tajima, N.,Yamada, H., Sawada, H., Ishikawa, H., Mimura, T., Kitada, M., Suzuki, Y., Ide, C.,2004. Specific induction of neuronal cells from bone marrow stromal cells andapplication for autologous transplantation. J. Clin. Invest. 113, 17011710.

    Diener, P.S., Bregman, B.S., 1998. Fetal spinal cord transplants support growth ofsupraspinal and segmental projections after cervical spinal cord hemisection inthe neonatal rat. J. Neurosci. 18, 779793.

    Dixon, C.E., Flinn, P., Bao, J., Venya, R., Hayes, R.L., 1997. Nerve growth factorattenuates cholinergic deficits following traumatic brain injury in rats. Exp.Neurol. 146, 479490.

    Dominici,M., Le Blanc,K., Mueller, I.,Slaper-Cortenbach, I.,Marini, F.C., Krause, D.S.,Deans, R.J., Keating, A., Prockop, D.J., Horwitz, E.M., 2006. Minimal criteria for

    defining multipotent mesenchymal stromal cells. The International Society forCellular Therapy position statement. Cytotherapy 8, 315317.Falci, S., Holtz, A., Akesson, E., Azizi, M., Ertzgaard, P., Hultling, C., Kjaeldgaard, A.,

    Levi, R., Ringden, O., Westgren, M., Lammertse, D., Seiger, A., 1997. Obliterationof a posttraumatic spinal cord cyst with solid human embryonic spinal cordgrafts: first clinical attempt. J. Neurotrauma 14, 875884.

    Fernandez, E., Pallini, R., Minciacchi, D., Sbriccoli, A., 1986. Peripheral nerve auto-grafts to the rat spinal cord: study on the origin and course of regeneratingfibres. Acta Neurochir. (Wien) 82, 5763.

    Garca, R., Aguiar, J., Alberti, E., de la Cuetara, K., Pavon, N., 2004. Bone marrowstromal cells produce nerve growth factor and glial cell line-derived neuro-trophic factor. Biochem. Biophys. Res. Commun. 316, 753754.

    Geffner, L.F., Santacruz, P., Izurieta, M., Flor, L., Maldonado, B., Auad, A.H., Mon-tenegro, X., Gonzalez, R., Silva, F., 2008. Administration of autologous bonemarrowstemcellsintospinal cord injury patientsvia multipleroutes issafe andimproves their quality of life: comprehensive case studies. Cell Transplant. 17,12771293.

    Harting, M.T., Baumgartner, J.E., Worth, L.L., Ewing-Cobbs, L., Gee, A.P., Day, M.C.,Cox, C.S., 2008. Cell therapies for traumatic brain injury. Neurosurg. Focus 24,

    E17.Hermann, A., Gastl, R., Liebau, S., Popa, M.O., Fiedler, J., Boehm, B.O., Maisel, M.,

    Lerche, H.,Schwarz,J., Brenner, R., Storch, A.,2004. Efficient generationof neural

    J. Vaquero, M. Zurita / Progress in Neurobiology 93 (2011) 341349 347

  • 8/3/2019 Progr Neurobiology Con Portada

    9/10

    Author's personal copy

    stem cell-likecellsfrom adult human bone marrowstromal cells.J. Cell Sci. 117,44114422.

    Himes, B.T., Neuhuber, B., Coleman, C., Kushner, R., Swanger, S.A., Kopen, G.C.,Wagner, J., Shumsky, J.S., Fischer, I., 2006. Recovery of function followinggrafting of human bone marrow-derived stromal cells into the injured spinalcord. Neurorehabil. Neural Repair 20, 278296.

    Hoffer, B.J., Olson, L., 1991. Ethical issues in brain-cell transplantation. TrendsNeurosci. 14, 384388.

    Hofstetter, C.P., Schwarz, E.J., Hess, D., Widenfalk, J., El Manira, A., Prockop, D.J.,Olson, L., 2002. Marrow stromal cells form guiding strands in the injuredspinal cord and promote recovery. Proc. Natl. Acad. Sci. U.S.A. 99, 21992204.

    Horwitz, E.M., Le Blanc, K., Dominici, M., Mueller, I., Slaper-Cortenbach, I., Marini,F.C.,Deans, R.J., Krause, D.S., Keating, A., 2005. Clarification of thenomenclaturefor MSC: the International Society for Cellular Therapy position statement.Cytotherapy 7, 393395.

    Hung, S., Cheng, H., Pan, C., Tsai, M.J., Kao, L., Ma, H., 2002. In vitro differentiation ofsize-sieved stem cells into electrically active neural cells. Stem Cells 20, 522529.

