Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(2)2024 Jan 06.
Article in English | MEDLINE | ID: mdl-38255817

ABSTRACT

Demyelination in the central nervous system (CNS) resulting from injury or disease can cause loss of nerve function and paralysis. Cell therapies intended to promote remyelination of axons are a promising avenue of treatment, with mesenchymal stromal cells (MSCs) a prominent candidate. We have previously demonstrated that MSCs derived from human olfactory mucosa (hOM-MSCs) promote myelination to a greater extent than bone marrow-derived MSCs (hBM-MSCs). However, hOM-MSCs were developed using methods and materials that were not good manufacturing practice (GMP)-compliant. Before considering these cells for clinical use, it is necessary to develop a method for their isolation and expansion that is readily adaptable to a GMP-compliant environment. We demonstrate here that hOM-MSCs can be derived without enzymatic tissue digestion or cell sorting and without culture antibiotics. They grow readily in GMP-compliant media and express typical MSC surface markers. They robustly produce CXCL12 (a key secretory factor in promoting myelination) and are pro-myelinating in in vitro rodent CNS cultures. GMP-compliant hOM-MSCs are comparable in this respect to those grown in non-GMP conditions. However, when assessed in an in vivo model of demyelinating disease (experimental autoimmune encephalitis, EAE), they do not significantly improve disease scores compared with controls, indicating further pre-clinical evaluation is necessary before their advancement to clinical trials.


Subject(s)
Anti-Bacterial Agents , Mesenchymal Stem Cells , Humans , Culture Techniques , Axons , Biological Transport
2.
Glia ; 71(7): 1683-1698, 2023 07.
Article in English | MEDLINE | ID: mdl-36945189

ABSTRACT

There is an urgent need for therapies that target the multicellular pathology of central nervous system (CNS) disease. Modified, nonanticoagulant heparins mimic the heparan sulfate glycan family and are known regulators of multiple cellular processes. In vitro studies have demonstrated that low sulfated modified heparin mimetics (LS-mHeps) drive repair after CNS demyelination. Herein, we test LS-mHep7 (an in vitro lead compound) in experimental autoimmune encephalomyelitis (EAE) and cuprizone-induced demyelination. In EAE, LS-mHep7 treatment resulted in faster recovery and rapidly reduced inflammation which was accompanied by restoration of animal weight. LS-mHep7 treatment had no effect on remyelination or on OLIG2 positive oligodendrocyte numbers within the corpus callosum in the cuprizone model. Further in vitro investigation confirmed that LS-mHep7 likely mediates its pro-repair effect in the EAE model by sequestering inflammatory cytokines, such as CCL5 which are upregulated during immune-mediated inflammatory attacks. These data support the future clinical translation of this next generation modified heparin as a treatment for CNS diseases with active immune system involvement.


Subject(s)
Central Nervous System Diseases , Encephalomyelitis, Autoimmune, Experimental , Animals , Mice , Cuprizone/toxicity , Sulfates/adverse effects , Oligodendroglia/pathology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Corpus Callosum/pathology , Central Nervous System Diseases/pathology , Heparitin Sulfate/therapeutic use , Mice, Inbred C57BL , Disease Models, Animal , Myelin Sheath/pathology
3.
Biology (Basel) ; 12(3)2023 Mar 04.
Article in English | MEDLINE | ID: mdl-36979099

ABSTRACT

Therapies that target the multicellular pathology of central nervous system (CNS) disease/injury are urgently required. Modified non-anticoagulant heparins mimic the heparan sulphate (HS) glycan family and have been proposed as therapeutics for CNS repair since they are effective regulators of numerous cellular processes. Our in vitro studies have demonstrated that low-sulphated modified heparan sulphate mimetics (LS-mHeps) drive CNS repair. However, LS-mHeps are derived from pharmaceutical heparin purified from pig intestines, in a supply chain at risk of shortages and contamination. Alternatively, cellular synthesis of heparin and HS can be achieved using mammalian cell multiplex genome engineering, providing an alternative source of recombinant HS mimetics (rHS). TEGA Therapeutics (San Diego) have manufactured rHS reagents with varying degrees of sulphation and we have validated their ability to promote repair in vitro using models that mimic CNS injury, making comparisons to LS-mHep7, a previous lead compound. We have shown that like LS-mHep7, low-sulphated rHS compounds promote remyelination and reduce features of astrocytosis, and in contrast, highly sulphated rHS drive neurite outgrowth. Cellular production of heparin mimetics may, therefore, offer potential clinical benefits for CNS repair.

