Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
bioRxiv ; 2024 Apr 11.
Article En | MEDLINE | ID: mdl-38370637

Microelectrode array (MEA) recordings are commonly used to compare firing and burst rates in neuronal cultures. MEA recordings can also reveal microscale functional connectivity, topology, and network dynamics-patterns seen in brain networks across spatial scales. Network topology is frequently characterized in neuroimaging with graph theoretical metrics. However, few computational tools exist for analyzing microscale functional brain networks from MEA recordings. Here, we present a MATLAB MEA network analysis pipeline (MEA-NAP) for raw voltage time-series acquired from single- or multi-well MEAs. Applications to 3D human cerebral organoids or 2D human-derived or murine cultures reveal differences in network development, including topology, node cartography, and dimensionality. MEA-NAP incorporates multi-unit template-based spike detection, probabilistic thresholding for determining significant functional connections, and normalization techniques for comparing networks. MEA-NAP can identify network-level effects of pharmacologic perturbation and/or disease-causing mutations and, thus, can provide a translational platform for revealing mechanistic insights and screening new therapeutic approaches.

2.
Commun Biol ; 6(1): 678, 2023 06 29.
Article En | MEDLINE | ID: mdl-37386082

Genome-wide association studies identified several disease-causing mutations in neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). However, the contribution of genetic variants to pathway disturbances and their cell type-specific variations, especially in glia, is poorly understood. We integrated ALS GWAS-linked gene networks with human astrocyte-specific multi-omics datasets to elucidate pathognomonic signatures. It predicts that KIF5A, a motor protein kinesin-1 heavy-chain isoform, previously detected only in neurons, can also potentiate disease pathways in astrocytes. Using postmortem tissue and super-resolution structured illumination microscopy in cell-based perturbation platforms, we provide evidence that KIF5A is present in astrocyte processes and its deficiency disrupts structural integrity and mitochondrial transport. We show that this may underly cytoskeletal and trafficking changes in SOD1 ALS astrocytes characterised by low KIF5A levels, which can be rescued by c-Jun N-terminal Kinase-1 (JNK1), a kinesin transport regulator. Altogether, our pipeline reveals a mechanism controlling astrocyte process integrity, a pre-requisite for synapse maintenance and suggests a targetable loss-of-function in ALS.


Amyotrophic Lateral Sclerosis , Proteogenomics , Humans , Amyotrophic Lateral Sclerosis/genetics , Astrocytes , Genome-Wide Association Study , Kinesins/genetics
3.
Neurotrauma Rep ; 3(1): 415-420, 2022.
Article En | MEDLINE | ID: mdl-36204389

Traumatic brain injury and aneurysmal subarachnoid haemorrhage are a major cause of morbidity and mortality worldwide. Treatment options remain limited and are hampered by our understanding of the cellular and molecular mechanisms, including the inflammatory response observed in the brain. Mitochondrial DNA (mtDNA) has been shown to activate an innate inflammatory response by acting as a damage-associated molecular pattern (DAMP). Here, we show raised circulating cell-free (ccf) mtDNA levels in both cerebrospinal fluid (CSF) and serum within 48 h of brain injury. CSF ccf-mtDNA levels correlated with clinical severity and the interleukin-6 cytokine response. These findings support the use of ccf-mtDNA as a biomarker after acute brain injury linked to the inflammatory disease mechanism.

5.
Nat Neurosci ; 24(11): 1542-1554, 2021 11.
Article En | MEDLINE | ID: mdl-34675437

Amyotrophic lateral sclerosis overlapping with frontotemporal dementia (ALS/FTD) is a fatal and currently untreatable disease characterized by rapid cognitive decline and paralysis. Elucidating initial cellular pathologies is central to therapeutic target development, but obtaining samples from presymptomatic patients is not feasible. Here, we report the development of a cerebral organoid slice model derived from human induced pluripotent stem cells (iPSCs) that recapitulates mature cortical architecture and displays early molecular pathology of C9ORF72 ALS/FTD. Using a combination of single-cell RNA sequencing and biological assays, we reveal distinct transcriptional, proteostasis and DNA repair disturbances in astroglia and neurons. We show that astroglia display increased levels of the autophagy signaling protein P62 and that deep layer neurons accumulate dipeptide repeat protein poly(GA), DNA damage and undergo nuclear pyknosis that could be pharmacologically rescued by GSK2606414. Thus, patient-specific iPSC-derived cortical organoid slice cultures are a reproducible translational platform to investigate preclinical ALS/FTD mechanisms as well as novel therapeutic approaches.