    Jiang, Y., Jahagirdar, B.N.,Reinhardt, R.L.,Schwartz,R.E., Keene,C.D., Ortiz-Gonzalez,X.R., Reyes, M., Lenvik, T., Lund, T., Blackstad, M., Du, J., Aldrich, S., Lisberg, A.,Low, W.C., Largaespada, D.A., Verfaille, C.M., 2002. Pluripotency of mesenchy-mal stem cells derived from adult marrow. Nature 418, 4149.

    Kao, C.,1974. Comparison of healing process in transected spinalcordsgrafted withautogenous brain tissue, sciatic nerve, and no dose ganglion. Exp. Neurol. 44,424439.

    Le Blanc, K., Ringden, O., 2005. Immunobiology of humam mesenchymmal stem

    cells and future use in hematopoietic stem cell transplantation. Biol. BloodMarrow Transplant. 11, 321334.

    Lee, J., Kuroda, S., Shichinohe, H., Ikeda, J., Seki, T., Hida, K., Tada, M., Sawada, K.,Iwasaki, Y., 2003. Migration and differentiation of nuclear fluorescence-labeledbone marrow stromal cells after transplantation into cerebral infarct and spinalcord injury in mice. Neuropathology 23, 169180.

    Longhi, L., Watson, D.J., Saatman, K.E., Thompson, H.J., Zhang, C., Fujimoto, S., Royo,N., Castelbuono, D., Raghupathi, R., Trojanowski, J.Q., Lee, V.M., Wolfe, J.H.,Stocchetti, N., McIntosh, T.K., 2004. Ex vivo gene therapy using targetedengraftment of NGF-expressing human NT2N neurons attenuates cognitivedeficits following traumatic brain injury in mice. J. Neurotrauma 21, 17231736.

    Lu, D., Mahmood, A., Wang, L., Li, Y., Lu, M., Chopp, M., 2001. Adult bone marrowstromal cells administered intravenously to rats after traumatic brain injurymigrate into brain and improve neurological outcome. Neuroreport 12, 559563.

    Lu, J., Moochhala, S., Moore, X.L., Ng, C.K., Tan, M.H., Lee, L.K.H., He, B., Wong, M.C.,Ling, E.A., 2006. Adult bone marrow cells differentiate into neural phenotypesand improve functional recovery in rats following traumatic brain injury.

    Neurosci. Lett. 398, 1217.Mackay-Sim, A., Feron, F., Cochrane, J., Bassingthwaighte, L., Bayliss, C., Davies, W.,

    Fronek, P., Gray, C., Kerr, G., Licina, P., Nowitzke, Perry, C., Silburn, P.A.S.,Urquhart, S., Geraghthy, T., 2008. Autologous olfactory ensheathing celltransplantation in human paraplegia: a 3-year clinical trial. Brain 131, 23762386.

    Mahmood, A., Dunyue, L., Changsheng, Q., Goussev, A., Chopp, M., 2007. Treatmentof traumatic brain injury with a combination therapy of marrow stromal cellsand atorvastatin in rats. Neurosurgery 60, 546554.

    Mahmood, A., Lu, D., Chopp, M., 2004a. Intravenous administration of marrowstromal cells (MSCs)increases theexpressionof growthfactorsin ratbrainaftertraumatic brain injury. J. Neurotrauma 21, 3339.

    Mahmood, A., Lu, D., Chopp, M., 2004b. Marrow stromal cell transplantation aftertraumatic brain injury promotes cellular proliferation within the brain. Neuro-surgery 55, 11851193.

    Mahmood, A., Lu, D., Lu, M., Chopp, M., 2003. Treatment of traumatic brain injury inadult rats with intravenous administration of human bone marrow stromalcells. Neurosurgery 53, 697703.

    Mahmood, A., Lu, D., Qu, C., Goussev, A., Chopp, M., 2005. Human marrow stromalcell treatment provides long-lasting benefit after traumatic brain injury in rats.Neurosurgery 57, 10261031.

    Mahmood, A., Lu, D., Qu, C., Goussev, A., Chopp, M., 2006. Long-term recovery afterbone marrow stromal cell treatment of traumatic brain injury in rats. J.Neurosurg. 104, 272277.

    Mahmood, A., Lu, D., Wang, L., Chopp, M., 2002. Intracerebral transplantation ofmarrow stromal cells cultured with neurotrophic factors promotes functionalrecovery in adult rats subjected to traumatic brain injury. J. Neurotrauma 19,16091617.