4.
Methods Mol Biol ; 2429: 333-344, 2022.
Article in English | MEDLINE | ID: mdl-35507171

ABSTRACT

Striatum-derived neural stem cells have been used to generate a variety of neural cell populations. They are composed of free-floating clusters of clonal neural stem cells, termed neurospheres, and can be expanded under growth factor stimulation in vitro. The multipotent nature of neurospheres means that under certain growth conditions they can differentiate into neuronal and glial progenitors of the central nervous system (CNS).Here, we describe a method for creating a population of astrocytes derived from rat striatum neurospheres, which in turn can be used to generate astrocytes with different reactivity phenotypes. Several methods and techniques are already available for the generation of neurospheres, but the method detailed herein provides an accessible, reproducible protocol for large numbers of astrocyte cultures, that can then be manipulated in an experimental format for further investigation.


Subject(s)
Astrocytes , Neural Stem Cells , Animals , Cell Differentiation/physiology , Cells, Cultured , Immunohistochemistry , Phenotype , Rats
5.
Exp Neurol ; 354: 114113, 2022 08.
Article in English | MEDLINE | ID: mdl-35569511

ABSTRACT

Iron released from oligodendrocytes during demyelination or derived from haemoglobin breakdown products is believed to amplify oxidative tissue injury in multiple sclerosis (MS). However, the pathophysiological significance of iron-containing haemoglobin breakdown products themselves is rarely considered in the context of MS and their cellular specificity and mode of action remain unclear. Using myelinating cell cultures, we now report the cytotoxic potential of hemin (ferriprotoporphyrin IX chloride), a major degradation product of haemoglobin, is 25-fold greater than equimolar concentrations of free iron in myelinating cultures; a model that reproduces the complex multicellular environment of the CNS. At low micro molar concentrations (3.3 - 10 µM) we observed hemin preferentially binds to myelin and axons to initiate a complex detrimental response that results in targeted demyelination and axonal loss but spares neuronal cell bodies, astrocytes and the majority of oligodendroglia. Demyelination and axonal loss in this context are executed by a combination of mechanisms that include iron-dependent peroxidation by reactive oxygen species (ROS) and ferroptosis. These effects are microglial-independent, do not require any initiating inflammatory insult and represent a direct effect that compromises the structural integrity of myelinated axons in the CNS. Our data identify hemin-mediated demyelination and axonal loss as a novel mechanism by which intracerebral degradation of haemoglobin may contribute to lesion development in MS.


Subject(s)
Hemin , Multiple Sclerosis , Axons/pathology , Central Nervous System/pathology , Hemin/metabolism , Hemin/pharmacology , Humans , Iron/metabolism , Multiple Sclerosis/pathology , Myelin Sheath/pathology , Oligodendroglia/metabolism , Oxidative Stress
6.
Acta Neuropathol Commun ; 10(1): 12, 2022 01 29.
Article in English | MEDLINE | ID: mdl-35093166

