Abstract
Stem cell therapies have been explored as a new avenue for the treatment of neurologic disease and damage within the CNS in part due to their native ability to mimic repair mechanisms in the brain. Mesenchymal stem cells have been of particular clinical interest due to their ability to release beneficial neurotrophic factors and their ability to foster a neuroprotective microenviroment. While early stem cell transplantation therapies have been fraught with technical and political concerns as well as limited clinical benefits, mesenchymal stem cell therapies have been shown to be clinically beneficial and derivable from nonembryonic, adult sources. The focus of this review will be on emerging and extant stem cell therapies for juvenile and adult-onset Huntington’s disease.
Keywords: Huntington’s disease, regenerative medicine, stem cell, transplantation
Significant advances in stem cell therapies
The clinical use of stem cell therapies has gained approval for a variety of injuries and diseases of the CNS. While much work is still needed before the widespread use of stem cells in a clinical setting can be realized, this mode of therapy may be advantageous to treat neurological disorders than many others because of the ability of stem cells to accurately mimic the normal cell repair and development process in the brain [1]. Although cell transplantation therapies have been fraught with technical and political problems, there are signs that this approach has considerable potential. Early work with Parkinson’s disease, where the first clinical trials were performed in the mid-1980s and a total of 300—400 patients have been treated subsequently with fetal cell transplantation and in the open label studies, has yielded evidence of some functional improvement [for review [2,3]] as measured by withdrawal of anti-parkinsonian medications. Patients with Huntington’s disease (HD) have received clinical benefits from implants of fetal/embryonic stem cells as well, however, these effects have been shown to be temporal [4–6].
Another type of cells, mesenchymal stem cells (MSCs), have emerged for clinical transplantation studies due to their capacity to release neurotrophic factors and their ability to create a neuroprotective microenvironment through the release of specific ILs and cytokines. Clinical trials using MSCs in the CNS are now also underway, and are focused on the safety of the cells. MSCs have been autologously transplanted into the subventricular zone in patients with advanced Parkinson’s disease [7], intravenously in patients that had suffered a stroke [8,9], and umbilical cord MSCs have been administered intravenously in children with cerebral palsy [10] with no adverse side effects from the cells and observed clinical efficacy as measured by improvements in neurological domains and fractional anisotropy values in brain MRI-DTI.
Stem cell clinical trials for stroke, spinal cord injury and amyotrophic lateral sclerosis are already underway while additional studies utilizing adult stem cells are nearing clinical trials for Parkinson’s and Alzheimer’s and HD.
The goal of stem cell transplantation should focus on providing therapeutic benefit through two main mechanisms. Successful cell transplantation should be able to work synergistically with the endogenous microenvironment to upregulate intrinsic cell proliferation or neuroprotection via trophic factor secretion and immune modulation, potentially enhancing the overall regenerative capacity of the transplanted tissue [11], or by being capable of integrating into the endogenous host network and replacing or repairing the lost neurons. This review will focus on the potential of adult stem cells to provide neuroprotection and immune modulation in adult-onset and juvenile HD.
Huntington’s disease
HD is an autosomal-dominant disorder caused by an expanded and unstable CAG trinucleotide repeat that causes a progressive degeneration of neurons, primarily in the putamen, caudate nucleus and cerebral cortex [12]. In the USA, there is estimated to be approximately 30,000 individuals with HD while the Europe Union has a slightly higher prevalence of individuals with symptomatic HD with an estimated 45,000 patients [13]. Juvenile HD (JHD) is defined by disease onset before the age of 20 years and occurs in less than 10% of all HD cases [14]; however, JHD may be further subdivided into patients that have disease onset prior to the age of 10 years or between 10 and 20 years of age as they present with different clinical characteristics [15].
HD occurs when the gene that codes for the htt protein, located on the short arm of chromosome 4, shows an increased number of CAG repeats [16]. Typically, greater than 38 CAG repeats correlate with an onset of the illness in adulthood. JHD is typically transmitted from the paternal allele and usually have more than 60 CAG repeats [14], although there is a reported case of a JHD patient with 250 CAG repeats [17]. Disease onset prior to the age of 20 years is clearly dominated by paternal transmission (about 3:1 paternal–maternal), and paternal transmission is, to date, solely responsible for disease onset prior to the age of 10 years [18].
Adult HD is dominated by chorea and other involuntary movements in the initial and middle stages of the disease, but it is becoming clearer that HD patients have cognitive and emotional deficits including slowing of psychomotor speed, impairment of attention and memory as well as executive and visuospatial functions that eventually degrade into dementia along with depression and apathy, although the emotional features are more variable than the motor or cognitive features [19,20]. Typically, HD eventually culminates in death around 15–20 years after the onset of motor symptoms. The disease progression is more rapid in children than in adults and has been described in three phases: initial phase of behavioural disorder, learning difficulty, gait disturbance and mild chorea; a florid phase with signs of mental deterioration, rigidity, speech disturbance and seizures; and a terminal phase of bed confinement, hypotonia and increasing seizures [15]. JHD patients typically have less chorea than adult onset HD with rigidity reported as the dominant clinical manifestation [14].
Historically, the neuroanatomical changes in the striatum have been the focus of neuropathological and neuroimaging studies, but recently, the presence of abnormalities throughout the cerebellum, specifically in JHD [21], including cortical thinning and decreased white matter volumes, in the prefrontal cortex, have gained significant interest [20,22]. Striatal atrophy as well as white matter loss, as measured by MRI studies, can detect HD-like degeneration 15 years prior to the onset of motor symptoms [23,24], suggesting that once the clinical onset of motor symptoms appear, significant striatal loss has already occurred.
Although HD and JHD have a single genetic cause, HD as a whole has a very complex pathology, with detrimental effects on a wide variety of cellular processes [25]. It has recently been uncovered that while conditional knockout of mutant huntingtin in the striatum of transgenic mice leads to partial motor and psychiatric recovery, silencing of mutant huntingtin in both the cortex and striatum is needed to ameliorate HD-like symptoms [26]. This is suggestive that symptomology is due to widespread dysfunction of the brain and even possibly in other organs as well [27–30].
Currently, only symptomatic treatments are available. Pharmacotherapy is difficult in HD due to the complexity and amount of damage to the brain. The symptomatology of JHD is complex and causes suffering in all domains of life and the pharmacological treatment is difficult as there are no studies to guide the current trial-and-error approach to treating these patients [21]. Clinical outlooks for HD patients and the care given to their family members have improved due to the increased recognition of the disorder, better access to genetic counseling, and more availability to specialized care programs that utilize behavioral, neurological and psychiatric rehabilitation programs [13]. Treatment for patients suffering from HD generally comprises neuroleptics, anticonvulsants [31] or tetra-benazine. The latter of which involves a complicated prescribing process, specialty pharmacies for delivering the drug, strictly managed doses and annual costs exceeding US$70,000 which makes it prohibitively expensive for many patients [32].
Thus, due to the time and nature in diagnosing HD following neuronal loss and motor deficits, restorative therapies should focus on creating a neuroprotective environment to slow the loss of endogenous neurons as well as replacing lost neurons through either stimulating endogenous neurogenesis or transplanting cells capable of differentiating, integrating and replacing lost cells.
MSCs for HD
At the time of manuscript preparation, 16 published articles have implicated improvement of either behavioral or neuropathological deficits in rodent models of HD following treatment with MSCs (Table 1). These studies have used MSCs from multiple sources including autologous transplantation of unpurified whole bone marrow from rats [33], purified rat MSCs [34–38], mouse bone marrow-derived MSCs [39,40], mouse umbilical cord-derived MSCs [41], human adipose derived MSCs [42–44] and human bone marrow MSCs [45–47].
Table 1.
Study (year) |
Animal model |
Type of cell |
Behavioral outcome |
Histology | Trophic support | Time course | Immunosuppression | Transplantation | Ref. |
Mouse animal model | |||||||||
Dey et al. (2010) | YAC128 Mouse | Genetically engineered mouse bone marrow MSCs | YAC128 mice receiving bone marrow transplants demonstrated reduced clasping behavior and longer latencies on the rotarod task | YAC128 mice receiving grafts had significantly more NeuN-positive cells in the striatum compared with untreated YAC128 mice | BDNF-engineered Cells had greater behavioral and histological recovery than NGF- or non-engineered cells | Cells were not observed at the conclusion of study, but behavioral and histological effects were still observed | None | Allogeneic | [39] |
Im et al. (2010) | YAC128 mouse | HD Human adipose MSC | YAC128 mice receiving cell transplants showed a delay in motor deficits up to 4 weeks (measured on the rotarod) following transplantation | Normal adipose cells were able to reduce striatal atrophy while HD adipose cells were unable to prevent atrophy | Cells expressed BDNF in vitro | Cells transplanted at 8 or 12 months old. Cells were not detectable 4 months post-transplantation | None | Xenogeneic (hMSC) | [43] |
Lee et al. (2009) | R6/2 mouse | Human adipose MSC | In R6/2 mice receiving transplants, there was increased life-span, rotarod performance and decreased limb clasping | Grafted R6/2 mice showed a decrease of striatal neuron loss and reduced huntingtin aggregation | Adipose stem Cells secreted BDNF, believed to be mechanism of recovery | Cells transplanted at 8 weeks tested for four additional weeks (approximately 13.5% cell survival). Behavioral effects seen after 2 weeks post-operation | None | Xenogeneic (hMSC) | [44] |
Fink et al. (2013) | R6/2 mouse | mouse umbilical cord-derived MSC | Transient behavioral sparing was observed following transplantation | MSC transplantation significantly reduced striatal atrophy | UC MSC expressed BDNF in vitro and was speculated to promote neuropathological sparing | MSCs survived 6 weeks post-transplantation | None | Allogeneic | [41] |
Rossignol et al. (2015) | R6/2 mouse | Mouse bone marrow MSC | Delay in the onset of motor and cognitive deficits in rotarod, clasping and Morris Water Maze | MSC transplantation significantly reduced striatal atrophy | MSC transplantation upregulated expression of BDNF in vivo. | MSCs survived 6 weeks post-transplantation | None | Allogeneic | [40] |
Lin et al. (2009) | QA Mouse model, R6/2 mouse model | hMSCs | QA mice receiving MSC transplants demonstrated significant motor recovery on the rotarod task and increased the survivability of the mice | QA mice receiving transplants showed partial striatal recovery in terms of striatal volume | MSC improved neuronal differentiation, motor deficits and cell loss through trophic support | Small number of cells survived up to 8 weeks and could induce endogenous cell proliferation up to 16 weeks | None | Xenogeneic (hMSC) | [45] |
Snyder et al. (2012) | N171–82Q (knockin) mouse | hMSCs | No behavioral analysis was performed | Human MSCs were rapidly rejected from the host. However, mice receiving transplants had increased proliferation and neural differentiation of endogenous stem cells. Mice receiving grafts also displayed decreased striatal atrophy and increased neurotrophic signaling | Transplanted MSC stimulated neurotrophic signaling | Transplanted human MSC disappeared over 15 days; however, endogenous cell proliferation, neural differentiation, neurotrophic signaling and decreased atrophy persisted up to 30 days | Cyclosporine A | Xenogeneic (hMSC) | [47] |
Rat animal model | |||||||||
