125 results on '"Martin Grumet"'
Search Results
2. Anti-Inflammatory Effects of Encapsulated Human Mesenchymal Stromal/Stem Cells and a Method to Scale-Up Cell Encapsulation
- Author
-
Suneel Kumar, Maciej Kabat, Sayantani Basak, Joanne Babiarz, Francois Berthiaume, and Martin Grumet
- Subjects
mesenchymal stem cells ,spinal cord injury ,encapsulation ,alginate ,human IL10 ,Microbiology ,QR1-502 - Abstract
Mesenchymal stem/stromal cells (MSC) promote recovery in a wide range of animal models of injury and disease. They can act in vivo by differentiating and integrating into tissues, secreting factors that promote cell growth and control inflammation, and interacting directly with host effector cells. We focus here on MSC secreted factors by encapsulating the cells in alginate microspheres, which restrict cells from migrating out while allowing diffusion of factors including cytokines across the capsules. One week after intrathecal lumbar injection of human bone marrow MSC encapsulated in alginate (eMSC), rat IL-10 expression was upregulated in distant rat spinal cord injury sites. Detection of human IL-10 protein in rostrally derived cerebrospinal fluid (CSF) indicated distribution of this human MSC-secreted cytokine throughout rat spinal cord CSF. Intraperitoneal (IP) injection of eMSC in a rat model for endotoxemia reduced serum levels of inflammatory cytokines within 5 h. Detection of human IL-6 in sera after injection of human eMSC indicates rapid systemic distribution of this human MSC-secreted cytokine. Despite proof of concept for eMSC in various disorders using animal models, translation of encapsulation technology has not been feasible primarily because methods for scale-up are not available. To scale-up production of eMSC, we developed a rapid, semi-continuous, capsule collection system coupled to an electrosprayer. This system can produce doses of encapsulated cells sufficient for use in clinical translation.
- Published
- 2022
- Full Text
- View/download PDF
3. Trends in mesenchymal stem cell clinical trials 2004‐2018: Is efficacy optimal in a narrow dose range?
- Author
-
Maciej Kabat, Ivan Bobkov, Suneel Kumar, and Martin Grumet
- Subjects
ClinicalTrials.gov ,dose ,intravenous ,mesenchymal stromal cell ,Medicine (General) ,R5-920 ,Cytology ,QH573-671 - Abstract
Abstract The number of clinical trials using mesenchymal stem cells (MSCs) has increased since 2008, but this trend slowed in the past several years and dropped precipitously in 2018. Previous reports have analyzed MSC clinical trials by disease, phase, cell source, country of origin, and trial initiation date, all of which can be downloaded directly from ClinicalTrials.gov. We have extended analyses to a larger group of 914 MSC trials reported through 2018. To search for potential factors that may influence the design of new trials, we extracted data on routes of administration and dosing from individual ClinicalTrials.gov records as this information cannot be downloaded directly from the database. Intravenous (IV) injection is the most common, least invasive and most reproducible method, accounting for 43% of all trials. The median dose for IV delivery is 100 million MSCs/patient/dose. Analysis of all trials using IV injection that reported positive outcomes indicated minimal effective doses (MEDs) ranging from 70 to 190 million MSCs/patient/dose in 14/16 trials with the other two trials administering much higher doses of at least 900 million cells. Dose‐response data showing differential efficacy for improved outcomes were reported in only four trials, which indicated a narrower MED range of 100‐150 million MSCs/patient with lower and higher IV doses being less effective. The results suggest that it may be critical to determine MEDs in early trials before proceeding with large clinical trials.
- Published
- 2020
- Full Text
- View/download PDF
4. System for Cleavable Fc Fusion Proteins Using Tobacco Etch Virus (TEV) Protease
- Author
-
Jeffrey Haspel, Cy Blanco, Jeffrey Jacob, and Martin Grumet
- Subjects
Biology (General) ,QH301-705.5 - Abstract
We describe a novel Fc fusion protein system that can be cleaved by tobacco etch virus (TEV) protease. This system is desirable because it takes advantage of the high specificity of TEV protease and its activity at 4°C. We produced two TEV-Fc fusion proteins that contain the first three Ig domains and all six Ig domains of the cell adhesion molecule L1. Both proteins were efficiently cleaved by TEV protease at 4°C. Functional analysis of the cleavage products in neurite outgrowth assays showed they had similar activities to their parental Fc fusion proteins. Therefore, TEV-Fc fusion proteins may increase the utility and flexibility of the Fc fusion protein system.
- Published
- 2001
- Full Text
- View/download PDF
5. Purification of Ig-Fusion Proteins from Medium Containing Ig
- Author
-
Takeshi Sakurai, Chanland Roonprapunt, and Martin Grumet
- Subjects
Biology (General) ,QH301-705.5 - Published
- 1998
- Full Text
- View/download PDF
6. Efficacy of MSC in Patients with Severe COVID-19: Analysis of the Literature and a Case Study
- Author
-
Martin Grumet, Jason Sherman, and Barry S Dorf
- Subjects
Male ,Aged, 80 and over ,Respiratory Distress Syndrome ,SARS-CoV-2 ,Humans ,COVID-19 ,Cytokines ,Mesenchymal Stem Cells ,Cell Biology ,General Medicine ,Cytokine Release Syndrome ,Mesenchymal Stem Cell Transplantation ,Developmental Biology - Abstract
Patients with severe COVID-19 experience cytokine storm, an uncontrolled upregulation of pro-inflammatory cytokines, which if unresolved leads to acute respiratory distress syndrome (ARDS), organ damage, and death. Treatments with mesenchymal stromal cells (MSC) [Viswanathan S, Shi Y, Galipeau J, et al. Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy Mesenchymal Stromal Cell committee position statement on nomenclature. Cytotherapy. 2019;21:1019-1024] appear to be effective in reducing morbidity and mortality. MSC respond to pro-inflammatory cytokines by releasing anti-inflammatory factors and mobilizing immune cells. We analyzed 82 COVID-19 clinical trials registered at ClinicalTrials.gov to determine MSC dosing, routes of administration, and outcome measures. Nearly all trials described the use of intravenous delivery with most doses ranging between 50 and 125 million MSC/treatment, which overlaps with a minimal effective dose range that we described previously. We also searched the literature to analyze clinical trial reports that used MSC to treat COVID-19. MSC were found to improve survival and oxygenation, increase discharge from intensive care units and hospitals, and reduce levels of pro-inflammatory markers. We report on a 91-year-old man with severe COVID-19 who responded rapidly to MSC treatment with transient reductions in several pro-inflammatory markers and delayed improvement in oxygenation. The results suggest that frequent monitoring of pro-inflammatory markers for severe COVID-19 will provide improved treatment guidelines by determining relationships between cytokine storms and ARDS. We propose that markers for cytokine storm are leading indicators for ARDS and that measurement of cytokines will indicate earlier treatment with MSC than is performed now for ARDS in severe COVID-19.
- Published
- 2022
- Full Text
- View/download PDF
7. Microfibrous substrate geometry as a critical trigger for organization, self‐renewal, and differentiation of human embryonic stem cells within synthetic 3‐dimensional microenvironments
- Author
-
Aaron L. Carlson, Joseph J. Kim, Thomas Neubauer, Rick I. Cohen, Joachim Kohn, Jennifer C. Moore, Prabhas V. Moghe, Charles A. Florek, and Martin Grumet
- Subjects
Cellular differentiation ,Cell Culture Techniques ,Nanotechnology ,Biochemistry ,Research Communications ,Extracellular matrix ,Biopolymers ,Directed differentiation ,Tissue engineering ,Cell Adhesion ,Genetics ,Humans ,Polylysine ,Cell adhesion ,Molecular Biology ,Embryonic Stem Cells ,Cell Proliferation ,Matrigel ,Tissue Scaffolds ,Chemistry ,Cell Differentiation ,Stereoisomerism ,Embryonic stem cell ,Cell biology ,Transplantation ,Drug Combinations ,Tyrosine ,Proteoglycans ,Collagen ,Laminin ,Biotechnology - Abstract
Substrates used to culture human embryonic stem cells (hESCs) are typically 2-dimensional (2-D) in nature, with limited ability to recapitulate in vivo-like 3-dimensional (3-D) microenvironments. We examined critical determinants of hESC self-renewal in poly-d-lysine-pretreated synthetic polymer-based substrates with variable microgeometries, including planar 2-D films, macroporous 3-D sponges, and microfibrous 3-D fiber mats. Completely synthetic 2-D substrates and 3-D macroporous scaffolds failed to retain hESCs or support self-renewal or differentiation. However, synthetic microfibrous geometries made from electrospun polymer fibers were found to promote cell adhesion, viability, proliferation, self-renewal, and directed differentiation of hESCs in the absence of any exogenous matrix proteins. Mechanistic studies of hESC adhesion within microfibrous scaffolds indicated that enhanced cell confinement in such geometries increased cell-cell contacts and altered colony organization. Moreover, the microfibrous scaffolds also induced hESCs to deposit and organize extracellular matrix proteins like laminin such that the distribution of laminin was more closely associated with the cells than the Matrigel treatment, where the laminin remained associated with the coated fibers. The production of and binding to laminin was critical for formation of viable hESC colonies on synthetic fibrous scaffolds. Thus, synthetic substrates with specific 3-D microgeometries can support hESC colony formation, self-renewal, and directed differentiation to multiple lineages while obviating the stringent needs for complex, exogenous matrices. Similar scaffolds could serve as tools for developmental biology studies in 3-D and for stem cell differentiation in situ and transplantation using defined humanized conditions.—Carlson, A. L., Florek, C. A., Kim, J. J., Neubauer, T., Moore, J. C., Cohen, R. I., Kohn, J., Grumet, M., Moghe, P. V. Microfibrous substrate geometry as a critical trigger for organization, self-renewal, and differentiation of human embryonic stem cells within synthetic 3-dimensional microenvironments.
- Published
- 2012
- Full Text
- View/download PDF
8. E-Cadherin-Expressing Feeder Cells Promote Neural Lineage Restriction of Human Embryonic Stem Cells
- Author
-
Jocie F. Cherry, Martin Grumet, Rebecca N. Moore, Prabhas V. Moghe, Vani Mathur, and Rick I. Cohen
- Subjects
Cellular differentiation ,Cell Culture Techniques ,Gene Expression ,Nerve Tissue Proteins ,Biology ,Mice ,Neurosphere ,Animals ,Humans ,Cell Lineage ,Progenitor cell ,Cell Shape ,Embryonic Stem Cells ,Neurons ,Feeder Cells ,Membrane Proteins ,Cell Differentiation ,Cell Biology ,Hematology ,Fibroblasts ,Cadherins ,Antigens, Differentiation ,Molecular biology ,Coculture Techniques ,Neural stem cell ,Cell biology ,Neuroepithelial cell ,Cytoskeletal Proteins ,embryonic structures ,Neural cell adhesion molecule ,Stem cell ,Developmental Biology ,Adult stem cell - Abstract
Human embryonic stem cells (hESCs) represent a promising source of tissues of different cell lineages because of their high degree of self-renewal and their unique ability to give rise to most somatic cell lineages. In this article, we report on a new approach to differentiate hESCs into neural stem cells that can be differentiated further into neuronal restricted cells. We have rapidly and efficiently differentiated hESCs into neural stem cells by presenting the cell adhesion molecule, E-cadherin, to undifferentiated hESCs via E-cadherin transfected fibroblast monolayers. The neural restricted progenitor cells rapidly express nestin and beta-III-tubulin, but not glial fibrillary acidic protein (GFAP) during the 1-week E-cadherin induction phase, suggesting that E-cadherin promotes rapid neuronal differentiation. Further, these cells are able to achieve enhanced neuronal differentiation with the addition of exogenous growth factors. Cadherin-induced hESCs show a loss in Oct4 and nestin expression associated with positive staining for vimentin, neurofilament, and neural cell adhesion molecule. Moreover, blocking by functional E-cadherin antibody and failure of paracrine stimulation suggested that direct E-cadherin engagement is necessary to induce neural restriction. By providing hESCs with molecular cues to promote differentiation, we are able to utilize a specific cell-cell adhesion molecule, E-cadherin, to influence the nature and degree of neural specialization.
- Published
- 2012
- Full Text
- View/download PDF
9. Microscale plasma-initiated patterning of electrospun polymer scaffolds
- Author
-
Christopher L. Ricupero, Jeremy Griffin, Martin Grumet, Kathryn E. Uhrich, Sally Meiners, and Roberto Delgado-Rivera
- Subjects
Materials science ,Polymers ,Surface Properties ,Nanotechnology ,Substrate (printing) ,Article ,Plasma ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,Cell Adhesion ,Animals ,Dimethylpolysiloxanes ,Physical and Theoretical Chemistry ,Microscale chemistry ,chemistry.chemical_classification ,Polydimethylsiloxane ,Biomolecule ,Surfaces and Interfaces ,General Medicine ,Polymer ,Rats ,chemistry ,Nanofiber ,Polyamide ,Microscopy, Electron, Scanning ,Laminin ,Biotechnology ,Micropatterning - Abstract
Microscale plasma-initiated patterning (μPIP) is a novel micropatterning technique used to create biomolecular micropatterns on polymer surfaces. The patterning method uses a polydimethylsiloxane (PDMS) stamp to selectively protect regions of an underlying substrate from oxygen plasma treatment resulting in hydrophobic and hydrophilic regions. Preferential adsorption of the biomolecules onto either the plasma-exposed (hydrophilic) or plasma-protected (hydrophobic) regions leads to the biomolecular micropatterns. In the current work, laminin-1 was applied to an electrospun polyamide nanofibrillar matrix following plasma treatment. Radial glial clones (neural precursors) selectively adhered to these patterned matrices following the contours of proteins on the surface. This work demonstrates that textured surfaces, such as nanofibrillar scaffolds, can be micropatterned to provide external chemical cues for cellular organization.