ABSTRACT

One of the therapeutic approaches for the treatment of the autoimmune demyelinating disease, multiple sclerosis (MS) is bone marrow mesenchymal stromal cell (hBM-MSCs) transplantation. However, given their capacity to enhance myelination in vitro, we hypothesised that human olfactory mucosa-derived MSCs (hOM-MSCs) may possess additional properties suitable for CNS repair. Herein, we have examined the efficacy of hOM-MSCs versus hBM-MSCs using the experimental autoimmune encephalomyelitis (EAE) model. Both MSC types ameliorated disease, if delivered during the initial onset of symptomatic disease. Yet, only hOM-MSCs improved disease outcome if administered during established disease when animals had severe neurological deficits. Histological analysis of spinal cord lesions revealed hOM-MSC transplantation reduced blood-brain barrier disruption and inflammatory cell recruitment and enhanced axonal survival. At early time points post-hOM-MSC treatment, animals had reduced levels of circulating IL-16, which was reflected in both the ability of immune cells to secrete IL-16 and the level of IL-16 in spinal cord inflammatory lesions. Further in vitro investigation revealed an inhibitory role for IL-16 on oligodendrocyte differentiation and myelination. Moreover, the availability of bioactive IL-16 after demyelination was reduced in the presence of hOM-MSCs. Combined, our data suggests that human hOM-MSCs may have therapeutic benefit in the treatment of MS via an IL-16-mediated pathway, especially if administered during active demyelination and inflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Interleukin-16/metabolism , Mesenchymal Stem Cell Transplantation/methods , Myelin Sheath/metabolism , Olfactory Mucosa/cytology , Animals , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Mice , Neurogenesis/physiology
7.
Cells ; 10(4)2021 04 14.
Article in English | MEDLINE | ID: mdl-33919910

ABSTRACT

The use of mesenchymal stem/stromal cells (MSCs) for transplant-mediated repair represents an important and promising therapeutic strategy after spinal cord injury (SCI). The appeal of MSCs has been fuelled by their ease of isolation, immunosuppressive properties, and low immunogenicity, alongside the large variety of available tissue sources. However, despite reported similarities in vitro, MSCs sourced from distinct tissues may not have comparable biological properties in vivo. There is accumulating evidence that stemness, plasticity, immunogenicity, and adaptability of stem cells is largely controlled by tissue niche. The extrinsic impact of cellular niche for MSC repair potential is therefore important, not least because of its impact on ex vivo expansion for therapeutic purposes. It is likely certain niche-targeted MSCs are more suited for SCI transplant-mediated repair due to their intrinsic capabilities, such as inherent neurogenic properties. In addition, the various MSC anatomical locations means that differences in harvest and culture procedures can make cross-comparison of pre-clinical data difficult. Since a clinical grade MSC product is inextricably linked with its manufacture, it is imperative that cells can be made relatively easily using appropriate materials. We discuss these issues and highlight the importance of identifying the appropriate niche-specific MSC type for SCI repair.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Nerve Regeneration/physiology , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy , Stem Cell Niche , Humans
8.
Biomater Sci ; 8(13): 3611-3627, 2020 Jul 07.
Article in English | MEDLINE | ID: mdl-32515439

ABSTRACT

The limited regenerative capacity of the CNS poses formidable challenges to the repair of spinal cord injury (SCI). Two key barriers to repair are (i) the physical gap left by the injury, and (ii) the inhibitory milieu surrounding the injury, the glial scar. Biomaterial implantation into the injury site can fill the cavity, provide a substrate for cell migration, and potentially attenuate the glial scar. We investigated the biological viability of a biocompatible and biodegradable poly-ε-lysine based biomaterial, Proliferate®, in low and high cross-linked forms and when coated with IKVAV peptide, for SCI implantation. We demonstrate altered astrocyte morphology and nestin expression on Proliferate® compared to conventional glass cell coverslips suggesting a less reactive phenotype. Moreover Proliferate® supported myelination in vitro, with myelination observed sooner on IKVAV-coated constructs compared with uncoated Proliferate®, and delayed overall compared with maintenance on glass coverslips. For in vivo implantation, parallel-aligned channels were fabricated into Proliferate® to provide cell guidance cues. Extensive vascularisation and cellular infiltration were observed in constructs implanted in vivo, along with an astrocyte border and microglial response. Axonal ingrowth was observed at the construct border and inside implants in intact channels. We conclude that Proliferate® is a promising biomaterial for implantation following SCI.