    Mahmood, A., Lu, D., Wang, L., Li, Y., Lu, M., Chopp, M., 2001a. Treatment oftraumatic brain injury in female rats with intravenous administration of bonemarrow stromal cells. Neurosurgery 49, 11961203.

    Mahmood, A., Lu, D., Yi, L., Chen, J.L., Chopp, M., 2001b. Intracranial bone marrowtransplantation after traumatic brain injury improving functional outcome inadult rats. J. Neurosurg. 94, 589595.

    McMahon, S.S., Albermann, S., Rooney, G.E., Shaw,G., Garcia, Y., Sweeney, E., Hynes, J., Dockery, P., OBrien, T., Windebank, A.J., Allsopp, T.E., Barry, F.P., 2010.Engrafment. Migration and differentiation of neural stem cells in the rat spinal

    cord following contusion injury. Cytotherapy 12, 313325.Mezey, E., Chandross, K.J., 2000. Bone marrow: a possible alternative source of cells

    in the adult nervous system. Eur. J. Pharmacol. 405, 297302.

    Nygren, L.G., Olson, L., Seiger, A., 1977. Monoaminergic reinnervation of thetransected spinal cord by homologous fetal brain grafts. Brain Res. 129, 227235.

    Ohta, M.,Suzuki, Y.,Noda,T., Ejiri, Y.,Dezawa, M.,Kataoka,K., Chou,H., Ishikawa,N.,Matsumoto, N., Iwashita, Y., Mizuta, E., Kuno, S., Ide, C., 2004. Bone marrowstromal cellsinfusedinto the cerebrospinal fluidpromote functionalrecoveryofthe injured rat spinal cord with reduced cavity formation. Exp. Neurol. 187,266278.

    Opydo-Chanek, M., 2007. Bone marrow stromal cells in traumatic brain injury(TBI) therapy: true perspective or false hope? Acta Neurobiol. Exp. 67, 187195.

    Pal, R., Venkataramana, N.K., Bansal, A., Balaraju, S., Jan, M., Chandra, R., Dixit, A.,Rauthan, A., Murgod, U., Totey, S., 2009. Ex vivo-expanded autologous bonemarrow-derived mesenchymal stromalcells in human spinal cord injury/para-plegia: a pilot clinical study. Cytotherapy 11, 897911.

    Pallini, R., Fernandez, E., Gangitano, C., Del Fa, A., Olivieri-Sangiacomo, C., Sbriccoli,A.,1989.Studies onembryonic transplantsto thetransectedspinal cord of adultrats. J. Neurosurg. 70, 454462.

    Park, H.C., Shims, Y.S., Ha, Y., Yoon, S.H., Park, S.R., Choi, B.H., Park, H.S., 2005.Treatment of complete spinal cord injury patients by autologous bone marrowcell transplantation and administration of granulocytemacrophage colonystimulating factor. Tissue Eng. 11, 913922.

    Park,K.I.,Liu,S., Flax,J.D.,Nissim,S., Stieg,P.E.,Snyder,E.Y.,1999.Transplantation ofneural progenitors and stem cells: developmental insights may suggest newtherapies for spinal cord and other CNS dysfunction. J. Neurotrauma 16, 675687.

    Parr, A.M., Tator, C.H., Keating, A., 2007. Bone marrow-derived mesenchymal

    stromal cells for the repair of central nervous system injury. Bone MarrowTransplant. 40, 609619.

    Philips, M.F., Mattiasson, G., Wieloch, T., Bjorklund, A., Johansson, B.B., Tomasevic,G., Martnez-Serrano, A., Lenzlinger,P.M., Sinson, G., Grady, M.S., McIntosh,T.K.,2001. Neuroprotective and behavioral efficacy of nerve growth factor-trans-fected hippocampal progenitor cell transplants after experimental traumaticbrain injury. J. Neurosurg. 94, 765774.

    Raedt, R., Pinxteren, J., Van Dycke, A., Waeytems, A., Craeye, D., Timmermans, F.,Vonck, K., Vandekerckhove, B., Plum, J., Boon, P., 2007. Differentiation assays ofbone marrow-derived multipotent adult progenitor cell (MAPC)-like cellstowards neural cells cannot depend on morphology and a limited set of neuralmarkers. Exp. Neurol. 203, 542554.

    Ramon-Cueto, A., Cordero, M.I., Santos-Benito, F.F., Avila, J., 2000. Functionalrecovery of paraplegic rats and motor axon regeneration in their spinal cordsby olfactory ensheating glia. Neuron 25, 425435.