- Published
- 2011
- Full Text
- View/download PDF
10. Delayed Intrathecal Delivery of RhoA siRNA to the Contused Spinal Cord Inhibits Allodynia, Preserves White Matter, and Increases Serotonergic Fiber Growth
- Author
-
Hagit Ashush, Jennie Ayer, Ron Lahav, Elena Feinstein, Kurt M. Gibbs, Hagar Kalinski, Martin Grumet, Crista Adamson, Venkatachalam Sankar, Seiji Otsuka, Evgenia Alpert, Noriko Kane-Goldsmith, and Joanne Babiarz
- Subjects
Serotonin ,RHOA ,Pharmacology ,Serotonergic ,Rats, Sprague-Dawley ,Animals ,Medicine ,RNA, Small Interfering ,Spinal cord injury ,Injections, Spinal ,Spinal Cord Injuries ,biology ,business.industry ,Genetic Therapy ,medicine.disease ,Spinal cord ,Actin cytoskeleton ,Nerve Regeneration ,Rats ,Up-Regulation ,Disease Models, Animal ,Allodynia ,medicine.anatomical_structure ,Hyperalgesia ,Anesthesia ,Corticospinal tract ,biology.protein ,Female ,Neurology (clinical) ,medicine.symptom ,rhoA GTP-Binding Protein ,business - Abstract
RhoA is a key regulator of the actin cytoskeleton that is upregulated after spinal cord injury (SCI). We analyzed different methods for siRNA delivery and developed siRNAs targeting RhoA (siRhoA) for SCI treatment. Cy 3.5-labeled siRNA delivered at the time of SCI yielded fluorescence in several cell types in the injury site. Intraspinal injections of chemically stabilized siRhoA into the spinal cord of injured rats reduced RhoA protein levels after 1 week and improved hindlimb walking over 6 weeks. To explore a less invasive route, we tested intrathecal injection of Cy 3.5-labeled siRNA via lumbar puncture 1 day after SCI, which resulted in robust uptake in the T9-T10 injury site. Lumbar injection of siRhoA 1 day after SCI reduced RhoA mRNA and protein levels 3 days after injection. Although siRhoA treatment did not yield significant improvement in locomotion, it decreased tactile hypersensitivity significantly compared to controls. Histological analysis at 8 weeks showed significant improvement in white matter sparing with siRhoA compared to control siRNA. siRhoA treatment also resulted in less accumulation of ED1+macrophages, increased PKC-γ immunoreactivity in the corticospinal tract rostral to the injury site, and increased serotonergic fiber growth 12 mm caudal to the contusion site. The ability of siRhoA to preserve white matter and promote serotonergic axonal regrowth caudal to the injury site is likely to suppress allodynia. This provides justification for considering clinical development of RhoA inhibitors to treat SCI sub-acutely to reduce allodynia, which occurs frequently in SCI patients.
- Published
- 2011
- Full Text
- View/download PDF
11. F3/contactin and TAG1 play antagonistic roles in the regulation of sonic hedgehog-induced cerebellar granule neuron progenitor proliferation
- Author
-
Andrew J.W. Furley, Takeshi Sakurai, Dia Xenaki, Martin Grumet, Kyoji Ohyama, Gianfranco Gennarini, Indira B. Martin, and Lynn Yoshida
- Subjects
Contactin 1 ,animal structures ,Cellular differentiation ,Morphogenesis ,Biology ,Mice ,Paracrine signalling ,Cerebellum ,Contactin 2 ,Animals ,Hedgehog Proteins ,Progenitor cell ,Sonic hedgehog ,Molecular Biology ,Cells, Cultured ,Cell Proliferation ,Neurons ,Cell adhesion molecule ,Cell Differentiation ,Anatomy ,Mice, Mutant Strains ,Cell biology ,embryonic structures ,biology.protein ,Signal transduction ,Signal Transduction ,Research Article ,Developmental Biology - Abstract
Modulation of the sonic hedgehog (SHH) pathway is a crucial factor in cerebellar morphogenesis. Stimulation of granule neuron progenitor (GNP) proliferation is a central function of SHH signalling, but how this is controlled locally is not understood. We show that two sequentially expressed members of the contactin (CNTN) family of adhesion molecules, TAG1 and F3, act antagonistically to control SHH-induced proliferation: F3 suppresses SHH-induced GNP proliferation and induces differentiation, whereas TAG1 antagonises F3. Production of GNPs in TAG1-null mice is delayed and reduced. F3 and TAG1 colocalise on GNPs with the related L1-like adhesion molecule NrCAM, and F3 fails to suppress the SHH-induced proliferation of NrCAM-deficient GNPs. We show that F3 and SHH both primarily affect a group of intermediate GNPs (IPs), which, though actively dividing, also express molecules associated with differentiation, including β-tubulin III (TuJ1) and TAG1. In vivo, intermediate progenitors form a discrete layer in the middle of the external germinal layer (mEGL), while F3 becomes expressed on the axons of postmitotic granule neurons as they leave the inner EGL (iEGL). We propose, therefore, that F3 acts as a localised signal in the iEGL that induces SHH-stimulated cells in the overlying mEGL to exit cell cycle and differentiate. By contrast, expression of TAG1 on GNPs antagonises this signal in the mEGL, preventing premature differentiation and sustaining GNP expansion in a paracrine fashion. Together, these findings indicate that CNTN and L1-like proteins play a significant role in modulating SHH-induced neuronal precursor proliferation.
- Published
- 2011
- Full Text
- View/download PDF
12. Small Molecule Activation of Adaptive Gene Expression
- Author
-
Robert S. Freeman, Harley I. Kornblum, Ksenia Karpisheva, Ambreena Siddiq, Ronald P. Hart, Hsin Hwa Lee, Michael S. Beattie, Sashwati Roy, Thomas S. Carmichael, Irina G. Gazaryan, Stephen J. McConoughey, Martin Grumet, Brett Langley, Ahmet Hoke, Rajiv R. Ratan, Gregg L. Semenza, Chandan K. Sen, Cameron Rink, Dongming Sun, N. A. Smirnova, Leila Aminova, Daniel H. Geschwind, and Giovanni Coppola
- Subjects
Male ,Central nervous system ,Gene Expression ,Tilorone ,Biology ,Pharmacology ,Neuroprotection ,Article ,General Biochemistry, Genetics and Molecular Biology ,Rats, Sprague-Dawley ,History and Philosophy of Science ,Gene expression ,medicine ,Animals ,Cells, Cultured ,Spinal Cord Injuries ,Reporter gene ,Activator (genetics) ,General Neuroscience ,Hypoxia (medical) ,Hypoxia-Inducible Factor 1, alpha Subunit ,Rats ,Stroke ,medicine.anatomical_structure ,Hypoxia-inducible factors ,medicine.symptom ,medicine.drug - Abstract
A major challenge for neurological therapeutics is the development of small molecule drugs that can activate a panoply of downstream pathways without toxicity. Over the past decade our group has shown that a family of enzymes that regulate posttranscriptional and transcriptional adaptive responses to hypoxia are viable targets for neuronal protection and repair. The family is a group of iron, oxygen, and 2-oxoglutarate-dependent dioxygenases, known as the HIF prolyl 4-hydroxylases (HIF PHDs). We have previously shown that pluripotent protection offered by iron chelators is mediated, in part, via the ability of these agents to inhibit the HIF PHDs. Our group and others have implicated the transcriptional activator HIF-1 in some of the salutary effects of iron chelation-induced PHD inhibition. While some iron chelators are currently employed in humans for conditions such as hemochromatosis, the diverse utilization of iron in physiological processes in the brain makes the development of HIF activators that do not bind iron a high priority. Here we report the development of a high throughput screen to develop novel HIF activators and/or PHD inhibitors for therapeutic use in the central nervous system (CNS). We show that tilorone, a low-molecular weight, antiviral, immunomodulatory agent is the most effective activator of the HIF pathway in a neuronal line. We also show that tilorone enhances HIF protein levels and increases the expression of downstream target genes independent of iron chelation and HIF PHD inhibition in vitro. We further demonstrate that tilorone can activate an HIF-regulated reporter gene in the CNS. These studies confirm that tilorone can penetrate the blood–brain barrier to activate HIF in the CNS. As expected from these findings, we show that tilorone provides effective prophylaxis against permanent ischemic stroke and traumatic spinal cord injury in male rodents. Altogether these findings identify tilorone as a novel and potent modulator of HIF-mediated gene expression in neurons with neuroprotective properties.
- Published
- 2008
- Full Text
- View/download PDF
13. Functional differentiation of a clone resembling embryonic cortical interneuron progenitors
- Author
-
Loyal A. Goff, Cynthia Camarillo, Ronald P. Hart, Kevin Thompson, Yu R. Han, Mavis R. Swerdel, Jonathan Davila, Hedong Li, Caixia Bi, Martin Grumet, Mark R. Plummer, and Christopher L. Ricupero
- Subjects
Patch-Clamp Techniques ,Interneuron ,Cellular differentiation ,Clone (cell biology) ,Action Potentials ,Bone Morphogenetic Protein 2 ,Gene Expression ,Nerve Tissue Proteins ,Cell Separation ,Biology ,Calbindin ,Article ,Magnetics ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Interneurons ,Tubulin ,medicine ,Animals ,Cells, Cultured ,Embryonic Stem Cells ,Cerebral Cortex ,Glutamate receptor ,Cell Differentiation ,Embryo, Mammalian ,Molecular biology ,Clone Cells ,Rats ,medicine.anatomical_structure ,nervous system ,Cerebral cortex ,Culture Media, Conditioned ,Forebrain ,Fibroblast Growth Factor 2 ,Calretinin - Abstract
We have generated clones (L2.3 and RG3.6) of neural progenitors with radial glial properties from rat E14.5 cortex that differentiate into astrocytes, neurons, and oligodendrocytes. Here, we describe a different clone (L2.2) that gives rise exclusively to neurons, but not to glia. Neuronal differentiation of L2.2 cells was inhibited by bone morphogenic protein 2 (BMP2) and enhanced by Sonic Hedgehog (SHH) similar to cortical interneuron progenitors. Compared with L2.3, differentiating L2.2 cells expressed significantly higher levels of mRNAs for glutamate decarboxylases (GADs), DLX transcription factors, calretinin, calbindin, neuropeptide Y (NPY), and somatostatin. Increased levels of DLX-2, GADs, and calretinin proteins were confirmed upon differentiation. L2.2 cells differentiated into neurons that fired action potentials in vitro, and their electrophysiological differentiation was accelerated and more complete when cocultured with developing astroglial cells but not with conditioned medium from these cells. The combined results suggest that clone L2.2 resembles GABAergic interneuron progenitors in the developing forebrain.
- Published
- 2008
- Full Text
- View/download PDF
14. Activated Notch1 maintains the phenotype of radial glial cells and promotes their adhesion to laminin by upregulating nidogen
- Author
-
Hui Wen Su, Yu Wen Chang, Ajoeb Baridi, Ronald P. Hart, Hedong Li, Martin Grumet, Christopher L. Ricupero, and Kriti Mohan
- Subjects
Green Fluorescent Proteins ,Clone (cell biology) ,Nerve Tissue Proteins ,Fatty Acid-Binding Proteins ,Transfection ,Article ,Cellular and Molecular Neuroscience ,Laminin ,Cell Adhesion ,Animals ,Receptor, Notch1 ,Progenitor cell ,Cell adhesion ,Cells, Cultured ,Oligonucleotide Array Sequence Analysis ,Cerebral Cortex ,Membrane Glycoproteins ,biology ,Cell adhesion molecule ,Stem Cells ,Adhesion ,Embryo, Mammalian ,Embryonic stem cell ,Molecular biology ,Neural stem cell ,Rats ,Up-Regulation ,Phenotype ,Spinal Cord ,Neurology ,biology.protein ,Fatty Acid-Binding Protein 7 ,Neuroglia ,Stem Cell Transplantation - Abstract
Radial glia are neural stem cells that exist only transiently during central nervous system (CNS) development, where they serve as scaffolds for neuronal migration. Their instability makes them difficult to study, and therefore we have isolated stabilized radial glial clones from E14.5 cortical progenitors (e.g., L2.3) after expression of v-myc. Activated Notch1 intracellular region (actNotch1) promotes radial glia in the embryonic mouse forebrain (Gaiano et al., (2000), and when it was introduced into E14.5 cortical progenitors or radial glial clone L2.3, the cells exhibited enhanced radial morphology and increased expression of the radial glial marker BLBP. A representative clone of L2.3 cells expressing actNotch1 called NL2.3-4 migrated more extensively than L2.3 cells in culture and in white matter of the adult rat spinal cord. Microarray and RT-PCR comparisons of mRNAs expressed in these closely related clones showed extensive similarities, but differed significantly for certain mRNAs including several cell adhesion molecules. Cell adhesion assays demonstrated significantly enhanced adhesion to laminin of NL2.3-4 by comparison to L2.3 cells. The laminin binding protein nidogen was the most highly induced adhesion molecule in NL2.3-4, and immunological analyses indicated that radial glia synthesize and secrete nidogen. Adhesion of NL2.3-4 cells to laminin was inhibited by anti-nidogen antibodies and required the nidogen binding region in laminin, indicating that nidogen promotes cell adhesion to laminin. The combined results indicate that persistent expression of activated Notch1 maintains the phenotype of radial glial cells, inhibits their differentiation, and promotes their adhesion and migration on a laminin/nidogen complex. © 2008 Wiley-Liss, Inc.
- Published
- 2008
- Full Text
- View/download PDF
15. Single, High-Dose Intraspinal Injection of Chondroitinase Reduces Glycosaminoglycans in Injured Spinal Cord and Promotes Corticospinal Axonal Regrowth after Hemisection but Not Contusion
- Author
-
Noriko Kane-Goldsmith, Sameer Ahmed, Tetsuhito Okuda, Martin Grumet, Wise Young, Yu Wen Chang, Marlon Mathew, and Tsutomu Iseda
- Subjects
Pathology ,medicine.medical_specialty ,Time Factors ,animal structures ,Central nervous system ,Pyramidal Tracts ,Chondroitin ABC Lyase ,Rats, Sprague-Dawley ,Lesion ,chemistry.chemical_compound ,Injury Site ,medicine ,Animals ,Chondroitin sulfate ,Injections, Spinal ,Spinal Cord Injuries ,Dose-Response Relationship, Drug ,business.industry ,Chondroitin Sulfates ,Chondroitinase treatment ,Anatomy ,Spinal cord ,Axons ,Nerve Regeneration ,Rats ,medicine.anatomical_structure ,chemistry ,Gliosis ,embryonic structures ,Corticospinal tract ,Female ,Neurology (clinical) ,medicine.symptom ,business - Abstract
Chondroitin sulfate proteoglycans (CSPGs) inhibit axonal growth, and treatment with chondroitinase ABC promotes axonal regeneration in some models of central nervous system (CNS) injury. The aims of this study were (1) to compare the spatiotemporal appearance of CSPG expression between spinal cord contusion and hemisection models, and (2) to evaluate chondroitinase treatment effects on axonal regrowth in the two injury models. After hemisection, CSPG-immunoreactivity (IR) in the injury site rose to peak levels at 18 days but then decreased dramatically by 49 days; in contrast, CSPG-IR remained high for at least 49 days after contusion. After hemisection, many anterogradely labeled corticospinal tract (CST) axons remained close to CSPG-rich lesion sites, but after contusion, most CST axons retracted by approximately 1 mm rostral from the rostral-most CSPG-rich cyst. Intraspinal injection of chondroitinase at 0, 1, 2, and 4 weeks following injury dramatically reduced CSPG-IR in both injury models within 4 days, and CSPG-IR remained low for at least 3 weeks. After the chondroitinase treatment, many axons grew around the lesion site in hemisected spinal cords but not in contused spinal cords. We propose that improved axonal growth in hemisected spinal cords is due to decreased inhibition resulting from degradation of CSPGs located adjacent to severed CST axons. However, in spinal cord contusions, retracted CST axons fail to grow across gliotic regions that surround CSPG-rich injury sites despite efficient degradation with chondroitinase, suggesting that other inhibitors of axonal growth persist in the gliotic regions.
- Published
- 2008
- Full Text
- View/download PDF
16. Differential expression and functions of neuronal and glial neurofascin isoforms and splice variants during PNS development
- Author
-
Martin Grumet, Noriko Kane-Goldsmith, Sayantani Basak, Darshan Koticha, Karthik Raju, and Joanne Babiarz
- Subjects
Gene isoform ,Cell Adhesion Molecules, Neuronal ,PNS ,Schwann cell ,Biology ,Myelin ,Ganglia, Spinal ,Glia ,Paranode ,Cell Adhesion ,medicine ,Animals ,Humans ,Protein Isoforms ,Nerve Growth Factors ,RNA, Small Interfering ,Cell adhesion ,Molecular Biology ,Cells, Cultured ,Myelin Sheath ,Neurons ,Node of Ranvier ,Cell adhesion molecule ,Cell Biology ,Embryo, Mammalian ,Coculture Techniques ,Fibronectins ,Rats ,Cell biology ,Fibronectin ,Alternative Splicing ,Nerve development ,Ig CAM ,medicine.anatomical_structure ,nervous system ,Immunology ,biology.protein ,Neuroglia ,Schwann Cells ,Cell Adhesion Molecules ,Developmental Biology - Abstract
The cell adhesion molecule neurofascin (NF) has a major neuronal isoform (NF186) containing a mucin-like domain followed by a fifth fibronectin type III repeat while these domains are absent from glial NF155. Neuronal NF isoforms lacking one or both of these domains are expressed transiently in embryonic dorsal root ganglia (DRG). These two domains are co-expressed in mature NF186, which peaks in expression prior to birth and then persists almost exclusively at nodes of Ranvier on myelinated axons. In contrast, glial NF155 is only detected postnatally with the onset of myelination. All these forms of NF bound homophilically and to Schwann cells but only the mature NF186 isoform inhibits cell adhesion, and this activity may be important in formation of the node of Ranvier. Schwann cells deficient in NF155 myelinated DRG axons in a delayed manner and they showed significantly decreased clustering of both NF and Caspr in regions where paranodes normally form. The combined results suggest that NF186 is expressed prenatally on DRG neurons and it may modulate their adhesive interactions with Schwann cells, which express NF155 postnatally and require it for development of axon–glial paranodal junctions.
- Published
- 2007
- Full Text
- View/download PDF
17. Nodes of Ranvier and axon initial segments are ankyrin G–dependent domains that assemble by distinct mechanisms
- Author
-
Martin Grumet, Yulia Dzhashiashvili, Isabel Lam, James L. Salzer, Jolanta Galinska, and Yanqing Zhang
- Subjects
Ankyrins ,animal structures ,Biology ,Article ,Sodium Channels ,03 medical and health sciences ,0302 clinical medicine ,Ranvier's Nodes ,Ankyrin ,Animals ,Spectrin ,Nerve Growth Factors ,Cytoskeleton ,Research Articles ,Cells, Cultured ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Cell adhesion molecule ,Sodium channel ,Cell Biology ,Axon initial segment ,Axons ,Cell biology ,Protein Structure, Tertiary ,Rats ,chemistry ,Ectodomain ,nervous system ,Immunology ,NODAL ,Cell Adhesion Molecules ,030217 neurology & neurosurgery - Abstract
Axon initial segments (AISs) and nodes of Ranvier are sites of action potential generation and propagation, respectively. Both domains are enriched in sodium channels complexed with adhesion molecules (neurofascin [NF] 186 and NrCAM) and cytoskeletal proteins (ankyrin G and βIV spectrin). We show that the AIS and peripheral nervous system (PNS) nodes both require ankyrin G but assemble by distinct mechanisms. The AIS is intrinsically specified; it forms independent of NF186, which is targeted to this site via intracellular interactions that require ankyrin G. In contrast, NF186 is targeted to the node, and independently cleared from the internode, by interactions of its ectodomain with myelinating Schwann cells. NF186 is critical for and initiates PNS node assembly by recruiting ankyrin G, which is required for the localization of sodium channels and the entire nodal complex. Thus, initial segments assemble from the inside out driven by the intrinsic accumulation of ankyrin G, whereas PNS nodes assemble from the outside in, specified by Schwann cells, which direct the NF186-dependent recruitment of ankyrin G.
- Published
- 2007
18. BMP and LIF signaling coordinately regulate lineage restriction of radial glia in the developing forebrain
- Author
-
Hedong Li and Martin Grumet
- Subjects
PAX6 Transcription Factor ,Bone Morphogenetic Protein 2 ,Nerve Tissue Proteins ,Neural Cell Adhesion Molecule L1 ,Ciliary neurotrophic factor ,Fatty Acid-Binding Proteins ,Leukemia Inhibitory Factor ,Bone morphogenetic protein 2 ,Cellular and Molecular Neuroscience ,Prosencephalon ,Transforming Growth Factor beta ,Animals ,Paired Box Transcription Factors ,Cell Lineage ,Secretion ,Eye Proteins ,Neural cell ,Cells, Cultured ,Embryonic Stem Cells ,Janus Kinases ,Homeodomain Proteins ,Neurons ,biology ,Polysialic acid ,Stem Cells ,Antibodies, Monoclonal ,Cell Differentiation ,Sialyltransferases ,Neural stem cell ,Rats ,Cell biology ,Repressor Proteins ,Neurology ,Antigens, Surface ,Bone Morphogenetic Proteins ,Choroid Plexus ,embryonic structures ,Forebrain ,Sialic Acids ,biology.protein ,Choroid plexus ,Fatty Acid-Binding Protein 7 ,Neuroglia ,Neuroscience ,Biomarkers ,Signal Transduction - Abstract
The earliest radial glia are neural stem cells that guide neural cell migration away from ventricular zones. Subsequently, radial glia become lineage restricted during development before they differentiate into more mature cell types in the CNS. We have previously shown that subpopulations of radial glial cells express markers for glial and neuronal restricted precursors (GRPs and NRPs) in expression patterns that are temporally and spatially regulated during CNS development. To characterize further the mechanism of this regulation in rat forebrain, we tested whether secreted factors that are present during development effect lineage restriction of radial glia. We show here that in radial glial cultures LIF/CNTF up-regulates, whereas BMP2 down-regulates GRP antigens recognized by monoclonal antibodies A2B5/4D4. These activities combined with secretion of BMPs dorsally and LIF/CNTF from the choroid plexus provide an explanation for the graded distribution pattern of A2B5/4D4 in dorso-lateral ventricular regions in vivo. The regulation by LIF/CNTF of A2B5/4D4 is mediated through the JAK-STAT pathway. BMP2 promotes expression on radial glial cells of the NRP marker polysialic acid most likely by regulating N-CAM expression itself, as well as at least one polysialyl transferase responsible for synthesis of polysialic acid on N-CAM. Taken together, these results suggest that generation of lineage-restricted precursors is coordinately regulated by gradients of the secreted factors BMPs and LIF/CNTF during development of dorsal forebrain.
- Published
- 2007
- Full Text
- View/download PDF
19. Transcriptional Regulation of Notch1 Expression by Nkx6.1 in Neural Stem/Progenitor Cells during Ventral Spinal Cord Development
- Author
-
Martin Grumet, Ying Li, Kelvin Y. Kwan, Evangeline Tzatzalos, and Li Cai
- Subjects
0301 basic medicine ,Transcription, Genetic ,Cellular differentiation ,Neurogenesis ,Cell Count ,Biology ,Models, Biological ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neural Stem Cells ,Genes, Reporter ,Interneurons ,Animals ,Progenitor cell ,Receptor, Notch1 ,Regulation of gene expression ,Homeodomain Proteins ,Motor Neurons ,Multidisciplinary ,Spinal Cord Ventral Horn ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Embryonic stem cell ,Immunohistochemistry ,Neural stem cell ,Cell biology ,Neuroepithelial cell ,Endothelial stem cell ,030104 developmental biology ,Enhancer Elements, Genetic ,embryonic structures ,cardiovascular system ,030217 neurology & neurosurgery ,Adult stem cell ,Protein Binding - Abstract
Notch1 signaling plays a critical role in maintaining and determining neural stem/progenitor cell (NSPC) fate, yet the transcriptional mechanism controlling Notch1 specific expression in NSPCs remains incomplete. Here, we show transcription factor Nkx6.1 interacts with a cis-element (CR2, an evolutionarily conserved non-coding fragment in the second intron of Notch1 locus) and regulates the expression of Notch1 in ventral NSPCs of the developing spinal cord. We show that the Notch1 expression is modulated by the interaction of Nkx6.1 with a 139 bp enhancer sequence within CR2. Knockdown or overexpression of Nkx6.1 leads to down- or up-regulated Notch1 expression, respectively. In CR2-GFP transgenic mouse, GFP expression was found prominent in the ventricular zone and neural progenitor cells from embryonic day 9.5 to postnatal day 7. GFP+ cells were mainly neural progenitors for interneurons and not for motoneurons or glial cells. Moreover, GFP expression persisted in a subset of ependymal cells in the adult spinal cord, suggesting that CR2 is active in both embryonic and adult NSPCs. Together our data reveal a novel mechanism of Notch1 transcriptional regulation in the ventral spinal cord by Nkx6.1 via its binding with Notch1 enhancer CR2 during embryonic development.
- Published
- 2015
20. Neurofascin interactions play a critical role in clustering sodium channels, ankyrinG and βIV spectrin at peripheral nodes of Ranvier
- Author
-
Sayantani Basak, Noriko Kane-Goldsmith, Martin Grumet, Patrice Maurel, James L. Salzer, Joanne Babiarz, Darshan Koticha, and George Zanazzi
- Subjects
Ankyrins ,Recombinant Fusion Proteins ,PNS ,Schwann cell ,Nerve Tissue Proteins ,Cell Communication ,Receptors, Fc ,Biology ,Sodium Channels ,Myelin ,Glia ,Ranvier's Nodes ,medicine ,Animals ,Ankyrin ,Spectrin ,Nerve Growth Factors ,Molecular Biology ,chemistry.chemical_classification ,Cell adhesion molecule ,Sodium channel ,Cell Biology ,Axon initial segment ,Axons ,Coculture Techniques ,Growth Inhibitors ,Rats ,Cell biology ,Nerve development ,Ig CAM ,medicine.anatomical_structure ,nervous system ,chemistry ,Immunology ,Schwann Cells ,Neuron ,Cell Adhesion Molecules ,Developmental Biology - Abstract
The Ig cell adhesion molecules (CAM) neurofascin (NF) and Nr-CAM are localized at developing nodes of Ranvier in peripheral myelinated axons prior to clustering of Na + channels. Different isoforms of NF are expressed on neurons and glia, and NF binding on both cells has been suggested to play roles in node and paranode formation. To clarify the role of NF further, we analyzed effects of NF-Fc fusion proteins in Schwann cell–DRG neuron myelinating cocultures. NF-Fc significantly inhibited nodal clustering of Na + channels, ankyrin G , and βIV spectrin, and modestly reduced Caspr clustering at paranodal junctions; it did not significantly affect lengths or numbers of myelin-positive segments, axon initial segments, or accumulations of phosphorylated-ERM proteins in Schwann cell nodal microvilli. NF-Fc binds to Schwann cells but little or no binding to DRG neurons was detected. The results suggest a critical early role for axonal NF in clustering of Na + channels at nodes of Ranvier via interactions with receptors on Schwann cells.
- Published
- 2006
- Full Text
- View/download PDF
21. A Role for Nr-CAM in the Patterning of Binocular Visual Pathways
- Author
-
Mark Henkemeyer, David R. Colman, Carol A. Mason, Scott E. Williams, Takeshi Sakurai, and Martin Grumet
- Subjects
Retinal Ganglion Cells ,Neurite ,genetic structures ,Neuroscience(all) ,Optic chiasm ,Mice, Transgenic ,DEVBIO ,In situ hybridization ,Visual system ,Biology ,Functional Laterality ,MOLNEURO ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Visual Pathways ,In Situ Hybridization ,030304 developmental biology ,Mice, Knockout ,Vision, Binocular ,0303 health sciences ,Retina ,Cell adhesion molecule ,General Neuroscience ,Cell Adhesion Molecules, Neuron-Glia ,Anatomy ,Immunohistochemistry ,eye diseases ,medicine.anatomical_structure ,Retinal ganglion cell ,Optic Chiasm ,CELLBIO ,sense organs ,Neuroscience ,Binocular vision ,030217 neurology & neurosurgery - Abstract
SummaryRetinal ganglion cell (RGC) axons diverge within the optic chiasm to project to opposite sides of the brain. In mouse, contralateral RGCs are distributed throughout the retina, whereas ipsilateral RGCs are restricted to the ventrotemporal crescent (VTC). While repulsive guidance mechanisms play a major role in the formation of the ipsilateral projection, little is known about the contribution of growth-promoting interactions to the formation of binocular visual projections. Here, we show that the cell adhesion molecule Nr-CAM is expressed by RGCs that project contralaterally and is critical for the guidance of late-born RGCs within the VTC. Blocking Nr-CAM function causes an increase in the size of the ipsilateral projection and reduces neurite outgrowth on chiasm cells in an age- and region-specific manner. Finally, we demonstrate that EphB1/ephrin-B2-mediated repulsion and Nr-CAM-mediated attraction comprise distinct molecular programs that each contributes to the proper formation of binocular visual pathways.
- Published
- 2006
- Full Text
- View/download PDF
22. Cell adhesion and neurite outgrowth are promoted by neurofascin NF155 and inhibited by NF186
- Author
-
Noriko Kane-Goldsmith, Jeffrey Jacob, Joanne Babiarz, Martin Grumet, Karthik Raju, and Darshan Koticha
- Subjects
Neurite ,Kidney ,Cell Line ,Cellular and Molecular Neuroscience ,Ranvier's Nodes ,Cell Adhesion ,Neurites ,Humans ,Nerve Growth Factors ,Cell adhesion ,Molecular Biology ,Neural cell ,RGD motif ,Neurons ,biology ,Cell adhesion molecule ,Alternative splicing ,Cell Biology ,Molecular biology ,Protein Structure, Tertiary ,Cell biology ,Fibronectin ,Alternative Splicing ,Mutagenesis ,biology.protein ,Neural cell adhesion molecule ,Cell Adhesion Molecules ,Neuroglia - Abstract
Neurofascin (NF) is a neural cell adhesion molecule in the L1-family containing six Ig domains and multiple fibronectin type III (FnIII) repeats in its extracellular region. NF has many splicing variants and two of these are exemplars that have different cellular patterns of expression during development. NF186, which is expressed on neurons, contains an unusual mucin-like region and NF155, which is expressed on glia, contains a unique FnIII repeat with an RGD motif. Analysis of Fc fusion proteins representing different extracellular regions of NF indicate that NF186 inhibits cell adhesion and neurite outgrowth, and the inhibition is associated with the region containing the mucin-like domain. NF155 promotes neural cell adhesion and neurite outgrowth, and the RGD motif in its third FnIII repeat is critical for cell spreading and neurite outgrowth. The results suggest that different splicing variants of NF expressed on neurons and glia play distinct roles during neural development.
- Published
- 2005
- Full Text
- View/download PDF
23. NrCAM in Addiction Vulnerability: Positional Cloning, Drug-Regulation, Haplotype-Specific Expression, and Altered Drug Reward in Knockout Mice
- Author
-
Takeshi Sakurai, F. S. Hall, Hiroshi Ujike, Jian Ping Gong, Hiroki Ishiguro, Martin Grumet, Marisela Morales, Qing-Rong Liu, and George R. Uhl
- Subjects
Male ,Narcotics ,DNA, Complementary ,Genotype ,Positional cloning ,Substance-Related Disorders ,media_common.quotation_subject ,Amphetamine-Related Disorders ,Molecular Sequence Data ,Biology ,Rats, Sprague-Dawley ,Cocaine-Related Disorders ,Mice ,Cocaine ,Reward ,Animals ,Humans ,RNA, Messenger ,Cloning, Molecular ,Gene ,In Situ Hybridization ,media_common ,Brain Chemistry ,Mice, Knockout ,Pharmacology ,Genetics ,Regulation of gene expression ,Differential display ,Morphine ,Genome, Human ,Addiction ,Haplotype ,Chromosome Mapping ,Blotting, Northern ,Rats ,Mice, Inbred C57BL ,Psychiatry and Mental health ,Gene Expression Regulation ,Haplotypes ,Knockout mouse ,Cell Adhesion Molecules ,Addiction vulnerability - Abstract
Several lines of evidence support roles for the cell adhesion molecule NrCAM in addictions. Fine mapping within a chromosome 7 region that contains previously linked and associated genomic markers identifies NrCAM haplotypes that are associated with substance abuse vulnerabilities in four samples of abusers and controls. Differential display identifies NrCAM as a drug regulated gene. NrCAM is expressed in neurons linked to reward and memory. NrCAM displays haplotype-specific gene expression in human post-mortem brain samples. Knockout mice display reduced opiate- and stimulant-conditioned place preferences. These observations support NrCAM as a positionally cloned and drug-regulated gene whose variants are likely to change expression and alter substance abuse vulnerabilities in human addictions and animal models of drug reward.
- Published
- 2005
- Full Text
- View/download PDF
24. Embryonic radial glia bridge spinal cord lesions and promote functional recovery following spinal cord injury
- Author
-
Osamu Ikeda, Hedong Li, Yu Wen Chang, Noriko Kane-Goldsmith, Koichi Hasegawa, Martin Grumet, and Yana Berlin
- Subjects
Male ,Pathology ,Indoles ,Time Factors ,Fluorescent Antibody Technique ,Nestin ,Rats, Sprague-Dawley ,Intermediate Filament Proteins ,Neurofilament Proteins ,Tubulin ,Spinal cord injury ,Cells, Cultured ,Behavior, Animal ,Stem Cells ,Chondroitin Sulfates ,Cell Differentiation ,Ectodysplasins ,Neural stem cell ,medicine.anatomical_structure ,Neurology ,Neuroglia ,Female ,Proteoglycans ,Stem cell ,Fatty Acid-Binding Protein 7 ,medicine.medical_specialty ,Green Fluorescent Proteins ,Central nervous system ,Nerve Tissue Proteins ,Motor Activity ,Biology ,Fatty Acid-Binding Proteins ,Prosencephalon ,Developmental Neuroscience ,Neurosphere ,Glial Fibrillary Acidic Protein ,medicine ,Animals ,Antigens ,Spinal Cord Injuries ,Membrane Proteins ,Recovery of Function ,Fibroblasts ,Embryo, Mammalian ,medicine.disease ,Spinal cord ,Clone Cells ,Rats ,nervous system ,Neuron ,Carrier Proteins ,Neuroscience ,Stem Cell Transplantation - Abstract
Radial glial cells are neural stem cells (NSC) that are transiently found in the developing CNS. To study radial glia, we isolated clones following immortalization of E13.5 GFP rat neurospheres with v-myc. Clone RG3.6 exhibits polarized morphology and expresses the radial glial markers nestin and brain lipid binding protein. Both NSC and RG3.6 cells migrated extensively in the adult spinal cord. However, RG3.6 cells differentiated into astroglia slower than NSC, suggesting that immortalization can delay differentiation of radial glia. Following spinal cord contusion, implanted RG3.6 cells migrated widely in the contusion site and into spared white matter where they exhibited a highly polarized morphology. When injected immediately after injury, RG3.6 cells formed cellular bridges surrounding spinal cord lesion sites and extending into spared white matter regions in contrast to GFP fibroblasts that remained in the lesion site. Behavioral analysis indicated higher BBB scores in rats injected with RG3.6 cells than rats injected with fibroblasts or medium as early as 1 week after injury. Spinal cords transplanted with RG3.6 cells or dermal fibroblasts exhibited little accumulation of chondroitin sulfate proteoglycans (CSPG) including NG2 proteoglycans that are known to inhibit axonal growth. Reduced levels of CSPG were accompanied by little accumulation in the injury site of activated macrophages, which are a major source of CSPG. However, increased staining and organization of neurofilaments were found in injured rats transplanted with RG3.6 cells suggesting neuroprotection or regrowth. The combined results indicate that acutely transplanted radial glia can migrate to form bridges across spinal cord lesions in vivo and promote functional recovery following spinal cord injury by protecting against macrophages and secondary damage.
- Published
- 2005
- Full Text
- View/download PDF
25. Signal transduction pathways implicated in neural recognition molecule L1 triggered neuroprotection and neuritogenesis
- Author
-
Melitta Schachner, Suzhen Chen, Martin Grumet, and Gabriele Loers
- Subjects
biology ,Kinase ,Biochemistry ,Neuroprotection ,Cell biology ,Cellular and Molecular Neuroscience ,Mitogen-activated protein kinase ,biology.protein ,Phosphorylation ,Src family kinase ,Signal transduction ,Protein kinase A ,Protein kinase B - Abstract
The signal transduction pathways involved in adhesion molecule L1-triggered neuritogenesis and neuroprotection were investigated using the extracellular domain of mouse or human L1 in fusion with the Fc portion of human immunoglobulin G or L1 purified from mouse brain by affinity chromatography. Substrate L1-triggered neuritogenesis and neuroprotection depended on distinct but also overlapping signal transduction pathways and on the expression of L1 at the neuronal cell surface. PI3 kinase inhibitors, Src family kinase inhibitors as well as mitogen-activated protein kinase kinase inhibitors reduced both L1-triggered neuritogenesis and neuroprotection. In contrast, fibroblast growth factor receptor inhibitors, a protein kinase A inhibitor, and an inhibitor of cAMP-mediated signal transduction pathways, blocked neuritogenesis, but did not affect L1-triggered neuroprotection. Proteolytic cleavage of L1 or its interaction partners is necessary for both L1-mediated neuritogensis and neuroprotection. Furthermore, L1-triggered neuroprotection was found to be associated with increased phosphorylation of extracellular signal-regulated kinases 1/2, Akt and Bad, and inhibition of caspases. These observations suggest possibilities of differentially targeting signal transduction pathways for L1-dependent neuritogenesis and neuroprotection.
- Published
- 2005
- Full Text
- View/download PDF
26. The Role of the Ankyrin-Binding Protein NrCAM in Node of Ranvier Formation
- Author
-
Peter Shrager, Takeshi Sakurai, William Simon, Xiaorong Xu, Katia Kazarinova-Noyes, Andrew W. Custer, and Martin Grumet
- Subjects
Ankyrins ,Action Potentials ,Schwann cell ,In Vitro Techniques ,Biology ,Ankyrin binding ,Sodium Channels ,Mice ,Ranvier's Nodes ,medicine ,Animals ,Ankyrin ,Remyelination ,Axon ,Cytoskeleton ,Cells, Cultured ,Myelin Sheath ,Mice, Knockout ,chemistry.chemical_classification ,Node of Ranvier ,General Neuroscience ,Binding protein ,Lysophosphatidylcholines ,Optic Nerve ,Sciatic Nerve ,Axons ,Rats ,Cell biology ,medicine.anatomical_structure ,nervous system ,chemistry ,Schwann Cells ,Cell Adhesion Molecules ,Neuroscience ,Cellular/Molecular - Abstract
Molecular events involved in Na+channel clustering at the node of Ranvier have been investigated during early development. NrCAM, an ankyrinG-binding protein, precedes Na+channels at cluster sites adjacent to the tips of Schwann cell processes. Both Na+channel and ankyrinGsequestration at developing nodes are delayed in NrCAM null mutants. The action of NrCAM is manifest locally at individual nodes, rather than affecting overall neuronal expression, and is linked to glial interactions. During remyelination, Na+channel clusters at new nodes are initially labile, and anchoring to the cytoskeleton appears to grow progressively with time. The distance between Na+channel clusters across remyelinating Schwann cells (nascent internodes) increases markedly from 83 to 274 μm during node formation, arguing against schemes in which the loci of nodes are fixed in advance by the axon. A hypothesis for node formation in which axonal Na+channels move by lateral diffusion from regions of Schwann cell contact, with clustering dependent on linkage to the cytoskeleton by ankyrinG, is proposed and tested in a computational model. To match experimental measurements, this latter reaction needs fast kinetics, and the early arrival of NrCAM is postulated to contribute to this requirement.
- Published
- 2003
- Full Text
- View/download PDF
27. Microtubules are critical for radial glial morphology: Possible regulation by MAPs and MARKs
- Author
-
Ronald P. Hart, Martin Grumet, Hedong Li, and Yana Berlin
- Subjects
Cytochalasin D ,Microtubule-associated protein ,Population ,Protein Serine-Threonine Kinases ,Biology ,Microfilament ,Microtubules ,Rats, Sprague-Dawley ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Microtubule ,medicine ,Animals ,RNA, Messenger ,education ,Cytoskeleton ,Cells, Cultured ,Cell Size ,education.field_of_study ,Nocodazole ,Stem Cells ,Brain ,Cell Polarity ,Cell Differentiation ,Rats ,Cell biology ,medicine.anatomical_structure ,Animals, Newborn ,nervous system ,Neurology ,chemistry ,Neuroglia ,Microtubule-Associated Proteins ,Neuroscience - Abstract
Radial glia are a polarized cell type that in most neural regions appear only transiently during development. They have long been recognized as glia or glial progenitors that support neuronal migration. Recent evidence indicates that radial glia also give rise to neurons and appear to be a major population of dividing precursor cells in the embryonic cortical ventricular zone. Radial glia extend long processes from the ventricular zone to the pial surface that provide guides for neuronal migration. We reasoned that the unique morphology of radial glia is due to the composition and organization of their cytoskeleton. In this present study, we have used C6-R, a radial glial-like cell line and isolated perinatal cerebellar radial glia to ask what are the critical cytoskeletal elements in radial glial cells and how they are regulated. Treatments with nocodazole and cytochalasin D showed that microtubules, but not microfilaments, are critical for the polarized morphology of radial glia. In addition, quantitative real-time PCR indicated that certain mRNAs specific for microtubule-associated proteins (MAPs) are selectively expressed in radial glia. These results together with the known ability of microtubule affinity-regulating kinases to regulate microtubule organization suggest that microtubules and MAPs are critical for the morphology of radial glia.
- Published
- 2003
- Full Text
- View/download PDF
28. Trauma-induced tumorigenesis of cells implanted into the rat spinal cord
- Author
-
Koichi Hasegawa and Martin Grumet
- Subjects
Pathology ,medicine.medical_specialty ,Central nervous system ,medicine.disease_cause ,Green fluorescent protein ,Rats, Sprague-Dawley ,Central nervous system disease ,Glial Fibrillary Acidic Protein ,Tumor Cells, Cultured ,medicine ,Animals ,Spinal Cord Neoplasms ,Spinal Cord Injuries ,business.industry ,medicine.disease ,Spinal cord ,Pathophysiology ,Rats ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Spinal Cord ,Cell culture ,Female ,Glioblastoma ,Carcinogenesis ,business - Abstract
Object. Findings in several clinical cases have suggested a correlation between tumor formation and previous injury to the central nervous system (CNS); however, the relationship between trauma and tumorigenesis has not been investigated well experimentally. In this study the authors provide evidence correlating tumorigenesis with trauma in the rat spinal cord. Methods. A glial cell line, C6R-G/H, which expresses green fluorescent protein (GFP) and hygromycin phosphotransferase (HPT), was implanted into normal and injured rat spinal cords. In all rats in which the cells were implanted into an injured site, locomotor function deteriorated and histological analysis demonstrated glioblastoma multiforme by 6 weeks; tumorigenesis was correlated with a loss of both GFP expression and resistance to hygromycin treatment. In contrast, no evidence of tumor formation was found at 6 weeks in rats in which the cells were implanted into healthy tissue. When C6R-G/H cells were treated with contused spinal cord extract in culture before implantation, they lost GFP expression and hygromycin resistance, and later formed tumors after implantation into normal spinal cord. Conclusions. The findings of this study indicate that trauma can induce tumorigenesis. Implantation of C6R-G/H cells into traumatized spinal cords resulted in their transformation, which was signaled by loss of GFP expression and hygromycin resistance accompanied by tumor formation. Exposure to extracts derived from injured spinal cord produced similar transformation and gene expression changes, as well as tumor formation after such cells were implanted into normal cords. Care, therefore, should be taken when cells are implanted into an injured CNS because of potential mutagenesis due to trauma-induced factors.
- Published
- 2003
- Full Text
- View/download PDF
29. Nr-CAM and TAG-1 are expressed in distinct populations of developing precerebellar and cerebellar neurons
- Author
-
Takeshi Sakurai, Constantino Sotelo, Martin Grumet, Evelyne Bloch-Gallego, and S. Backer
- Subjects
Neurons ,Cerebellum ,Cell Adhesion Molecules, Neuronal ,General Neuroscience ,Pontine nuclei ,Gene Expression Regulation, Developmental ,Biology ,Spinal cord ,Pons ,Rats ,Mice ,medicine.anatomical_structure ,Reticular connective tissue ,Contactin 2 ,medicine ,Animals ,RNA, Messenger ,Brainstem ,Cell Adhesion Molecules ,Neuroscience ,Medulla ,Floor plate - Abstract
Nr-CAM and TAG-1 interact at the floor-plate during the formation of spinal cord commissural projections [Stoeckli, E.T., Landmesser, L.T., Sci. 274 (1995) 1123-1133; Fitzli, D., Stoeckli, E.T., Kunz, S., Siribour, K., Rader, C., Kunz, B., Kozlov, S.V., Buchstaller, A., Lane, R.P., Suter, D.M., Dreyer, W.J., Sonderegger, P., J. Cell. Biol. 149 (2000) 951-968]. We report here the spatio-temporal patterns of expression of these two adhesion molecules during the development of the lower brainstem (medulla and pons) and cerebellum. Nr-CAM and Tag-1 label distinct populations of precerebellar neurons at key steps of their development. Nr-CAM expression starts at E11.5–E12 in the floor-plate, that constitutes an intermediate target during axon outgrowth and nuclear migration of precerebellar neurons. At E13–E14, it is expressed in both floor-plate and inferior olivary nuclei (ION) neurons before being strictly restricted to ION neurons from E15 onwards. Furthermore Nr-CAM , which is widely expressed in the cerebellum during embryonic development, becomes strictly confined to Purkinje and Golgi cells in postnatal cerebellum, suggesting a possible role of Nr-CAM for the maturation or stabilization of the synaptic contacts, in particular between climbing fibers and Purkinje cells. On the other hand, Tag-1 is expressed by migrating neurons that will form the lateral reticular and basilar pontine nuclei. These results emphasize the possibility that TAG-1/Nr-CAM interactions are also involved in the development of the cerebellar system (precerebellar and cerebellar neurons). However, the pattern of cerebellar expression of TAG-1 – early migrating Purkinje cells up to E14 and external granular cells – prevents the implication of this adhesion molecule in the organization of extracerebellar projections.
- Published
- 2002
- Full Text
- View/download PDF
30. Long-term maintenance of Na+ channels at nodes of Ranvier depends on glial contact mediated by gliomedin and NrCAM
- Author
-
Martin Grumet, Konstantin Feinberg, Yael Eshed-Eisenbach, Veronique Amor, Shahar Frechter, Elior Peles, Anya Vainshtein, and Jack Rosenbluth
- Subjects
Ankyrins ,Male ,Cell Adhesion Molecules, Neuronal ,Schwann cell ,Action Potentials ,Voltage-Gated Sodium Channels ,Biology ,Myelin ,Mice ,Ranvier's Nodes ,medicine ,Ankyrin ,Animals ,Spectrin ,Nerve Growth Factors ,chemistry.chemical_classification ,Node of Ranvier ,General Neuroscience ,Sodium channel ,Cell Membrane ,Articles ,Axolemma ,Cell biology ,Protein Transport ,medicine.anatomical_structure ,chemistry ,nervous system ,Female ,NODAL ,Neuroscience ,Cell Adhesion Molecules ,Neuroglia ,Gene Deletion - Abstract
Clustering of Na+channels at the nodes of Ranvier is coordinated by myelinating glia. In the peripheral nervous system, axoglial contact at the nodes is mediated by the binding of gliomedin and glial NrCAM to axonal neurofascin 186 (NF186). This interaction is crucial for the initial clustering of Na+channels at heminodes. As a result, it is not clear whether continued axon-glial contact at nodes of Ranvier is required to maintain these channels at the nodal axolemma. Here, we report that, in contrast to mice that lack either gliomedin or NrCAM, absence of both molecules (and hence the glial clustering signal) resulted in a gradual loss of Na+channels and other axonal components from the nodes, the formation of binary nodes, and dysregulation of nodal gap length. Therefore, these mice exhibit neurological abnormalities and slower nerve conduction. Disintegration of the nodes occurred in an orderly manner, starting with the disappearance of neurofascin 186, followed by the loss of Na+channels and ankyrin G, and then βIV spectrin, a sequence that reflects the assembly of nodes during development. Finally, the absence of gliomedin and NrCAM led to the invasion of the outermost layer of the Schwann cell membrane beyond the nodal area and the formation of paranodal-like junctions at the nodal gap. Our results reveal that axon-glial contact mediated by gliomedin, NrCAM, and NF186 not only plays a role in Na+channel clustering during development, but also contributes to the long-term maintenance of Na+channels at nodes of Ranvier.
- Published
- 2014
31. Disulfide-mediated dimerization of L1 Ig domains
- Author
-
Martin Grumet, Gregor Schürmann, Harold P. Erickson, Jeffrey Haspel, and Jeffrey Jacob
- Subjects
Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,medicine.anatomical_structure ,Monomer ,chemistry ,Neurite ,Cell ,medicine ,Disulfide bond ,Biophysics ,Extracellular ,Neural Cell Adhesion Molecule L1 ,Fusion protein - Abstract
The neural cell adhesion molecule L1 contains immunoglobulin-like (Ig) domains in its extracellular region that mediate homophilic binding, neurite outgrowth and other activities relevant to CNS development. To correlate conformations of these domains to biological function, several L1-Fc fusion proteins whose bioactivities were previously characterized were analyzed by rotary shadowing electron microscopy. We found that bioactive L1-Fcs containing Ig domains 1–4 or 1–6 exhibited extended, branched structures. In contrast, inactive L1-Fcs containing only the first two or three Ig domains assumed compact shapes that suggested interactions between the L1 arms of these proteins. Analysis of an untagged L1 fragment composed of Ig domains 1-3 demonstrated a mixture of monomeric and dimeric forms. Surprisingly, these dimers were stabilized by intermolecular disulfide bonds. Finally, cell surface L1-GFP fusion proteins containing only the first two or three Ig domains in the extracellular region also engaged in disulfide-mediated dimerization. These results suggest a novel mechanism by which mutations in L1 could interfere with its biological functioning. J. Neurosci. Res. 66:347–355, 2001. © 2001 Wiley-Liss, Inc.
- Published
- 2001
- Full Text
- View/download PDF
32. Cell Adhesion Molecule L1 in Folded (Horseshoe) and Extended Conformations
- Author
-
Martin Grumet, Harold P. Erickson, Gregor Schürmann, and Jeffrey Haspel
- Subjects
Protein Folding ,Glycosylation ,Protein Conformation ,Bent molecular geometry ,Immunoglobulins ,Centrifugation ,Biology ,Article ,Cell Line ,law.invention ,Protein structure ,law ,Cell Adhesion ,Humans ,Molecule ,Cell adhesion ,Neural Cell Adhesion Molecules ,Molecular Biology ,Horseshoe (symbol) ,Membrane Glycoproteins ,Proteins ,Cell Biology ,Negative stain ,Recombinant Proteins ,Protein Structure, Tertiary ,Microscopy, Electron ,Crystallography ,Biochemistry ,Insect Proteins ,Electrophoresis, Polyacrylamide Gel ,Protein folding ,Electron microscope ,Leukocyte L1 Antigen Complex - Abstract
We have investigated the structure of the cell adhesion molecule L1 by electron microscopy. We were particularly interested in the conformation of the four N-terminal immunoglobulin domains, because x-ray diffraction showed that these domains are bent into a horseshoe shape in the related molecules hemolin and axonin-1. Surprisingly, rotary-shadowed specimens showed the molecules to be elongated, with no indication of the horseshoe shape. However, sedimentation data suggested that these domains of L1 were folded into a compact shape in solution; therefore, this prompted us to look at the molecules by an alternative technique, negative stain. The negative stain images showed a compact shape consistent with the expected horseshoe conformation. We speculate that in rotary shadowing the contact with the mica caused a distortion of the protein, weakening the bonds forming the horseshoe and permitting the molecule to extend. We have thus confirmed that the L1 molecule is primarily in the horseshoe conformation in solution, and we have visualized for the first time its opening into an extended conformation. Our study resolves conflicting interpretations from previous electron microscopy studies of L1.
- Published
- 2001
- Full Text
- View/download PDF
33. Reduced tumorigenicity of rat glioma cells in the brain when mediated by hygromycin phosphotransferase
- Author
-
Adı́lia Hormigo, Martin Grumet, Perry A. Brittis, David R. Friedlander, and David Zagzag
- Subjects
Graft Rejection ,Pathology ,medicine.medical_specialty ,Angiogenesis ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,Drug Resistance ,Mitosis ,Hygromycin-B kinase ,Biology ,Transfection ,medicine.disease_cause ,Rats, Inbred WKY ,Rats, Nude ,Glioma ,Tumor Cells, Cultured ,medicine ,Animals ,Genes, Tumor Suppressor ,Neovascularization, Pathologic ,Brain Neoplasms ,Cell growth ,medicine.disease ,Pathophysiology ,Rats ,Transplantation ,Luminescent Proteins ,Phosphotransferases (Alcohol Group Acceptor) ,Phenotype ,Cinnamates ,Cell culture ,Mutation ,Cancer research ,Female ,Hygromycin B ,Carcinogenesis ,Cell Division ,Neoplasm Transplantation - Abstract
Object. A variant of C6 glioma cells, C6R-G/H cells express hygromycin phosphotransferase (HPT) and appear to have reduced tumorigenicity in the embryonic brain. The goal of this study was to investigate their reduced capacity to generate tumors in the adult rat brain. Methods. Cell lines were implanted into rat brains and tumorigenesis was evaluated. After 3 weeks, all rats with C6 cells showed signs of neurological disease, whereas rats with C6R-G/H cells did not and were either killed then or allowed to survive until later. Histological studies were performed to analyze tumor size, malignancy, angiogenesis, and cell proliferation. Cells isolated from rat brain tumors were analyzed for mutation to HPT by testing their sensitivity to hygromycin. Conclusions. The results indicate that HPT suppresses tumor formation. Three weeks after implantation, only 44% of animals implanted with C6R-G/H cells developed tumors, whereas all animals that received C6 glioma cells developed high-grade gliomas. The C6R-G/H cells filled a 20-fold smaller maximal cross-sectional area than the C6 cells, and exhibited less malignant characteristics, including reduced angiogenesis, mitosis, and cell proliferation. Similar results were obtained in the brain of nude rats, indicating that the immune system did not play a significant role in suppressing tumor growth. The combination of green fluorescent protein (GFP) and HPT was more effective in suppressing tumorigenesis than either plasmid by itself, indicating that the GFP may protect against inactivation of the HPT. Interestingly, hygromycin resistance was lost in tumor cells that were recovered from a group of animals in which C6R-G/H cells formed tumors, confirming the correlation of HPT with reduced tumorigenicity.
- Published
- 2001
- Full Text
- View/download PDF
34. Nr-CAM expression in the developing mouse nervous system: Ventral midline structures, specific fiber tracts, and neuropilar regions
- Author
-
Carol A. Mason, Martin Grumet, Marc Lustig, Takeshi Sakurai, and Lynda Erskine
- Subjects
Nervous system ,Neuropil ,animal structures ,Central nervous system ,Anterior commissure ,Chick Embryo ,Biology ,Hippocampus ,Nervous System ,Embryonic and Fetal Development ,Mice ,Nerve Fibers ,medicine ,Animals ,In Situ Hybridization ,Floor plate ,General Neuroscience ,Embryo, Mammalian ,Spinal cord ,Immunohistochemistry ,Olfactory Bulb ,medicine.anatomical_structure ,Spinal Cord ,Forebrain ,Axon guidance ,Cell Adhesion Molecules ,Neuroscience - Abstract
Nr-CAM is a member of the L1 subfamily of cell adhesion molecules (CAMs) that belong to the immunoglobulin superfamily. To explore the role of Nr-CAM in the developing nervous system, we prepared specific antibodies against both chick and mouse Nr-CAM using recombinant Fc fusion proteins of chick Nr-CAM and mouse Nr-CAM, respectively. First, we show the specificity of the new anti-chick Nr-CAM antibody compared with a previously employed antibody using the expression patterns of Nr-CAM in the chick spinal cord and floor plate and on commissural axons, where Nr-CAM has been implicated in axon guidance. Using the anti-mouse Nr-CAM antibody, we then studied the expression patterns of Nr-CAM in the developing mouse nervous system along with the patterns of two related CAMs, L1, which labels most growing axons, and TAG-1, which binds to Nr-CAM and has a more restricted distribution. Major sites that are positive for Nr-CAM are specialized glial formations in the ventral midline, including the floor plate in the spinal cord, the hindbrain and midbrain, the optic chiasm, and the median eminence in the forebrain. Similar to what is seen in the chick spinal cord, Nr-CAM is expressed on crossing fibers as they course through these areas. In addition, Nr-CAM is found in crossing fiber pathways, including the anterior commissure, corpus callosum, and posterior commissure, and in nondecussating pathways, such as the lateral olfactory tract and the habenulointerpeduncular tract. Nr-CAM, for the most part, is colocalized with TAG-1 in all of these systems. Based on in vitro studies indicating that the Nr-CAM-axonin-1/TAG-1 interaction is involved in peripheral axonal growth and guidance in the spinal cord [Lustig et al. (1999) Dev Biol 209:340–351; Fitzli et al. (2000) J Cell Biol 149:951–968], the expression patterns described herein implicate a role for this interaction in central nervous system axon growth and guidance, especially at points of decussation. Nr-CAM also is expressed in cortical regions, such as the olfactory bulb. In the hippocampus, however, TAG-1-positive areas are segregated from Nr-CAM-positive areas, suggesting that, in neuropilar regions, Nr-CAM interacts with molecules other than TAG-1. J. Comp. Neurol. 434:13–28, 2001. © 2001 Wiley-Liss, Inc.
- Published
- 2001
- Full Text
- View/download PDF
35. Ventral midline cells are required for the local control of commissural axon guidance in the mouse spinal cord
- Author
-
Martin Grumet, Alexandra L. Joyner, Marc Lustig, Takeshi Sakurai, and Michael P. Matise
- Subjects
animal structures ,Central nervous system ,Kruppel-Like Transcription Factors ,Mice, Transgenic ,Nerve Tissue Proteins ,Zinc Finger Protein Gli2 ,Biology ,Mice ,Posterior commissure ,Interneurons ,medicine ,Animals ,Nerve Growth Factors ,RNA, Messenger ,Axon ,Molecular Biology ,Body Patterning ,Floor plate ,Mice, Knockout ,Tumor Suppressor Proteins ,Gene Expression Regulation, Developmental ,Anatomy ,Netrin-1 ,Commissure ,Spinal cord ,Slit ,Axons ,medicine.anatomical_structure ,Spinal Cord ,nervous system ,Axon guidance ,Transcription Factors ,Developmental Biology - Abstract
Specialized cells at the midline of the central nervous system have been implicated in controlling axon projections in both invertebrates and vertebrates. To address the requirement for ventral midline cells in providing cues to commissural axons in mice, we have analyzed Gli2 mouse mutants, which lack specifically the floor plate and immediately adjacent interneurons. We show that a Dbx1 enhancer drives tau-lacZ expression in a subpopulation of commissural axons and, using a reporter line generated from this construct, as well as DiI tracing, we find that commissural axons projected to the ventral midline in Gli2−/− embryos. Netrin1 mRNA expression was detected in Gli2−/− embryos and, although much weaker than in wild-type embryos, was found in a dorsally decreasing gradient. This result demonstrates that while the floor plate can serve as a source of long-range cues for C-axons in vitro, it is not required in vivo for the guidance of commissural axons to the ventral midline in the mouse spinal cord. After reaching the ventral midline, most commissural axons remained clustered in Gli2−/− embryos, although some were able to extend longitudinally. Interestingly, some of the longitudinally projecting axons in Gli2−/− embryos extended caudally and others rostrally at the ventral midline, in contrast to normal embryos in which virtually all commissural axons turn rostrally after crossing the midline. This finding indicates a critical role for ventral midline cells in regulating the rostral polarity choice made by commissural axons after they cross the midline. In addition, we provide evidence that interactions between commissural axons and floor plate cells are required to modulate the localization of Nr-CAM and TAG-1 proteins on axons at the midline. Finally, we show that the floor plate is not required for the early trajectory of motoneurons or axons of the posterior commissure, whose projections are directed away from the ventral midline in both WT and Gli2−/− embryos, although they are less well organized in Gli2−/−mutants.
- Published
- 1999
- Full Text
- View/download PDF
36. Multi-ligand interactions with receptor-like protein tyrosine phosphatase β: implications for intercellular signaling
- Author
-
Joseph Schlessinger, Elior Peles, and Martin Grumet
- Subjects
Cell signaling ,animal structures ,biology ,Receptor-Like Protein Tyrosine Phosphatases, Class 5 ,Cell adhesion molecule ,Nerve Tissue Proteins ,Cell Communication ,Protein phosphatase 2 ,Protein tyrosine phosphatase ,Ligands ,Nervous System ,Biochemistry ,Receptor tyrosine kinase ,Extracellular Matrix ,Cell biology ,Extracellular matrix ,Receptor-Like Protein Tyrosine Phosphatases ,biology.protein ,Animals ,Humans ,Protein Tyrosine Phosphatases ,Signal transduction ,Neural Cell Adhesion Molecules ,Molecular Biology ,Signal Transduction - Abstract
Receptor-like protein tyrosine phosphatase beta (RPTP beta) shows structural and functional similarity to cell adhesion molecules (CAMs). It binds to several neuronal CAMs and extracellular matrix (ECM) proteins that combine to form cell-recognition complexes. Here, the authors discuss the implications of such complexes for intercellular signaling, and the regulation of RPTP activity by cell-cell and cell-ECM contact.
- Published
- 1998
- Full Text
- View/download PDF
37. Nr-CAM: A cell adhesion molecule with ligand and receptor functions
- Author
-
Martin Grumet
- Subjects
Nervous system ,animal structures ,Histology ,Cell Adhesion Molecules, Neuronal ,Receptors, Cell Surface ,Biology ,Ligands ,Proteomics ,Nervous System ,Pathology and Forensic Medicine ,medicine ,Animals ,Humans ,Nervous System Physiological Phenomena ,Receptors, Immunologic ,Receptor ,Cell adhesion ,Cell adhesion molecule ,Ligand ,Cell Biology ,Cell biology ,medicine.anatomical_structure ,nervous system ,biology.protein ,Antibody ,Cell Adhesion Molecules ,Neural development ,Neuroscience - Abstract
Nr-CAM Ng-CAM-related cell adhesion molecule) is expressed only in the nervous system on a subset of neurons and non-neuronal cells, including floor-plate cells in the spinal cord and Schwann cells. It is a member of the immunoglobulin (Ig) superfamily and can bind by a homophilic mechanism but its heterophilic interactions may be of greater biological significance. Nr-CAM functions as a neuronal receptor for neurite-growth-promoting stimuli provided by contactin/F11, neurofascin, and RPTPbeta. In addition, by binding to neuronal receptors such as axonin-1, it can modulate axonal guidance. Nr-CAM can also interact laterally with contactin/F11 within the plasma membrane in the form of a complex that may transmit signals to regulate axonal growth. This review summarizes the structure and expression of Nr-CAM and discusses its potential functions as a ligand and as a receptor during neural development.
- Published
- 1997
- Full Text
- View/download PDF
38. Heterophilic interactions of the neural cell adhesion molecules Ng-CAM and Nr-CAM with neural receptors and extracellular matrix proteins
- Author
-
Martin Grumet and Takeshi Sakurai
- Subjects
Axonal fasciculation ,animal structures ,nervous system ,L1 ,Nectin ,Chemistry ,Cell adhesion molecule ,General Neuroscience ,Neural cell adhesion molecule ,Intercellular adhesion molecule ,Cell adhesion ,Floor plate ,Cell biology - Abstract
Ng-CAM/L1 and Nr-CAM are closely related neural cell adhesion molecules expressed on neurons and Schwann cells. Ng-CAM/L1 is prevalent on axons and is a potent promoter of neurite growth and axonal fasciculation. Although Nr-CAM does not appear to play a major role in these functions, its transient expression in the developing floor plate of the spinal cord is important for guidance of commissural axons. Ng-CAM and Nr-CAM can bind homophilicly and heterophilicly to several adhesion molecules. Different complexes of adhesion molecules are likely to control axonal growth and guidance as a consequence of their differential localizationsin vivo.
- Published
- 1996
- Full Text
- View/download PDF
39. Expression of polypeptide variants of receptor-type protein tyrosine phosphatase ?: The secreted form, phosphacan, increases dramatically during embryonic development and modulates glial cell behavior in vitro
- Author
-
David R. Friedlander, Takeshi Sakurai, and Martin Grumet
- Subjects
Cell adhesion molecule ,Phosphatase ,Protein tyrosine phosphatase ,Biology ,Molecular biology ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,chemistry ,Proteoglycan ,Chondroitin sulfate proteoglycan ,Cytoplasm ,biology.protein ,Receptor ,Cell adhesion - Abstract
Glial cells express three splicing variants of a receptor-type protein tyrosine phosphatase called RPTP beta. Two are receptor forms that differ in a large extracellular domain. The third is a secreted proteoglycan called phosphacan that lacks the cytoplasmic phosphatase domains. We have now identified, by immunoblotting, proteins corresponding to these three forms of RPTP beta in rat C6 glioma cells and brain. The short receptor form is much more prevalent than the full-length receptor in C6 glioma cells. Phosphacan is much more abundant than either of the receptor forms in rat brain, and its expression increases progressively during embryonic development, while the receptor forms show only moderate changes. In contrast to the long form and phosphacan that were detected as proteoglycans, the short receptor form, lacking the large alternatively spliced domain, was not detected as a chondroitin sulfate proteoglycan. We recently showed that phosphacan binds to the neuron-glia cell adhesion molecule, Ng-CAM, and we now report that glia expressing RPTP beta adhere and extend processes on substrates coated with Ng-CAM. After one day in culture, however, the glia retract their processes and often lift off the substrate. Conditioned medium from glial cells, which contains large amounts of phosphacan, inhibits glial adhesion to Ng-CAM, and depletion of phosphacan from the conditioned medium by immunoadsorption reduces the inhibitory activity. The results show that phosphacan increases dramatically during development, and indicate that secreted forms of RPTP beta can modulate glial cell adhesion and behavior.
- Published
- 1996
- Full Text
- View/download PDF
40. NrCAM-regulating neural systems and addiction-related behaviors
- Author
-
Hiroki, Ishiguro, Frank S, Hall, Yasue, Horiuchi, Takeshi, Sakurai, Akitoyo, Hishimoto, Martin, Grumet, George R, Uhl, Emmanuel S, Onaivi, and Tadao, Arinami
- Subjects
Male ,Mice, Knockout ,Alcohol Drinking ,Ethanol ,Morphine ,Central Nervous System Depressants ,Anxiety ,MSH Release-Inhibiting Hormone ,Article ,Analgesics, Opioid ,Behavior, Addictive ,Mice, Inbred C57BL ,Adaptation, Psychological ,Conditioning, Psychological ,Exploratory Behavior ,Reaction Time ,Animals ,Maze Learning ,Social Behavior ,Cell Adhesion Molecules - Abstract
We have previously shown that a haplotype associated with decreased NrCAM expression in brain is protective against addiction vulnerability for polysubstance abuse in humans and that Nrcam knockout mice do not develop conditioned place preferences for morphine, cocaine or amphetamine. In order to gain insight into NrCAM involvement in addiction vulnerability, which may involve specific neural circuits underlying behavioral characteristics relevant to addiction, we evaluated several behavioral phenotypes in Nrcam knockout mice. Consistent with a potential general reduction in motivational function, Nrcam knockout mice demonstrated less curiosity for novel objects and for an unfamiliar conspecific, showed also less anxiety in the zero maze. Nrcam heterozygote knockout mice reduced alcohol preference and buried fewer marbles in home cage. These observations provide further support for a role of NrCAM in substance abuse including alcoholism vulnerability, possibly through its effects on behavioral traits that may affect addiction vulnerability, including novelty seeking, obsessive compulsion and responses to aversive or anxiety-provoking stimuli. Additionally, in order to prove glutamate homeostasis hypothesis of addiction, we analyzed glutamatergic molecules regulated by NRCAM expression. Glutaminase appears to be involved in NrCAM-related molecular pathway in two different tissues from human and mouse. An inhibitor of the enzyme, prolyl-leucyl-glycinamide, treatment produced, at least, some of the phenotypes of mice shown in alcohol preference and in anxiety-like behavior. Thus, NrCAM could affect addiction-related behaviors via at least partially modulation of some glutamatergic pathways and neural function in brain.
- Published
- 2012
41. A cis-element in the Notch1 locus is involved in the regulation of gene expression in interneuron progenitors
- Author
-
Ying Li, Sung Tae Doh, Martin Grumet, Shannon M. Smith, Li Cai, Alson Wu, Hailing Hao, and Evangeline Tzatzalos
- Subjects
Embryo, Nonmammalian ,Ganglionic eminence ,Interneuron ,Chick Embryo ,Biology ,Article ,Avian Proteins ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neural Stem Cells ,Cell Movement ,Interneurons ,Gene expression ,medicine ,Animals ,Neural progenitor cells ,Receptor, Notch1 ,Enhancer ,Gene ,Molecular Biology ,Embryonic Stem Cells ,030304 developmental biology ,Genetics ,Regulation of gene expression ,Notch1 ,0303 health sciences ,Reporter gene ,Neurogenesis ,Gene Expression Regulation, Developmental ,Cell Biology ,Embryo, Mammalian ,cis-element ,Cell biology ,medicine.anatomical_structure ,Enhancer Elements, Genetic ,Genetic Loci ,Gene expression Interneuron ,GABAergic ,Chickens ,030217 neurology & neurosurgery ,Developmental Biology ,Protein Binding - Abstract
Interneurons comprise approximately one third of the total cortical neurons in the mammalian cerebral cortex. Studies have revealed many details in the generation of this cell type. However, the mechanism that defines interneuron-lineage specific gene expression is not well understood. Gene regulatory elements, e.g., promoters, enhancers, and trans-acting factors, are essential for the proper control of gene expression. Here, we report that a novel evolutionarily conserved cis-element in the second intron of the Notch1 locus plays an important role in regulating gene expression in interneuron progenitors. The spatiotemporal activity of the cis-element in the developing central nervous system (CNS) was determined by both transient reporter expression in the developing chick and a transgenic mouse model. Its activity is well correlated with neurogenesis in both the chick and mouse and restricted to neural progenitor cells in the ganglionic eminence that are fated to differentiate into GABAergic interneurons of the neocortex. We further demonstrate that the cis-element activity requires the binding motif for trans-acting factors Gsh1/Barx2/Brn3. Deletion of this binding motif abolishes reporter gene expression. Together, these data provide new insights into the regulatory mechanisms of interneuron development in the vertebrate CNS.
- Published
- 2012
42. Isolation of a Novel Rat Neural Progenitor Clone that Expresses Dlx Family Transcription Factors and Gives Rise to Functional GABAergic Neurons in Culture
- Author
-
Ronald P. Hart, Joanne Babiarz, Christopher L. Ricupero, Yu R. Han, Anna T. Hader, Sasha B. Godfrey, Joseph A. Wong, Martin Grumet, Mark R. Plummer, Hedong Li, John P. Corradi, and Myles Fennell
- Subjects
Ganglionic eminence ,Interneuron ,Neurogenesis ,Biology ,Inhibitory postsynaptic potential ,Article ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Neural Stem Cells ,medicine ,Animals ,GABAergic Neurons ,Cells, Cultured ,Cerebral Cortex ,Homeodomain Proteins ,Gene Expression Profiling ,Glutamate receptor ,Bicuculline ,Neural stem cell ,Rats ,medicine.anatomical_structure ,nervous system ,GABAergic ,Neuroscience ,medicine.drug ,Transcription Factors - Abstract
Gamma-aminobutyric acid (GABA) ergic interneurons are lost in conditions including epilepsy and central nervous system injury, but there are few culture models available to study their function. Toward the goal of obtaining renewable sources of GABAergic neurons, we used the molecular profile of a functionally incomplete GABAergic precursor clone to screen 17 new clones isolated from GFP(+) rat E14.5 cortex and ganglionic eminence (GE) that were generated by viral introduction of v-myc. The clones grow as neurospheres in medium with FGF2, and after withdrawal of FGF2, they exhibit varying patterns of differentiation. Transcriptional profiling and quantitative reverse transcriptase polymerase chain reaction (RT-PCR) indicated that one clone (GE6) expresses high levels of mRNAs encoding Dlx1, 2, 5, and 6, glutamate decarboxylases, and presynaptic proteins including neuropeptide Y and somatostatin. Protein expression confirmed that GE6 is a progenitor with restricted differentiation giving rise mostly to neurons with GABAergic markers. In cocultures with hippocampal neurons, GE6 neurons became electrically excitable and received both inhibitory and excitatory synapses. After withdrawal of FGF2 in cultures of GE6 alone, neurons matured to express βIII-tubulin, and staining for synaptophysin and vesicular GABA transporter were robust after 1-2 weeks of differentiation. GE6 neurons also became electrically excitable and displayed synaptic activity, but synaptic currents were carried by chloride and were blocked by bicuculline. The results suggest that the GE6 clone, which is ventrally derived from the GE, resembles GABAergic interneuron progenitors that migrate into the developing forebrain. This is the first report of a relatively stable fetal clone that can be differentiated into GABAergic interneurons with functional synapses.
- Published
- 2012
43. Interactions of the chondroitin sulfate proteoglycan phosphacan, the extracellular domain of a receptor-type protein tyrosine phosphatase, with neurons, glia, and neural cell adhesion molecules
- Author
-
David R. Friedlander, M. Flad, Martin Grumet, Laina Karthikeyan, Renée K. Margolis, Peter Milev, Richard U. Margolis, and Takeshi Sakurai
- Subjects
Cell Adhesion Molecules, Neuronal ,Nerve Tissue Proteins ,Receptors, Cell Surface ,Chick Embryo ,Protein tyrosine phosphatase ,Biology ,Nervous System ,Radioligand Assay ,chemistry.chemical_compound ,Laminin ,Neurocan ,Cell Adhesion ,Neurites ,Animals ,Chondroitin sulfate ,Neurons ,Extracellular Matrix Proteins ,Dose-Response Relationship, Drug ,Receptor-Like Protein Tyrosine Phosphatases, Class 5 ,Cell adhesion molecule ,Tenascin ,Glioma ,Articles ,Cell Biology ,Rats ,Cell biology ,Fibronectin ,Chondroitin Sulfate Proteoglycans ,Biochemistry ,chemistry ,Chondroitin sulfate proteoglycan ,biology.protein ,Neural cell adhesion molecule ,Protein Tyrosine Phosphatases ,Neuroglia ,Protein Binding - Abstract
Phosphacan is a chondroitin sulfate proteoglycan produced by glial cells in the central nervous system, and represents the extracellular domain of a receptor-type protein tyrosine phosphatase (RPTP zeta/beta). We previously demonstrated that soluble phosphacan inhibited the aggregation of microbeads coated with N-CAM or Ng-CAM, and have now found that soluble 125I-phosphacan bound reversibly to these neural cell adhesion molecules, but not to a number of other cell surface and extracellular matrix proteins. The binding was saturable, and Scatchard plots indicated a single high affinity binding site with a Kd of approximately 0.1 nM. Binding was reduced by approximately 15% after chondroitinase treatment, and free chondroitin sulfate was only moderately inhibitory, indicating that the phosphacan core glycoprotein accounts for most of the binding activity. Immunocytochemical studies of embryonic rat spinal phosphacan, Ng-CAM, and N-CAM have overlapping distributions. When dissociated neurons were incubated on dishes coated with combinations of phosphacan and Ng-CAM, neuronal adhesion and neurite growth were inhibited. 125I-phosphacan bound to neurons, and the binding was inhibited by antibodies against Ng-CAM and N-CAM, suggesting that these CAMs are major receptors for phosphacan on neurons. C6 glioma cells, which express phosphacan, adhered to dishes coated with Ng-CAM, and low concentrations of phosphacan inhibited adhesion to Ng-CAM but not to laminin and fibronectin. Our studies suggest that by binding to neural cell adhesion molecules, and possibly also by competing for ligands of the transmembrane phosphatase, phosphacan may play a major role in modulating neuronal and glial adhesion, neurite growth, and signal transduction during the development of the central nervous system.
- Published
- 1994
- Full Text
- View/download PDF
44. Receptor tyrosine phosphatase beta is expressed in the form of proteoglycan and binds to the extracellular matrix protein tenascin
- Author
-
Peter Milev, Gilad Barnea, Olli Silvennoinen, Martin Grumet, Jan Sap, Joseph Schlessinger, and Joan B. Levy
- Subjects
animal structures ,Cell Adhesion Molecules, Neuronal ,Tenascin ,Nerve Tissue Proteins ,Receptors, Cell Surface ,Protein tyrosine phosphatase ,Transfection ,Biochemistry ,Mice ,chemistry.chemical_compound ,Methionine ,Extracellular ,Animals ,Molecular Biology ,Extracellular Matrix Proteins ,biology ,Receptor-Like Protein Tyrosine Phosphatases, Class 5 ,Sulfates ,Brain ,Cell Biology ,Molecular biology ,Transmembrane protein ,Extracellular Matrix ,Alternative Splicing ,Chondroitin Sulfate Proteoglycans ,Proteoglycan ,Receptor-Like Protein Tyrosine Phosphatases ,chemistry ,Chondroitin sulfate proteoglycan ,embryonic structures ,biology.protein ,Proteoglycans ,Protein Tyrosine Phosphatases ,Signal transduction ,Protein Binding - Abstract
The extracellular domain of receptor type protein tyrosine phosphatase beta (RPTP beta) exhibits striking sequence similarity with a soluble, rat brain chondroitin sulfate proteoglycan (3F8 PG). Immunoprecipitation experiments of cells transfected with RPTP beta expression vector and metabolically labeled with [35S]sulfate and [35S]methionine indicate that the transmembrane form of RPTP beta is indeed a chondroitin sulfate proteoglycan. The 3F8 PG is therefore a variant form composed of the entire extracellular domain of RPTP beta probably generated by alternative RNA splicing. Previous immunohistochemical studies indicated that both RPTP beta and the extracellular matrix protein tenascin are localized in similar regions of the central nervous system. We have performed co-aggregation assays with red and green Co-vaspheres coated with tenascin and 3F8 PG, respectively, showing that the extracellular domain of RPTP beta (3F8 PG) binds specifically to tenascin. The interaction between a receptor tyrosine phosphatase and an extracellular matrix protein may have a role in development of the mammalian central nervous system.
- Published
- 1994
- Full Text
- View/download PDF
45. Interactions with tenascin and differential effects on cell adhesion of neurocan and phosphacan, two major chondroitin sulfate proteoglycans of nervous tissue
- Author
-
Mario Bourdon, Richard U. Margolis, Renée K. Margolis, Peter Milev, Takeshi Sakurai, Martin Grumet, and Laina Karthikeyan
- Subjects
animal structures ,Cell Adhesion Molecules, Neuronal ,Tenascin ,Nerve Tissue Proteins ,Biochemistry ,Cell Line ,Extracellular matrix ,chemistry.chemical_compound ,Neurocan ,Cerebellum ,Cell Adhesion ,Tumor Cells, Cultured ,Animals ,Lectican ,Lectins, C-Type ,Chondroitin sulfate ,Molecular Biology ,Neurons ,Extracellular Matrix Proteins ,biology ,Receptor-Like Protein Tyrosine Phosphatases, Class 5 ,Cell adhesion molecule ,Antibodies, Monoclonal ,Brain ,Glioma ,Cell Biology ,musculoskeletal system ,Immunohistochemistry ,Rats ,Cell biology ,Fibronectin ,Kinetics ,Chondroitin Sulfate Proteoglycans ,Proteoglycan ,chemistry ,embryonic structures ,biology.protein - Abstract
We have studied interactions of tenascin with two chondroitin sulfate proteoglycans, neurocan and phosphacan. Neurocan is a multi-domain proteoglycan with a 136-kDa core protein that is synthesized by neurons and binds to hyaluronic acid, whereas the 173-kDa core protein of phosphacan, which is synthesized by glia, represents an extracellular variant of the receptor-type protein tyrosine phosphatase RPTP zeta/beta. Keratan sulfate-containing glycoforms of phosphacan (designated phosphacan-KS) are also present in brain. Immunocytochemical studies of early postnatal rat cerebellum demonstrated that the localization of neurocan, phosphacan, and phosphacan-KS all overlap extensively with that of tenascin, an extracellular matrix protein that modulates cell adhesion and migration. Binding studies using purified proteins covalently attached to fluorescent microbeads demonstrated that proteoglycan-coated beads co-aggregated with differently fluorescing beads coated with tenascin. The co-aggregation was specifically inhibited by Fab' fragments of antibodies against tenascin or the proteoglycans and by soluble neurocan, phosphacan, and tenascin. A solid phase radioligand binding assay confirmed that neurocan, phosphacan, and phosphacan-KS bind to tenascin but not to laminin and fibronectin. Chondroitinase treatment of the proteoglycans or addition of free chondroitin sulfate had no significant effect, indicating that the binding activity is mediated largely via the core glycoproteins. Scatchard analysis demonstrated high affinity binding of 125I-phosphacan, phosphacan-KS, and neurocan to a single site in tenascin, and neurocan and various glycoforms of phosphacan all inhibited binding of 125I-phosphacan to tenascin. In studies of cell adhesion to proteins adsorbed to Petri dishes, phosphacan inhibited adhesion of C6 glioma cells to tenascin whereas neurocan had no effect. Our results suggest that tenascin binds phosphacan and neurocan in vivo and that interactions between chondroitin sulfate proteoglycans and tenascin may play important roles in nervous tissue histogenesis, possibly by modulating signal transduction across the plasma membrane.
- Published
- 1994
- Full Text
- View/download PDF
46. Regions of the Human Neurokinin A Receptor Involved in the Generation of Second Messengers and in Receptor Desensitization
- Author
-
C. R. Cyr, J. P. Gardner, Martin Grumet, S. M. Josiah, R. M. Kris, and Vi Chu
- Subjects
Protein Conformation ,Neurokinin A ,Molecular Sequence Data ,Mutant ,Biophysics ,CHO Cells ,Biology ,Transfection ,Polymerase Chain Reaction ,Second Messenger Systems ,Biochemistry ,chemistry.chemical_compound ,Cricetinae ,Animals ,Humans ,Amino Acid Sequence ,Codon ,Receptor ,Molecular Biology ,DNA Primers ,Sequence Deletion ,chemistry.chemical_classification ,Chinese hamster ovary cell ,Wild type ,Receptors, Neurokinin-2 ,Cell Biology ,Recombinant Proteins ,Amino acid ,Kinetics ,chemistry ,Mutagenesis ,Second messenger system ,Calcium ,Signal transduction ,Signal Transduction - Abstract
Deletion analysis was used to study sites of human Neurokinin A receptor (HNKAR) necessary for signal transduction in CHO cells. Deletion of 62 and 81 amino acids from the c-terminus of HNKAR forms mutant receptors HNKAR delta 62 and HNKAR delta 81, which bind neurokinin A with high affinity but are functionally different. Wild type HNKAR and HNKAR delta 62 are functionally active whereas HNKAR delta 81 is functionally inactive. In addition, HNKAR and HNKAR delta 62 were both desensitized to the neurokinin A signal within 5 minutes. The data indicates: 1) an intact cytoplasmic tail of the HNKAR is not critical for signal transduction, but the n-terminal amino acids of the cytoplasmic tail are necessary for signaling and 2) the c-terminal 62 amino acids are not necessary for desensitization.
- Published
- 1994
- Full Text
- View/download PDF
47. Receptor tyrosine phosphatase R-PTP-kappa mediates homophilic binding
- Author
-
Jan Sap, David R. Friedlander, Joseph Schlessinger, Martin Grumet, and Ying-Ping Jiang
- Subjects
Cell signaling ,DNA, Complementary ,animal structures ,Protein Conformation ,Gene Expression ,Cell Communication ,Protein tyrosine phosphatase ,Immunoglobulin domain ,Biology ,Transfection ,environment and public health ,Cell Line ,Mice ,chemistry.chemical_compound ,Cell–cell interaction ,Cell surface receptor ,Cell Adhesion ,Animals ,Humans ,Tyrosine ,Molecular Biology ,Cell Aggregation ,Receptor-Like Protein Tyrosine Phosphatases, Class 2 ,Tyrosine phosphorylation ,Cell Biology ,Recombinant Proteins ,Cell biology ,enzymes and coenzymes (carbohydrates) ,Drosophila melanogaster ,Receptor-Like Protein Tyrosine Phosphatases ,Biochemistry ,chemistry ,Protein Tyrosine Phosphatases ,Extracellular Space ,Research Article - Abstract
Receptor tyrosine phosphatases (R-PTPases) feature PTPase domains in the context of a receptor-like transmembrane topology. The R-PTPase R-PTP-kappa displays an extracellular domain composed of fibronectin type III motifs, a single immunoglobulin domain, as well as a recently defined MAM domain (Y.-P. Jiang, H. Wang, P. D'Eustachio, J.M. Musacchio, J. Schlessinger, and J. Sap, Mol. Cell. Biol. 13:2942-2951, 1993). We report here that R-PTP-kappa can mediate homophilic intercellular interaction. Inducible expression of the R-PTP-kappa protein in heterologous cells results in formation of stable cellular aggregates strictly consisting of R-PTP-kappa-expressing cells. Moreover, the purified extracellular domain of R-PTP-kappa functions as a substrate for adhesion by cells expressing R-PTP-kappa and induces aggregation of coated synthetic beads. R-PTP-kappa-mediated intercellular adhesion does not require PTPase activity or posttranslational proteolytic cleavage of the R-PTP-kappa protein and is calcium independent. The results suggest that R-PTPases may provide a link between cell-cell contact and cellular signaling events involving tyrosine phosphorylation.
- Published
- 1994
- Full Text
- View/download PDF
48. The neuronal chondroitin sulfate proteoglycan neurocan binds to the neural cell adhesion molecules Ng-CAM/L1/NILE and N-CAM, and inhibits neuronal adhesion and neurite outgrowth
- Author
-
Martin Grumet, Richard U. Margolis, David R. Friedlander, Peter Milev, Renée K. Margolis, and Laina Karthikeyan
- Subjects
Neurite ,Cell Adhesion Molecules, Neuronal ,Nerve Tissue Proteins ,Chick Embryo ,Biology ,chemistry.chemical_compound ,Neurocan ,Cell Adhesion ,Neurites ,Lectican ,Animals ,Lectins, C-Type ,Chondroitin sulfate ,Aggrecans ,Cell adhesion ,Neurons ,Extracellular Matrix Proteins ,Cell adhesion molecule ,Brain ,Cell Differentiation ,Tenascin ,Cell Biology ,Articles ,Cell biology ,Rats ,chemistry ,Biochemistry ,Chondroitin Sulfate Proteoglycans ,Chondroitin sulfate proteoglycan ,Neural cell adhesion molecule ,Proteoglycans ,Collagen ,Protein Binding - Abstract
We have previously shown that aggregation of microbeads coated with N-CAM and Ng-CAM is inhibited by incubation with soluble neurocan, a chondroitin sulfate proteoglycan of brain, suggesting that neurocan binds to these cell adhesion molecules (Grumet, M., A. Flaccus, and R. U. Margolis. 1993. J. Cell Biol. 120:815). To investigate these interactions more directly, we have tested binding of soluble 125I-neurocan to microwells coated with different glycoproteins. Neurocan bound at high levels to Ng-CAM and N-CAM, but little or no binding was detected to myelin-associated glycoprotein, EGF receptor, fibronectin, laminin, and collagen IV. The binding to Ng-CAM and N-CAM was saturable and in each case Scatchard plots indicated a high affinity binding site with a dissociation constant of approximately 1 nM. Binding was significantly reduced after treatment of neurocan with chondroitinase, and free chondroitin sulfate inhibited binding of neurocan to Ng-CAM and N-CAM. These results indicate a role for chondroitin sulfate in this process, although the core glycoprotein also has binding activity. The COOH-terminal half of neurocan was shown to have binding properties essentially identical to those of the full-length proteoglycan. To study the potential biological functions of neurocan, its effects on neuronal adhesion and neurite growth were analyzed. When neurons were incubated on dishes coated with different combinations of neurocan and Ng-CAM, neuronal adhesion and neurite extension were inhibited. Experiments using anti-Ng-CAM antibodies as a substrate also indicate that neurocan has a direct inhibitory effect on neuronal adhesion and neurite growth. Immunoperoxidase staining of tissue sections showed that neurocan, Ng-CAM, and N-CAM are all present at highest concentration in the molecular layer and fiber tracts of developing cerebellum. The overlapping localization in vivo, the molecular binding studies, and the striking effects on neuronal adhesion and neurite growth support the view that neurocan may modulate neuronal adhesion and neurite growth during development by binding to neural cell adhesion molecules.
- Published
- 1994
49. Encapsulated Mesenchymal Stromal Cells for In-Vivo Transplantation
- Author
-
Martin Grumet, Martin L. Yarmush, Seiji Otsuka, Jeffrey Barminko, Andrea Gray, Jae Hwan Kim, and Rene S. Schloss
- Subjects
Alginates ,Cell Survival ,Population ,Gene Expression ,Nitric Oxide Synthase Type II ,Bioengineering ,Applied Microbiology and Biotechnology ,Article ,Glucuronic Acid ,In vivo ,medicine ,Macrophage ,Animals ,Humans ,education ,Spinal cord injury ,Spinal Cord Injuries ,education.field_of_study ,Transplantation ,business.industry ,Histocytochemistry ,Gene Expression Profiling ,Hexuronic Acids ,Macrophages ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cells, Immobilized ,M2 Macrophage ,medicine.disease ,equipment and supplies ,In vitro ,Microspheres ,Rats ,Disease Models, Animal ,Immunology ,Cancer research ,Cytokines ,business ,Biotechnology - Abstract
Immunomodulatory human mesenchymal stromal cells (hMSC) have been incorporated into therapeutic protocols to treat secondary inflammatory responses post-spinal cord injury (SCI) in animal models. However, limitations with direct hMSC implantation approaches may prevent effective translation for therapeutic development of hMSC infusion into post-SCI treatment protocols. To circumvent these limitations, we investigated the efficacy of alginate microencapsulation in developing an implantable vehicle for hMSC delivery. Viability and secretory function were maintained within the encapsulated hMSC population, and hMSC secreted anti-inflammatory cytokines upon induction with the pro-inflammatory factors, TNF-α and IFN-γ. Furthermore, encapsulated hMSC modulated inflammatory macrophage function both in-vitro and in-vivo, even in the absence of direct hMSC-macrophage cell contact and promoted the alternative M2 macrophage phenotype. In-vitro, this was evident by a reduction in macrophage iNOS expression with a concomitant increase in CD206, a marker for M2 macrophages. Finally, Sprague-Dawley rat spinal cords were injured at vertebra T10 via a weight drop model (NYU model) and encapsulated hMSC were administered via lumbar puncture 24 hours post- injury. Encapsulated hMSC localized primarily in the cauda equina of the spinal cord. Histological assessment of spinal cord tissue 7 days post SCI indicated that as few as 5×104 encapsulated hMSC yielded increased numbers of CD206-expressing macrophages, consistent with our in-vitro studies. The combined findings support the inclusion of immobilized hMSC in post-CNS trauma tissue protective therapy, and suggest that conversion of macrophages to the M2 subset is responsible, at least in part, for tissue protection.
- Published
- 2011
50. NrCAM Deletion Causes Topographic Mistargeting of Thalamocortical Axons to the Visual Cortex and Disrupts Visual Acuity
- Author
-
Galina P. Demyanenko, Patricia F. Maness, Eli P B Darnell, Jasbir S. Dalal, Thorfinn T. Riday, Tracy S. Tran, Benjamin D. Philpot, Martin Grumet, Leann H. Brennaman, and Takeshi Sakurai
- Subjects
Male ,genetic structures ,L1 family ,Thalamus ,Growth Cones ,Visual Acuity ,Nerve Tissue Proteins ,Biology ,Somatosensory system ,Article ,Visual processing ,Mice ,medicine ,Animals ,Axon ,Visual Cortex ,Mice, Knockout ,Cerebrum ,General Neuroscience ,Motor Cortex ,Membrane Proteins ,Somatosensory Cortex ,Axons ,Neuropilin-2 ,medicine.anatomical_structure ,Visual cortex ,Evoked Potentials, Visual ,Female ,Neuroscience ,Cell Adhesion Molecules ,Motor cortex - Abstract
NrCAM is a neural cell adhesion molecule of the L1 family that has been linked to autism spectrum disorders (ASDs), a disease spectrum in which abnormal thalamocortical connectivity may contribute to visual processing defects. Here we show that NrCAM interaction with Neuropilin-2 (Npn-2) is critical for Semaphorin3F (Sema3F)-induced guidance of thalamocortical axon subpopulations at the ventral telencephalon (VTe), an intermediate target for thalamic axon sorting. Genetic deletion of NrCAM or Npn-2 caused contingents of embryonic thalamic axons to misproject caudally in the VTe, away from a caudal-high Sema3F gradient. The resultant thalamocortical map of NrCAM null mutants showed striking mistargeting of motor and somatosensory thalamic axon contingents to the primary visual cortex, but retino-geniculate targeting and segregation were normal. NrCAM formed a molecular complex with Npn-2 in brain and neural cells, and was required for Sema3F-induced growth cone collapse in thalamic neuron cultures, consistent with a vital function for NrCAM in Sema3F-induced axon repulsion. NrCAM null mice displayed reduced responses to visual evoked potentials (VEPs) recorded from layer IV in the binocular zone of primary visual cortex (V1), particularly when evoked from the ipsilateral eye, indicating abnormal visual acuity and ocularity. These results demonstrate that NrCAM is required for normal maturation of cortical visual acuity, and suggest that the aberrant projection of thalamic motor and somatosensory axons to the visual cortex in NrCAM null mutant mice impairs cortical functions.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.