Sadan et al. (2008) | QA rat model | Rat MSCs | No behavioral analysis was performed | Cells were capable of migrating toward the lesion site and aided in decreasing the lesion volume | BDNF was the main contributor for migration to lesion site | Cells survived for 19 days in vivo | None | Allogeneic | [37] |
Lescaudron et al. (2003) | QA rat model | Rat whole bone marrow | QA rats receiving bone marrow transplants demonstrated a reduction of cognitive deficits in the radial arm water maze when compared with untreated QA rats | No neuronal differentiation of transplanted cells | Behavioral recovery was speculated to be due to BDNF release | Tested 10 days following bone marrow administration | None | Autologous | [33] |
Lee et al. (2009) | QA rat model | Human adipose MSC | In QA rats receiving adipose stem cells, there was a reduction in apomophine-induced rotation behavior | Grafted QA rats had decreased lesion volume and striatal apoptosis | Adipose stem cells secreted BDNF, believed to be mechanism of recovery | Cells transplanted at 8 weeks tested for 4 additional weeks (approximately 13.5% cell survival). Behavioral effects seen after 2 weeks post-operation | None | Xenogeneic (hMSC) | [44] |
Edalatmanesh et al. (2010) | QA rat model | Rat bone marrow MSCs | In QA rats receiving MSCs, there was a reduction in apomophine-induced rotation behavior, increased performance in the cylinder test, improvement of motor function as measured by beam walking and hanging wire test and memory improvement as measured in the Morris Water Maze when compared with untreated animals | Histological analysis was not performed | Recovery was speculated to be due to the release of neurotrophic factors, specifically BDNF | Cells transplanted 1 week post QA lesion, behavior testing performed one week following transplantation. Behavioral sparing observed immediately | None | Allogeneic | [48] |
Jiang et al. (2010) | QA rat model | Rat bone marrow MSCs | Rats receiving transplants exhibited reduced apomorphine-induced rotational behavior and longer latencies on the rotarod when compared with untreated animals | Grafted cells survived for 8 weeks, significantly reduced the amount of striatal loss observed and elevated the levels of NGF, BDNF, GDNF and CNTF in the brain | Behavioral recovery due to the release of neurotrophic factors, including BDNF | Cells, transplanted 1 week following QA lesion, were detectable for 2 weeks; however, few cells were observed at 8 weeks post | None | Allogeneic | [34] |
Sadan et al.(2010) | QA rat model | Neurotrophic-factor-treated hMSCs | Reduction in apomorphine-induced rotations | Grafted cells survived for 42 days and reduced lesion size | Neurotrophic factor secreting cells reduce lesion size and behavior abnormalities | Neurotrophic factortreated MSCs (specifically BDNF) survived better than nontreated cells | Cyclosporin A | Allogeneic | [46] |
Serrano Sánchez et al. (2014) | QA rat | Rat bone marrow MSC | None performed | Significant reductions of BDNF levels in the cortex and striatum following lesion | MSC transplantation increased brain BDNF levels | MSC transplanted 4 weeks following QA lesion. BDNF levels were measured 30 days posttransplantation and were elevated in the group receiving MSC | None | Allogeneic | [38] |
Hosseini et al. (2015) | QA rat | Human adipose MSC | Improvement in rotarod, hanging wire, reduced apomophine-induced rotations and reduction in anxiety-like behaviors | Cells survived for at least 7 weeks in the brain | Recovery was speculated to be due to the release of neurotrophic factors | Rats were transplanted 7 days following QA lesion. Behavioral testing was conducted over a 10-week period | None | Xenogeneic (hMSC) | [42] |
Rossignol et al. (2011) | 3-NP rat model | Rat bone marrow MSCs | 3-NP rats receiving transplants showed reduction of deficits in paw placement, stepping test and hindlimb retraction when compared with untreated animals | Trend toward lesion size reduction in rats receiving transplant. No neuronal differentiation of transplanted cells | Behavioral and histological recovery thought to be due to increase of BDNF immunoreactivity in the area around the transplant | Small transplants were observed 72 days post-transplantation | None | Allogeneic | [35] |
Rossignol et al. (2013) | tgHD rat | Rat bone marrow MSC and aNSC | Co-transplantation of MSC and aNSC reduced motor dysfunction as measured on the Accelerod | MSC significantly increased aNSC graft survival | MSC likely secreted trophic support and created a local immunomodulatory environment in the striatum | MSCs survived up to 12 weeks post-transplantation | None | Allogeneic | [36] |
aNSC: Adult neural stem cell; HD: Huntingdon’s disease, hMSC: Human bone marrow mesenchymal stem cell; MSC: Mesenchymal stem cell; QA: Quinolinic acid; tp :transplantation.
These studies have demonstrated improvement in motor function [34–36,39–40,42–46,48], cognition [33,40,48], anxiety-like behaviors [42] and the ability to extend the lifespan of these animals [44]. Decreases in the striatal lesion size, less neuronal and medium spiny neuron loss, stimulation of endogenous neurogenesis and reduction of huntingtin aggregation has also been observed following transplantation of MSC [34,37,39–41,43–47].
Several groups have reported that MSCs have the ability to differentiate into neuronal lineages in vitro [49–52] and following transplantation into the brain [53–58]. However, the stance that MSCs have the ability to transdifferentiate into mature neuronal phenotypes in vitro or in vivo remains controversial [59] and none of the aforementioned studies observed neuronal differentiation of the transplanted MSC.
There are several possible mechanisms that MSCs may provide in lieu of neuronal differentiation such as trophic support and immunomodulation. These hypotheses are supported from studies of other neurological disorders (Huang et al., [60]; Lin et al., [61]; Han et al., [62] Uccelli et al. [63]) and were observed in many HD studies following MSC transplantation (Table 1).
One of the most common mechanism of action postulated following MSC transplantation in HD is that the cells are capable of providing trophic support, specifically BDNF [33–35,37–41,43–44,46,48]. As a reduction in BDNF levels has been noted in HD patients [64,65] and BDNF targeted therapies have shown to ameliorate partial disease pathology [66–93] upregulating BDNF in the HD brain has become a lead therapeutic candidate.
As trophic support is speculated to be the main contributor to behavioral and histological recovery following transplantation of MSC, the potential of MSCs as a delivery vehicle for gene therapy has been examined [94–97]. Due to the nature in which MSC can be engineered in vitro, a study tested MSCs that overproduced either BDNF, nerve growth factor or a combination of both [39]. YAC128 transgenic mice that received transplantations of the MSC to overexpress BDNF displayed a reduction in motor deficits and had significantly more NeuN- and Darpp32-positive cells (mature and medium spiny neurons, respectively) in the striatum than all other YAC128 groups [39]. The results from this study, along with the previously discussed literature of successful pre-clinical trials has led to translational studies using engineered human MSC in the preparation of a clinical trial [98]. However, many of the successful pre-clinical studies only examine the efficacy of the MSC treatment for a period of days to weeks (refer to Table 1), and the long-term efficacy of this strategy needs to be examined.
Clinical cell transplantation in HD
As mentioned previously, several clinical studies have been conducted to assess the viability of fetal cells as a therapeutic treatment for HD. However, there have been varying results for the long-term viability of fetal cells for HD (Table 2). Bachloud-Levi, et al., found that three out of five patients transplanted with ganglionic eminence cells showed metabolically active graft cells 10 years following transplantation [99] and these results correlated with a slowing of the progressive nature of the disease, with even some functional recovery observed at the early time points; however, in another study the transplanted ganglionic eminence underwent a similar neurodegeneration associated with HD [5], likely due to accumulation of mutant huntingtin in the neuronal graft [100,101]. It has since been postulated that mutant huntingtin is transneuronally propagated along neuronal networks, likely contributing to the pathophysiology of HD [102]. This theory has been reported in other disease models and it is thought that the mutant protein is transferred from the host into the transplanted fetal neurons via retrograde transfer [103]. Even in studies where the transplanted cells were still viable, their effect on behavioral recovery began to diminish between 2 and 4 years following the treatment [99,101,104]. While ganglionic eminence transplantations into HD patients have shown considerable promise as a treatment for HD there are many problems with the continued use of fetal cells for transplantation therapies such as ethical, logistical and availability issues [105–107].
Table 2.
Study (year) | Clinical size | Type of cell | Clinical outcome | Negative effects | Ref. |
---|---|---|---|---|---|
Bachoud-Levi et al. (2006, 2009, 2000) | Five patients | Whole ganglionic eminence | Three of five patients showed stability of symptoms or clinical improvement for 4–6 years | One patient showed development of a putaminial cyst | [4,99,108] |
Capetian et al. (2009) | One patient | Whole ganglionic eminence | UHDRS score stability for 6 months. Survival and differentiation of grafted cells | None reported (patient died from unrelated causes) | [109] |
Cicchetti et al. (2009, 2014) | Three patients | Lateral ventricular eminence containing striatal primordia | Improvement of UHDRS in two of three patients for up to 18 months before returning to presurgical levels | Grafts underwent disease-like neuronal degeneration. Cortical hemorrhage, subdural hematoma following surgery | [5,100] |
Freeman et al. (2000) | One patient | Lateral ventricular eminence containing striatal primordia | Stability of UHDRS 15 months following transplantation. Transplants integrated into the host tissue | None reported | [110] |
Furtado et al. (2005) | Seven patients | Fetal striatal tissue | Transplants failed to restore fluorodeoxyglucose uptake and D1 and D2 receptor binding in subjects | Possible technical issues with regards to the ganglionic eminence and in targeting the striatum | [111] |
Hauser et al. (2002) | Seven patients | Fetal striata | Grafts developed striatal morphology, UHDRS improved significantly 12 months following surgery | Three subjects developed subdural hemorrhages, one patient died 18 months following surgery from probable cardiac arrhythmia | [112] |
Keene et al. (2007) | Two patients | Fetal lateral ganglionic eminence | Improved ambulation 3 months following transplant in one patient. In both patients, transplanted cells displayed morphology of neurons and astrocytes | One patient reported chronic headaches following surgery and was treated for bilateral subdural hematomas. Reported that transplants did not have an effect on the course of HD | [113] |
Keene et al. (2009) | One patient | Fetal neuronal tissue | Clinical improvement for UHDRS for 2 years. Patient died 121 months following surgery from complications of advanced HD | Three mass lesions and one large cyst were present on the left caudate and putamen. Five mass lesions and two cysts were present on the right caudate and putamen | [6] |
Kopyov et al. (1998) | Three patients | Lateral ganglionic eminence | Clinical improvement for UHDRS for all three patients 12 months following surgery. Graft survival and growth within the striatum without displacing host tissue | None reported | [114] |
Krystkowiak et al. (2007) | 13 patients | Fetal neuronal tissue | Pre- and post-UHDRS were not reported. Four of the 13 patients had grafts that did not display signs of rejection | Biological, radiological and clinical rejection of grafts in other subjects (reversible under immunosuppressive treatment) | [115] |
Reuter et al. (2008) | Two Patients | Whole ganglionic Eminence | Clinical improvement for UHDRS over 5-year period for one patient. Increased striatal D2 receptor binding, suggesting long-term survival and efficacy of grafts | None reported | [101] |
Rosser et al. (2002) | Four Patients | Whole ganglionic Eminence | Stability of UHDRS as well as cognitive ability up to 6 months following surgery. Graft survival without Overgrowth | None reported | [116] |
Philpott et al(1997) | Three Patients | Lateral ganglionic Eminence | Increased cognitive functioning 6 months following surgery | None reported | [117] |
Gallina et al. (2010) | Four Patients | Whole ganglionic Eminence | Stability or improvement in motor, behavioral and functional scores up to 24 months following surgery | None reported | [104] |
Madrazo et al. (1995) | Two patients | Whole ganglionic eminence | Stability or improvement on functional capacity for up to 25 months following surgery when a slow progression of HD was observed | None reported | [118] |
HD: Huntingdon’s disease; UHDRS: Unified Huntingdon’s disease rating scale.