    Ramon-Cueto, A., Plant, G.W., Avila, J., Bunge, M.B., 1998. Long-distance axonalregeneration in the transected adult rat spinal cord is promoted by olfactoryensheathing glia transplants. J. Neurosci. 18, 38033815.

    Ramon y Cajal, S., 1914. Studies on the degeneration and regeneration of Nervous

    System, vol. II. Printing Nicholas Moya Sons, Madrid (in Spanish).Reier, P.J., 2004. Cellular transplantation strategies for spinal cord injury and

    translational neurobiology. Neurorx 1, 424451.Reier, P.J., 1985. Neural tissue grafts and repair of the injured spinal cord. Neuro-

    pathol. Appl. Neurobiol. 11, 81104.Saatman, K.E., Contreras, P.C., Smith, D.H., Raghupathi, R., McDermott, K.L., Fernan-

    dez, S.C., Sanderson, K.L., Voddi, M., McIntosh, T.K., 1997. Insulin-like growthfactor-1 (IGF-1) improves both neurological motor and cognitive outcomefollowing experimental brain injury. Exp. Neurol. 147, 418427.

    Saito, F., Nakatani, T., Iwase, M., Maeda, Y., Hirakawa, A., Murao, Y., Suzuki, Y.,Onodera, R., Fukushima, M., Chizuka, I., 2008. Spinalcord injury treatment withintrathecal autologous bone marrow stromal cell transplantation: the firstclinical trial case report. J. Trauma 64, 5359.

    Sanchez-Ramos, J., Song, S., Cardozo-Pelaez, F., Hazzi, C., Stedeford, T., Willing, A.,Freeman,T.B.,Saporta,S., Janssen, W.,Patel, N.,Cooper,D.R.,Sanberg, P.R., 2000.Adult bone marrow stromal cells differentiate into neural cells in vitro. Exp.Neurol. 164, 247256.

    Sateke, K., Lou, J., Lenke, L.G., 2004. Migration of mesenchymal stem cells through

    cerebrospinal fluid into injured spinal cord tissue. Spine 29, 19711979.Sinson, G., Voddi, M., McIntosh, T.K., 1996. Combined fetal neural transplantationand nerve growth factor infusion: effects on neurological outcome followingfluid-percussion brain injury in the rat. J. Neurosurg. 84, 655662.

    Sykova, E.,Homola, A.,Mazanec, R.,Lachmann, H.,Konradova, S.L., Kobylka, P.,Padr,R., Neuwirth, J.,Komrska,V., Vavra, V.,Stulik,J., Bojar,M., 2006. Autologousbonemarrow transplantation in patients with subacute and chronic spinal cordinjury. Cell Transplant. 15, 675687.

    Tsuji, O., Miura, K., Okada, Y., Fujiyoshi, K., Mukaino, M., Nagoshi, N., Kitamura, K.,Kumagai, G.,Nishino, M.,Tomisato, S.,Higashi,H., Nagai,T., Katoh,H., Kohda,K.,Matsuzaki, Y., Yuzaki, M., Ikeda, E., Toyama, Y., Nakamura, M., Yamanaka, S.,Okano, H., 2010. Therapeutic potential of appropiately evaluated safe-inducedpluripotent stem cells for spinal cord injury. PNAS 107, 1270412709.

    Vaquero, J., Arias, A., Martnez, R., Oya, S., Zurita, M., 1992a. Maturation of embry-onic cerebral tissue grafted into injured spinal cord.Acta Neurochir.(Wien) 117,84 (Abstract).

    Vaquero, J., Arias, A., Oya, S., Coca, S., Zurita, M., 1992b. Delayed transplantation offoetal cerebral tissue into injured spinal cord of adult rats. Acta Neurochir.(Wien) 115, 133142.

    Vaquero, J., Zurita, M., Oya, S., Santos, M., 2006. Cell therapy using bone marrowstromal cells in chronic paraplegic rats: systemic or local administration?Neurosci. Lett. 398, 129134.

    J. Vaquero, M. Zurita / Progress in Neurobiology 93 (2011) 341349348

  • 8/3/2019 Progr Neurobiology Con Portada

    10/10

    Author's personal copy

    Widner, H., Brundin, P., 1988. Immunological aspects of grafting in the mammaliancentral nervous system. A review and speculative synthesis. Brain Res. 472,287324.

    Wirth III, E.D., Reier, P.J., Fessler, R.G., Thompson, F.J., Uthman, B., Behrman, A.,Beard, J., Vierck, C.J.,Anderson,D.K., 2001. Feasibility and safety of neural tissuetransplantation in patients with syringomyelia. J. Neurotrauma 18, 911929.

