CA2322747A1 - Human spinal cord cell lines and methods of use therefor - Google Patents

Human spinal cord cell lines and methods of use therefor Download PDF

Info

Publication number
CA2322747A1
CA2322747A1 CA002322747A CA2322747A CA2322747A1 CA 2322747 A1 CA2322747 A1 CA 2322747A1 CA 002322747 A CA002322747 A CA 002322747A CA 2322747 A CA2322747 A CA 2322747A CA 2322747 A1 CA2322747 A1 CA 2322747A1
Authority
CA
Canada
Prior art keywords
cell
spinal cord
cells
human spinal
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002322747A
Other languages
French (fr)
Inventor
Ronghao Li
Dinah W.Y. Sah
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Signal Pharmaceuticals LLC
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2322747A1 publication Critical patent/CA2322747A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0623Stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2503/00Use of cells in diagnostics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/04Immortalised cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Neurology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Neurosurgery (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Developmental Biology & Embryology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Conditionally-immortalized human spinal cord cell lines are provided. Such cell lines, which may be clonal, may be used to generate neurons, including motoneurons. The cell lines and/or differentiated cells may be used for the development of therapeutic agents to prevent and treat a variety of spinal cord-related diseases and injuries. The cell lines and/or differentiated cells may also be used in assays and for the general study of spinal cord cell development and differentiation.

