Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 117(16): 9094-9100, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32253308

ABSTRACT

Stem cell transplantation can improve behavioral recovery after stroke in animal models but whether stem cell-derived neurons become functionally integrated into stroke-injured brain circuitry is poorly understood. Here we show that intracortically grafted human induced pluripotent stem (iPS) cell-derived cortical neurons send widespread axonal projections to both hemispheres of rats with ischemic lesions in the cerebral cortex. Using rabies virus-based transsynaptic tracing, we find that at 6 mo after transplantation, host neurons in the contralateral somatosensory cortex receive monosynaptic inputs from grafted neurons. Immunoelectron microscopy demonstrates myelination of the graft-derived axons in the corpus callosum and that their terminals form excitatory, glutamatergic synapses on host cortical neurons. We show that the stroke-induced asymmetry in a sensorimotor (cylinder) test is reversed by transplantation. Light-induced inhibition of halorhodopsin-expressing, grafted neurons does not recreate the impairment, indicating that its reversal is not due to neuronal activity in the graft. However, we find bilateral decrease of motor performance in the cylinder test after light-induced inhibition of either grafted or endogenous halorhodopsin-expressing cortical neurons, located in the same area, and after inhibition of endogenous halorhodopsin-expressing cortical neurons by exposure of their axons to light on the contralateral side. Our data indicate that activity in the grafted neurons, probably mediated through transcallosal connections to the contralateral hemisphere, is involved in maintaining normal motor function. This is an example of functional integration of efferent projections from grafted neurons into the stroke-affected brain's neural circuitry, which raises the possibility that such repair might be achievable also in humans affected by stroke.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Infarction, Middle Cerebral Artery/therapy , Motor Activity/physiology , Neurons/transplantation , Somatosensory Cortex/physiopathology , Action Potentials/physiology , Animals , Behavior Observation Techniques , Behavior, Animal/physiology , Cell Differentiation/physiology , Cell Line , Disease Models, Animal , Humans , Infarction, Middle Cerebral Artery/etiology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Neurons/physiology , Optogenetics , Patch-Clamp Techniques , Rats , Recovery of Function , Somatosensory Cortex/cytology , Somatosensory Cortex/pathology
3.
Proc Natl Acad Sci U S A ; 113(23): 6544-9, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27140603

ABSTRACT

Clinical trials using cells derived from embryonic ventral mesencephalon have shown that transplanted dopaminergic neurons can survive and function in the long term, as demonstrated by in vivo brain imaging using (18)F-fluorodopa and (11)C-raclopride positron emission tomography. Here we report the postmortem analysis of a patient with Parkinson's disease who 24 y earlier underwent unilateral transplantation of embryonic dopaminergic neurons in the putamen and subsequently exhibited major motor improvement and recovery of striatal dopaminergic function. Histopathological analysis showed that a dense, near-normal graft-derived dopaminergic reinnervation of the putamen can be maintained for a quarter of a century despite severe host brain pathology and with no evidence of immune response. In addition, ubiquitin- and α-synuclein-positive inclusions were seen, some with the appearance of typical Lewy bodies, in 11-12% of the grafted dopaminergic neurons, reflecting the spread of pathology from the host brain to the transplants. Because the clinical benefits induced by transplantation in this patient were gradually lost after 14 y posttransplantation, our findings provide the first reported evidence, to our knowledge, that even a viable dopaminergic graft giving rise to extensive striatal reinnervation may lose its efficacy if widespread degenerative changes develop in the host brain.


Subject(s)
Dopamine , Fetal Tissue Transplantation , Corpus Striatum , Humans , Mesencephalon/embryology , Neurons , Parkinson Disease , Putamen , alpha-Synuclein
4.
Brain ; 140(3): 692-706, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28115364