Amyotrophic Lateral Sclerosis/pathology , Astrocytes/pathology , Frontotemporal Dementia/pathology , Neurons/pathology , Organoids/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Neurons/metabolism , Organ Culture Techniques/methods , Organoids/metabolism
6.
Nat Neurosci ; 24(3): 312-325, 2021 03.
Article En | MEDLINE | ID: mdl-33589835

Reactive astrocytes are astrocytes undergoing morphological, molecular, and functional remodeling in response to injury, disease, or infection of the CNS. Although this remodeling was first described over a century ago, uncertainties and controversies remain regarding the contribution of reactive astrocytes to CNS diseases, repair, and aging. It is also unclear whether fixed categories of reactive astrocytes exist and, if so, how to identify them. We point out the shortcomings of binary divisions of reactive astrocytes into good-vs-bad, neurotoxic-vs-neuroprotective or A1-vs-A2. We advocate, instead, that research on reactive astrocytes include assessment of multiple molecular and functional parameters-preferably in vivo-plus multivariate statistics and determination of impact on pathological hallmarks in relevant models. These guidelines may spur the discovery of astrocyte-based biomarkers as well as astrocyte-targeting therapies that abrogate detrimental actions of reactive astrocytes, potentiate their neuro- and glioprotective actions, and restore or augment their homeostatic, modulatory, and defensive functions.


Aging/pathology , Astrocytes/pathology , Brain/pathology , Spinal Cord/pathology , Animals , Brain Diseases/pathology , Brain Injuries/pathology , Humans , Spinal Cord Injuries/pathology
7.
Nat Neurosci ; 22(4): 669-679, 2019 04.
Article En | MEDLINE | ID: mdl-30886407

Neural organoids have the potential to improve our understanding of human brain development and neurological disorders. However, it remains to be seen whether these tissues can model circuit formation with functional neuronal output. Here we have adapted air-liquid interface culture to cerebral organoids, leading to improved neuronal survival and axon outgrowth. The resulting thick axon tracts display various morphologies, including long-range projection within and away from the organoid, growth-cone turning, and decussation. Single-cell RNA sequencing reveals various cortical neuronal identities, and retrograde tracing demonstrates tract morphologies that match proper molecular identities. These cultures exhibit active neuronal networks, and subcortical projecting tracts can innervate mouse spinal cord explants and evoke contractions of adjacent muscle in a manner dependent on intact organoid-derived innervating tracts. Overall, these results reveal a remarkable self-organization of corticofugal and callosal tracts with a functional output, providing new opportunities to examine relevant aspects of human CNS development and disease.


Cerebral Cortex/growth & development , Neurons/physiology , Organoids/growth & development , Tissue Culture Techniques/methods , Axons/physiology , Cell Survival , Cerebral Cortex/cytology , Female , Humans , Male , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/cytology , Organoids/cytology , Pluripotent Stem Cells/physiology
8.
Neuron ; 98(2): 306-319.e7, 2018 04 18.
Article En | MEDLINE | ID: mdl-29606582