Description

Description HUMAN SPINAL CORD CELL LINES AND METHODS OF USE THEREFOR
TECHNICAL FIELD
The present invention relates generally to human spinal cord cell lines. The invention is more particularly related to conditionally-immortalized spinal cord cell lines capable of differentiation into neurons, and to differentiated cells derived from such cell lines.
Such cell lines and/or differentiated cells may be used in the development of therapeutic agents for the prevention and treatment of spinal cord-related diseases and injuries, as well as pain. The present invention is also related to the use of such cell lines and/or differentiated cells within various assays and for the study of spinal cord cell development and differentiation.
BACKGROUND OF THE INVENTION
The spinal cord plays a critical role in the functioning of the central nervous system (CNS). Although the spinal cord is the simplest region of the CNS, it contains diverse neural cell types which are interconnected in intricate patterns. During embryonic development, multi-potent stem cells in the neural tube proliferate and then ultimately differentiate into neurons and glia according to extrinsic cues and intrinsic determinants.
Diseases and injuries that affect the functioning of spinal cord neural cells are often debilitating, and generally remain difficult to treat effectively.
To develop improved therapies for such disorders, further study of human spinal cord neurons and the differentiation process is needed. To date, such study has been hampered by the difficulty of obtaining sufficient primary human CNS tissue for research and the limited lifespan of primary cultures. In fact, research to date has failed to even demonstrate the existence of neuronal-restricted precursor cells in the human fetal spinal cord. As a result of the difficulties associated with performing human spinal cord research, the differentiation process remains poorly understood and the development of therapies for spinal cord diseases and injuries has been impeded.
2 Accordingly, there is a need in the art for stable spinal cord lines that can be readily differentiated, and can be used in the development of therapies for spinal cord-related conditions. The present invention fulfills these needs and further provides other related advantages.
SUMMARY OF THE INVENTION
Briefly stated, the present invention provides conditionally-immortalized human spinal cord cell lines capable of differentiation into neurons. In one aspect, the present invention provides methods for producing a conditionally-immortalized human spinal cord neural precursor cell, comprising the steps of: (a) transfecting human spinal cord cells plated on a first surface and in a first growth medium that permit proliferation with DNA encoding a selectable marker and an externally regulatable growth-promoting gene; and (b) selecting the transfected cells on a second surface and in a second growth medium that permit attachment and proliferation, and therefrom producing a conditionally-immortalized human spinal cord neural precursor cell. Within certain embodiments, the first and second surfaces are independently selected from the group consisting of substrates comprising one or more of a polyamino acid (e.g., polylysine or polyornithine), fibronectin, laminin or tissue culture plastic. Similarly, in certain embodiments, the growth-promoting gene may be an oncogene, such as v-myc, and expression of the growth-promoting gene may, but need not, be inhibited by tetracycline.
Within other aspects, the present invention provides conditionally-immortalized human spinal cord neural precursor cells capable of differentiation into neurons such as motoneurons.
The present invention further provides methods for producing neurons, comprising culturing a conditionally-immortalized human spinal cord neural precursor cell as described above under conditions inhibiting expression of the growth-promoting gene.
Within certain embodiments, the cell may be cultured in medium comprising tetracycline and/or in the presence of one or more differentiating agents such as NGF, CNTF
and BDNF.
Within related aspects, the present invention provides neurons, such as motoneurons, produced as described above.
3 PCTNS99/04535 The present invention further provides methods for transplanting a human spinal cord cell into a mammal, comprising administering to a mammal a cell, including neurons, produced as described above. Within related aspects, methods are provided for treating a spinal cord-related condition in a patient, comprising administering to a patient a cell, including neurons, produced as described above.
In other aspects, methods are provided for screening for an agent that modulates an activity of a protein produced by a human spinal cord cell, comprising: (a) contacting a cell produced as described above with a candidate agent; and (b) subsequently measuring the ability of the candidate agent to modulate an activity of a protein produced by the cell.
Within further aspects, the present invention provides methods for detecting the presence or absence of a protein in a sample, comprising: (a) contacting a sample with a cell produced as described above; and (b) subsequently detecting a response in the cell, and therefrom detecting the presence of a protein in the sample.
The present invention also provides methods for identifying a human spinal cord gene or protein, comprising detecting the presence of a gene or protein within a culture of cells produced as described above.
In further aspects, methods are provided for screening for an agent that affects human spinal cord cell death, comprising: (a) contacting a cell produced as described above with a candidate agent under conditions that, in the absence of candidate agent, result in death of the cell; and (b) subsequently measuring the ability of the candidate agent to affect the death of the cell.
The present invention also provides methods for screening for a protein that regulates human spinal cord cell death, comprising: (a) altering the level of expression of a protein within a cell produced as described above; and (b) subsequently measuring the effect of the alteration on the death of the cell, and therefrom identifying a protein that regulates human spinal cord neural precursor cell death.
Within still other aspects, the present invention provides conditionally immortalized human spinal cord neural precursor cells produced as described above. Such cells may be present within a clonal cell line.
4 These and other aspects of the present invention will become apparent upon reference to the following detailed description and attached drawings. All references disclosed herein are hereby incorporated by reference in their entirety as if each was incorporated individually.
BRIEF DESCRIPTION OF THE DRAWINGS
Figures 1 A and 1 B are live phase contrast micrographs of immortalized human spinal cord cells. Figure lA shows the cells after growth in proliferative conditions. Figure 1 B shows the cells after 5 days of differentiation with DMEM/F 12 medium containing N2 supplements, chick embryo extract (0.3%), laminin (5 pg/ml), tetracycline (1 p,g/ml), CNTF
(ciliary neurotrophic factor; 10 ng/ml) and NGF (nerve growth factor; 100 ng/ml).
Figure 2 is a set of live phase contrast micrographs (phase) and photographs showing labeling for v-myc (v-myc) in representative immortalized human spinal cord clonal cell lines HSP1, HSP2 and HSP4, as indicated. The clones HSPI, HSP2 and HSP4 were cultured for 3 days in the proliferative growth condition with (+Tc) or without (-Tc) daily tetracycline (1 pg/ml) administration, as indicated, before being processed for v-myc immunofluorescence. For each field, a phase photograph (post-fixation) is shown immediately above the corresponding immunofluorescence photograph.
Figures 3A-3D are graphs showing the effects of FGF-2 (basic fibroblast growth factor), EGF (epidermal growth factor) and fibronectin on the proliferation of the representative clones HSP1 (Figures 3A and 3B) and HSP2 (Figures 3C and 3D).
In Figures 3A and 3C, the cells were plated in DMEM/F12 medium containing N2 supplements, fibronectin (5 p,g/ml) and chick embryo extract (0.3%). FGF-2 or EGF was added at the concentrations indicated. In Figures 3B and 3D, the cells were plated in DMEM/F12 medium containing N2 supplements, FGF-2 (50 ng/ml), EGF (50 ng/ml) and chick embryo extract (0.3%) in 24-well plates pre-coated with fibronectin at the concentrations indicated. HSPI
was plated at a density of 2x103 cells/well and the cell number was counted on day 4. HSP2 was plated at a density of 2x104 cells/well and the cell number was counted on day 3. Data are shown as the mean ~ standard deviation of 4 replicates, with the Student's t-test for WO 99!45103 PCT/US99l04535 significance indicated for differences between cultures grown in the presence and absence of growth factor (a: p < 0.05; b: p < 0.01 ).
Figure 4 is a series of photographs showing immunofluorescence labeling for NCAM (neural cell adhesion molecule) and NF200 (neurofilament 200 kD) in the
5 representative immortalized human spinal cord clonal cell lines HSP1, HSP2 and HSP4, as indicated. For each cell line, a phase photograph (post-fixation) is shown to the left of the corresponding immunofluorescence photographs, as indicated.
Figure 5 depicts an electrophysiological recording of an action potential in an HSP1 cell after differentiation for 4 days. The top trace shows the injected current pulse whereas the lower trace shows the membrane potential of the cell. The dashed line indicates zero membrane potential.
Figures 6A-6D are photographs showing the immunoreactivities for cell-type specific markers in representative clone HSP2 after 2 weeks of differentiation. Figures 6A
and 6B show labeling for GFAP (glial fibrillary acidic protein; Figure 6A) and A2B5 (6B) in the same field. Figure 6C shows immunofluorescence for A2B5 in differentiated neurons in a different field. Figure 6D shows double-labeling for GFAP (FITC) and neurofilament 200kD
(Texas Red).
Figures 7A-7D are photographs illustrating the immunoreactivities for tau, MAP2a&b (microtubule associated protein), peripherin and synaptophysin, as indicated, in representative clone HSP1 after 17 days of differentiation.
Figures 8A-8D are photographs illustrating the immunoreactivities for NGFR
(low affinity nerve growth factor receptor), peripherin, NGFR/Islet-1 (homeodomain transcription factor) and ChAT (choline acetyltransferase), as indicated, in representative clone HSP1 after 3, 3, 15 or 17 days respectively.
Figure 9 is a set of photographs illustrating the immunoreactivities for ChAT, glutamate and GABA (y-aminobutyric acid), as indicated, in representative clone HSP 1 after 7 days of differentiation. For each field, a phase photograph (post-fixation) is shown immediately to the right of the corresponding immunostaining photograph.
Examples of positively- and negatively-stained cells are indicated by arrows and arrowheads, respectively.
6 DETAILED DESCRIPTION OF THE INVENTION
As noted above, the present invention is generally directed to conditionally-immortalized human spinal cord cell lines, differentiated cells generated from such cell lines and various methods employing such cells. In particular, the present invention is directed to conditionally-immortalized human spinal cord neural precursor cells that are capable of differentiation into neurons, and to the use of such cells for drug discovery and development, transplantation studies, therapeutic methods and a variety of assays.
Conditionally-immortalized human spinal cord neural precursor cell lines of the present invention may, but need not, be clonal cell lines. The cell lines described herein provide an infinite, renewable supply of homogeneous cells and facilitate drug development for spinal cord disorders.
Conditionally-immortalized human spinal cord neural precursor cells may generally be prepared from human spinal cord tissue (e.g., fetal spinal cord tissue). Such tissue is preferably dissected from the spinal column, with the meninges and peripheral nerves removed. Spinal cord tissue is then dissociated, washed and plated on a surface and in 1 S a growth medium that permits proliferation (i. e., the surface and medium permit at least about 1 % of the cells to double in a 24 hour period). One preferred growth medium contains DMEM/F-12 with N2 and B27 supplements (GIBCO, Baltimore, MD), FGF-2 (human recombinant, 50 ng/ml, Boehringer Mannheim, Indianapolis, IN), EGF (human recombinant, 50 ng/ml, GIBCO, Baltimore, MD), IGF-I (human recombinant insulin-like growth factor, 10 ng/ml, R & D Systems), PDGF (human recombinant platelet-derived growth factor, 10 ng/ml, Boehringer Mannheim, Indianapolis, IN), chick embryo extract (0.3%, GIBCO, Baltimore, MD, ultrafiltered), and bovine plasma fibronectin (5 ~g/ml, Sigma, St. Louis, MO). Suitable surfaces include, but are not limited to, one or a combination of polyamino acids (e.g., polylysine and/or polyornithine), tissue culture plastic and surfaces treated with laminin or fibronectin. Cells may generally be plated at a density ranging from 103 to 105 cells/cm2, preferably at a density of approximately 104 cells/cm2.
Human spinal cord neural precursor cells may be conditionally immortalized by transfection of the plated cells with any suitable vector containing a growth-promoting gene (i.e., a gene encoding a protein that, under appropriate conditions, promotes growth of the transfected cell) such that the production and/or activity of the growth-promoting protein WO 99/45103 PC"T/US99/04535
7 is regulatable by an external factor. In a preferred embodiment the growth-promoting gene is an oncogene such as, but not limited to, v-myc. Other oncogenes that may be used as growth promoting genes include N-myc, c-myc, p53, SV40 large T antigen, polyoma large T antigen, E 1 a adenovirus and E7 protein of human papillomavirus. In general, a "growth promoting gene" is any gene that, when employed within a tet-controlled expression system as described herein, results in the generation of cultures of neural precursor cells which can be differentiated into neurons and motoneurons.
External regulation of the growth-promoting protein may be achieved by placing the growth-promoting gene under the control of an externally-regulatable promoter (i.e., a promoter whose activity may be controlled by, for example, modifying the temperature of the transfected cells or the composition of the medium in contact with the cells). In general, regulation of expression of the growth-promoting gene should be relatively tight (i.e., expression of the growth-promoting gene should generally be undetectable by immunofluorescent techniques as described herein when the promoter is repressed. For example, a tetracycline (tet)-controlled gene expression system may be employed (see Gossen et al., Proc. Natl. Acad. Sci. USA 89:5547-5551, 1992; Hoshimaru et al., Proc.
Natl. Acad.
Sci. USA 93:1518-1523, 1996). In the absence of tet, a tet-controlled transactivator (tTA) within this vector strongly activates transcription from ph~MV*-1, a minimal promoter from human cytomegalovirus fused to tet operator sequences. tTA is a fusion protein of the repressor (tetR) of the transposon-10-derived tet resistance operon of E. coli and the acidic domain of VP16 of herpes simplex virus. Low, non-toxic concentrations of tet (0.01-1.0 pg/mL) almost completely abolish transactivation by tTA (i.e., v-myc is no longer detectable using an immunofluorescence assay as provided herein).
In a preferred embodiment, the vector further contains a gene encoding a selectable marker (e.g., a protein that confers drug resistance). The bacterial neomycin resistance gene (neon) is one such marker that may be employed within the present invention.
Cells carrying neon may be selected by means known to those of ordinary skill in the art, such as the addition of 100-200 p.g/mL 6418 to the growth medium. It will be readily apparent that other markers may be employed, and appropriate selections may be readily performed by those of ordinary skill in the art.
8 Transfection may be achieved by any of a variety of means known to those of ordinary skill in the art including, but not limited to, retroviral infection.
In general, plated cells may be transfected by infection with a suitable retrovirus (e.g., VSV-G
pseudotyped LINX v-myc retrovirus, as described further below). The use of VSV-G
pseudotyped S retrovirus is preferred, in order to obtain higher stock concentrations of virus, to obtain stocks in the medium of choice (after centrifugation) and to increase the infectivity of human cells.
Recently developed (nontraditional) VSV-G pseudotyped retroviral vectors may be especially useful for the infection of human cells, since the receptor for the VSV-glycoprotein is more abundant and less species-specific than the receptors for traditional amphotropic envelope proteins. Moreover, VSV-G pseudotyped viral particles have been reported to withstand ultracentrifugation, allowing concentration of virus and resuspension in growth medium compatible with neural progenitor cell growth (Burns et al., Proc. Natl. Acad.
Sci. USA
90:8033-8037, 1993).
For example, a human spinal cord neural precursor cell culture prepared as described above may be infected within two days after plating by incubation for about 12-24 hours (e.g., overnight) with retrovirus in the presence of polybrene (4-8 pg/mL). Retrovirus may then be removed by washing with fresh growth medium. Transfected cells carrying a selectable marker may generally be selected on a surface and in a growth medium that permit attachment and proliferation. Proliferation may be assessed as described above. The ability of a surface to permit attachment may be determined using visual microscopic inspection. In general, at least about 20% of the cells should adhere to the surface. One preferred growth medium contains DMEM/F-12 with N2 and B27 supplements (GIBCO, Baltimore, MD), FGF-2 (human recombinant, 50 ng/ml, Boehringer Mannheim, Indianapolis, IN), EGF
(human recombinant, 50 ng/ml, GIBCO, Baltimore, MD), IGF-I (human recombinant insulin-like growth factor, 10 ng/ml, R & D Systems), PDGF (human recombinant platelet-derived growth factor, 10 ng/ml, Boehringer Mannheim, Indianapolis, IN), chick embryo extract (0.3%, GIBCO, Baltimore, MD, ultrafiltered), and bovine plasma fibronectin (5 wg/ml, Sigma, St. Louis, MO). Suitable surfaces include, but are not limited to, one or a combination of a polyamino acid (e.g., polylysine and/or polyornithine), tissue culture plastic and surfaces treated with laminin or fibronectin, as described above, and should grow as an
9 adherent monolayer. The majority of the cells are generally polyconal, with very short processes.
Following transfection, cultures may be maintained in a simplified growth medium containing, for example, DMEM/F-12 with N2 supplement, FGF-2 (50 ng/ml), EGF
(50 ng/ml), chick embryo extract (0.3%), and bovine plasma fibronectin (5 p,g/ml). Cultures approaching confluence may be passaged by trypsinization and split 1:5.
Typically, one T75 flask near confluence may yield 10' cells, and cultures may be passaged every 3 to 4 days.
Cells may also be frozen in liquid nitrogen for long-term storage.
Clonal cell lines may be isolated from a conditionally-immortalized human spinal cord neural precursor cell line prepared as described above. In general, such clonal cell lines may be isolated using standard techniques, such as by limit dilution or using cloning rings, and expanded. Clonal cell lines may generally be fed and passaged as described above.
Genomic Southern blots may be performed to confirm clonality.
Conditionally-immortalized human spinal cord neural precursor cell lines (which may, but need not, be clonal) may generally be induced to differentiate into neurons (and, in some instances, glia) by inhibiting the expression of the growth-promoting gene (i.e., suppressing the production and/or activity of the growth-promoting protein).
For example, if the gene encoding the growth-promoting protein is under the control of an externally-regulatable promoter, the conditions (e.g., temperature or composition of medium) may be modified to suppress transcription of the growth-promoting gene. For the tetracycline-controlled gene expression system discussed above, differentiation may be achieved by the addition of tetracycline to suppress transcription of the growth-promoting gene. In general, 1-S p.g/mL tetracycline for 48 hours is sufficient to begin neuronal morphological differentiation, and the number of differentiated neurons increases during subsequent days.
Such differentiation may be performed, for example, by plating the cells on a suitable substrate (e.g., laminin or tissue culture plastic) in a medium consisting of containing N2 supplements, chick embryo extract (0.3%), natural mouse laminin (5 pg/ml), FGF-2 ( 1-10 ng/ml) and tetracycline ( 1-5 p,g/ml). In addition, agents that raise cyclic AMP, such as forskolin (10 pM) and/or neurotrophic factors may be included in the medium to enhance motoneuron differentiation (e.g., CNTF (10 ng/ml), NGF (100 ng/ml) and BDNF

(brain-derived neurotrophic factor; 10 ng/ml)). Culture medium may then be refreshed (e.g., every 5 days) using medium without FGF-2.
Characterization of both progenitor and differentiated cell lines may generally be performed using techniques well known to those of ordinary skill in the art, including 5 morphological analysis of cell type, immunocytochemistry and PCR (to identify cell type specific markers and receptors and to confirm the presence of the growth-promoting gene) and electrophysiological analysis of voltage- and ligand-gated currents.
Briefly, neuronal cells may be identified morphologically based on the presence of phase bright cell bodies and long, thin processes. Neuronal markers include NCAM, (3III-tubulin and neurofilament (160
10 and 200 kD). Glial markers include GFAP (astrocyte) and the oligodendrocyte markers O1, 04 and GaIC (galactose cerebroside). The presence or absence of such markers may be readily determined using standard immunofluorescence techniques (employing, for example, commercially available primary antibodies and fluorescent reagents) and the levels of mRNA
encoding such markers may be determined using PCR or hybridization techniques.
Electrophysiological analyses familiar to those of ordinary skill in the art may be employed to evaluate the ability of the cells to fire action potentials and to express sodium, calcium and potassium currents, as well as ligand-gated currents (e.g., N-methyl-D-aspartate (NMDA), kainate (KA) and y-amino-n-butyric acid (GABA)), thereby determining the levels of functional channels and receptors.
Within certain aspects, the conditionally-immortalized human spinal cord neural precursor cell lines described herein may be introduced into a mammal for transplantation studies or for treatment of a patient. For example, cells may be introduced by grafting into the nervous system of animals such as rats, mice or monkeys.
Studies may address the differentiation of the cells when transplanted into the developing or adult spinal cord or brain. The ability of the cells to serve as therapeutic agents in pathological conditions can also be examined. In particular, the cells themselves may have the capacity to functionally replace neurons that die in neurodegenerative disorders or due to injury, or may serve as sources of agents (such as trophic factors) that have therapeutic benefit. Such agents may be produced by endogenous genes or by genes transfected into the cells.
11 For treatment of a patient, conditionally-immortalized human spinal cord neural precursor cell lines and/or modulating agents (which may inhibit or enhance an activity of a protein produced by a human spinal cord neural precursor cell, as described below) may be administered to a patient (either prophylactically or for treatment of an S existing condition). Conditions that may be prevented and/or treated using such cells and/or modulating agents include, but are not limited to, motoneuron diseases (e.g., amyotrophic lateral sclerosis and spinal muscular atrophy), myelin-related diseases (e.g., multiple sclerosis), spinal cord injuries and pain. Cells may generally be introduced into the patient (i.e., transplanted) as described above. Modulating agents may be administered by any of a variety of routes known to those of ordinary skill in the art. Such agents may be administered in their active form, as prodrugs (i.e., compounds that are converted to the active form within the patient) or as nucleic acid sequences encoding the modulating agent or prodrug.
Conditionally-immortalized human spinal cord neural precursor cells for use in this aspect of the present invention may, but need not, be further transfected such that they express one or more additional proteins (such as modulating agents) within the patient.
For administration to a patient, one or more conditionally-immortalized human spinal cord neural precursor cells (and/or modulating agents) are generally formulated as a pharmaceutical composition. A pharmaceutical composition may be a sterile aqueous or non-aqueous solution, suspension or emulsion, which additionally comprises a physiologically acceptable carrier (i. e., a non-toxic material that does not interfere with the activity of the active ingredient). Any suitable Garner known to those of ordinary skill in the art may be employed in the pharmaceutical compositions of the present invention. The selection of a carrier will depend, in part, on the nature of the substance (i.e., cells or chemical compounds) being administered. Representative carriers include physiological saline solutions, gelatin, water, alcohols, natural or synthetic oils, saccharide solutions, glycols, injectable organic esters such as ethyl oleate or a combination of such materials. Optionally, a pharmaceutical composition may additionally contain preservatives and/or other additives such as, for example, antimicrobial agents, anti-oxidants, chelating agents and/or inert gases, and/or other active ingredients.
12 Routes and frequency of administration, as well as doses, will vary from patient to patient and on the nature of the substance being administered. In general, the pharmaceutical compositions may be administered intravenously, intramuscularly, subcutaneously or intracavity. Doses are preferably administered daily. A
suitable dose is an S amount that is sufficient to show improvement in the symptoms of a patient afflicted with a spinal cord-related condition or to inhibit the onset of such a condition.
Symptom improvement may be detected based on improvement and/or delay in clinical symptoms associated with the disease. In general, the amount of modulating agent present in a dose, or produced in situ by DNA present in a dose, ranges from about 1 mg to about 200 mg per kg of host. Suitable dose sizes will vary with the size of the patient, but will typically range from about 0.1 mL to about SmL for 10-60 kg animal.
Within certain aspects of the present invention, conditionally-immortalized human spinal cord neural precursor cell lines may be used in a variety of in vitro assays and screens. It will be apparent that the cell lines described herein may be used within numerous well known assays and screens, and that specific parameters and criteria for performing these methods will depend upon the assay being performed. Those of ordinary skill in the art can readily design specific assays and screens based upon well known methods and upon the desired properties of the compounds to be identified.
Within certain assays, the differentiated or undifferentiated conditionally-immortalized human spinal cord neural precursor cell lines described herein may be used to detect the presence or absence of a nucleic acid molecule or protein of interest in such cells.
To detect a particular nucleic acid sequence (i. e., DNA and/or RNA) within such cells, the well known methods of PCR and/or various hybridization techniques may be employed.
Such assays may be readily designed and performed using methods described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratories, Cold Spring Harbor, NY, 1989. To detect a protein, the detection reagent is typically an antibody, which may be prepared as described below. There are a variety of assay formats known to those of ordinary skill in the art for using an antibody to detect a protein in a sample. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988. Antibodies for use in such assays may be polyclonal or
13 monoclonal. Antibodies may be prepared by any of a variety of techniques known to those of ordinary skill in the art and monoclonal antibodies specific for a protein of interest may be prepared, for example, using the technique of Kohler and Milstein, Eur. J.
Immunol. 6:51 I -519, 1976, and improvements thereto. Alternatively, a protein may be detected based upon its activity, using any suitable assay known in the art.
Such assays may generally be used, for example, within methods to evaluate the ability of an agent to modulate an activity of a protein produced by a human spinal cord neural precursor cell or a neuron. Within such assays, differentiated or undifferentiated cells may be contacted with a candidate agent under conditions and for a time sufficient to permit an agent to modulate protein activity. Following contact, the ability of the candidate agent to modulate an activity of a protein produced by the cell is measured using standard techniques, such as PCR or hybridization (for evaluating levels of mRNA) or any of a variety of immunoassays or functional assays appropriate for the protein of interest. For example, calcium-sensitive or voltage-sensitive dye coupled assays, cAMP measurements and/or receptor binding assays may be employed to evaluate the effect of a candidate modulating agent. In general, a suitable amount of antibody or other agent for use in such a screen will vary depending on the particular protein, but will range from about 10 pg to about 100 mg.
The term "modulation," as used herein, includes the suppression or enhancement of an activity of a protein of interest. Such modulation may occur at the transcriptional or translational level, or may be the result of altering the activity of the intact protein. Modulation of protein activity may be direct (i. e., the modulating agent may interact directly with the protein of interest) or rnay be indirect (i. e., the modulating agent may alter the expression and/or activity of one or more other proteins which in turn modulate the activity of the protein of interest). Modulating agents may be antibodies (e.g., monoclonal), polynucleotides or other drugs. Agents that modulate the activity of any cellular protein may be identified within such screens. In certain embodiments, modulating agents may be identified for proteins such as neurotransmitter receptors (e.g., AMPA-preferring receptors, kainate receptors, GABA receptors, adenosine receptors and/or 5-HT receptors), growth factor receptors (e.g., receptors for FGF-2, EGF, BDNF, NGF, CNTF, NT-3 and/or GDNF) or ion channels (e.g., sodium channels, calcium channels and/or potassium channels).
14 Preferred modulating agents are able to suppress or enhance the activity of a protein by at least a factor of 2.
In another aspect of the present invention, the cell lines described herein may be used within a system for studying protein and/or gene expression in a native cellular S environment. For example, receptor expression and/or activity may be assayed, and the effect of various modifications on such expression and/or activity may be evaluated using methods well known to those of ordinary skill in the art. In one such method, cell lines may be permanently or transiently transfected with one or more genes of interest such as, but not limited to, genes that produce or modify membrane proteins, secreted proteins or intracellular proteins of interest. Such genes include ion channels, neurotransmitter receptors, (3-amyloid and/or MAP kinases. The transfected genes may also be coupled to reporter genes for use in drug development. Within this and other aspects described herein, conditionally-immortalized human spinal cord neural precursor cells may be employed without differentiation, or differentiated cells may be used. In addition, cells of varying ages and grown in any of a variety of conditions may be employed. The cell lines of the present invention have many advantages over existing cell lines for such studies, including the ability to provide clonal cell lines capable of producing neurons, and the property of conditional-immortalization, which allows arrest at specific stages of development. The selection of particular cells for any given study will depend on the goals of the study, and those of ordinary skill in the art will be readily able to prepare appropriate cells using the techniques described herein.
Within further aspects, conditionally-immortalized human spinal cord neural precursor cell lines of the present invention may be used in assays to detect the presence or absence of a particular protein in a sample. In general, an assay may be performed by contacting such cells with a sample and then measuring a response induced by the protein within the cells using methods familiar to those of ordinary skill in the art.
For example, a response may be measured using differential display techniques.
In a further aspect, the conditionally-immortalized human spinal cord neural precursor cell lines described herein may be used in the identification of novel genes and proteins present in human spinal cord neural precursor cells. Techniques, such as PCR, differential display, hybridization, expression library screens, immunoassays and two-hybrid screens may be employed for such identification. A particularly useful technique is differential gene screening. Clonal cell lines as described herein are particularly suited to such studies because they are derived from a single parental cell and, therefore, human spinal 5 cord neural precursor cell-specific genes are amplified with respect to non-clonal cell lines.
Novel genes and proteins that are expressed upon experimental manipulation (e.g., induction of apoptosis) may also be identified.
The cell lines provided herein may also be used in in vitro models of neuronal cell death including, but not limited to, neuronal apoptosis induced by growth factor 10 withdrawal. Briefly, a clonal conditionally-immortalized human spinal cord neural precursor cell line may be differentiated under conditions designed to minimize the basal level of apoptosis. Suitable conditions may be readily identified by evaluating the percentage of apoptotic nuclei in cells grown under different test conditions. The percentage of apoptotic nuclei may generally be determined by methods well known to those of ordinary skill in the
15 art, such as by DAPI staining or the in situ nick end-labeling assay.
Suitable conditions for minimizing basal apoptosis include differentiation in the presence of 1 p,g/mL
tetracycline, CNTF ( 10 ng/mL), NGF ( 100 ng/mL) and BDNF ( 10 ng/mL). Cells should be maintained in suitable differentiation conditions for a time sufficient to allow differentiation, while minimizing the basal level of apoptosis (which generally increases during the first 10 days of differentiation).
Such differentiated neuronal cells may then be employed in any of several models of apoptosis. In one such model, growth factors and N2 supplement are withdrawn for an amount of time sufficient to significantly increase the percentage of apoptotic cells with condensed nuclei. Preferably, the percentage of such apoptotic cells increases by at least about two fold. Under the representative conditions described above, about 18 hours of withdrawal is generally sufficient. A significant percentage of the cells should be rendered nonviable. Preferably, at least about 50% of the neurons are no longer viable following withdrawal of growth factors.
Regardless of the particular model, the cells may be used to study the mechanisms of apoptosis, as well as the effect of various conditions and agents on the apoptosis of neuronal cells, using experimental techniques well known to those of ordinary skill in the art. For example, the cells may be used to screen for an agent that affects neuronal cell death. Such a screen may be performed by contacting the cells during growth factor withdrawal with a candidate agent and then evaluating the ability of the candidate agent to affect the subsequent level of apoptosis. Similarly, the cells may be used to screen for a protein that regulates neuronal cell death. In such a screen, the level of expression or activity of a candidate protein (e.g., an enzyme) is altered within the cells (using standard techniques) and then the affect of the alteration on the level of apoptosis following treatments (including, but not limited to, growth factor withdrawal) is measured.
The following Examples are offered by way of illustration and not by way of limitation.

EXAMPLES
Example 1 Preparation of Human Spinal Cord Progenitor Cell Lines This Example illustrates the conditional immortalization of human spinal cord progenitor cells.
Human spinal cord cells in primary culture were prepared from human fetal spinal columns, of 8 to 9 weeks gestational age (obtained through Advanced Bioscience Resources, Inc.). The tissue was procured in compliance with state and federal laws and regulations. Prior to dissection, spinal columns were maintained for approximately 24 hours at 4°C in Hank's balanced salt solution (with no calcium or magnesium) containing penicillin (50 LU./ml) and streptomycin (50 p,g/ml). The spinal cord was dissected from the spinal column, and meninges and peripheral nerves were removed. The spinal cord tissue was then dissociated by incubation at 37°C for 30 to 45 minutes in an enzyme solution containing 0.2% collagenase plus dispase, with occasional trituration. The dispersed spinal cord cells were washed 3 times in high-glucose Dulbecco's modified Eagle medium (DMEM)/F-(l:l). The cells were subsequently plated at a density of approximately 104 cells/cm2 in growth medium consisting of DMEM/F-12 with N2 and B27 supplements (GIBCO, Baltimore, MD), FGF-2 (human recombinant, SO ng/ml, Boehringer Mannheim, Indianapolis, IN), EGF (human recombinant, 50 ng/ml, GIBCO, Baltimore, MD), IGF-I (human recombinant, 10 ng/ml, R & D Systems), PDGF (human recombinant, 10 ng/ml, Boehringer Mannheim, Indianapolis, IN), chick embryo extract (0.3%, GIBCO, Baltimore, MD, ultrafiltered), and bovine plasma fibronectin (5 p,g/ml, Sigma, St. Louis, MO).
For retroviral infection, the LINX v-myc vector was employed (Hoshimaru et al., Proc. Natl. Acad. Sci. USA 93:1518-1523, 1996; Sah et al., Nature Biotechnol. 15:574-580, 1997). In this system, in the absence of tetracycline, a tetracycline-controlled transactivator (tTA) strongly activates transcription from ph~M~,_,, resulting in the expression of the downstream v-myc oncogene. Tetracycline (0.01-1.0 p,g/ml) almost completely abolishes transcriptional activation by tTA, thereby blocking transcription of the v-myc oncogene. A gene conferring neomycin resistance is also present in the vector.
Spinal cord cultures were retrovirally infected and 6418- selected using methods analogous to those described by Sah et al., Nature Biotechnol. 15:574-580, 1997.
After 6418 selection, cultures were maintained in a simplified growth medium consisting of DMEM/F-12 with N2 supplement, FGF-2 (50 ng/ml), EGF (50 ng/ml), chick embryo extract (0.3%) and bovine plasma fibronectin (5 pg/ml). Cultures approaching confluence were passaged by trypsinization and split 1:5. Typically, one T75 flask near confluence yielded 10' cells, and cultures were passaged every 3 to 4 days.
During 6418 selection, some cell death occurred; after the selection, v-myc+
cells became the predominant cell type in the culture. These 6418- resistant, v-myc+ cells grew as an adherent monolayer. The majority of the cells were polygonal with very short processes (Figure 1 A).
To isolate clonal cell lines, half a cell, on average, was seeded into each well of a 96-well plate. Out of 2000 wells, 279 wells contained single colonies, which were expanded by feeding and passaging them as described above.
Most of these single colonies grew as adherent monolayers; only 4 colonies formed aggregates loosely attached to the substrate. These four colonies were designated HSP1, HSP2, HSP3 and HSP4, and expanded further. All four putative clones grew as adherent monolayers when the cells were passaged frequently; however, with less frequent passaging, cell aggregates formed. Cells with fine neuronal processes were occasionally seen in HSP 1. Three of the putative clones (HSP l, HSP2 and HSP4) grew well and were subsequently characterized.
For immunocytochemistry, cultures were fixed with methanol, and were incubated with primary antibody in blocking buffer (PBS, 1 % normal donkey serum, 0.1 Triton X-100) for 2 hours at room temperature, rinsed with PBS, and then incubated with a fluorescein (FITC)- or Texas red-conjugated species- specific secondary antibody (Jackson Immunoresearch Laboratories, Inc., West Grove, PA) in blocking buffer for another hour at room temperature. Cultures were then rinsed three times with PBS and coverslipped with PVA/DABCO before scoring and photographing representative fields.

The v-myc oncoprotein was detected by immunofluorescence in the nuclei of HSP1, HSP2 and HSP4 cells when the cells were cultured in the absence of tetracycline (Figure 2, -Tc). As expected, after addition of tetracycline, v-myc immunoreactivity was no longer detectable (Figure 2, +Tc). These results indicate that the clonal cell lines described herein express v-myc in a tetracycline-regulatable manner.
Example 2 Differentiation of Conditionally-Immortalized Human Spinal Cord Progenitor Cells into Post-mitotic Neurons This Example illustrates the differentiation of the retrovirally infected progenitor cells into neurons.
Bulk-infected cultures were plated at a density of 2x 104 cells/cm2 into differentiation medium (DMEM/F12 medium containing N2 supplements, FGF-2 (1-10 ng/ml), chick embryo extract (0.3%), natural mouse laminin (5 ~.g/ml), tetracycline (l-5 pg/ml) and a combination of neurotrophic factors [CNTF ( 10 ng/ml) and NGF ( 100 nglml)]
known to enhance motoneuron differentiation). The culture medium was subsequently refreshed every 5 days with differentiation medium lacking FGF-2. Within 48 hours, a few cells with neuronal morphology were apparent. The number of differentiated neurons increased during the subsequent days. These neurons exhibited round somata with long processes (Figure 1 B). Most of these differentiated cells did not die upon fluorodeoxyuridine (80 pM) treatment for 10 days, indicating that they were post-mitotic.
Cultures were generally differentiated for 7-14 days prior to immunocytochemical staining and for 4-20 days prior to electrophysiological recording.
Example 3 Growth Requirements of Human Spinal Cord Cell Lines This Example illustrates the effect of growth factors on cell lines as provided herein.

WO 99/45103 PC'f/US99/04535 The proliferation of HSP1, HSP2 and HSP4 cell lines required growth factors;
FGF-2 stimulated proliferation strongly whereas EGF increased proliferation to only a small extent (Figure 3A). Unlike neuronal restricted precursors from the rat spinal cord (Mayer-Proschel et al., Neuron 19:773-785, 1997), the growth rates of HSP1, HSP2 and 5 HSP4 were unaffected by the presence of NT-3 (10 ng/ml). The threshold for the growth promoting activity of FGF-2 was 0.1 ng/ml whereas maximal growth promoting activity occurred at 10 ng/ml (Figures 3A and 3C). Stimulation of proliferation by EGF
was maximal at 1 ng/ml (Figures 3A and 3C). Although the human spinal cord cell lines could attach, survive and proliferate without additional attachment factors, fibronectin enhanced cell 10 proliferation (Figures 3B and 3D) by promoting cell attachment. The growth promoting effect of fibronectin saturated at 3 ~g/ml.
Example 4 1 S Characterization of Human final Cord Cell Lines This Example illustrates characterization of HSP1, HSP2 and HSP4 cell lines, as well as differentiated cells generated by these cell lines.
Neuronal markers were detected using immunocytochemical assays. The cultures were incubated with primary antibodies at 37°C for 1 hour before methanol fixation.
20 Primary antibodies used for immunocytochemistry included polyclonal antibodies against neurofilament 200 kD ( 1:800, Chemicon, Temecula, CA) and neurofilament 68 kD
( I :400, Chemicon, Temecula, CA), and monoclonal antibodies against MAP2a/b ( 1:100, Chemicon, Temecula, CA), neurofilament 68 kD (1:40, Sigma, St. Louis, MO), neurofilament 160 kD
( 1:400, Sigma, St. Louis, MO), neurofilament 200 kD ( 1:40, Sigma), NCAM ( 1:1, a gift from Dr. Manhendra Rao, also available from Developmental Studies Hybridoma Bank) and ~3III
tubulin (1:400, Sigma, St. Louis, MO). All primary antibodies were validated on positive and negative control cultures. Cultures were incubated with primary antibody in blocking buffer (PBS, 1% normal donkey serum, 0.1% Triton X-100) for 2 hours at room temperature, rinsed with PBS, and then incubated with a fluorescein (FITC)- or Texas red-conjugated species-specific secondary antibody (Jackson Immunoresearch Laboratories, Inc., West Grove, PA) in blocking buffer for another hour at room temperature. Cultures were then rinsed three times with PBS and coverslipped with PVA/DABCO before scoring and photographing representative fields.
In the proliferative condition, over 90% of HSP1, HSP2 and HSP4 cells expressed neuronal markers such as neurofilament (both 200 and 68 kD), (3III-tubulin and NCAM (Figure 4) whereas MAP2a/b- immunoreactivity was absent. Neurofilament (160 kD)- immunoreactivity was present to a lesser extent, in approximately 30% of HSP1, HSP2 and HSP4 cells. After differentiation with DMEM/F12 medium containing N2 supplements, CNTF, NGF, BDNF, chick embryo extract, laminin and tetracycline (3 p.g/ml), neurofilament (200, 160 and 68 kD)- and MAP2a/b- (Figure 7B) immunoreactivities were present in the cytoplasm and processes. The cellular distribution of all neurofilaments shifted from the cytoplasm to the processes after differentiation. Moreover, after differentiation, HSPI cells expressed mature neuronal markers such as tau, peripherin and synaptophysin (Figures 7A, 7C and 7D).
These results indicate that neuronal-restricted precursor cell lines described herein express the neuronal markers neurofilament ( 160 and 200 kD), [3III-tubulin and NCAM in the proliferative growth condition. The presence of neurofilament (200 kD) has been described previously in proliferating spinal cord neuroblasts (Ray and Gage, J.
Neurosci. 14:354$-3564, 1994). However, in contrast to the observations reported herein, proliferating neuronal-restricted neural precursors from the rat spinal cord that are NCAM-immunoreactive do not express (3III-tubulin or neurofilament ( 160 kD) (Mayer-Proschel et al., Neuron 19:773-785, 1997). Taken together, these findings suggest that human and rat neuronal-restricted precursor cells differ with respect to at least 2 cytoskeletal components.
After differentiation, not only did cells with neuronal morphology exhibit staining for all neuronal markers examined, but they also fired action potentials upon current injection (13 of 14 cells). The whole-cell configuration of the patch-clamp method was used to record action potentials in current clamp mode. The bath solution was Tyrode's (in mM, 150 NaCI, 4 KCI, 2 MgClz, 10 Glucose, 10 HEPES, pH adjusted to 7.4 with NaOH;
2 mM
CaClz and 4 mM BaClz were added). Pipettes (2-4 MSZ resistance) were pulled from Boralex glass (Rochester Scientific, Rochester, NY), and filled with internal solution containing (in mM) 130 KCI, 5 MgClz, 10 HEPES, 10 EGTA, pH adjusted to 7.4 with KOH. Action potentials were recorded using an Axopatch 200A patch clamp amplifier and the BASIC-FASTLAB interface system (INDEC Systems, Capitols, CA). Figure 5 shows an example of an action potential in an HSP 1 cell after 4 days of differentiation; in current clamp mode, a 150 msec, 104 pA current pulse elicited an action potential that reached a peak of +61 mV.
To determine if HSP1, HSP2 or HSP4 could give rise to glial cells, the cultures were stained for astrocyte (GFAP) and oligodendrocyte (Oi, 04 and GaIC) markers after differentiation with DMEM/F 12 medium containing N2 supplements, CNTF, NGF, BDNF, chick embryo extract, laminin and tetracycline (3 p,g/ml).
Immunocytochemistry was performed as described above, except that primary antibodies were polyclonal antibodies against glutamate (1:15,000, Sigma, St. Louis, MO) and monoclonal antibodies against 04 (a gift from Dr. Manhendra Rao, also available from Chemicon, Temecula, CA), O1 ( 1:5, Boehringer Mannheim, Indianapolis, IN) and GaIC (a gift from Dr. Manhendra Rao, also available from Chemicon, Temecula, CA, and Sigma, St. Louis, MO). Only HSP2 gave rise to cells exhibiting GFAP immunoreactivity (<0.1 % of cells, Figure 6A) which did not co-localize with A2B5- or neurofilament 200kD- immunoreactivities (Figures 6B-D).
Oligodendrocyte markers (O1, 04 and GaIC) were not detectable with immunocytochemistry in any of the cell lines under these conditions. Even the addition to the differentiation medium of CNTF and NT-3, which strongly promote oligodendrocyte differentiation in CNS
stem cells (Lachyankar et al., Exp. Neurol. 144:350-360, 1997), did not result in the expression of oligodendrocyte markers.
HSP 1. and HSP4 were differentiated in the presence of 5% fetal bovine serum to determine if the cell lines could be induced to differentiate into astrocytes under these conditions. After 3 days of differentiation with tetracycline plus 5% fetal bovine serum, virtually no cells remained viable, indicating that this human spinal cord cell line responds to serum in a manner similar to that of neuronal-restricted precursors from the rat spinal cord.
The human spinal cord cell lines differentiated into multiple types of spinal cord neurons, including motoneurons, and expressed different excitatory and inhibitory neurotransmitters. To examine the neuron subsets obtained from the cell lines described herein, cultures were examined by immunofluorescence for glutamate, GABA, choline acetyltransferase (ChAT), NGFR and Islet-1. The cultures were fixed with 1%
glutaraldehyde in a 3% sucrose solution for glutamate and GABA detection by immuno-alkaline phosphatase methods (Li et al., Endocrine 5:205-217, 1996).
ChAT was detected by the APAAP method; briefly, after incubation with anti-ChAT
antibody at 4°C
overnight, the cultures were incubated with anti-mouse IgG (Jackson Immunoresearch Laboratories, Inc., West Grove, PA, 1:2000) for 1 hour, followed by incubation with APAAP
(Sigma, St. Louis, MO, 1:40) for 1 hour at 37°C. NTBBCIP (Boehringer Mannheim, Indianapolis, IN) or Fast Red (Sigma, St. Louis, MO) was used as the substrate for alkaline phosphatase. Primary antibodies used for immunocytochemistry included polyclonal antibodies against glutamate ( 1:15,000, Sigma, St. Louis, MO) and GABA ( 1:1000, Chemicon, Temecula, CA) and monoclonal antibodies against choline acetyltransferase (1:5, Boehringer Mannheim, Indianapolis, IN).
Immunoreactivity for ChAT, the synthetic enzyme for acetylcholine (which is specific to the motoneuron lineage), was detected in 23 ~ 9% of differentiated HSP 1 neurons (Figures 8D and 9), consistent with motoneuron phenotype. The presence of immunoreactivity for NGFR and Islet-1 (Figure 8) further confirmed the motoneuron phenotype. After HSP 1 differentiation, glutamate-immunoreactivity was detected in every neuron whereas GABA-immunoreactivity, consistent with interneuron phenotype, was present in 45 ~ 6% of the neurons (Figure 7). These results indicate that neuronal restricted precursor cell lines from the human spinal cord are useful for examining extrinsic and intrinsic signals that direct the differentiation of specific subsets of spinal cord neurons.
Moreover, these results establish that these immortalized human spinal cord cell lines can be differentiated into motoneurons.
From the foregoing, it will be appreciated that, although specific embodiments of the invention have been described herein for the purpose of illustration, various modifications may be made without deviating from the spirit and scope of the invention.

Claims (23)

Claims
1. A method for producing a conditionally-immortalized human spinal cord neural precursor cell, comprising:
(a) transfecting human spinal cord cells plated on a first surface and in a first growth medium that permit proliferation with DNA encoding a selectable marker and an externally regulatable growth-promoting gene; and (b) selecting the transfected cells on a second surface and in a second growth medium that permit attachment and proliferation, and therefrom producing a conditionally-immortalized human spinal cord neural precursor cell.
2. The method of claim 1 wherein the first and second surfaces are independently selected from substrates comprising one or more of a polyamino acid, fibronectin, laminin or tissue culture plastic.
3. The method of claim 1 wherein the growth-promoting gene is an oncogene.
4. The method of claim 3 wherein the oncogene is v-myc.
5. The method of claim 1 wherein expression of the growth-promoting gene is inhibited by tetracycline.
6. A conditionally-immortalized human spinal cord neural precursor cell capable of differentiation into neurons.
7. A conditionally-immortalized human spinal cord neural precursor cell according to claim 6, wherein the cell is further capable of differentiation into motoneurons.
8. A method for producing neurons, comprising culturing a cell produced according to claim 1 under conditions inhibiting expression of the growth-promoting gene.
9. A method according to claim 8, wherein the cell is cultured in medium comprising tetracycline.
10. A method according to claim 8, wherein the cells are cultured in the presence of one or more differentiating agents.
11. A method according to claim 10, wherein the differentiating agent is selected from the group consisting of NGF, CNTF and BDNF.
12. A neuron produced according to the method of claim 8.
13. A motoneuron produced according to the method of claim 8.
14. A cell produced according to the method of claim 1 or claim 8, for use in the manufacture of a medicament for transplanting a human spinal cord cell into a mammal.
15. A cell produced according to the method of claim 1 or claim 8, for use in the manufacture of a medicament for treating a spinal cord-related condition in a patient.
16. A cell according to claim 1 S, wherein the condition is selected from the group consisting of pain, spinal cord injury, motoneuron diseases and myelination-related diseases.
17. A method for screening for an agent that modulates an activity of a protein produced by a human spinal cord cell, comprising:
(a) contacting a cell produced according to the method of claim 1 or claim 8 with a candidate agent; and (b) subsequently measuring the ability of the candidate agent to modulate an activity of a protein produced by the cell.
18. A method for detecting the presence or absence of a protein in a sample, comprising:
(a) contacting a sample with a cell produced according to the method of claim 1 or claim 8; and (b) subsequently detecting a response in the cell, and therefrom detecting the presence of a protein in the sample.
19. A method for identifying a human spinal cord gene or protein, comprising detecting the presence of a gene or protein within a culture of cells produced according to the method of claim 1 or claim 8.
20. A method for screening for an agent that affects human spinal cord cell death, comprising:
(a) contacting a cell produced according to the method of claim 1 or claim 8 with a candidate agent under conditions that, in the absence of candidate agent, result in death of the cell; and (b) subsequently measuring the ability of the candidate agent to affect the death of the cell.
21. A method for screening for a protein that regulates human spinal cord cell death, comprising:
(a) altering the level of expression of a protein within a cell produced according to claim 1 of claim 8; and (b) subsequently measuring the effect of the alteration on the death of the cell, and therefrom identifying a protein that regulates human spinal cord neural precursor cell death.
22. A conditionally-immortalized human spinal cord neural precursor cell produced according to the method of claim 1.
23. A cell according to claim 22, wherein the cell is present within a clonal cell line.
CA002322747A 1998-03-02 1999-03-02 Human spinal cord cell lines and methods of use therefor Abandoned CA2322747A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US7656798P 1998-03-02 1998-03-02
US60/076,567 1998-03-02
US09/059,790 US6225122B1 (en) 1998-03-02 1998-04-13 Human spinal cord cell lines and methods of use therefor
US09/059,790 1998-04-13
PCT/US1999/004535 WO1999045103A2 (en) 1998-03-02 1999-03-02 Human spinal cord cell lines and methods of use therefor

Publications (1)

Publication Number Publication Date
CA2322747A1 true CA2322747A1 (en) 1999-09-10

Family

ID=26739180

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002322747A Abandoned CA2322747A1 (en) 1998-03-02 1999-03-02 Human spinal cord cell lines and methods of use therefor

Country Status (7)

Country Link
US (1) US6225122B1 (en)
EP (1) EP1060245A2 (en)
CN (1) CN1297477A (en)
AU (1) AU3065599A (en)
BR (1) BR9908443A (en)
CA (1) CA2322747A1 (en)
WO (1) WO1999045103A2 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7037493B2 (en) 2000-05-01 2006-05-02 Cornell Research Foundation, Inc. Method of inducing neuronal production in the brain and spinal cord
US20030077564A1 (en) * 2001-10-02 2003-04-24 Board Of Trustees Of Southern Illinois University Nutrient medium for maintaining neural cells in injured nervous system
US6770521B2 (en) * 2001-11-30 2004-08-03 Texas Instruments Incorporated Method of making multiple work function gates by implanting metals with metallic alloying additives
CA2470853A1 (en) * 2001-12-13 2003-06-19 Japan Science And Technology Agency Human cell culture medium and culture method
WO2003070189A2 (en) * 2002-02-15 2003-08-28 Cornell Research Foundation, Inc. Enhancing neurotrophin-induced neurogenesis by endogenous neural progenitor cells by concurrent overexpression of brain derived neurotrophic factor and an inhibitor of a pro-gliogenic bone morphogenetic protein
US8932620B2 (en) * 2005-06-17 2015-01-13 Drexel University Three-dimensional scaffolds for tissue engineering made by processing complex extracts of natural extracellular matrices
WO2015017354A1 (en) * 2013-07-29 2015-02-05 University Of Houston System Volatile organic gases as bioindicators for transplant rejection
US11156595B2 (en) * 2015-05-28 2021-10-26 Axogen Corporation Organotypic DRG-peripheral nerve culture system
EP3332695B1 (en) * 2016-12-09 2021-05-26 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Measurement device

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996014400A1 (en) 1994-11-08 1996-05-17 Bradley Michael John Stringer Human cell-lines
US5770414A (en) 1996-02-20 1998-06-23 The Regents Of The University Of California Regulatable retrovirus system for genetic modification of cells
US6713247B1 (en) 1996-09-03 2004-03-30 Signal Pharmaceuticials, Inc. Human CNS cell lines and methods of use therefor

Also Published As

Publication number Publication date
WO1999045103A2 (en) 1999-09-10
US6225122B1 (en) 2001-05-01
BR9908443A (en) 2002-01-15
EP1060245A2 (en) 2000-12-20
AU3065599A (en) 1999-09-20
CN1297477A (en) 2001-05-30
WO1999045103A3 (en) 1999-11-25

Similar Documents

Publication Publication Date Title
US6913925B1 (en) Human mesencephalon cell lines and methods of use therefor
Tang et al. Long-term culture of purified postnatal oligodendrocyte precursor cells: evidence for an intrinsic maturation program that plays out over months
Chambers et al. Spatiotemporal selectivity of response to Notch1 signals in mammalian forebrain precursors
Belachew et al. Postnatal NG2 proteoglycan–expressing progenitor cells are intrinsically multipotent and generate functional neurons
Gage et al. Isolation, characterization, and use of stem cells from the CNS
Learish et al. Inhibition of mitogen-activated protein kinase kinase blocks proliferation of neural progenitor cells
US6713247B1 (en) Human CNS cell lines and methods of use therefor
US6900054B2 (en) Lineage restricted glial precursors from the central nervous system
Piper et al. Identification and characterization of neuronal precursors and their progeny from human fetal tissue
US6835567B1 (en) PNS cell lines and methods of use therefor
Thompson et al. Neurogenin2 identifies a transplantable dopamine neuron precursor in the developing ventral mesencephalon
Pereiro et al. Optimization of a method to isolate and culture adult porcine, rats and mice Müller glia in order to study retinal diseases
US6225122B1 (en) Human spinal cord cell lines and methods of use therefor
Nobre et al. Characterization and differentiation potential of rat ventral mesencephalic neuronal progenitor cells immortalized with SV40 large T antigen
DUTT et al. Establishment of a human retinal cell line by transfection of SV40 T antigen gene with potential to undergo neuronal differentiation
Marone et al. An immortalized mouse neuroepithelial cell line with neuronal and glial phenotypes
Richard et al. Early, middle, and late stages of neural cells from ovine embryo in primary cultures
Eubanks et al. Three distinct neuronal phenotypes exist in embryonic rat hippocampal neurons cultured in basic fibroblast growth factor
Li et al. Culture of pluripotent neural epithelial progenitor cells from e9 rat embryo
Giordano et al. Development of Immortalized Cell Lines for Transplantation in Central Nervous System Injury and Degeneration Models
Shi Studies of mitogenic and morphogenic signals during mouse limb development
Dedesma Expression pattern and functional role of Tctex-1, a dynein light chain, in neural proliferative cells and young neurons
US20050186184A1 (en) Mammalian pluripotent neural cells and uses thereof
Hartley Characterization of human NT2N neurons for transplantation therapeutics
Giordano et al. Injury and Degeneration Models

Legal Events

Date Code Title Description
FZDE Dead