    Whittemore, S.R., Onifer, S.M., 2000. Immortalized neural cell lines for CNS trans-plantation. Prog. Brain Res. 127, 4965.

    Woodbury,D., Schwar, E.J., Prockop, D.J., Black,I.B.,2000.Adultrat andhumanbonemarrow stromal cells differentiate into neurons. J. Neurosci. Res. 61, 364370.

    Wrathall, J.R., Rigamonti, D.D., Braford, M.R., Kao, C.C., 1982. Reconstruction of thecontused cat spinal cord by the delayed nerve graft technique and culturedperipheral non-neuronal cells. Acta Neuropathol. (Berl.) 57, 5969.

    Wu, D.C., Boyd, A.S., Wood, K.J., 2007. Embryonic stem cell transplantation: poten-tial applicability in cell replacement therapy and regenerative medicine. Front.Biosci. 12, 45254535.

    Ye, M., Chen, S., Wang, X., Qi, C., Lu, G., Liang, L., Xu, J., 2005. Glial cell line-derivedneurotrophic factor in bone marrow stromal cells of rat. Neuroreport 16,581584.

    Yoon, S.H., Shim, Y.S., Park, Y.H., Chung, J.K., Nam, J.H., Kim, M.O., Park, H.C., Park,S.R., Min, B.H., Kim, E.Y., Choi, B.H., Park, H., Ha, Y., 2007. Complete spinal cordinjury treatment using autologous bone marrow cell transplantation and bonemarrow stimulation with granulocytemacrophage-colony stimulating factor:phase I/II clinical trial. Stem Cells 25, 20662073.

    Yoshimura, S., Teramoto, T., Whalen, M.J., Irizarry, M.C., Takagi, Y., Qiu, J., Harada, J.,Waeber, C., Breakefield, X.O., Moskowitz, M., 2003. FGF-2 regulates neurogen-

    esis and degeneration in the dentate gyrus after traumatic brain injury. J. Clin.Invest. 112, 12021210.

    Zeng, L., Rahrmann, E., Hu, Q., Lund, T., Sandquist, L., Felten, M., OBrien, T.D., Zhang, J., Verfaille, C., 2006. Multipotent adult progenitor cells from swine bonemarrow. Stem Cells 24, 23552366.

    Zhou, Z., Chen, H., Zhang, K., Yang, H., Liu, J., Huang, Q., 2003. Protective effect ofnerve growth factor on neurons after traumatic brain injury. J. Basic Clin.Physiol. Pharmacol. 14, 217224.

    Zompa, E.A., Cain, L.D., Everhart, A.W., Moyer, M.P., Hulsebosch, C.E., 1997. Trans-plant therapy: recovery of function after spinal cord injury. J. Neurotrauma 14,479506.

    Zurita, M., Otero, L., Aguayo, C., Bonilla, C., Ferreira, E., Parajon, A., Vaquero, J., 2010.Celltherapy for spinal cordrepair: optimizationof biologicscaffoldsfor survivaland neural differentiation of human bone marrow stromal cells. Cytotherapy12, 522537.

    Zurita, M., Vaquero, J., 2004. Functional recovery in chronic paraplegia after bonemarrow stromal cells transplantation. Neuroreport 15, 11051108.

    Zurita, M., Vaquero, J., 2006. Bone marrow stromal cells can achieve cure of chronicparaplegic rats: functional and morphological outcome one year after trans-plantation. Neurosci. Lett. 402, 5156.

    Zurita, M., Vaquero, J., Bonilla, C., Santos, M., De Haro, J., Oya, S., Aguayo, C., 2008.Functional recovery of chronic paraplegic pigs after autologous transplantationof bone marrow stromal cells. Transplantation 86, 845853.

    Zurita, M., Vaquero, J., Oya, S., Miguel, M., 2005. Schwann cells induce neuronaldifferentiation of bone marrow stromal cells. Neuroreport 16, 505508.

    Zurita, M., Vaquero, J., Oya, S., Montilla, J., 2001. Functional recovery in chronicparaplegic rats after co-grafts of fetal brain and adult peripheral nerve tissue.Surg. Neurol. 55, 249254.

    Zurita, M., Vaquero, J., Oya, S., 2000. Grafting of neural tissue in chronically injured

    spinal cord: influence of the donor tissue on regenerative activity. Surg. Neurol.54, 117125.

    J. Vaquero, M. Zurita / Progress in Neurobiology 93 (2011) 341349 349