ABSTRACT

Transplanted neurons derived from stem cells have been proposed to improve function in animal models of human disease by various mechanisms such as neuronal replacement. However, whether the grafted neurons receive functional synaptic inputs from the recipient's brain and integrate into host neural circuitry is unknown. Here we studied the synaptic inputs from the host brain to grafted cortical neurons derived from human induced pluripotent stem cells after transplantation into stroke-injured rat cerebral cortex. Using the rabies virus-based trans-synaptic tracing method and immunoelectron microscopy, we demonstrate that the grafted neurons receive direct synaptic inputs from neurons in different host brain areas located in a pattern similar to that of neurons projecting to the corresponding endogenous cortical neurons in the intact brain. Electrophysiological in vivo recordings from the cortical implants show that physiological sensory stimuli, i.e. cutaneous stimulation of nose and paw, can activate or inhibit spontaneous activity in grafted neurons, indicating that at least some of the afferent inputs are functional. In agreement, we find using patch-clamp recordings that a portion of grafted neurons respond to photostimulation of virally transfected, channelrhodopsin-2-expressing thalamo-cortical axons in acute brain slices. The present study demonstrates, for the first time, that the host brain regulates the activity of grafted neurons, providing strong evidence that transplanted human induced pluripotent stem cell-derived cortical neurons can become incorporated into injured cortical circuitry. Our findings support the idea that these neurons could contribute to functional recovery in stroke and other conditions causing neuronal loss in cerebral cortex.


Subject(s)
Brain Injuries/surgery , Evoked Potentials, Somatosensory/physiology , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/transplantation , Synapses/physiology , Action Potentials , Afferent Pathways/physiology , Animals , Brain/cytology , Brain/ultrastructure , Brain Injuries/etiology , Cell Line, Transformed , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/ultrastructure , Disease Models, Animal , Humans , Lysine/analogs & derivatives , Lysine/metabolism , Male , Neurons/physiology , Neurons/ultrastructure , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Nude , Rats, Sprague-Dawley , Stroke/complications , Synapses/ultrastructure , Ventral Thalamic Nuclei/cytology
5.
J Neurosci ; 36(15): 4182-95, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-27076418

ABSTRACT

Stroke is a leading cause of disability and currently lacks effective therapy enabling long-term functional recovery. Ischemic brain injury causes local inflammation, which involves both activated resident microglia and infiltrating immune cells, including monocytes. Monocyte-derived macrophages (MDMs) exhibit a high degree of functional plasticity. Here, we determined the role of MDMs in long-term spontaneous functional recovery after middle cerebral artery occlusion in mice. Analyses by flow cytometry and immunocytochemistry revealed that monocytes home to the stroke-injured hemisphere., and that infiltration peaks 3 d after stroke. At day 7, half of the infiltrating MDMs exhibited a bias toward a proinflammatory phenotype and the other half toward an anti-inflammatory phenotype, but during the subsequent 2 weeks, MDMs with an anti-inflammatory phenotype dominated. Blocking monocyte recruitment using the anti-CCR2 antibody MC-21 during the first week after stroke abolished long-term behavioral recovery, as determined in corridor and staircase tests, and drastically decreased tissue expression of anti-inflammatory genes, including TGFß, CD163, and Ym1. Our results show that spontaneously recruited monocytes to the injured brain early after the insult contribute to long-term functional recovery after stroke. SIGNIFICANCE STATEMENT: For decades, any involvement of circulating immune cells in CNS repair was completely denied. Only over the past few years has involvement of monocyte-derived macrophages (MDMs) in CNS repair received appreciation. We show here, for the first time, that MDMs recruited to the injured brain early after ischemic stroke contribute to long-term spontaneous functional recovery through inflammation-resolving activity. Our data raise the possibility that inadequate recruitment of MDMs to the brain after stroke underlies the incomplete functional recovery seen in patients and that boosting homing of MDMs with an anti-inflammatory bias to the injured brain tissue may be a new therapeutic approach to promote long-term improvement after stroke.


Subject(s)
Macrophages , Monocytes , Recovery of Function , Stroke/physiopathology , Animals , Antibodies, Blocking/pharmacology , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/biosynthesis , Antigens, Differentiation, Myelomonocytic/genetics , Behavior, Animal/drug effects , Chimera , Functional Laterality , Infarction, Middle Cerebral Artery/physiopathology , Inflammation/pathology , Lectins/biosynthesis , Lectins/genetics , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Monocytes/pathology , Neuronal Plasticity/physiology , Psychomotor Performance/drug effects , Receptors, CCR2/antagonists & inhibitors , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Recovery of Function/drug effects , Stroke/pathology , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , beta-N-Acetylhexosaminidases/biosynthesis , beta-N-Acetylhexosaminidases/genetics
6.
EMBO J ; 32(11): 1489-95, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23644381