Subject(s)
Biocompatible Materials/chemistry , Central Nervous System Diseases/therapy , Polylysine/chemistry , Prostheses and Implants , Spinal Cord Injuries/therapy , Animals , Biocompatible Materials/chemical synthesis , Cells, Cultured , Polylysine/chemical synthesis , Rats , Rats, Sprague-Dawley
9.
Nat Rev Neurol ; 16(4): 229-240, 2020 04.
Article in English | MEDLINE | ID: mdl-32099190

ABSTRACT

Spinal cord injury (SCI) remains one of the biggest challenges in the development of neuroregenerative therapeutics. Cell transplantation is one of numerous experimental strategies that have been identified and tested for efficacy at both preclinical and clinical levels in recent years. In this Review, we briefly discuss the state of human olfactory cell transplantation as a therapy, considering both its current clinical status and its limitations. Furthermore, we introduce a mesenchymal stromal cell derived from human olfactory tissue, which has the potential to induce multifaceted reparative effects in the environment within and surrounding the lesion. We argue that no single therapy will be sufficient to treat SCI effectively and that a combination of cell-based, rehabilitation and pharmaceutical interventions is the most promising approach to aid repair. For this reason, we also introduce a novel pharmaceutical strategy based on modifying the activity of heparan sulfate, an important regulator of a wide range of biological cell functions. The multi-target approach that is exemplified by these types of strategies will probably be necessary to optimize SCI treatment.


Subject(s)
Heparitin Sulfate/therapeutic use , Mesenchymal Stem Cell Transplantation/methods , Olfactory Mucosa/cytology , Spinal Cord Injuries/therapy , Spinal Cord Regeneration , Cell Transplantation/methods , Chondroitin Sulfate Proteoglycans/metabolism , Heparan Sulfate Proteoglycans/metabolism , Heparin/therapeutic use , Heparitin Sulfate/analogs & derivatives , Humans , Mesenchymal Stem Cells/cytology , Nerve Regeneration , Neuroglia , Olfactory Mucosa/physiology , Olfactory Receptor Neurons
10.
Biology (Basel) ; 8(3)2019 Jun 28.
Article in English | MEDLINE | ID: mdl-31261710

ABSTRACT

In vitro cell-based assays have been fundamental in modern drug discovery and have led to the identification of novel therapeutics. We have developed complex mixed central nervous system (CNS) cultures, which recapitulate the normal process of myelination over time and allow the study of several parameters associated with CNS damage, both during development and after injury or disease. In particular, they have been used as a reliable screen to identify drug candidates that may promote (re)myelination and/or neurite outgrowth. Previously, using these cultures, we demonstrated that a panel of low sulphated heparin mimetics, with structures similar to heparan sulphates (HSs), can reduce astrogliosis, and promote myelination and neurite outgrowth. HSs reside in either the extracellular matrix or on the surface of cells and are thought to modulate cell signaling by both sequestering ligands, and acting as co-factors in the formation of ligand-receptor complexes. In this study, we have used these cultures as a screen to address the repair potential of numerous other commercially available sulphated glycomolecules, namely heparosans, ulvans, and fucoidans. These compounds are all known to have certain characteristics that mimic cellular glycosaminoglycans, similar to heparin mimetics. We show that the N-sulphated heparosans promoted myelination. However, O-sulphated heparosans did not affect myelination but promoted neurite outgrowth, indicating the importance of structure in HS function. Moreover, neither highly sulphated ulvans nor fucoidans had any effect on remyelination but CX-01, a low sulphated porcine intestinal heparin, promoted remyelination in vitro. These data illustrate the use of myelinating cultures as a screen and demonstrate the potential of heparin mimetics as CNS therapeutics.