Embryonic cell transplantation in HD
Preclinical research using ganglionic eminence transplanted into rodent models of HD has yielded similar results in that the cells can differentiate into mature neurons and astrocytes [119], rescue the behavioral deficits [120], but that these effects are not long lasting [121]. It has been observed that HD animals receiving pluripotent embryonic stem cells (ESCs) show transient recovery of motor deficits, but this effect rarely extends beyond 8 weeks [121]. Similar to what is observed in animals receiving transplants of fetal tissue, ESCs are either rejected by the host immune system or overpro-liferate, disrupting the host cytoarchitechure and causing teratoma formation [122]. This short-term effect of the cells is likely due to a failure of the graft to successfully rebuild or replace the lost cellular connections, or due to the grafts being systematically rejected by the host immune system. Induced pluripotent stem cells have recently been transplanted into a 3-nitropropionic acid (3-NP) toxic lesion models of HD [123] and in the transgenic YAC128 [124] with both studies reporting significant behavioral improvement and that the transplanted cells were capable of differentiating into neuronal phenotypes. However, more work is still needed to characterize the safety and immunological profile of these cells following transplantation before they could be considered for clinical use.
Neuronal cell transplantation in HD
Another stem cell type that has been shown to be a potential avenue for cell replacement therapy in HD is neuronal stem cells (NSCs). Immortalized human [125], mouse [126] and rat [127] embryonic NSCs have all shown considerable promise when transplanted into various models of HD. In both transgenic mice and toxic lesion rat models of HD, NSCs have been shown to survive up to 8 weeks following transplantation, differentiate into mature neurons and astrocytes and show behavioral recovery, specifically in apomorphine-induced rotational tests [121,127–130], beam walking [126] and in the amount of time on the rotarod [120]. However, Johann et al., found that NSCs were rapidly rejected after 28 days in the R6/2 and after 14 days in a quinolinic acid (QA) mouse model of HD [131]. A large inflammatory immune response was observed following NSCs transplantation in a transgenic rat model of HD 40 weeks following transplantation [36], suggesting that these cells elicit an extended immune response. While it is possible to globally suppress the immune system with cyclosporine or other immune-suppressors to enhance the graft survival, there are several side effects associated with long-term immunosuppressive treatments [132]. For pluripotent or adult NSCs to be a viable therapeutic option for HD, local immune suppression or genetically engineering the cells to avoid rejection from the host is necessary along with the ability to direct the cells into the correct lineage following transplantation (Table 3).
Table 3.
Study (year) | Animal model | Type of cell | Behavioral outcome | Histology | Ref. |
---|---|---|---|---|---|
Mouse animal model | |||||
Yang & Yu (2009) | R6/2 mouse | Mouse NSCs | R6/2 mice receiving cells had increased life spans and improved motor function on the beam walking and rotarod task when compared with untreated animals | NSCs transplanted into R6/2 mice differentiated into neurons, reduced striatal loss and reduced ubiquitin-positive aggregation in the striatum | [126] |
Dunnett et al. (1998) | R6/2 mouse | Mouse lateral ganglionic eminence | R6/2 mice receiving transplants demonstrated increased locomotion in the open field test | Grafts were capable of survival, integration and differentiation into neurons | [119] |
Yang & Yu (2009) | R6/2 mouse | Mouse NSCs | R6/2 mice receiving cells had increased life spans and improved motor function on the beam walking and rotarod task when compared with untreated animals | NSCs transplanted into R6/2 mice differentiated into neurons, reduced striatal loss and reduced ubiquitin-positive aggregation in the striatum | [126] |
Johann et al. (2007) | QA mouse model, R6/2 mouse | Mouse embryonic NSCs | No behavioral analysis was performed | Cells differentiated into astrocytes and were rejected after 14 (QA mouse) and 28 days (R6/2) | [131] |
Bernreuther et al.(2006) | QA mouse model | Mouse ESCs | Mice receiving transplants of cells exhibited reduced amphetamine-induced rotational behavior when compared with untreated animals up to 4 weeks following surgery, but returned to sham levels at 8 weeks | Transplanted mice showed an increase in the number of neurons in the striatum and differentiated into astrocytes and GABAergic neurons | [121] |
Pineda et al. (2007) | QA mouse model | Genetically engineered mouse NSCs | Mice receiving transplants of cells exhibited reduced amphetamine-induced rotational behavior when compared with untreated animals | Cells were able to survive and proliferate in the mouse brain. Mice receiving transplants showed less striatal loss when compared with untreated animals | [129] |
Shin et al. (2012) | QA mouse model | Mouse embryonic NSCs | No behavioral analysis was performed | Grafted cells survived for 28 days and differentiated into mature neurons expressing DARPP32 | [133] |
Rat animal model | |||||
Aubry et al. (2008) | QA rat model | Striatal progenitors derived from human ESCs | No behavioral analysis was performed. | Cells transplanted at the ganglionic eminence stage were capable of survival,differentiation into striatal neurons, but resulted in tumor-like overproliferation | [122] |
Song et al. (2007) | QA rat model | Human ESC neural precursors | Rats receiving transplants exhibited reduced apomorhine-induced rotational behavior when compared with untreated animals | Cells were positive for early neuronal markers and no tumor formation was observed at 3 weeks post-transplantation | [130] |
Kordower et al. (1997) | QA rat model | Genetically engineered mouse embryonic NSCs | No behavioral analysis was performed | Rats receiving grafts displayed sparing of striatal neurons after QA injection | [134] |
Hurlbert et al. (1999) | QA rat model | Human teratocarcinoma neural precursors | Rats receiving transplants exhibited reduced methamphetamine-induced rotational behavior and improved forelimb use in a staircase task when compared with untreated animals | Cells survived for 12 weeks and displayed markers of mature neurons but did not differentiate into medium spiny neurons (DARPP32) | [135] |
Armstrong et al. (2000) | QA rat model | Rat embryonic NSCs | No behavioral analysis was performed | Grafted cells survived for 12 weeks following surgery and some differentiated into mature phenotypes expressing DARPP32. It was also observed that grafts exhibited neuronal fibers outgrowth | [136] |
Vazey et al. (2006) | QA rat model | Rat adult NSCs | Rats receiving transplants exhibited reduced apomorhine-induced rotational behavior and increased forelimb exploratory behavior when compared with untreated animals | Cells survived for up to 8 weeks following surgery, migrated throughout the striatum and differentiated into astrocytes, mature neurons and striatal medium spiny neurons | [127] |
Visnyei et al. (2006) | QA rat model | Rat embryonic NSCs | No behavioral recovery was observed in QA rats receiving cells in apomorphine-induced rotation tests | Cells survived, migrated toward the lesion site and olfactory bulbs and differentiated into astrocytes and neurons | [137] |
Bosch et al. (2004) | QA rat model | Immortalized NSCs | Rats receiving transplants exhibited reduced apomorhine-induced rotational behavior when compared with untreated animals | Transplanted cells maintained a GABAergic phenotype, had elaborate neurite processes and formed synaptic connections with endogenous neurons | [128] |
Ryu et al. (2004) | 3-NP rat model | Immortalized human embryonic NSCs | Rats that received cell transplantation prior to administration of 3-NP demonstrated improved motor function on a rotarod task when compared with 3-NP animals not receiving cells | Transplanted cells expressed primarily immature neuronal markers with few cells expressing intermediate neurons or astrocytes | [120] |
3-NP: 3-nitropropionic acid; ESC: Embryonic stem cell; NSC:Nueral stem cell; QA: Quinolinic acid.
While transplantation of embryonic, neural and mesenchymal stem cells have shown to be effective both clinically and experimentally, they are not effective cures for the natural progression of HD due to the gene mutation and the ability of the mutant protein to propagate into the transplanted cells, specifically neuronal linages and are thought to only delay the onset or change the trajectory of the disease.
Ongoing challenges
An ongoing challenge to the clinical development of stem cell therapies for HD and JHD is navigating the immune response to the transplant. Although the brain has often been considered an ‘immune privileged’ organ, there are several reported cases suggesting a strong immune response with the brain that can lead to the rejection of the graft and the subsequent halting of beneficial effects [138] While it has been suggested in previous work that MSC provide immune modulation in the area around the transplant, many of these studies use an allotransplantation paradigm, thus reducing the extent of neuroinflammation [139]. While this can be addressed by using species-specific cells to avoid rejection of the xenograft, this strategy includes several caveats that impede the clinical relevancy of these studies. It is known that mouse stem cells express many different surface expression markers than human cells [140], and behave differently following in vitro expansion protocols [141–143].
The challenge of the immune response following transplantation into the brain raises an interesting dichotomy when developing stem cell therapies for clinical trials. While the ideal candidate for preclinical studies would be the type of cell planned to be used in a theoretical trial, the immune response following xenotransplantation may potentially mask some of the beneficial effects. On the contrary, conducting studies using an allotransplantation paradigm to avoid the immune response to the xenograft may lead to false discovery as cells isolated from mice or rats may be inherently different than human cells.
A second challenge that exists with translating successful stem cell therapies for HD or any other neurodegenerative disease is the accuracy of the animal model in recapitulating the human disease phenotypes. HD is a unique disease in that it is caused by a single gene mutation that can be mimicked in transgenic animals (Table 4). Transgenic mouse models can be useful tools for the study of biochemical, morphological and functional changes associated with the mutant htt [16]. The R6/2, with the N-terminal portion of human htt, containing a highly expanded glutamine repeat (145—155; [144], the yeast artificial chromosome (YAC) with the full-length human mutant htt gene carrying 128 CAG repeats [16,25] and knock-in (KI) mice, typically with 92–140 CAG repeats generated by the insertion in the endogenous htt gene, mimic the disease manifestation and show several phenotypical alterations, resembling those observed in HD patients [16]. While these mouse models capture some of the phenotypes of HD, none of the mouse models recapitulates the substantial striatal neuronal cell loss that is characteristic in HD patients, thereby limiting the effectiveness of translational research [145]. Specifically in the human disease, approximately 50% atrophy of the caudate and putamen is observed prior to the onset of clinically classified motor dysfunction [146,147]. 3-NP crosses the blood–brain–barrier and can be administered systematically to induce cell death in the brain, through excitatory mechanisms closely correlated with HD [148] and create the neuropathology and behavioral abnormalities of HD [149]. QA administration recapitulates many histopathological and neurochemical features of HD neuropathy and also causes memory deficits, leading many researchers to use QA models to explore striatal neurodegeneration as well as to evaluate neuroprotective strategies against HD [48,150–152]. The 3-NP and QA models of HD are useful tools for studying the motor dysfunction associated with clinical or late stage HD, but may not be appropriate to study the early cognitive deficits and presymptomatic pathology associated with HD patients. The number of transgenic rat models recapitulating key pathological hallmarks of HD is still limited [153–156] and these models have many of the same limitations of the transgenic mice.