ABSTRACT

At the time of writing, the Italian Parliament is debating a new law that would make it legal to practice an unproven stem cell treatment in public hospitals. The treatment, offered by a private non-medical organization, may not be safe, lacks a rationale, and violates current national laws and European regulations. This case raises multiple concerns, most prominently the urgent need to protect patients who are severely ill, exposed to significant risks, and vulnerable to exploitation. The scientific community must consider the context-social, financial, medical, legal-in which stem cell science is currently situated and the need for stringent regulation. Additional concerns are emerging. These emanate from the novel climate, created within science itself, and stem cell science in particular, by the currently prevailing model of 'translational medicine'. Only rigorous science and rigorous regulation can ensure translation of science into effective therapies rather than into ineffective market products, and mark, at the same time, the sharp distinction between the striving for new therapies and the deceit of patients.


Subject(s)
Stem Cell Transplantation/legislation & jurisprudence , Europe , Humans , Italy , Patient Safety , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/standards , Stem Cells , Translational Research, Biomedical
7.
J Neuroinflammation ; 14(1): 153, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28754163

ABSTRACT

BACKGROUND: Choroid plexus (CP) supports the entry of monocyte-derived macrophages (MDMs) to the central nervous system in animal models of traumatic brain injury, spinal cord injury, and Alzheimer's disease. Whether the CP is involved in the recruitment of MDMs to the injured brain after ischemic stroke is unknown. METHODS: Adult male C57BL/6 mice were subjected to focal cortical ischemia by permanent occlusion of the distal branch of the right middle cerebral artery. Choroid plexus tissues were collected and analyzed for Vcam1, Madcam1, Cx3cl1, Ccl2, Nt5e, and Ifnγ expression at different timepoints after stroke using qPCR. Changes of MDMs in CP and cerebrospinal fluid (CSF) at 1 day and 3 days after stroke were analyzed using flow cytometry. Infiltration of MDMs into CP and CSF were validated using ß-actin-GFP chimeric mice and Fgd5-CreERT2 x Lox-stop-lox-Tomato mice. CD115+ monocytes were isolated using a magnetic cell separation system from bone marrow of Cx3cr1-GFP or wild-type C57BL/6 donor mice. The freshly isolated monocytes or M2-like MDMs primed in vitro with IL4 and IL13 were stereotaxically injected into the lateral ventricle of stroke-affected mice to trace for their migration into ischemic hemisphere or to assess their effect on post-stroke recovery using open field, corridor, and active avoidance behavioral tests. RESULTS: We found that CP responded to cortical stroke by upregulation of gene expression for several possible mediators of MDM trafficking and, concomitantly, MDMs increased in CP and cerebrospinal fluid (CSF). We then confirmed that MDMs infiltrated from blood into CP and CSF after the insult using ß-actin-GFP chimeric mice and Fgd5-CreERT2 x Lox-stop-lox-Tomato mice. When MDMs were directly administered into CSF following stroke, they homed to the ischemic hemisphere. If they had been primed in vitro prior to their administration to become M2-like macrophages, they promoted post-stroke recovery of motor and cognitive function without influencing infarct volume. CONCLUSIONS: Our findings suggest the possibility that autologous transplantation of M2-like MDMs into CSF might be developed into a new strategy for promoting recovery also in patients with stroke.


Subject(s)
Cerebrospinal Fluid/metabolism , Choroid Plexus/pathology , Macrophages/pathology , Monocytes/pathology , Stroke/pathology , Actins/genetics , Actins/metabolism , Animals , Antigens, CD/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Monocytes/metabolism , Nerve Tissue Proteins/metabolism , Stroke/physiopathology , Time Factors , Zonula Occludens-1 Protein/metabolism
8.
Neurobiol Dis ; 83: 1-15, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26299391