11.
Glia ; 65(4): 639-656, 2017 04.
Article in English | MEDLINE | ID: mdl-28144983

ABSTRACT

Autologous cell transplantation is a promising strategy for repair of the injured spinal cord. Here we have studied the repair potential of mesenchymal stromal cells isolated from the human olfactory mucosa after transplantation into a rodent model of incomplete spinal cord injury. Investigation of peripheral type remyelination at the injury site using immunocytochemistry for P0, showed a more extensive distribution in transplanted compared with control animals. In addition to the typical distribution in the dorsal columns (common to all animals), in transplanted animals only, P0 immunolabelling was consistently detected in white matter lateral and ventral to the injury site. Transplanted animals also showed reduced cavitation. Several functional outcome measures including end-point electrophysiological testing of dorsal column conduction and weekly behavioural testing of BBB, weight bearing and pain, showed no difference between transplanted and control animals. However, gait analysis revealed an earlier recovery of co-ordination between forelimb and hindlimb stepping in transplanted animals. This improvement in gait may be associated with the enhanced myelination in ventral and lateral white matter, where fibre tracts important for locomotion reside. Autologous transplantation of mesenchymal stromal cells from the olfactory mucosa may therefore be therapeutically beneficial in the treatment of spinal cord injury. GLIA 2017 GLIA 2017;65:639-656.


Subject(s)
Gait Disorders, Neurologic/etiology , Gait Disorders, Neurologic/surgery , Mesenchymal Stem Cell Transplantation/methods , Olfactory Mucosa/cytology , Remyelination/physiology , Spinal Cord Injuries/complications , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Disease Models, Animal , Electroencephalography , Evoked Potentials, Somatosensory/physiology , Exploratory Behavior/physiology , Humans , Locomotion/physiology , Male , Myelin P0 Protein/metabolism , Nerve Tissue Proteins/metabolism , Pain Measurement , Rats , Rats, Sprague-Dawley , Weight-Bearing
12.
Neurochem Int ; 106: 101-107, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27498150

ABSTRACT

In recent years there has been a great deal of research within the stem cell field which has led to the definition and classification of a range of stem cells from a plethora of tissues and organs. Stem cells, by classification, are considered to be pluri- or multipotent and have both self-renewal and multi-differentiation capabilities. Presently there is a great deal of interest in stem cells isolated from both embryonic and adult tissues in the hope they hold the therapeutic key to restoring or treating damaged cells in a number of central nervous system (CNS) disorders. In this review we will discuss the role of mesenchymal stromal cells (MSCs) isolated from human olfactory mucosa, with particular emphasis on their potential role as a candidate for transplant mediated repair in the CNS. Since nestin expression defines the entire population of olfactory mucosal derived MSCs, we will compare these cells to a population of neural crest derived nestin positive population of bone marrow-MSCs.


Subject(s)
Central Nervous System Diseases/metabolism , Central Nervous System Diseases/therapy , Mesenchymal Stem Cell Transplantation/trends , Mesenchymal Stem Cells/metabolism , Nestin/biosynthesis , Animals , Cell Differentiation/physiology , Central Nervous System Diseases/genetics , Humans , Nestin/genetics , Olfactory Mucosa/metabolism
13.
Glia ; 65(1): 19-33, 2017 01.
Article in English | MEDLINE | ID: mdl-27535874

ABSTRACT

Schwann cell (SC) transplantation following spinal cord injury (SCI) may have therapeutic potential. Functional recovery is limited however, due to poor SC interactions with host astrocytes and the induction of astrogliosis. Olfactory ensheathing cells (OECs) are closely related to SCs, but intermix more readily with astrocytes in culture and induce less astrogliosis. We previously demonstrated that OECs express higher levels of sulfatases, enzymes that remove 6-O-sulfate groups from heparan sulphate proteoglycans, than SCs and that RNAi knockdown of sulfatase prevented OEC-astrocyte mixing in vitro. As human OECs are difficult to culture in large numbers we have genetically engineered SCs using lentiviral vectors to express sulfatase 1 and 2 (SC-S1S2) and assessed their ability to interact with astrocytes. We demonstrate that SC-S1S2s have increased integrin-dependent motility in the presence of astrocytes via modulation of NRG and FGF receptor-linked PI3K/AKT intracellular signaling and do not form boundaries with astrocytes in culture. SC-astrocyte mixing is dependent on local NRG concentration and we propose that sulfatase enzymes influence the bioavailability of NRG ligand and thus influence SC behavior. We further demonstrate that injection of sulfatase expressing SCs into spinal cord white matter results in less glial reactivity than control SC injections comparable to that of OEC injections. Our data indicate that sulfatase-mediated modification of the extracellular matrix can influence glial interactions with astrocytes, and that SCs engineered to express sulfatase may be more OEC-like in character. This approach may be beneficial for cell transplant-mediated spinal cord repair. GLIA 2016 GLIA 2017;65:19-33.