Table 4.
Animal model | Name | CAG repeat length | Strengths | Weaknesses | Ref. |
---|---|---|---|---|---|
Mouse | R6/2 | 144 | Rapid, progressive behavioral deficits | Limited neuropathology, short lifespan | [157] |
N171-Q82 | 82 | Accumulation of mutant Htt aggregates | Subtle motor changes | [158] | |
YAC128 | 128 | Striatal atrophy | Late onset, subtle and transient behavior deficits | [159] | |
BACHD | 97 | Striatal atrophy and behavioral deficits | Weight gain, late onset | [160] | |
Hdh (CAG)150 | 150 | Striatal atrophy and behavioral deficits | Late onset | [161] | |
Rat | TgHD51 | 51 | Progressive behavior deficits | Late-onset and limited neuropathology | [155] |
BACHD | 97 | Striatal atrophy and behavioral deficits | Limited availability and late onset | [156] | |
Quinolinic acid | N/A | Reproducible behavioral deficits and striatal cell loss | Not progressive, does not have the mutant Htt gene or produce mutant protein | [162] | |
3-nitropropionic acid | N/A | Reproducible behavioral deficits and striatal cell loss | Does not have the mutant Htt gene or produce mutant protein | [163] | |
Mini-pig | N208 | 105 | HD-like apoptotic neurons and DNA fragmentation | Limited behavioral tests and availability | [164] |
Sheep | OVT73 | 73 | Reduction in striatal GABA A receptor | Limited behavioral tests and availability | [165] |
Nonhuman primate | Exon 1 HTT | 84 | Dystonia, chorea, neuronal inclusions and neuropil aggregates | Extremely limited availability | [166] |
HD: Huntingdon’s disease.
While these models can provide a great deal of information on the behavioral-, histological- and molecular-level abnormalities associated with HD, no singular model can fully capture the diverse phenotypes associated with the disease. Many of the transgenic models currently available are unable to recapitulate both behavioral deficits and the associated neuropathology. While it is possible to study the progressive behavioral deficits in several of the transgenic mouse models, typically these models do not display neuronal loss that correlates to the human condition. Alternatively, the animal models that provide reproducible neuronal cell loss and striatal atrophy are either toxic lesion models not carrying the mutant gene or transgenic animals that have late disease onset (greater than 12 months) and display subtle motor deficits. The usefulness of rodent models is also limited by other translational constraints. Namely, the brains of rodents differ significantly from humans in both their small size and their neuroanatomical organization [167]. The second major concern using a transgenic animal model to study a prodromal disease that extends over a long period of time is that the animals have a significantly shorter lifespan [167]. Due to these specific shortfalls, large animal models of HD have been created and are now being studied. A transgenic minipig carrying 105 CAG repeats displays some neuropathology associated with HD, specifically apoptotic neurons in the striatum [164]. However, behavioral testing for minipigs has not been well established. Transgenic sheep have also been created carrying 73 CAG repeats. These animals showed reduction of GABA A receptors and expression of medium spiny neuron marker DARPP-32 in the striatum but behavior deficits have not been reported and are not well established in ovine models [165]. The use of large animals raises housing issues and a limited number of labs are capable of performing studies on sheep or minipigs, but they do present relevant large animal models for studying distribution and pharmacokinetics of therapeutic modalities.
Transgenic nonhuman primates have also been created by microinjection of a lentivirus carrying the human exon 1 fragment with 84 CAG repeats [166]. These nonhuman primates have shown behavioral deficits similar to the human condition such as chorea and dystonia and evidence of widespread mutant htt inclusions upon histological analysis. Furthermore, nonhuman primates have established cognitive and motor tests, albeit these have not been optimized for HD. However, the availability of these animals is at a premium and would prove to be cost prohibitive for most studies.
While HD is advanced in terms of creating rodent and large-scale models that recapitulate the genetic mutation known to cause the disease, the models need to be refined to better mimic the cognitive, motor and emotional phenotypes along with the associated neuropathology. As more therapies near clinical trials for HD, the need for animal models that more accurately predict clinical efficacy in humans is needed. While the initial costs of nonhuman primate studies may be prohibitive, they may prove more valuable for predicting promising therapeutics to take forward to clinical trials.
Unmet needs
Translational research for HD could benefit by having standardized tests and endpoints, agreed upon by the HD research community, for the different animal models on what would constitute a promising therapeutic study. While several behavioral tests, such as the rotarod, are generally accepted as a reliable measure of motor dysfunction in HD, other tests such as the limb clasping response are vague in their external validity to HD. Other histological and molecular analyses also differ between various animal models and the relative effect size observed is often difficult to extrapolate to the human condition. The rate of disease progression also plays a large role to the extent in which the respective animal model can be used to test therapeutic products. For example, many studies utilize the R6/2 mouse model to characterize behavioral deficits and the ability of a target therapy or compound to extend the lifespan of these animals; however it is widely accepted that this mouse recapitulates JHD and as such, therapies aimed at preventing neuronal loss would be unsuccessful due to the lack of neuropathology in this model. Conversely, therapies aimed at the metabolic dysfunction or at extending the lifespan of the mice might be unsuccessful in either the YAC128 or bacteria artificial chromosome HD mouse models as they exhibit weight gain uncharacteristic of the human condition and have a normal lifespan when compared with nongene carrying littermates.
As mentioned above, many genetic large animal models of HD are being developed. These new animal models should create an avenue for large animal safety and toxicology studies. The rodent brain lacks some of the major neuroanatomical characteristics relevant to the human HD brain; specifically mice and rats do not have separate caudate and putamen or the dark pigment, neuromelanin, in the substantia nigra [167]. Mice also have smooth (lisencephalic) cortices whereas the human cortex has convoluted (gyrencephalic) anatomy [167], which contributes to targeting difficulties if the planned therapeutic involving intracranial transplantation. These issues indicate a need to conduct large animal safety studies to accurately assess the delivery of the stem cells and to perform long-term toxicology studies.
Conclusion
In conclusion, stem cell therapies, particularly engineered MSC transplantation, holds great promise to slow the progression of HD. While many advances are being made in the field of stem cell research, the strong clinical safety profile of MSC make them a strong candidate to move forward with clinical trials for this devastating disease.
Future perspective
With the initiation of several clinical trials for the use of MSC in the CNS, the future of this therapy will focus on the clinical follow-up of these patients to demonstrate the safety and feasibility of such a trial. If these cells follow the same safety profile that they have demonstrated preclinically, the initiation of Phase II and III trials will hopefully be underway with larger cohorts of patients to test the efficacy of these treatments. It is likely that following the initial trials of MSC treatments that the preclinical focus will be on the development and optimization to improve the efficacy of these cells. The ease in which MSC can be engineered will likely shape the transplantation field in the next 5–10 years. The ability for MSCs to act as a biological delivery system will enable researchers to test different therapeutic targets for gene delivery using a reliable delivery platform. Several clinical trials have initiated testing the potential safety of adult stem cells in the CNS.
Conversely, the sustained engraftment of MSCs may be a potential obstacle in development of longterm cellular therapy. Allogeneic MSC engraftments in macaque monkeys have been shown to have varying success as a result of immunogenicity. Special care must be taken into account for future MSC engraftment studies in this regard. Transient engraftment of MSCs may prove to be a potential boon rather than a limitation insofar as a potential safeguard from a prolonged immune response [168–170].
As adult and juvenile HD have subtle but significant differences in disease progression and symptoms, it is important to consider these when developing a stem cell therapy. This review has focused mainly on the concept of neuroprotection in adult HD with the use of genetically engineered MSC, but in specific cases of juvenile HD, where the disease progression is too rapid; there may be too widespread neuronal loss for neuroprotection to be effective. It is likely that a polytherapy or multiple types of cell transplantation would be needed to address the multifaceted nature of the disease.
The company Brain-Storm Cell Therapeutics, Inc., based at the Hadassah University Medical Center in Jerusalem, reported in early 2015 that it treated the first patients with amyotrophic lateral sclerosis with a modified stem cell (NurOwn) isolated from the bone marrow and enhanced to resemble glial-derived neurotrophic factor astrocyte-like cells by exposure to specific growth factors [171].
In December 2014, Athersys concluded patient enrolment of a Phase IIa clinical study for ischemic stroke patients treated with an MSC-like stem cell therapy referred to as MultiStem. This stem cell trial has the potential to substantially improve neurological and functional recovery following ischemic stroke by providing neuroprotection to the damaged host neurons, immune-modulation, releasing factors that support neuronal recovery and regrowth and restoring immune system homeostasis [172].
Asterias Biotherapeutics, Inc. received approval by the US FDA in 2014 to begin a Phase I/IIa clinical trial to test the safety and efficacy of oligodendrocyte progenitor cells (AST-OPC1) for patients who have suffered spinal cord injuries. This study is an extension of a trial started by Geron in 2010, in which five patients treated showed no serious side effects [173].
Executive summary.
Significant advances in stem cell therapies
Stem cell therapies for diseases of the CNS are underway and hold significant clinical benefit.
Huntington’s disease
Stem cell therapies hold great potential for adult and juvenile Huntington’s disease (HD).
Mesenchymal stem cells for HD
Mesenchymal stem cells (MSCs) have a long, robust history in animal models of HD for providing behavioral and histological benefits.
Ongoing challenges
A major hurdle in developing stem cell therapies is addressing/managing the immune response following transplantation.
Unmet needs
Transgenic animal models need to be improved to help facilitate translational research to get to clinical trials.
MSCs have long displayed promising therapeutic effects and strong safety profiles in preclinical studies and are now gaining US FDA approval to clinically test for diseases and disorders of the CNS. Intrastriatal transplantation of MSCs in rodent models of HD has led to improvements of behavioral function and has proven capable of slowing the rate of neurodegeneration by creating a neuroprotective environment, likely through the release of trophic factors. These positive results have led to the proposed clinical use of MSCs engineered to release BDNF. However, more work is needed to optimize the safety and delivery of these cells in large animal models that more closely resemble the human brain.
Footnotes
Financial & competing interests disclosure
The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.
No writing assistance was utilized in the production of this manuscript.