ABSTRACT

Ischemic stroke triggers neurogenesis from neural stem/progenitor cells (NSPCs) in the subventricular zone (SVZ) and migration of newly formed neuroblasts toward the damaged striatum where they differentiate to mature neurons. Whether it is the injury per se or the associated inflammation that gives rise to this endogenous neurogenic response is unknown. Here we showed that inflammation without corresponding neuronal loss caused by intrastriatal lipopolysaccharide (LPS) injection leads to striatal neurogenesis in rats comparable to that after a 30 min middle cerebral artery occlusion, as characterized by striatal DCX+ neuroblast recruitment and mature NeuN+/BrdU+ neuron formation. Using global gene expression analysis, changes in several factors that could potentially regulate striatal neurogenesis were identified in microglia sorted from SVZ and striatum of LPS-injected and stroke-subjected rats. Among the upregulated factors, one chemokine, CXCL13, was found to promote neuroblast migration from neonatal mouse SVZ explants in vitro. However, neuroblast migration to the striatum was not affected in constitutive CXCL13 receptor CXCR5(-/-) mice subjected to stroke. Infarct volume and pro-inflammatory M1 microglia/macrophage density were increased in CXCR5(-/-) mice, suggesting that microglia-derived CXCL13, acting through CXCR5, might be involved in neuroprotection following stroke. Our findings raise the possibility that the inflammation accompanying an ischemic insult is the major inducer of striatal neurogenesis after stroke.


Subject(s)
Corpus Striatum/physiopathology , Encephalitis/physiopathology , Infarction, Middle Cerebral Artery/physiopathology , Neural Stem Cells/physiology , Neurogenesis , Neurons/physiology , Stroke/physiopathology , Animals , Cell Death , Cell Movement/drug effects , Chemokine CXCL13/pharmacology , Chemokine CXCL13/physiology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Doublecortin Protein , Encephalitis/chemically induced , Encephalitis/metabolism , Gene Expression , Infarction, Middle Cerebral Artery/pathology , Inflammation Mediators/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Lateral Ventricles/physiopathology , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/metabolism , Neurons/pathology , Rats , Rats, Wistar , Receptors, CXCR5/genetics , Receptors, CXCR5/physiology , Stroke/pathology
9.
Brain ; 136(Pt 12): 3561-77, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24148272

ABSTRACT

Stem cell-based approaches to restore function after stroke through replacement of dead neurons require the generation of specific neuronal subtypes. Loss of neurons in the cerebral cortex is a major cause of stroke-induced neurological deficits in adult humans. Reprogramming of adult human somatic cells to induced pluripotent stem cells is a novel approach to produce patient-specific cells for autologous transplantation. Whether such cells can be converted to functional cortical neurons that survive and give rise to behavioural recovery after transplantation in the stroke-injured cerebral cortex is not known. We have generated progenitors in vitro, expressing specific cortical markers and giving rise to functional neurons, from long-term self-renewing neuroepithelial-like stem cells, produced from adult human fibroblast-derived induced pluripotent stem cells. At 2 months after transplantation into the stroke-damaged rat cortex, the cortically fated cells showed less proliferation and more efficient conversion to mature neurons with morphological and immunohistochemical characteristics of a cortical phenotype and higher axonal projection density as compared with non-fated cells. Pyramidal morphology and localization of the cells expressing the cortex-specific marker TBR1 in a certain layered pattern provided further evidence supporting the cortical phenotype of the fated, grafted cells, and electrophysiological recordings demonstrated their functionality. Both fated and non-fated cell-transplanted groups showed bilateral recovery of the impaired function in the stepping test compared with vehicle-injected animals. The behavioural improvement at this early time point was most likely not due to neuronal replacement and reconstruction of circuitry. At 5 months after stroke in immunocompromised rats, there was no tumour formation and the grafted cells exhibited electrophysiological properties of mature neurons with evidence of integration in host circuitry. Our findings show, for the first time, that human skin-derived induced pluripotent stem cells can be differentiated to cortical neuronal progenitors, which survive, differentiate to functional neurons and improve neurological outcome after intracortical implantation in a rat stroke model.