Diversified neurons are essential for sensorimotor function, but whether astrocytes become specialized to optimize circuit performance remains unclear. Large fast α-motor neurons (FαMNs) of spinal cord innervate fast-twitch muscles that generate peak strength. We report that ventral horn astrocytes express the inward-rectifying K+ channel Kir4.1 (a.k.a. Kcnj10) around MNs in a VGLUT1-dependent manner. Loss of astrocyte-encoded Kir4.1 selectively altered FαMN size and function and led to reduced peak strength. Overexpression of Kir4.1 in astrocytes was sufficient to increase MN size through activation of the PI3K/mTOR/pS6 pathway. Kir4.1 was downregulated cell autonomously in astrocytes derived from amyotrophic lateral sclerosis (ALS) patients with SOD1 mutation. However, astrocyte Kir4.1 was dispensable for FαMN survival even in the mutant SOD1 background. These findings show that astrocyte Kir4.1 is essential for maintenance of peak strength and suggest that Kir4.1 downregulation might uncouple symptoms of muscle weakness from MN cell death in diseases like ALS.


Astrocytes/metabolism , Motor Neurons/metabolism , Potassium Channels, Inwardly Rectifying/biosynthesis , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Animals, Newborn , Astrocytes/chemistry , Astrocytes/pathology , Cells, Cultured , Female , Humans , Induced Pluripotent Stem Cells/chemistry , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Transgenic , Motor Neurons/chemistry , Motor Neurons/pathology , Organ Culture Techniques , Potassium Channels, Inwardly Rectifying/analysis
9.
Nat Commun ; 8(1): 1164, 2017 10 27.
Article En | MEDLINE | ID: mdl-29079839

Astrocyte responses to neuronal injury may be beneficial or detrimental to neuronal recovery, but the mechanisms that determine these different responses are poorly understood. Here we show that ephrin type-B receptor 1 (EphB1) is upregulated in injured motor neurons, which in turn can activate astrocytes through ephrin-B1-mediated stimulation of signal transducer and activator of transcription-3 (STAT3). Transcriptional analysis shows that EphB1 induces a protective and anti-inflammatory signature in astrocytes, partially linked to the STAT3 network. This is distinct from the response evoked by interleukin (IL)-6 that is known to induce both pro inflammatory and anti-inflammatory processes. Finally, we demonstrate that the EphB1-ephrin-B1 pathway is disrupted in human stem cell derived astrocyte and mouse models of amyotrophic lateral sclerosis (ALS). Our work identifies an early neuronal help-me signal that activates a neuroprotective astrocytic response, which fails in ALS, and therefore represents an attractive therapeutic target.


Astrocytes/cytology , Interleukin-6/metabolism , Neurons/metabolism , Receptor, EphB1/metabolism , STAT3 Transcription Factor/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Astrocytes/metabolism , Axons/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Inflammation , Male , Mice , Mice, Inbred C57BL , Motor Neurons/metabolism , Neuroprotection , Sciatic Nerve/metabolism , Signal Transduction , Transcriptome
10.
Cell Rep ; 19(9): 1739-1749, 2017 05 30.
Article En | MEDLINE | ID: mdl-28564594

Motor neurons (MNs) and astrocytes (ACs) are implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS), but their interaction and the sequence of molecular events leading to MN death remain unresolved. Here, we optimized directed differentiation of induced pluripotent stem cells (iPSCs) into highly enriched (> 85%) functional populations of spinal cord MNs and ACs. We identify significantly increased cytoplasmic TDP-43 and ER stress as primary pathogenic events in patient-specific valosin-containing protein (VCP)-mutant MNs, with secondary mitochondrial dysfunction and oxidative stress. Cumulatively, these cellular stresses result in synaptic pathology and cell death in VCP-mutant MNs. We additionally identify a cell-autonomous VCP-mutant AC survival phenotype, which is not attributable to the same molecular pathology occurring in VCP-mutant MNs. Finally, through iterative co-culture experiments, we uncover non-cell-autonomous effects of VCP-mutant ACs on both control and mutant MNs. This work elucidates molecular events and cellular interplay that could guide future therapeutic strategies in ALS.


Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Astrocytes/pathology , Models, Biological , Motor Neurons/pathology , Valosin Containing Protein/metabolism , Cell Survival , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum Stress , Humans , Induced Pluripotent Stem Cells/metabolism , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondria/ultrastructure , Mutation/genetics , Nerve Degeneration/pathology , Neurogenesis , Oxidative Stress , Phenotype , Synapses/pathology
12.
Curr Stem Cell Rep ; 2: 236-247, 2016.
Article En | MEDLINE | ID: mdl-27547709

Astrocytes abound in the human central nervous system (CNS) and play a multitude of indispensable roles in neuronal homeostasis and regulation of synaptic plasticity. While traditionally considered to be merely ancillary supportive cells, their complex yet fundamental relevance to brain physiology and pathology have only become apparent in recent times. Beyond their myriad canonical functions, previously unrecognised region-specific functional heterogeneity of astrocytes is emerging as an important attribute and challenges the traditional perspective of CNS-wide astrocyte homogeneity. Animal models have undeniably provided crucial insights into astrocyte biology, yet interspecies differences may limit the translational yield of such studies. Indeed, experimental systems aiming to understand the function of human astrocytes in health and disease have been hampered by accessibility to enriched cultures. Human induced pluripotent stem cells (hiPSCs) now offer an unparalleled model system to interrogate the role of astrocytes in neurodegenerative disorders. By virtue of their ability to convey mutations at pathophysiological levels in a human system, hiPSCs may serve as an ideal pre-clinical platform for both resolution of pathogenic mechanisms and drug discovery. Here, we review astrocyte specification from hiPSCs and discuss their role in modelling human neurological diseases.

13.
Nat Commun ; 5: 4294, 2014 Jul 11.
Article En | MEDLINE | ID: mdl-25014177

The role of remote astrocyte (AC) reaction to central or peripheral axonal insult is not clearly understood. Here we use a transgenic approach to compare the direct influence of normal with diminished AC reactivity on neuronal integrity and synapse recovery following extracranial facial nerve transection in mice. Our model allows straightforward interpretations of AC-neuron signalling by reducing confounding effects imposed by inflammatory cells. We show direct evidence that perineuronal reactive ACs play a major role in maintaining neuronal circuitry following distant axotomy. We reveal a novel function of astrocytic signal transducer and activator of transcription-3 (STAT3). STAT3 regulates perineuronal astrocytic process formation and re-expression of a synaptogenic molecule, thrombospondin-1 (TSP-1), apart from supporting neuronal integrity. We demonstrate that, through this new pathway, TSP-1 is responsible for the remote AC-mediated recovery of excitatory synapses onto axotomized motor neurons in adult mice. These data provide new targets for neuroprotective therapies via optimizing AC-driven plasticity.


Astrocytes/physiology , Motor Neurons/physiology , Neuronal Plasticity/physiology , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Synapses/physiology , Thrombospondin 1/physiology , Animals , Astrocytes/cytology , Axotomy , Cells, Cultured , Facial Nerve Injuries/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Animal , Nerve Regeneration/physiology , Patch-Clamp Techniques , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , Thrombospondin 1/deficiency , Thrombospondin 1/genetics
14.
J Mol Neurosci ; 54(3): 494-511, 2014 Nov.
Article En | MEDLINE | ID: mdl-24696167

PACAP (ADCYAP1) was isolated from ovine hypothalami. PACAP activates three distinct receptor types: G-protein coupled PAC1, VPAC1, and VPAC2 with seven transmembrane domains. Eight splice variants of PAC1 receptor are described. A part of the hypothalamic PACAP is released into the hypophyseal portal circulation. Both hypothalamic and pituitary PACAP are involved in the dynamic control of gonadotropic hormone secretion. In female rats, PACAP in the paraventricular nucleus is upregulated in the morning and pituitary PACAP is upregulated in the late evening of the proestrus stage of the reproductive cycle. PACAP mRNA peak in the hypothalamic PVN precedes the LHRH release into the portal circulation. It is supposed that PACAP peak is evoked by the elevated estrogen on proestrous morning. At the beginning of the so-called critical period of the same day, PACAP level starts to decline allowing LHRH release into the portal circulation, resulting in the LH surge that evokes ovulation. Just before the critical period, icv-administered exogenous PACAP blocks the LH surge and ovulation. The blocking effect of PACAP is mediated through CRF and endogenous opioids. The effect of the pituitary-born PACAP depends on the intracellular cross-talk between PACAP and LHRH.