Subject(s)
Astrocytes/cytology , Astrocytes/enzymology , Cell Movement/physiology , Nerve Regeneration/physiology , Schwann Cells/cytology , Schwann Cells/enzymology , Sulfatases/metabolism , Animals , Cells, Cultured , Neuroglia/cytology , Rats, Sprague-Dawley , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy
14.
Exp Neurol ; 283(Pt B): 541-9, 2016 09.
Article in English | MEDLINE | ID: mdl-26988764

ABSTRACT

Astrocytes are the major glial cell of the central nervous system (CNS), providing both metabolic and physical support to other neural cells. After injury, astrocytes become reactive and express a continuum of phenotypes which may be supportive or inhibitory to CNS repair. This review will focus on the ability of astrocytes to influence myelination in the context of specific secreted factors, cytokines and other neural cell targets within the CNS. In particular, we focus on how astrocytes provide energy and cholesterol to neurons, influence synaptogenesis, affect oligodendrocyte biology and instigate cross-talk between the many cellular components of the CNS.


Subject(s)
Astrocytes/physiology , Myelin Sheath/physiology , Animals , Astrocytes/chemistry , Cell Differentiation , Central Nervous System , Cytokines/metabolism , Humans , Neurons/physiology
15.
Nat Cell Biol ; 17(12): 1556-68, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26595383

ABSTRACT

L-Glutamine (Gln) functions physiologically to balance the carbon and nitrogen requirements of tissues. It has been proposed that in cancer cells undergoing aerobic glycolysis, accelerated anabolism is sustained by Gln-derived carbons, which replenish the tricarboxylic acid (TCA) cycle (anaplerosis). However, it is shown here that in glioblastoma (GBM) cells, almost half of the Gln-derived glutamate (Glu) is secreted and does not enter the TCA cycle, and that inhibiting glutaminolysis does not affect cell proliferation. Moreover, Gln-starved cells are not rescued by TCA cycle replenishment. Instead, the conversion of Glu to Gln by glutamine synthetase (GS; cataplerosis) confers Gln prototrophy, and fuels de novo purine biosynthesis. In both orthotopic GBM models and in patients, (13)C-glucose tracing showed that GS produces Gln from TCA-cycle-derived carbons. Finally, the Gln required for the growth of GBM tumours is contributed only marginally by the circulation, and is mainly either autonomously synthesized by GS-positive glioma cells, or supplied by astrocytes.


Subject(s)
Brain Neoplasms/metabolism , Cell Proliferation , Glioblastoma/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamine/metabolism , Nucleotides/biosynthesis , Animals , Astrocytes/cytology , Astrocytes/metabolism , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Citric Acid Cycle , Coculture Techniques , Female , Glioblastoma/genetics , Glioblastoma/pathology , Glutamate-Ammonia Ligase/genetics , Glutamic Acid/metabolism , Humans , Male , Mice, Inbred NOD , Mice, SCID , Models, Biological , Neoplastic Stem Cells/metabolism , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
16.
Glia ; 61(3): 368-82, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23281012