Subject(s)
Cerebral Cortex/cytology , Induced Pluripotent Stem Cells/physiology , Infarction, Middle Cerebral Artery/surgery , Neurons/physiology , Recovery of Function/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Cerebral Cortex/transplantation , Disease Models, Animal , Electric Stimulation , Glutaminase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/transplantation , Infarction, Middle Cerebral Artery/pathology , Neurons/classification , Neurons/drug effects , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , Rats , Rats, Nude , Rats, Sprague-Dawley
10.
Exp Cell Res ; 319(18): 2790-800, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24075965

ABSTRACT

Ependymal cells in the lateral ventricular wall are considered to be post-mitotic but can give rise to neuroblasts and astrocytes after stroke in adult mice due to insult-induced suppression of Notch signaling. The transcription factor FoxJ1, which has been used to characterize mouse ependymal cells, is also expressed by a subset of astrocytes. Cells expressing FoxJ1, which drives the expression of motile cilia, contribute to early postnatal neurogenesis in mouse olfactory bulb. The distribution and progeny of FoxJ1-expressing cells in rat forebrain are unknown. Here we show using immunohistochemistry that the overall majority of FoxJ1-expressing cells in the lateral ventricular wall of adult rats are ependymal cells with a minor population being astrocytes. To allow for long-term fate mapping of FoxJ1-derived cells, we used the piggyBac system for in vivo gene transfer with electroporation. Using this method, we found that FoxJ1-expressing cells, presumably the astrocytes, give rise to neuroblasts and mature neurons in the olfactory bulb both in intact and stroke-damaged brain of adult rats. No significant contribution of FoxJ1-derived cells to stroke-induced striatal neurogenesis was detected. These data indicate that in the adult rat brain, FoxJ1-expressing cells contribute to the formation of new neurons in the olfactory bulb but are not involved in the cellular repair after stroke.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , DNA Transposable Elements/genetics , Electroporation , Forkhead Transcription Factors/metabolism , Neurogenesis/physiology , Prosencephalon/cytology , Prosencephalon/metabolism , Animals , Cell Differentiation , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Forkhead Transcription Factors/genetics , Immunohistochemistry , Male , Neurogenesis/genetics , Olfactory Bulb/cytology , Rats , Rats, Wistar
11.
Proc Natl Acad Sci U S A ; 108(25): 10343-8, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21646515

ABSTRACT

Recent reports demonstrate that somatic mouse cells can be directly converted to other mature cell types by using combined expression of defined factors. Here we show that the same strategy can be applied to human embryonic and postnatal fibroblasts. By overexpression of the transcription factors Ascl1, Brn2, and Myt1l, human fibroblasts were efficiently converted to functional neurons. We also demonstrate that the converted neurons can be directed toward distinct functional neurotransmitter phenotypes when the appropriate transcriptional cues are provided together with the three conversion factors. By combining expression of the three conversion factors with expression of two genes involved in dopamine neuron generation, Lmx1a and FoxA2, we could direct the phenotype of the converted cells toward dopaminergic neurons. Such subtype-specific induced neurons derived from human somatic cells could be valuable for disease modeling and cell replacement therapy.


Subject(s)
Cell Transdifferentiation/physiology , Dopamine/metabolism , Fibroblasts/physiology , Neurons/physiology , Action Potentials/physiology , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Neurons/cytology , POU Domain Factors/genetics , POU Domain Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
12.
J Neurosci ; 32(15): 5151-64, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22496561

ABSTRACT

Ischemic stroke causes transient increase of neural stem and progenitor cell (NSPC) proliferation in the subventricular zone (SVZ), and migration of newly formed neuroblasts toward the damaged area where they mature to striatal neurons. The molecular mechanisms regulating this plastic response, probably involved in structural reorganization and functional recovery, are poorly understood. The adaptor protein LNK suppresses hematopoietic stem cell self-renewal, but its presence and role in the brain are poorly understood. Here we demonstrate that LNK is expressed in NSPCs in the adult mouse and human SVZ. Lnk(-/-) mice exhibited increased NSPC proliferation after stroke, but not in intact brain or following status epilepticus. Deletion of Lnk caused increased NSPC proliferation while overexpression decreased mitotic activity of these cells in vitro. We found that Lnk expression after stroke increased in SVZ through the transcription factors STAT1/3. LNK attenuated insulin-like growth factor 1 signaling by inhibition of AKT phosphorylation, resulting in reduced NSPC proliferation. Our findings identify LNK as a stroke-specific, endogenous negative regulator of NSPC proliferation, and suggest that LNK signaling is a novel mechanism influencing plastic responses in postischemic brain.


Subject(s)
Brain Ischemia/pathology , Brain/cytology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Neural Stem Cells/physiology , Stroke/pathology , Adaptor Proteins, Signal Transducing , Animals , Antimetabolites , Bromodeoxyuridine , Cell Proliferation , Cell Survival , Cells, Cultured , Chromatin Immunoprecipitation , Electroporation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Male , Membrane Proteins , Mice , Mice, Knockout , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/physiology , Real-Time Polymerase Chain Reaction , Recovery of Function , Retroviridae/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/physiology , Transcription Factors/metabolism , Transfection/methods
13.
Neurobiol Dis ; 52: 191-203, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23276704

ABSTRACT

Neural stem/progenitor cells (NSPCs) in subventricular zone (SVZ) produce new striatal neurons during several months after stroke, which may contribute to recovery. Intracerebral grafts of NSPCs can exert beneficial effects after stroke through neuronal replacement, trophic actions, neuroprotection, and modulation of inflammation. Here we have explored whether human fetal striatum-derived NSPC-grafts influence striatal neurogenesis and promote recovery in stroke-damaged brain. T cell-deficient rats were subjected to 1h middle cerebral artery occlusion (MCAO). Human fetal NSPCs or vehicle were implanted into ipsilateral striatum 48 h after MCAO, animals were assessed behaviorally, and perfused at 6 or 14 weeks. Grafted human NSPCs survived in all rats, and a subpopulation had differentiated to neuroblasts or mature neurons at 6 and 14 weeks. Numbers of proliferating cells in SVZ and new migrating neuroblasts and mature neurons were higher, and numbers of activated microglia/macrophages were lower in the ischemic striatum of NSPC-grafted compared to vehicle-injected group both at 6 and 14 weeks. A fraction of grafted NSPCs projected axons from striatum to globus pallidus. The NSPC-grafted rats showed improved functional recovery in stepping and cylinder tests from 6 and 12 weeks, respectively. Our data show, for the first time, that intrastriatal implants of human fetal NSPCs exert a long-term enhancement of several steps of striatal neurogensis after stroke. The grafts also suppress striatal inflammation and ameliorate neurological deficits. Our findings support the idea that combination of NSPC transplantation and stimulation of neurogenesis from endogenous NSPCs may become a valuable strategy for functional restoration after stroke.


Subject(s)
Infarction, Middle Cerebral Artery/surgery , Motor Skills/physiology , Neural Stem Cells/transplantation , Neurogenesis/physiology , Recovery of Function/physiology , Animals , Cell Movement/physiology , Corpus Striatum/physiopathology , Humans , Infarction, Middle Cerebral Artery/physiopathology , Male , Neurons/physiology , Rats , Rats, Nude , Walking/physiology
14.
Neurobiol Dis ; 49: 118-27, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22940632

ABSTRACT

Human fetal midbrain tissue grafting has provided proof-of-concept for dopamine cell replacement therapy (CRT) in Parkinson's disease (PD). However, limited tissue availability has hindered the development and widespread use of this experimental therapy. Here we present a method for generating large numbers of midbrain dopaminergic (DA) neurons based on expanding and differentiating neural stem/progenitor cells present in the human ventral midbrain (hVM) tissue. Our results show that hVM neurospheres (hVMN) with low cell numbers, unlike their rodent counterparts, expand the total number of cells 3-fold, whilst retaining their capacity to differentiate into midbrain DA neurons. Moreover, Wnt5a promoted DA differentiation of expanded cells resulting in improved morphological maturation, midbrain DA marker expression, DA release and electrophysiological properties. This method results in cell preparations that, after expansion and differentiation, can contain 6-fold more midbrain DA neurons than the starting VM preparation. Thus, our results provide evidence that by improving expansion and differentiation of progenitors present in the hVM it is possible to greatly enrich cell preparations for DA neurons. This method could substantially reduce the amount of human fetal midbrain tissue necessary for CRT in patients with PD, which could have major implications for the widespread adoption of this approach.


Subject(s)
Cell Culture Techniques , Dopaminergic Neurons/physiology , Mesencephalon/embryology , Mesencephalon/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Cell Count , Chromatography, High Pressure Liquid , Dopamine/metabolism , Dopaminergic Neurons/cytology , Humans , Immunohistochemistry , Mesencephalon/cytology , Neural Stem Cells/cytology , Patch-Clamp Techniques , Polymerase Chain Reaction , Proto-Oncogene Proteins/administration & dosage , Wnt Proteins/administration & dosage , Wnt-5a Protein
15.
Nat Rev Neurosci ; 9(10): 741-5, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18769444

ABSTRACT

Neuropathological changes in Parkinson's disease progress slowly and spread according to a characteristic pattern. Recent papers have shed light on this progression of pathology by examining the fate of neurons grafted into the brains of patients with Parkinson's disease. Two of these studies demonstrate that grafted healthy neurons can gradually develop the same pathology as host neurons in the diseased brains. According to these studies, implanted neurons developed alpha-synuclein- and ubiquitin-positive Lewy bodies more than a decade after transplantation. We discuss the possible underlying mechanisms and their implications for how pathology spreads in Parkinson's disease.


Subject(s)
Brain Tissue Transplantation/adverse effects , Brain/physiopathology , Graft Survival/physiology , Nerve Degeneration/physiopathology , Parkinson Disease/physiopathology , Brain/pathology , Disease Progression , Humans , Lewy Bodies/metabolism , Lewy Bodies/pathology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Oxidative Stress/physiology , Parkinson Disease/pathology , Parkinson Disease/therapy , alpha-Synuclein/metabolism
16.
Stem Cells ; 30(12): 2657-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22961761

ABSTRACT

Transplantation of neural stem cells (NSCs) is a novel strategy to restore function in the diseased brain, acting through multiple mechanisms, for example, neuronal replacement, neuroprotection, and modulation of inflammation. Whether transplanted NSCs can operate by fusing with microglial cells or mature neurons is largely unknown. Here, we have studied the interaction of a mouse embryonic stem cell-derived neural stem (NS) cell line with rat and mouse microglia and neurons in vitro and in vivo. We show that NS cells spontaneously fuse with cocultured cortical neurons, and that this process requires the presence of microglia. Our in vitro data indicate that the NS cells can first fuse with microglia and then with neurons. The fused NS/microglial cells express markers and retain genetic and functional characteristics of both parental cell types, being able to respond to microglia-specific stimuli (LPS and IL-4/IL-13) and to differentiate to neurons and astrocytes. The NS cells fuse with microglia, at least partly, through interaction between phosphatidylserine exposed on the surface of NS cells and CD36 receptor on microglia. Transplantation of NS cells into rodent cortex results in fusion with mature pyramidal neurons, which often carry two nuclei, a process probably mediated by microglia. The fusogenic role of microglia could be even more important after NSC transplantation into brains affected by neurodegenerative diseases associated with microglia activation. It remains to be elucidated how the occurrence of the fused cells will influence the functional outcome after NSC transplantation in the diseased brain.


Subject(s)
Embryonic Stem Cells/cytology , Microglia/cytology , Neural Stem Cells/cytology , Neurons/cytology , Animals , Cell Differentiation/physiology , Cell Fusion , Cells, Cultured , Embryonic Stem Cells/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Microglia/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Rats , Rats, Wistar
17.
Stem Cells ; 30(6): 1120-33, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22495829

ABSTRACT

Reprogramming of adult human somatic cells to induced pluripotent stem cells (iPSCs) is a novel approach to produce patient-specific cells for autologous transplantation. Whether such cells survive long-term, differentiate to functional neurons, and induce recovery in the stroke-injured brain are unclear. We have transplanted long-term self-renewing neuroepithelial-like stem cells, generated from adult human fibroblast-derived iPSCs, into the stroke-damaged mouse and rat striatum or cortex. Recovery of forepaw movements was observed already at 1 week after transplantation. Improvement was most likely not due to neuronal replacement but was associated with increased vascular endothelial growth factor levels, probably enhancing endogenous plasticity. Transplanted cells stopped proliferating, could survive without forming tumors for at least 4 months, and differentiated to morphologically mature neurons of different subtypes. Neurons in intrastriatal grafts sent axonal projections to the globus pallidus. Grafted cells exhibited electrophysiological properties of mature neurons and received synaptic input from host neurons. Our study provides the first evidence that transplantation of human iPSC-derived cells is a safe and efficient approach to promote recovery after stroke and can be used to supply the injured brain with new neurons for replacement.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Neurons/cytology , Stem Cell Transplantation/methods , Stroke/pathology , Stroke/surgery , Aged , Animals , Brain/cytology , Brain/pathology , Cell Differentiation/physiology , Cells, Cultured , Female , Humans , Immunohistochemistry , Mice , Rats
18.
Mov Disord ; 28(3): 268-73, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23401015

ABSTRACT

Despite 3 decades of basic and clinical studies, there is still no dopaminergic cell therapy for Parkinson's disease. Several arguments have been put forward why this approach, so far tested with transplantation of human fetal mesencephalic dopamine-rich tissue, will never be of clinical use and should be abandoned: (1) Lack of efficacy in 2 sham surgery-controlled trials; (2) occurrence of troublesome off-medication dyskinesias in a subgroup of grafted patients; (3) disease process destroys grafted neurons; and (4) non-motor symptoms will not be influenced by intrastriatal dopaminergic grafts. Here, the author argues that, based on recent scientific advancements, the development of a dopaminergic cell therapy for Parkinson's disease should continue. Factors influencing the outcome after transplantation have now been identified, and dopaminergic neurons can be generated in large numbers from stem cells. Mechanisms of graft-induced dyskinesias are much better understood, and patients with well functioning grafts can exhibit long-term motor recovery of therapeutic value even in the presence of non-motor symptoms.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Dopaminergic Neurons/physiology , Parkinson Disease/surgery , Animals , Humans
19.
Stem Cell Reports ; 18(8): 1643-1656, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37236198

ABSTRACT

Neuronal loss and axonal demyelination underlie long-term functional impairments in patients affected by brain disorders such as ischemic stroke. Stem cell-based approaches reconstructing and remyelinating brain neural circuitry, leading to recovery, are highly warranted. Here, we demonstrate the in vitro and in vivo production of myelinating oligodendrocytes from a human induced pluripotent stem cell (iPSC)-derived long-term neuroepithelial stem (lt-NES) cell line, which also gives rise to neurons with the capacity to integrate into stroke-injured, adult rat cortical networks. Most importantly, the generated oligodendrocytes survive and form myelin-ensheathing human axons in the host tissue after grafting onto adult human cortical organotypic cultures. This lt-NES cell line is the first human stem cell source that, after intracerebral delivery, can repair both injured neural circuitries and demyelinated axons. Our findings provide supportive evidence for the potential future use of human iPSC-derived cell lines to promote effective clinical recovery following brain injuries.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Rats , Adult , Animals , Cell Differentiation/physiology , Neurons , Oligodendroglia/metabolism , Axons/physiology , Myelin Sheath/physiology
20.
Cell Stem Cell ; 30(10): 1299-1314.e9, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37802036

ABSTRACT

Cell replacement therapies for Parkinson's disease (PD) based on transplantation of pluripotent stem cell-derived dopaminergic neurons are now entering clinical trials. Here, we present quality, safety, and efficacy data supporting the first-in-human STEM-PD phase I/IIa clinical trial along with the trial design. The STEM-PD product was manufactured under GMP and quality tested in vitro and in vivo to meet regulatory requirements. Importantly, no adverse effects were observed upon testing of the product in a 39-week rat GLP safety study for toxicity, tumorigenicity, and biodistribution, and a non-GLP efficacy study confirmed that the transplanted cells mediated full functional recovery in a pre-clinical rat model of PD. We further observed highly comparable efficacy results between two different GMP batches, verifying that the product can be serially manufactured. A fully in vivo-tested batch of STEM-PD is now being used in a clinical trial of 8 patients with moderate PD, initiated in 2022.


Subject(s)
Human Embryonic Stem Cells , Parkinson Disease , Humans , Rats , Animals , Parkinson Disease/therapy , Tissue Distribution , Cell Differentiation/physiology , Stem Cell Transplantation/methods , Dopaminergic Neurons/physiology
SELECTION OF CITATIONS
SEARCH DETAIL