References
Papers of special note have been highlighted as:
•of interest;.••of considerable interest
- 1.Dutta S, Singh G, Sreejith S et al. Cell therapy: the final frontier for treatment of neurological diseases. CNS Neurosci. Ther 19(1), 5–11 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Barker RA. Stem cells and neurodegenerative diseases: where is it all going? Regen. Med 7(6 Suppl), 26–31 (2012). [DOI] [PubMed] [Google Scholar]
- 3.Karussis D, Petrou P, Kassis I. Clinical experience with stem cells and other cell therapies in neurological diseases. J. Neurol. Sci 324(1–2), 1–9 (2013). [DOI] [PubMed] [Google Scholar]
- 4.Bachoud-Lévi A-C, Gaura V, Brugières P et al. Effect of fetal neural transplants in patients with Huntington’s disease 6 years after surgery: a long-term follow-up study. Lancet Neurol. 5(4), 303–309 (2006). [DOI] [PubMed] [Google Scholar]
- 5.Cicchetti F, Saporta S, Hauser RA et al. Neural transplants in patients with Huntington’s disease undergo disease-like neuronal degeneration. Proc. Natl Acad. Sci. USA 106(30), 12483–12488 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Keene CD, Chang RC, Leverenz JB et al. A patient with Huntington’s disease and long-surviving fetal neural transplants that developed mass lesions. Acta Neuropathol. (Berl.). 117(3), 329–338 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Venkataramana NK, Kumar SKV, Balaraju S et al. Open-labeled study of unilateral autologous bone-marrow-derived mesenchymal stem cell transplantation in Parkinson’s disease. Transl. Res. J. Lab. Clin. Med 155(2), 62–70 (2010). [DOI] [PubMed] [Google Scholar]
- 8.Boncoraglio GB, Bersano A, Candelise L, Reynolds BA, Parati EA. Stem cell transplantation for ischemic stroke. Cochrane Database Syst. Rev. Online (9), CD007231 (2010). [DOI] [PubMed] [Google Scholar]
- 9.Lee JS, Hong JM, Moon GJ et al. A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem Cells Dayt. Ohio 28(6), 1099–1106 (2010). [DOI] [PubMed] [Google Scholar]
- 10.Lee Y-H, Choi KV, Moon JH et al. Safety and feasibility of countering neurological impairment by intravenous administration of autologous cord blood in cerebral palsy. J. Transl. Med 10, 10 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Madhavan L, Collier TJ. A synergistic approach for neural repair: cell transplantation and induction of endogenous precursor cell activity. Neuropharmacology 58(6), 835–844 (2010). [DOI] [PubMed] [Google Scholar]
- 12.The Huntington’s Collaborative Research Group. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes The Huntington’s Disease Collaborative Research Group; Cell 72(6), 971–983 (1993). [DOI] [PubMed] [Google Scholar]
- 13.Shoulson I, Young AB. Milestones in huntington disease. Mov. Disord 26(6), 1127–1133 (2011).• Excellent review of the major progress in the treatment, drug development, therapeutic targets and etiology of Hungtington’s disease (HD).
- 14.Ribaï P, Nguyen K, Hahn-Barma V et al. Psychiatric and cognitive difficulties as indicators of juvenile huntington disease onset in 29 patients. Arch. Neurol 64(6), 813–819 (2007). [DOI] [PubMed] [Google Scholar]
- 15.Gonzalez-Alegre P, Afifi AK. Clinical characteristics of childhood-onset (juvenile) Huntington disease: report of 12 patients and review of the literature. J. Child Neurol 21(3), 223–229 (2006). [DOI] [PubMed] [Google Scholar]
- 16.Estrada Sánchez AM, Mejía-Toiber J, Massieu L. Excitotoxic neuronal death and the pathogenesis of Huntington’s disease. Arch. Med. Res 39(3), 265–276 (2008). [DOI] [PubMed] [Google Scholar]
- 17.Nance MA, Mathias-Hagen V, Breningstall G, Wick MJ, McGlennen RC. Analysis of a very large trinucleotide repeat in a patient with juvenile Huntington’s disease. Neurology 52(2), 392–394 (1999). [DOI] [PubMed] [Google Scholar]
- 18.Biglan K, Shoulson I. Juvenile-onset huntington disease: a matter of perspective. Arch. Neurol 64(6), 783–784 (2007). [DOI] [PubMed] [Google Scholar]
- 19.Klempír J, Klempírová O, Stochl J, Spacková N, Roth J. The relationship between impairment of voluntary movements and cognitive impairment in Huntington’s disease. J. Neurol 256(10), 1629–1633 (2009). [DOI] [PubMed] [Google Scholar]
- 20.Ross CA, Aylward EH, Wild EJ et al. Huntington disease: natural history, biomarkers and prospects for therapeutics. Nat. Rev. Neurol 10(4), 204–216 (2014). [DOI] [PubMed] [Google Scholar]
- 21.Douglas I, Evans S, Rawlins MD, Smeeth L, Tabrizi SJ, Wexler NS. Juvenile Huntington’s disease: a population-based study using the General Practice Research Database. BMJ Open doi: 10.1136/bmjopen-2012-002085 (2013) (Epub ahead of print). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Stout JC, Weaver M, Solomon AC et al. Are cognitive changes progressive in prediagnostic HD? Cogn. Behav. Neurol 20(4), 212–218 (2007). [DOI] [PubMed] [Google Scholar]
- 23.Aylward EH, Rosenblatt A, Field K et al. Caudate volume as an outcome measure in clinical trials for Huntington’s disease: a pilot study. Brain Res. Bull 62(2), 137–141 (2003).•One of the first to establish robust clinical measurements to track the progression of HD. The results from this study help to design clinical trials aimed at slowing or at tenuating disease progression.
- 24.Tabrizi SJ, Langbehn DR, Leavitt BR et al. Biological and clinical manifestations of Huntington’s disease in the longitudinal TRACK-HD study: cross-sectional analysis of baseline data. Lancet Neurol. 8(9), 791–801 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Southwell AL, Ko J, Patterson PH. Intrabody gene therapy ameliorates motor, cognitive, and neuropathological symptoms in multiple mouse models of Huntington’s disease. J. Neurosci 29(43), 13589–13602 (2009).•One of the first to report significant in vivo data in transgenic mouse models of HD supporting the hypothesis that if mutant huntingtin was reduced the behavioral and neuropathological deficits would also be reduced.
- 26.Wang N, Gray M, Lu X-H et al. Neuronal targets for reducing mutant huntingtin expression to ameliorate disease in a mouse model of Huntington’s disease. Nat. Med 20(5), 536–541 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Mielcarek M, Inuabasi L, Bondulich MK et al. Dysfunction of the CNS-heart axis in mouse models of Huntington’s disease. PLoS Genet. 10(8), e1004550 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Buonincontri G, Wood NI, Puttick SG et al. Right ventricular dysfunction in the R6/2 transgenic mouse model of Huntington’s disease is unmasked by dobutamine. J. Huntingt. Dis 3(1), 25–32 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mihm MJ, Amann DM, Schanbacher BL, Altschuld RA, Bauer JA, Hoyt KR. Cardiac dysfunction in the R6/2 mouse model of Huntington’s disease. Neurobiol. Dis 25(2), 297–308 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zielonka D, Piotrowska I, Marcinkowski JT, Mielcarek M. Skeletal muscle pathology in Huntington’s disease. Front. Physiol 5, 5 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sari Y. Potential drugs and methods for preventing or delaying the progression of Huntington’s disease. Recent Patents CNS Drug Discov. 6(2), 80–90 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Venuto CS, McGarry A, Ma Q, Kieburtz K. Pharmacologic approaches to the treatment of Huntington’s disease. Mov. Disord 27(1), 31–41 (2012). [DOI] [PubMed] [Google Scholar]
- 33.Lescaudron L, Unni D, Dunbar GL. Autologous adult bone marrow stem cell transplantation in an animal model of hunrington’s disease: behavioral and morphological outcomes. Int. J. Neurosci 113(7), 945–956 (2003). [DOI] [PubMed] [Google Scholar]
- 34.Jiang Y, Lv H, Huang S, Tan H, Zhang Y, Li H. Bone marrow mesenchymal stem cells can improve the motor function of a Huntington’s disease rat model. Neurol. Res 33(3), 331–337 (2011). [DOI] [PubMed] [Google Scholar]
- 35.Rossignol J, Boyer C, Lévèque X et al. Mesenchymal stem cell transplantation and DMEM administration in a 3NP rat model of Huntington’s disease: morphological and behavioral outcomes. Behav. Brain Res 217(2), 369–378 (2011). [DOI] [PubMed] [Google Scholar]
- 36.Rossignol J, Fink K, Davis K et al. Transplants of adult mesenchymal and neural stem cells provide neuroprotection and behavioral sparing in a transgenic rat model of Huntington’s disease. Stem Cells Dayt. Ohio 32(2), 500–509 (2013).•Provided evidence for the use of mesenchymal stem cells to both modulate the host microenvironment to increase survival of normally rejected transplanted cells and to provide functional and histological benefits by themselves.
- 37.Sadan O, Shemesh N, Barzilay R et al. Migration of neurotrophic factors-secreting mesenchymal stem cells toward a quinolinic acid lesion as viewed by magnetic resonance imaging. Stem Cells Dayt. Ohio 26(10), 2542–2551 (2008). [DOI] [PubMed] [Google Scholar]
- 38.Serrano Sánchez T, Alberti Amador E, Lorigados Pedre L, Blanco Lezcano L, Diaz Armesto I, Bergado JA. BDNF in quinolinic acid lesioned rats after bone marrow cells transplant. Neurosci. Lett 559, 147–151 (2014). [DOI] [PubMed] [Google Scholar]
- 39.Dey ND, Bombard MC, Roland BP et al. Genetically engineered mesenchymal stem cells reduce behavioral deficits in the YAC 128 mouse model of Huntington’s disease. Behav. Brain Res 214(2), 193–200 (2010).••A proof-of-concept study that demonstrated the role of BDNF released from mesenchymal stem cells to provide significant recovery or prevention of deficits in a transgenic mouse model of HD.
- 40.Rossignol J, Fink KD, Crane AT et al. Reductions in behavioral deficits and neuropathology in the R6/2 mouse model of Huntington’s disease following transplantation of bone-marrow-derived mesenchymal stem cells is dependent on passage number. Stem Cell Res. Ther doi: 10.1186/scrt545 (2015) (Epub ahead of print). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Fink KD, Rossignol J, Crane AT et al. Transplantation of umbilical cord-derived mesenchymal stem cells into the striata of R6/2 mice: behavioral and neuropathological analysis. Stem Cell Res. Ther 4(5), 130 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hosseini M, Moghadas M, Edalatmanesh MA, Hashemzadeh MR. Xenotransplantation of human adipose derived mesenchymal stem cells in a rodent model of Huntington’s disease: motor and non-motor outcomes. Neurol. Res 37(4), 309–319 (2015). [DOI] [PubMed] [Google Scholar]
- 43.Im W, Lee S-T, Park JE et al. Transplantation of patient-derived adipose stem cells in YAC128 Huntington’s disease transgenic mice. PLoS Curr. 2 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lee S-T, Chu K, Jung K-H et al. Slowed progression in models of Huntington disease by adipose stem cell transplantation. Ann. Neurol 66(5), 671–681 (2009). [DOI] [PubMed] [Google Scholar]
- 45.Lin Y-T, Chern Y, Shen C-KJ et al. Human mesenchymal stem cells prolong survival and ameliorate motor deficit through trophic support in Huntington’s disease mouse models. PLoS ONE 6(8), e22924 (2011).•Further evidence of mesenchymal stem cells’ ability to release neurotrophic factors and reduce motor deficits and increase the lifespan following transplantation into transgenic mouse models of HD.
- 46.Sadan O, Shemesh N, Barzilay R et al. Mesenchymal stem cells induced to secrete neurotrophic factors attenuate quinolinic acid toxicity: a potential therapy for Huntington’s disease. Exp. Neurol 234(2), 417–427 (2012). [DOI] [PubMed] [Google Scholar]
- 47.Snyder BR, Chiu AM, Prockop DJ, Chan AWS. Human multipotent stromal cells (MSCs) increase neurogenesis and decrease atrophy of the striatum in a transgenic mouse model for Huntington’s disease. PLoS ONE 5(2), e9347 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Edalatmanesh M-A, Matin MM, Neshati Z, Bahrami A-R, Kheirabadi M. Systemic transplantation of mesenchymal stem cells can reduce cognitive and motor deficits in rats with unilateral lesions of the neostriatum. Neurol. Res 32(2), 166–172 (2010). [DOI] [PubMed] [Google Scholar]
- 49.Bae KS, Park JB, Kim HS, Kim DS, Park DJ, Kang SJ. Neuron-like differentiation of bone marrow-derived mesenchymal stem cells. Yonsei Med. J 52(3), 401–412 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Black IB, Woodbury D. Adult rat and human bone marrow stromal stem cells differentiate into neurons. Blood Cells. Mol. Dis 27(3), 632–636 (2001). [DOI] [PubMed] [Google Scholar]
- 51.Sanchez-Ramos J, Song S, Cardozo-Pelaez F et al. Adult bone marrow stromal cells differentiate into neural cells in vitro. Exp. Neurol 164(2), 247–256 (2000). [DOI] [PubMed] [Google Scholar]
- 52.Woodbury D, Schwarz EJ, Prockop DJ, Black IB. Adult rat and human bone marrow stromal cells differentiate into neurons. J. Neurosci. Res 61(4), 364–370 (2000). [DOI] [PubMed] [Google Scholar]
- 53.Brazelton TR, Rossi FM, Keshet GI, Blau HM. From marrow to brain: expression of neuronal phenotypes in adult mice. Science 290(5497), 1775–1779 (2000). [DOI] [PubMed] [Google Scholar]
- 54.Hung S-C, Cheng H, Pan C-Y, Tsai MJ, Kao L-S, Ma H-L. In vitro differentiation of size-sieved stem cells into electrically active neural cells. Stem Cells Dayt. Ohio 20(6), 522–529 (2002). [DOI] [PubMed] [Google Scholar]
- 55.Jiang Y, Jahagirdar BN, Reinhardt RL et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418(6893), 41–49 (2002). [DOI] [PubMed] [Google Scholar]
- 56.Kopen GC, Prockop DJ, Phinney DG. Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proc. Natl Acad. Sci. USA 96(19), 10711–10716 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mezey E, Chandross KJ, Harta G, Maki RA, McKercher SR. Turning blood into brain: cells bearing neuronal antigens generated in vivo from bone marrow. Science 290(5497), 1779–1782 (2000). [DOI] [PubMed] [Google Scholar]
- 58.Muñoz-Elias G, Marcus AJ, Coyne TM, Woodbury D, Black IB. Adult bone marrow stromal cells in the embryonic brain: engraftment, migration, differentiation, and long-term survival. J. Neurosci 24(19), 4585–4595 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Hardy SA, Maltman DJ, Przyborski SA. Mesenchymal stem cells as mediators of neural differentiation. Curr. Stem Cell Res. Ther 3(1), 43–52 (2008). [DOI] [PubMed] [Google Scholar]
- 60.Huang W et al. Transplantation of differentiated bone marrow stromal cells promotes motor functional recovery in rats with stroke. Neurol. Res 35, 320–328 (2013). [DOI] [PubMed] [Google Scholar]
- 61.Lin Q-M. Mesenchymal stem cells transplantation suppresses inflammatory responses in global cerebral ischemia: contribution of TNF-αa-induced protein 6. Acta Pharmacol. Sindoi: 10.1038/aps.2012.199 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Han EY, Chun MH, Kim ST, Lin D. Injection time-dependent effect of adult human bone marrow stromal cell transplantation in a rat model of severe traumatic brain injury. Curr. Stem CellRes. Ther 8, 172–181 (2013). [DOI] [PubMed] [Google Scholar]
- 63.Uccelli A, Benvenuto F, Laroni A, Giunti D et al. Neuroprotective features of mesenchymal stem cells. Best Pract. Res. Clin. Haematol 24, 59–64 (2011). [DOI] [PubMed] [Google Scholar]
- 64.Zuccato C, Ciammola A, Rigamonti D et al. Loss of huntingtin-mediated BDNF gene transcription in Huntington’s disease. Science 293(5529), 493–498 (2001). [DOI] [PubMed] [Google Scholar]
- 65.Zuccato C, Marullo M, Vitali B et al. Brain-derived neurotrophic factor in patients with Huntington’s disease. PLoS ONE 6(8), e22966 (2011).••An all-encompassing manuscript over the role of brain-derived neurotrophic factor’s role in HD. This study provides evidence that BDNF is decreased in Huntington’s patients and that BDNF is highly correlative with disease progression and decreases as the severity increases.
- 66.Almeida S, Laço M, Cunha-Oliveira T, Oliveira CR, Rego AC. BDNF regulates BIM expression levels in 3-nitropropionic acid-treated cortical neurons. Neurohiol. Dis 35(3), 448–456 (2009). [DOI] [PubMed] [Google Scholar]
- 67.Arregui L, Benítez JA, Razgado LF, Vergara P, Segovia J. Adenoviral astrocyte-specific expression of BDNF in the striata of mice transgenic for Huntington’s disease delays the onset of the motor phenotype. Cell. Mol. Neurobiol 31(8), 1229–1243 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bemelmans AP, Horellou P, Pradier L, Brunet I, Colin P, Mallet J. Brain-derived neurotrophic factor-mediated protection of striatal neurons in an excitotoxic rat model of Huntington’s disease, as demonstrated by adenoviral gene transfer. Hum. Gene Ther 10(18), 2987–2997 (1999). [DOI] [PubMed] [Google Scholar]
- 69.Benraiss A, Bruel-Jungerman E, Lu G, Economides AN, Davidson B, Goldman SA. Sustained induction of neuronal addition to the adult rat neostriatum by AAV4-delivered noggin and BDNF. Gene Ther. 19(5), 483–493 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Benraiss A, Toner MJ, Xu Q et al. Sustained mobilization of endogenous neural progenitors delays disease progression in a transgenic model of Huntington’s disease. Cell Stem Cell 12(6), 787–799 (2013).•It further strengthens the hypothesis that if BDNF can be increased in the brain of transgenic HD mice, behavioral deficits and neuropathology will be decreased. The manuscript also presents a mechanism by which BDNF increases endogenous neurogenesis.
- 71.Canals JM, Pineda JR, Torres-Peraza JF et al. Brain-derived neurotrophic factor regulates the onset and severity of motor dysfunction associated with enkephalinergic neuronal degeneration in Huntington’s disease. J. Neurosci 24(35), 7727–7739 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Cepeda C, Starling AJ, Wu N et al. Increased GABAergic function in mouse models of Huntington’s disease: reversal by BDNF. J. Neurosci. Res 78(6), 855–867 (2004). [DOI] [PubMed] [Google Scholar]
- 73.Gharami K, Xie Y, An JJ, Tonegawa S, Xu B. Brain-derived neurotrophic factor over-expression in the forebrain ameliorates Huntington’s disease phenotypes in mice. J. Neurochem 105(2), 369–379 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Giampà C, Montagna E, Dato C, Melone MAB, Bernardi G, Fusco FR. Systemic delivery of recombinant brain derived neurotrophic factor (BDNF) in the R6/2 mouse model of Huntington’s disease. PLoS ONE 8(5) e64037 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Giralt A, Friedman HC, Caneda-Ferrón B et al. BDNF regulation under GFAP promoter provides engineered astrocytes as a new approach for long-term protection in Huntington’s disease. Gene Ther. 17(10), 1294–1308 (2010). [DOI] [PubMed] [Google Scholar]
- 76.Giralt A, Carretón O, Lao-Peregrin C, Martín ED, Alberch J. Conditional BDNF release under pathological conditions improves Huntington’s disease pathology by delaying neuronal dysfunction. Mol. Neurodegener 6(1), 71 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Jiang M, Peng Q, Liu X et al. Small-molecule TrkB receptor agonists improve motor function and extend survival in a mouse model of Huntington’s disease. Hum. Mol. Genet 22(12), 2462–2470 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kells AP, Fong DM, Dragunow M, During MJ, Young D, Connor B. AAV-mediated gene delivery of BDNF or GDNF is neuroprotective in a model of Huntington disease. Mol. Ther. J. Am. Soc. Gene Ther 9(5), 682–688 (2004). [DOI] [PubMed] [Google Scholar]
- 79.Kells AP, Henry RA, Connor B. AAV–BDNF mediated attenuation of quinolinic acid-induced neuropathology and motor function impairment. Gene Ther. 15(13), 966–977 (2008). [DOI] [PubMed] [Google Scholar]
- 80.Lynch G, Kramar EA, Rex CS et al. Brain-derived neurotrophic factor restores synaptic plasticity in a knock-in mouse model of Huntington’s disease. J. Neurosci 27(16), 4424–4434 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Martire A, Pepponi R, Domenici MR, Ferrante A, Chiodi V, Popoli P. BDNF prevents NMDA-induced toxicity in models of Huntington’s disease: the effects are genotype specific and adenosine A(2A) receptor is involved. J. Neurochem doi: 10.1111/jnc.12177 (2013) (Epub ahead of print). [DOI] [PubMed] [Google Scholar]
- 82.Pérez-Navarro E, Alberch J, Neveu I, Arenas E. Brain-derived neurotrophic factor, neurotrophin-3 and neurotrophin-4/5 differentially regulate the phenotype and prevent degenerative changes in striatal projection neurons after excitotoxicity in vivo. Neuroscience 91(4), 1257–1264 (1999). [DOI] [PubMed] [Google Scholar]
- 83.Pérez-Navarro E, Canudas AM, Akerund P, Alberch J, Arenas E. Brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4/5 prevent the death of striatal projection neurons in a rodent model of Huntington’s disease. J. Neurochem 75(5), 2190–2199 (2000). [DOI] [PubMed] [Google Scholar]
- 84.Petersén AA, Larsen KE, Behr GG et al. Brain-derived neurotrophic factor inhibits apoptosis and dopamine-induced free radical production in striatal neurons but does not prevent cell death. Brain Res. Bull 56(3–4), 331–335 (2001). [DOI] [PubMed] [Google Scholar]
- 85.Reiner A, Wang HB, Del Mar N, Sakata K, Yoo W, Deng YP. BDNF may play a differential role in the protective effect of the mGluR2/3 agonist LY379268 on striatal projection neurons in R6/2 Huntington’s disease mice. Brain Res 1473, 161–172 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Saylor AJ, McGinty JF. An intrastriatal brain-derived neurotrophic factor infusion restores striatal gene expression in Bdnf heterozygous mice. Brain Struct. Funct 215(2), 97–104 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Simmons DA, Rex CS, Palmer L et al. Up-regulating BDNF with an ampakine rescues synaptic plasticity and memory in Huntington’s disease knockin mice. Proc. Natl Acad. Sci. USA 106(12), 4906–4911 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Simmons DA, Belichenko NP, Yang T et al. A small molecule TrkB ligand reduces motor impairment and neuropathology in R6/2 and BACHD mouse models of Huntington’s disease. J. Neurosci 33(48), 18712–18727 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Todd D, Gowers I, Dowler SJ et al. A monoclonal antibody TrkB receptor agonist as a potential therapeutic for Huntington’s disease. PLoS ONE 9(2), e87923 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Volpe BT, Wildmann J, Altar CA. Brain-derived neurotrophic factor prevents the loss of nigral neurons induced by excitotoxic striatal-pallidal lesions. Neuroscience 83(3), 741–748 (1998). [DOI] [PubMed] [Google Scholar]
- 91.Wu C-L, Hwang C-S, Yang D-I. Protective effects of brain-derived neurotrophic factor against neurotoxicity of 3-nitropropionic acid in rat cortical neurons. Neurotoxicology 30(4), 718–726 (2009). [DOI] [PubMed] [Google Scholar]
- 92.Xie Y, Hayden MR, Xu B. BDNF overexpression in the forebrain rescues Huntington’s disease phenotypes in YAC128 mice. J. Neurosci 30(44), 14708–14718 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Zuccato C, Liber D, Ramos C et al. Progressive loss of BDNF in a mouse model of Huntington’s disease and rescue by BDNF delivery. Pharmacol. Res 52(2), 133–139 (2005). [DOI] [PubMed] [Google Scholar]
- 94.Barzilay R, Ben-Zur T, Bulvik S, Melamed E, Offen D. Lentiviral delivery of LMX1a enhances dopaminergic phenotype in differentiated human bone marrow mesenchymal stem cells. Stem Cells Dev. 18(4), 591–601 (2009). [DOI] [PubMed] [Google Scholar]
- 95.Bauer G, Dao MA, Case SS et al. In vivo biosafety model to assess the risk of adverse events from retroviral and lentiviral vectors. Mol. Ther. J. Am. Soc. Gene Ther 16(7), 1308–1315 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Joyce N, Annett G, Wirthlin L, Olson S, Bauer G, Nolta JA. Mesenchymal stem cells for the treatment of neurodegenerative disease. Regen. Med 5(6), 933–946 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Meyerrose T, Olson S, Pontow S et al. Mesenchymal stem cells for the sustained in vivo delivery of bioactive factors. Adv. Drug Deliv. Rev 62(12), 1167–1174 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Pollock K, Stewart H, Cary W et al. Mesenchymal stem cells engineered to produce BDNF for the treatment of Huntington’s disease. Cell Transplant. 23(6), 780 (2014). [Google Scholar]
- 99.Bachoud-Lévi A-C. Neural grafts in Huntington’s disease: viability after 10 years. Lancet Neurol. 8(11), 979–981 (2009). [DOI] [PubMed] [Google Scholar]
- 100.Cicchetti F, Lacroix S, Cisbani G et al. Mutant huntingtin is present in neuronal grafts in Huntington disease patients. Ann. Neurol 76(1), 31–42 (2014). [DOI] [PubMed] [Google Scholar]
- 101.Reuter I, Tai YF, Pavese N et al. Long-term clinical and positron emission tomography outcome of fetal striatal transplantation in Huntington’s disease. J. Neurol. Neurosurg. Psychiatry 79(8), 948–951 (2008). [DOI] [PubMed] [Google Scholar]
- 102.Pecho-Vrieseling E, Rieker C, Fuchs S et al. Transneuronal propagation of mutant huntingtin contributes to non-cell autonomous pathology in neurons. Nat. Neurosci. 17(8), 1064–1072 (2014). [DOI] [PubMed] [Google Scholar]
- 103.Kordower JH, Dodiya HB, Kordower AM et al. Transfer of host-derived α synuclein to grafted dopaminergic neurons in rat. Neurohiol. Dis 43(3), 552–557 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Gallina P, Paganini M, Lombardini L et al. Human striatal neuroblasts develop and build a striatal-like structure into the brain of Huntington’s disease patients after transplantation. Exp. Neurol 222(1), 30–41 (2010). [DOI] [PubMed] [Google Scholar]
- 105.Brundin P, Barker RA, Parmar M. Neural grafting in Parkinson’s disease problems and possibilities. Prog. Brain Res 184, 265–294 (2010). [DOI] [PubMed] [Google Scholar]
- 106.Mathews DJH, Sugarman J, Bok H et al. Cell-based interventions for neurologic conditions: ethical challenges for early human trials. Neurology 71(4), 288–293 (2008). [DOI] [PubMed] [Google Scholar]
- 107.Pullicino PM, Burke WJ. Cell-based interventions for neurologic conditions: ethical challenges for early human trials. Neurology 72(19), 1709 (2009). [DOI] [PubMed] [Google Scholar]
- 108.Bachoud-Lévi AC, Rémy P, Nguyen JP et al. Motor and cognitive improvements in patients with Huntington’s disease after neural transplantation. Lancet 356(9246), 1975–1979 (2000). [DOI] [PubMed] [Google Scholar]
- 109.Capetian P, Knoth R, Maciaczyk J et al. Histological findings on fetal striatal grafts in a Huntington’s disease patient early after transplantation. Neuroscience 160(3), 661–675 (2009). [DOI] [PubMed] [Google Scholar]
- 110.Freeman TB, Cicchetti F, Hauser RA et al. Transplanted fetal striatum in Huntington’s disease: phenotypic development and lack of pathology. Proc. Natl Acad. Sci. USA 97(25), 13877–13882 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Furtado S, Sossi V, Hauser RA et al. Positron emission tomography after fetal transplantation in Huntington’s disease. Ann. Neurol 58(2), 331–337 (2005). [DOI] [PubMed] [Google Scholar]
- 112.Hauser RA, Furtado S, Cimino CR et al. Bilateral human fetal striatal transplantation in Huntington’s disease. Neurology 58(5), 687–695 (2002). [DOI] [PubMed] [Google Scholar]
- 113.Keene CD, Sonnen JA, Swanson PD et al. Neural transplantation in Huntington disease: long-term grafts in two patients. Neurology 68(24), 2093–2098 (2007). [DOI] [PubMed] [Google Scholar]
- 114.Kopyov OV, Jacques S, Lieberman A, Duma CM, Eagle KS. Safety of intrastriatal neurotransplantation for Huntington’s disease patients. Exp. Neurol 149(1), 97–108 (1998). [DOI] [PubMed] [Google Scholar]
- 115.Krystkowiak P, Gaura V, Labalette M et al. Alloimmunisation to donor antigens and immune rejection following foetal neural grafts to the brain in patients with Huntington’s disease. PLoS ONE 2(1), e166 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Rosser AE, Barker RA, Harrower T et al. Unilateral transplantation of human primary fetal tissue in four patients with Huntington’s disease: NEST-UK safety report ISRCTN no 36485475. J. Neurol. Neurosurg. Psychiatry 73(6), 678–685 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Philpott LM, Kopyov OV, Lee AJ et al. Neuropsychological functioning following fetal striatal transplantation in Huntington’s chorea: three case presentations. Cell Transplant 6(3), 203–212 (1997). [DOI] [PubMed] [Google Scholar]
- 118.Madrazo I, Franco-Bourland RE, Castrejon H, Cuevas C, Ostrosky-Solis F. Fetal striatal homotransplantation for Huntington’s disease: first two case reports. Neurol. Res 17(4), 312–315 (1995). [DOI] [PubMed] [Google Scholar]
- 119.Dunnett SB, Carter RJ, Watts C et al. Striatal transplantation in a transgenic mouse model of Huntington’s disease. Exp. Neurol 154(1), 31–40 (1998). [DOI] [PubMed] [Google Scholar]
- 120.Ryu JK, Kim J, Cho SJ et al. Proactive transplantation of human neural stem cells prevents degeneration of striatal neurons in a rat model of Huntington disease. Neurobiol. Dis 16(1), 68–77 (2004). [DOI] [PubMed] [Google Scholar]
- 121.Bernreuther C, Dihné M, Johann V et al. Neural cell adhesion molecule L1-transfected embryonic stem cells promote functional recovery after excitotoxic lesion of the mouse striatum. J. Neurosci 26(45), 11532–11539 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Aubry L, Bugi A, Lefort N, Rousseau F, Peschanski M, Perrier AL. Striatal progenitors derived from human ES cells mature into DARPP32 neurons in vitro and in quinolinic acid-lesioned rats. Proc. Natl Acad. Sci. USA 105(43), 16707–16712 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Fink KD, Crane AT, Lévêque X et al. Intrastriatal transplantation of adenovirus-generated induced pluripotent stem cells for treating neuropathological and functional deficits in a rodent model of Huntington’s disease. Stem Cells Transl. Med 3(5), 620–631 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Jeon I, Choi C, Lee N et al. In vivo roles of a patient-derived induced pluripotent stem cell line (HD72-iPSC) in the YAC128 model of Huntington’s disease. Int. J. Stem Cells 7(1), 43–47 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Lee S-T, Chu K, Park J-E et al. Intravenous administration of human neural stem cells induces functional recovery in Huntington’s disease rat model. Neurosci. Res 52(3), 243–249 (2005). [DOI] [PubMed] [Google Scholar]
- 126.Yang C-R, Yu RK. Intracerebral transplantation of neural stem cells combined with trehalose ingestion alleviates pathology in a mouse model of Huntington’s disease. J. Neurosci. Res 87(1), 26–33 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Vazey EM, Chen K, Hughes SM, Connor B. Transplanted adult neural progenitor cells survive, differentiate and reduce motor function impairment in a rodent model of Huntington’s disease. Exp. Neurol 199(2), 384–396 (2006). [DOI] [PubMed] [Google Scholar]
- 128.Bosch M, Pineda JR, Suñol C et al. Induction of GABAergic phenotype in a neural stem cell line for transplantation in an excitotoxic model of Huntington’s disease. Exp. Neurol 190(1), 42–58 (2004). [DOI] [PubMed] [Google Scholar]
- 129.Pineda JR, Rubio N, Akerud P et al. Neuroprotection by GDNF-secreting stem cells in a Huntington’s disease model: optical neuroimage tracking of brain-grafted cells. Gene Ther. 14(2), 118–128 (2007). [DOI] [PubMed] [Google Scholar]
- 130.Song J, Lee S-T, Kang W et al. Human embryonic stem cell-derived neural precursor transplants attenuate apomorphine-induced rotational behavior in rats with unilateral quinolinic acid lesions. Neurosci. Lett 423(1), 58–61 (2007). [DOI] [PubMed] [Google Scholar]
- 131.Johann V, Schiefer J, Sass C et al. Time of transplantation and cell preparation determine neural stem cell survival in a mouse model of Huntington’s disease. Exp. Brain Res 177(4), 458–470 (2007). [DOI] [PubMed] [Google Scholar]
- 132.Kawakami M, Yoshimoto T, Nakagata N, Yamamura K-I, Siesjo BK. Effects of cyclosporin A administration on gene expression in rat brain. Brain Inj. 25(6), 614–623 (2011). [DOI] [PubMed] [Google Scholar]
- 133.Shin E, Palmer MJ, Li M, Fricker RA. GABAergic neurons from mouse embryonic stem cells possess functional properties of striatal neurons in vitro, and develop into striatal neurons in vivo in a mouse model of Huntington’s disease. Stem Cell Rev. 8(2), 513–531 (2012). [DOI] [PubMed] [Google Scholar]
- 134.Kordower JH, Chen EY, Winkler C et al. Grafts of EGF-responsive neural stem cells derived from GFAP-hNGF transgenic mice: trophic and tropic effects in a rodent model of Huntington’s disease. J. Comp. Neurol 387(1), 96–113 (1997). [DOI] [PubMed] [Google Scholar]
- 135.Hurlbert MS, Gianani RI, Hutt C, Freed CR, Kaddis FG. Neural transplantation of hNT neurons for Huntington’s disease. Cell Transplant. 8(1), 143–151 (1999). [DOI] [PubMed] [Google Scholar]
- 136.Armstrong RJ, Watts C, Svendsen CN, Dunnett SB, Rosser AE. Survival, neuronal differentiation, and fiber outgrowth of propagated human neural precursor grafts in an animal model of Huntington’s disease. Cell Transplant. 9(1), 55–64 (2000). [DOI] [PubMed] [Google Scholar]
- 137.Visnyei K, Tatsukawa KJ, Erickson RI et al. Neural progenitor implantation restores metabolic deficits in the brain following striatal quinolinic acid lesion. Exp. Neurol 197(2), 465–474 (2006). [DOI] [PubMed] [Google Scholar]
- 138.Leveque X, Fink KD, Rossignol J, Dunbar GL, Lescaudron L. The use of xenotransplantation in neurodegenerative diseases: a way to go? Xenotransplantation 7, 93–107 (2012). [Google Scholar]
- 139.Lévêque X, Mathieux E, Nerrière-Daguin V et al. Local control of the host immune response performed with mesenchymal stem cells: perspectives for functional intracerebral xenotransplantation. J. Cell. Mol. Med 19(1), 124–134 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Chamberlain G, Wright K, Rot A, Ashton B, Middleton J. Murine mesenchymal stem cells exhibit a restricted repertoire of functional chemokine receptors: comparison with human. PLoS ONE 3(8), e2934 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Leten C, Roobrouck VD, Struys T et al. Controlling and monitoring stem cell safety in vivo in an experimental rodent model. Stem Cells Dayt. Ohio 32(11), 2833–2844 (2014). [DOI] [PubMed] [Google Scholar]
- 142.Liu AM, Qu WW, Liu X, Qu C-K. Chromosomal instability in in vitro cultured mouse hematopoietic cells associated with oxidative stress. Am. J. Blood Res 2(1), 71–76 (2012). [PMC free article] [PubMed] [Google Scholar]
- 143.Zhou YF, Bosch-Marce M, Okuyama H et al. Spontaneous transformation of cultured mouse bone marrow-derived stromal cells. Cancer Res. 66(22), 10849–10854 (2006). [DOI] [PubMed] [Google Scholar]
- 144.Murphy KP, Carter RJ, Lione LA et al. Abnormal synaptic plasticity and impaired spatial cognition in mice transgenic for exon 1 of the human Huntington’s disease mutation. J. Neurosci 20(13), 5115–5123 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Franich NR, Fitzsimons HL, Fong DM, Klugmann M, During MJ, Young D. AAV vector-mediated RNAi of mutant huntingtin expression is neuroprotective in a novel genetic rat model of Huntington’s disease. Mol. Ther 16(5), 947–956 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Paulsen JS, Langbehn DR, Stout JC et al. Detection of Huntington’s disease decades before diagnosis: the predict-HD study. J. Neurol. Neurosurg. Psychiatry 79(8), 874–880 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Aylward EH. Change in MRI striatal volumes as a biomarker in preclinical Huntington’s disease. Brain Res. Bull 72(2–3), 152–158 (2007). [DOI] [PubMed] [Google Scholar]
- 148.Túnez I, Tasset I, Pérez-De La Cruz V, Santamaría A. 3-nitropropionic acid as a tool to study the mechanisms involved in Huntington’s disease: past, present and future. Mol. Basel Switz 15(2), 878–916 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Shear DA, Haik KL, Dunbar GL. Creatine reduces 3-nitropropionic-acid-induced cognitive and motor abnormalities in rats. Neuroreport 11(9), 1833–1837 (2000). [DOI] [PubMed] [Google Scholar]
- 150.Ayalon L, Doron R, Weiner I, Joel D. Amelioration of behavioral deficits in a rat model of Huntington’s disease by an excitotoxic lesion to the globus pallidus. Exp. Neurol 186(1), 46–58 (2004). [DOI] [PubMed] [Google Scholar]
- 151.Kalonia H, Kumar P, Nehru B, Kumar A. Neuroprotective effect of MK-801 against intra-striatal quinolinic acid induced behavioral, oxidative stress and cellular alterations in rats. Indian J. Exp. Biol 47(11), 880–892 (2009). [PubMed] [Google Scholar]
- 152.Velloso NA, Dalmolin GD, Gomes GM et al. Spermine improves recognition memory deficit in a rodent model of Huntington’s disease. Neurobiol. Learn. Mem 92(4), 574–580 (2009). [DOI] [PubMed] [Google Scholar]
- 153.Fink KD, Rossignol J, Crane AT et al. Early cognitive dysfunction in the HD 51 CAG transgenic rat model of Huntington’s disease. Behav. Neurosci 126(3), 479–487 (2012). [DOI] [PubMed] [Google Scholar]
- 154.Jansson EKH, Clemens LE, Riess O, Nguyen HP. Reduced motivation in the BACHD rat model of Huntington disease is dependent on the choice of food deprivation strategy. PLoS ONE 9(8), e105662 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Von Hörsten S, Schmitt I, Nguyen HP et al. Transgenic rat model of Huntington’s disease. Hum. Mol. Genet 12(6), 617–624 (2003). [DOI] [PubMed] [Google Scholar]
- 156.Yu-Taeger L, Petrasch-Parwez E, Osmand AP et al. A novel BACHD transgenic rat exhibits characteristic neuropathological features of Huntington disease. J. Neurosci 32(44), 15426–15438 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Carter RJ, Lione LA, Humby T et al. Characterization of progressive motor deficits in mice transgenic for the human Huntington’s disease mutation. J. Neurosci 19(8), 3248–3257 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Schilling G, Becher MW, Sharp AH et al. Intranuclear inclusions and neuritic aggregates in transgenic mice expressing a mutant N-terminal fragment of huntingtin. Hum. Mol. Genet 8(3), 397–407 (1999). [DOI] [PubMed] [Google Scholar]
- 159.Slow EJ, van Raamsdonk J, Rogers D et al. Selective striatal neuronal loss in a YAC128 mouse model of Huntington disease. Hum. Mol. Genet 12(13), 1555–1567 (2003). [DOI] [PubMed] [Google Scholar]
- 160.Gray M, Shirasaki DI, Cepeda C et al. Full-length human mutant huntingtin with a stable polyglutamine repeat can elicit progressive and selective neuropathogenesis in BACHD mice. J. Neurosci 28(24), 6182–6195 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Lin CH, Tallaksen-Greene S, Chien WM et al. Neurological abnormalities in a knock-in mouse model of Huntington’s disease. Hum. Mol. Genet 10(2), 137–144 (2001). [DOI] [PubMed] [Google Scholar]
- 162.Beal MF, Kowall NW, Ellison DW, Mazurek MF, Swartz KJ, Martin JB. Replication of the neurochemical characteristics of Huntington’s disease by quinolinic acid. Nature 321(6066), 168–171 (1986). [DOI] [PubMed] [Google Scholar]
- 163.Hamilton BF, Gould DH. Nature and distribution of brain lesions in rats intoxicated with 3-nitropropionic acid: a type of hypoxic (energy deficient) brain damage. Acta Neuropathol. (Berl.). 72(3), 286–297 (1987). [DOI] [PubMed] [Google Scholar]
- 164.Yang D, Wang C-E, Zhao B et al. Expression of Huntington’s disease protein results in apoptotic neurons in the brains of cloned transgenic pigs. Hum. Mol. Genet 19(20), 3983–3994 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Jacobsen JC, Bawden CS, Rudiger SR et al. An ovine transgenic Huntington’s disease model. Hum. Mol. Genet 19(10), 1873–1882 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Yang S-H, Cheng P-H, Banta H et al. Towards a transgenic model of Huntington’s disease in a non-human primate. Nature 453(7197), 921–924 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Morton AJ, Howland DS. Large genetic animal models of Huntington’s Disease. J. Huntingt. Dis 2(1), 3–19 (2013). [DOI] [PubMed] [Google Scholar]
- 168.Isakova IA, Baker K, Dufour J, Gaupp D, Phinney DG. Preclinical evaluation of adult stem cell engraftment and toxicity in the CNS of rhesus macaques. Mol. Ther. J. Am. Soc. Gene Ther 13(6), 1173–1184 (2006). [DOI] [PubMed] [Google Scholar]
- 169.Isakova IA, Dufour J, Lanclos C, Bruhn J, Phinney DG. Cell-dose-dependent increases in circulating levels of immune effector cells in rhesus macaques following intracranial injection of allogeneic MSCs. Exp. Hematol 38(10), 957–967.e1 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Isakova IA, Lanclos C, Bruhn J et al. Allo-reactivity of mesenchymal stem cells in rhesus macaques is dose and haplotype dependent and limits durable cell engraftment in vivo. PLoS ONE 9(1), e87238 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Gothelf Y, Abramov N, Harel A, Offen D. Safety of repeated transplantations of neurotrophic factors-secreting human mesenchymal stromal stem cells. Clin. Transl. Med 3, 3 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.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 9(3), 381–386 (2014). [DOI] [PubMed] [Google Scholar]
- 173.Alper J Geron gets green light for human trial of ES cell-derived product. Nat. Biotechnol 27(3), 213–214 (2009). [DOI] [PubMed] [Google Scholar]