ABSTRACT

Spinal cord injury (SCI) is a devastating condition with limited capacity for repair. Cell transplantation is a potential strategy to promote SCI repair with cells from the olfactory system being promising candidates. Although transplants of human olfactory mucosa (OM) are already ongoing in clinical trials, the repair potential of this tissue remains unclear. Previously, we identified mesenchymal-like stem cells that reside in the lamina propria (LP-MSCs) of rat and human OM. Little is known about these cells or their interactions with glia such as olfactory ensheathing cells (OECs), which would be co-transplanted with MSCs from the OM, or endogenous CNS glia such as oligodendrocytes. We have characterized, purified, and assessed the repair potential of human LP-MSCs by investigating their effect on glial cell biology with specific emphasis on CNS myelination in vitro. Purified LP-MSCs expressed typical bone marrow MSC (BM-MSC) markers, formed spheres, were clonogenic and differentiated into bone and fat. LP-MSC conditioned medium (CM) promoted oligodendrocyte precursor cell (OPC) and OEC proliferation and induced a highly branched morphology. LP-MSC-CM treatment caused OEC process extension. Both LP and BM-MSCs promoted OPC proliferation and differentiation, but only myelinating cultures treated with CM from LP and not BM-MSCs had a significant increase in myelination. Comparison with fibroblasts and contaminating OM fibroblast like-cells showed the promyelination effect was LP-MSC specific. Thus LP-MSCs harvested from human OM biopsies may be an important candidate for cell transplantation by contributing to the repair of SCI.


Subject(s)
Bone and Bones/cytology , Mesenchymal Stem Cells/cytology , Myelin Sheath/pathology , Neuroglia/cytology , Olfactory Mucosa/cytology , Spinal Cord Injuries/pathology , Adolescent , Adult , Aged , Animals , Bone Transplantation , Cell Movement , Cell Proliferation , Female , Humans , Male , Mesenchymal Stem Cell Transplantation , Middle Aged , Neuroglia/transplantation , Olfactory Mucosa/transplantation , Rats , Wound Healing
17.
J Neurosci Res ; 90(3): 619-31, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22057888

ABSTRACT

Olfactory mucosa is a source of cells for transplant-mediated repair of spinal cord injury (SCI) and is currently being assessed in clinical trials. We previously reported that olfactory mucosa can generate two types of sphere-forming cells with stem cell-like properties. Here we have assessed the repair potential of these cells in a rodent SCI model. Sphere-forming cells transplanted into a dorsal column injury integrated with the host spinal cord, filling the injury cavity, but showed no evidence of differentiation in vivo. Moreover, transplants supported robust axonal regeneration, particularly when suspensions of smaller spheres, rather than large aggregates, were transplanted. However, tract-tracing of dorsal column fibers showed that regenerating axons did not extend beyond the transplant. These observations show that undifferentiated olfactory spheres, though capable of supporting axonal regeneration, do not show any advantage over olfactory ensheathing cells isolated from adult olfactory tissue. In addition, olfactory spheres induced a greater astrocytic hypertrophy at the injury site than previously observed for purified olfactory ensheathing cells.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , Olfactory Mucosa/transplantation , Spinal Cord Injuries/surgery , Spinal Cord/physiology , Animals , Cell Differentiation/physiology , Male , Olfactory Mucosa/cytology , Rats , Rats, Inbred F344
18.
Glia ; 58(2): 125-34, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19606497

ABSTRACT

Damage to the brain and spinal cord leads to permanent functional disability because of the very limited capacity of the central nervous system (CNS) for repair. Transplantation of cells into regions of CNS damage represents one approach to enhancing this repair. At present, the ideal cell type for transplant-mediated repair has not been identified but autologous transplantation would be advantageous. Olfactory tissue, in part because of its capacity for regeneration, has emerged as a promising source of cells and several clinical centers are using olfactory cells or tissues in the treatment of CNS damage. Until now, the olfactory ensheathing cell, a specialized glial cell of the olfactory system has been the main focus of attention. Transplants of this cell have been shown to have a neuroprotective function, support axonal regeneration, and remyelinate demyelinated axons. However, the olfactory mucosa is a heterogeneous tissue, composed of a variety of cells supporting both its normal function and its regenerative capacity. It is therefore possible that it contains several cell types that could participate in CNS repair including putative stem cells as well as glia. Here we review the cellular composition of the olfactory tissue and the evidence that equivalent cell types exist in both rodent and human olfactory mucosa suggesting that it is potentially a rich source of autologous cells for transplant-mediated repair of the CNS.