Gonadotropins/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Female , Hypothalamus/metabolism , Hypothalamus/physiology , Male , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Gland/metabolism , Pituitary Gland/physiology , Rats
15.
J Neurotrauma ; 22(11): 1282-93, 2005 Nov.
Article En | MEDLINE | ID: mdl-16305316

Intraspinal transplantation of olfactory glial cells (OGC) has produced well-defined beneficial effects in experimental rodent models of spinal cord injury (SCI) and therefore has considerable promise as a treatment for severe SCI in human patients. In this study, we used clinical canine cases of severe SCI to determine whether derivation and transplantation of OGC from an autologous source was feasible. From the nerve fiber layer of a single olfactory bulb, we were able to generate 5 x 10(6) cells from each patient within 3 weeks. Of this population, 72% were p75(+) OGC, 20% were meningeal cells, and the remainder mainly astrocytes. Intraspinal transplantation was not associated with any observable long- or short-term complications.


Cell Transplantation/methods , Neuroglia/transplantation , Olfactory Bulb/cytology , Spinal Cord Injuries/surgery , Spinal Cord Injuries/veterinary , Animals , Biopsy , Cell Transplantation/standards , Cells, Cultured , Defecation , Dogs , Gait , Magnetic Resonance Imaging , Neuroglia/cytology , Olfactory Bulb/surgery , Pain Threshold , Recovery of Function , Reflex , Reproducibility of Results , Spinal Cord Injuries/pathology , Tissue and Organ Harvesting/methods , Tissue and Organ Harvesting/standards , Transplantation, Autologous , Treatment Outcome , Urination
16.
J Neurosci ; 24(44): 9799-810, 2004 Nov 03.
Article En | MEDLINE | ID: mdl-15525765

Schwann cell (SC) and olfactory ensheathing cell (OEC) transplantation has been shown experimentally to promote CNS axonal regeneration and remyelination. To advance this technique into a clinical setting it is important to be able to follow the fates of transplanted cells by noninvasive imaging. Previous studies, using complex modification processes to enable uptake of contrast agents, have shown that cells labeled in vitro with paramagnetic contrast agents transplanted into rodent CNS can be visualized using magnetic resonance imaging (MRI). Here we show that SCs and OECs efficiently internalize dextran-coated superparamagnetic iron oxide (SPIO) from the culture medium by fluid phase pinocytosis. After transplantation into focal areas of demyelination in adult rat spinal cord both transplanted SPIO-labeled SCs and OECs produce a signal reduction using T(2)-weighted MRI in anesthetized rats that persists for up to 4 weeks. Although signal reduction was discernable after transplantation of unlabelled cells, this is nevertheless distinguishable from that produced by transplanted labeled cells. The region of signal reduction in SPIO-labeled cell recipients correlates closely with areas of remyelination. Because the retention of functional integrity by labeled cells is paramount, we also show that SPIO-labeled SCs and OECs are able to myelinate normally after transplantation into focal areas of demyelination. These studies demonstrate the feasibility of noninvasive imaging of transplanted SCs and OECs and represent a significant step toward the clinical application of promising experimental approaches.


Cell Transplantation/physiology , Demyelinating Diseases/surgery , Iron , Olfactory Bulb/cytology , Oxides , Schwann Cells/transplantation , Spinal Cord Injuries/surgery , Animals , Brain Tissue Transplantation/physiology , Demyelinating Diseases/pathology , Demyelinating Diseases/physiopathology , Ferrosoferric Oxide , Magnetic Resonance Imaging , Male , Myelin Sheath/physiology , Nanostructures , Nerve Regeneration/physiology , Pinocytosis , Rats , Rats, Inbred F344 , Schwann Cells/cytology , Schwann Cells/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
17.
Eur J Neurosci ; 18(8): 2253-64, 2003 Oct.
Article En | MEDLINE | ID: mdl-14622186

Glial growth factor-2 (GGF-2) is a neuronally derived isoform of neuregulin shown in vitro to promote proliferation and survival of oligodendrocytes, the myelinating cells of the CNS. Enhanced remyelination has been demonstrated in vivo following systemic delivery of human recombinant GGF-2 (rhGGF-2) in experimental autoimmune encephalomyelitis (EAE). However, it is uncertain whether this is the result of direct effects of rhGGF-2 on cells of the oligodendrocyte lineage or due to modulation of the immune or inflammatory response. If this enhanced remyelination was due to direct effects of rhGGF-2 on cells of the oligodendrocyte lineage then one would expect rhGGF-2 to induce a similar proremyelinating response in nonimmune, gliotoxin models of demyelination. Using a gliotoxin model of demyelination we were therefore able to ascertain the in vivo effect of rhGGF-2 following local CNS delivery in a model that is not confounded by the concurrent presence of an immune-mediated process. No significant alteration in the rate or character of remyelination was evident following local delivery as compared to controls, and indeed nor following systemic delivery in the gliotoxin model. The results of this study therefore indicate that both direct infusion and systemic delivery of rhGGF-2 do not alter remyelination in a nonimmune, gliotoxin model of demyelination. This suggests that the proremyelinating effects of systemically delivered rhGGF-2 in EAE are unlikely to be due to direct effects on the oligodendrocyte lineage, but may be mediated by rhGGF-2 inducing an environment more favourable to remyelination, possibly through modulation of the immune response.


Central Nervous System/drug effects , Demyelinating Diseases/pathology , Myelin Sheath/drug effects , Nerve Tissue Proteins , Neuregulin-1/administration & dosage , Animals , Bromodeoxyuridine/metabolism , Cell Count , Cell Division , Cells, Cultured , Central Nervous System/pathology , Demyelinating Diseases/chemically induced , Dose-Response Relationship, Drug , Drug Administration Routes/veterinary , Enzyme Inhibitors/toxicity , Ethidium/toxicity , Female , Immunohistochemistry/methods , In Vitro Techniques , Indoles/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , RNA, Messenger , Rats , Rats, Sprague-Dawley , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Schwann Cells , Time Factors
18.
Exp Neurol ; 184(1): 237-46, 2003 Nov.
Article En | MEDLINE | ID: mdl-14637095

Both Schwann cells and olfactory ensheathing cells (OECs) create an environment favorable to axon regeneration when transplanted into the damaged CNS. However, transplanted cells can also exert an effect on the host tissue that will influence the extent to which regenerating axons can grow beyond the transplanted area and reenter the host environment. In this study equivalent numbers of Lac-Z-labeled Schwann cells and OECs have been separately transplanted into normal white matter of adult rat spinal cord and the host astrocyte response to each compared. Schwann cell transplantation resulted in a greater area of increased glial fibrillary acidic protein (GFAP) expression compared to that associated with OEC transplantation. This was accompanied by a greater increase in the expression of axon growth inhibitory chrondroitin sulfate proteoglycans (CSPGs) following Schwann cell transplantation compared to OEC transplantation. However, no differences were detected in the increased expression of the specific CSPG neurocan following transplantation of the two cell types. These results mirror differences in the interactions between astrocytes and either Schwann cells or OECs observed in tissue culture models and reveal one aspect of the complex biology of creating regeneration-promoting environments by cell transplantation where transplanted OECs have favorable properties compared to transplanted Schwann cells.


Astrocytes/physiology , Brain/physiology , Chondroitin Sulfate Proteoglycans/biosynthesis , Neurons, Afferent/physiology , Schwann Cells/transplantation , Animals , Brain/cytology , Cells, Cultured , Chondroitin Sulfate Proteoglycans/metabolism , Genes, Reporter/genetics , Immunohistochemistry , Lac Operon/genetics , Lectins, C-Type , Nerve Regeneration/physiology , Nerve Tissue Proteins/metabolism , Neurocan , Neurons, Afferent/transplantation , Olfactory Mucosa/innervation , Rats , Schwann Cells/metabolism , Spinal Cord/pathology , Transduction, Genetic
19.
Brain ; 126(Pt 3): 598-609, 2003 Mar.
Article En | MEDLINE | ID: mdl-12566281

Olfactory ensheathing cells (OECs) are candidate cells for transplant-mediated repair of persistent demyelination in diseases such as multiple sclerosis. If this approach is to make the transition from laboratory to clinic, an important issue is the most suitable composition of the OEC transplant. Isolation of OECs involves concurrent isolation of other cell types, and specific selection techniques are required to produce purified OECs. In this study we address whether the purity of the OEC transplant affects their ability to remyelinate. Surprisingly, we find that a purified preparation of OECs, selected on the basis of low-affinity nerve growth factor receptor (p75) expression, results in less extensive remyelination than an unpurified preparation following transplantation into areas of persistent demyelination in rodent CNS in the X-irradiation/ethidium bromide (X-EB) model. A distinctive feature of the unpurified preparation both in vitro and following transplantation is the presence of meningeal cells. When meningeal cells are added to purified OECs there is a significant improvement in the extent of remyelination compared with the purified OECs, although if the cells are present in too great an abundance this beneficial effect is lost. These results highlight the important concept that the regenerative properties of OECs are profoundly influenced by the cells with which they are transplanted.


Central Nervous System , Meninges/cytology , Meninges/transplantation , Multiple Sclerosis/therapy , Olfactory Bulb/cytology , Olfactory Bulb/transplantation , Animals , Biomarkers/analysis , Cell Separation/methods , Fibronectins/analysis , Male , Models, Animal , Myelin Sheath/physiology , Rats , Rats, Inbred F344
20.
Curr Opin Neurol ; 15(6): 701-5, 2002 Dec.
Article En | MEDLINE | ID: mdl-12447108

PURPOSE OF REVIEW: This article reviews recent advances in the use of cell transplantation to promote recovery from traumatic injury of the CNS, focusing on axonal regeneration in the spinal cord. RECENT FINDINGS: The significant recent findings reported are: (1) the increased expression of inhibitory chondroitin sulphate-proteoglycans in host tissue following Schwann cell transplantation, highlighting the effects the transplant may have on the ability of the host tissue to support regeneration; (2) the ability of embryonic and neural stem cells to promote recovery following transplantation into experimental models of spinal cord injury; (3) that delayed grafting for several weeks after transplantation does not diminish the graft effectiveness and may be advantageous; (4) the use of transplanted fibroblasts engineered to express neurotrophic genes in a conditionally regulated manner using tetracycline-inducible promoters; and (5) the initial reports on phase 1 clinical trials of foetal spinal cord grafts into patients with post-traumatic syringomyelia demonstrating their feasibility and safety. SUMMARY: Recent advances largely involve experimental refinements of existing approaches and the emergent application of stem cell biology to overcome spinal cord injury. While most experimental studies concentrate on single or restricted combinations of approaches, the most effective clinical strategies will be multi-component. Their formulation will require the development of intermediate models for bridging the differences between experimental models in laboratory animals and naturally occurring traumatic injury in humans.


Brain Injuries/surgery , Fetal Tissue Transplantation/trends , Spinal Cord Injuries/surgery , Stem Cell Transplantation/trends , Animals , Axons , Brain Injuries/physiopathology , Fetal Tissue Transplantation/methods , Fibroblasts/transplantation , Humans , Macrophages/transplantation , Nerve Growth Factors/therapeutic use , Nerve Regeneration , Schwann Cells/transplantation , Spinal Cord Injuries/physiopathology , Stem Cell Transplantation/methods , Syringomyelia/therapy
...