Subject(s)
Brain Injuries/surgery , Olfactory Mucosa/transplantation , Spinal Cord Injuries/surgery , Animals , Brain Injuries/physiopathology , Humans , Nerve Regeneration/physiology , Olfactory Mucosa/cytology , Olfactory Mucosa/physiology , Olfactory Pathways/cytology , Olfactory Pathways/physiology , Olfactory Pathways/transplantation , Spinal Cord Injuries/physiopathology
19.
Stem Cells ; 27(9): 2196-208, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19544421

ABSTRACT

Olfactory mucosal (OM) tissue, a potential source of stem cells, is currently being assessed in the clinic as a candidate tissue for transplant-mediated repair of spinal cord injury. We examined the ability of embryonic rat OM tissue to generate stem cells using culture conditions known to promote neural stem cell proliferation. Primary spheres formed that proliferated and exhibited two main morphologies: (a) CNS neurosphere-like (OM-I) and (b) small, tight spheroid-like (OM-II). The OM-I spheres expressed the neural stem cell marker nestin but also markers of peripheral glia, neurons, and connective tissue. Further studies demonstrated the presence of multipotential mesenchymal-like stem cells within OM-I spheres that differentiated into bone, adipose, and smooth muscle cells. In contrast, the OM-II spheres contained mainly cytokeratin-expressing cells. Immunolabeling of rat olfactory tissue with Stro-1, CD90, and CD105 showed the presence of multipotent mesenchymal cells in the lamina propria, whereas cytokeratin was expressed by the epithelial cells of the olfactory epithelium. In addition, a comparable pattern of immunoreactivity was detected in human tissue using Stro-1 and cytokeratin, suggesting the presence of similar cells in this tissue. The identification of a nonepithelial multipotent cell in the OM may explain the varied reports on olfactory stem cell differentiation capacity in vitro and in vivo and illustrates the cellular complexity of this tissue as a potential source of stem cells for transplantation and translation to the clinic.


Subject(s)
Epithelial Cells/cytology , Multipotent Stem Cells/cytology , Olfactory Mucosa/cytology , Adipogenesis/physiology , Animals , Blotting, Western , Cell Differentiation/physiology , Embryo, Mammalian/cytology , Female , Flow Cytometry , Humans , Immunohistochemistry , In Vitro Techniques , Myocytes, Smooth Muscle/cytology , Neurons/cytology , Osteogenesis/physiology , Pregnancy , Rats , Rats, Sprague-Dawley
20.
J Cell Sci ; 120(Pt 17): 3011-21, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17684063

ABSTRACT

The initial step in directed cell movement is lamellipodial protrusion, an action driven by actin polymerization. Enabled/vasodilator-stimulated phosphoprotein (Ena/VASP) family proteins are key regulators of this actin polymerization and can control lamellipodial protrusion rate. Ena/VASP proteins are substrates for modification by cyclic-nucleotide-dependent protein kinases at a number of sites. Phosphorylation of Ser239 of VASP in vitro inhibits its anti-capping and filament-bundling activity but the effects of this modification on lamellipodial structure and function are unknown. To examine the functional effects of this modification in living cells, we studied VASP phosphorylation at Ser239 by nitric oxide (NO) stimulation of cGMP-dependent protein kinase. Using live cell imaging of primary cells transfected with GFP-VASP constructs, we found that NO produced rapid retraction of lamellipodia together with cell rounding that was dependent on guanylate cyclase and type II cGMP-dependent protein kinase. In cells expressing a mutant VASP (Ser239Ala) lacking the site preferentially phosphorylated by this kinase, NO had no effect. Phosphorylation of Ser239 of VASP results in loss of lamellipodial protrusions and cell rounding, and is a powerful means of controlling directed actin polymerization within lamellipodia.


Subject(s)
Cell Adhesion Molecules/metabolism , Microfilament Proteins/metabolism , Nitric Oxide/metabolism , Phosphoproteins/metabolism , Pseudopodia , Serine/metabolism , Actins/metabolism , Cell Adhesion Molecules/genetics , Cell Shape , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Cytoskeleton/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Guanylate Cyclase/metabolism , Humans , Kidney/cytology , Microfilament Proteins/genetics , Phosphoproteins/genetics , Phosphorylation , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL