This application contains an ST.26 compliant Sequence Listing, which is submitted concurrently in xml format via Patent Center and is hereby incorporated by reference in its entirety. The .xml copy, created on Aug. 21, 2024, is named Sequence Listing 009062-8511 US01 and is 97,490 bytes in size.
Worldwide, over 2.5 million people live with spinal cord injury, with over 100,000 new cases occurring annually. Spinal cord injury often causes motor dysfunction below the level of the injury. For example, thoracic and lumbar spinal cord injury can cause paraplegia and cervical spinal cord injury can cause quadriplegia. Such injury is permanent and often severe and there is no effective treatment. Various neurologic diseases also involve damaged or dysfunctional spinal cord neurons. Neural stem cell grafts have potential for treating such conditions. However, it has not been possible to obtain sufficient numbers of appropriately patterned neural stem cells that are karyotypically stable, having a spinal cord positional identity, for implanted cells to survive and functionally engraft.
Disclosed herein are methods of inducing and maintaining spinal cord neural stem cells (NSC) and spinal cord neural progenitor cells, staring with human pluripotent stem cells. In some embodiments the hPSC are human embryonic stem cells (hESC). In some embodiments the hPSC are induced pluripotent stem cells (iPSC).
Maintaining the spinal cord NSC includes expansion, that is, increasing their number. Maintaining the spinal cord NSC can include their differentiation into spinal cord neural progenitor cells. In some embodiments differentiation into spinal cord neural progenitor cells includes obtaining all three neuronal progenitor lineages: neuronal, astrocytic, and oligodendrocytic. Maintaining the spinal cord NSC includes retaining the spinal cord positional identity (patterning).
Inducing spinal cord NSC comprises culturing the cells in the presence of caudalizing morphogens (compounds causing development of spinal cord patterning, rather than, for example, brain patterning) and SMAD inhibitors (SMAD is an allusion to this family of proteins' homology with the Caenorhabditis elegans SMA (“small” worm phenotype) and Drosophila MAD (“Mothers Against Decapentaplegic”) family of genes). Some embodiments use dual SMAD inhibitors. In some embodiments, the SMAD inhibitors are inhibitors of bone morphogenetic protein (BMP), activin, and transforming growth factor β (TGF-β). In some embodiments, the caudalizing morphogens are fibroblast growth factor 2 (FGF2) and FGF8 (FGF2/8), and an activator of WNT signaling. In some embodiments, the activator of WNT signaling is CHIR99021. In some embodiments, the inhibitor of BMP is LDN-193189. In some embodiments, the inhibitor of activin and TGF-β is SB-431542.
Maintaining spinal cord NSC comprises culturing the NSC in the presence of inhibitors of activin and TGF-β, and activators of WNT signaling, and sonic hedgehog signaling (SHH). In some embodiments, the inhibitor of activin and TGF-β is SB-431542. In some embodiments, the activator of WNT signaling is CHIR99021. In some embodiments, the activator of SHH is Hh-Ag1.5.
Some embodiments comprise a method of inducing spinal cord NSC. Some embodiments comprise a method maintaining spinal cord NSC. Some embodiments comprise an integrated method of inducing and maintaining spinal cord NSC.
Some embodiments comprise an ex vivo population of cells generated by the herein disclosed methods. Some embodiments comprise an ex vivo population of cells enriched in spinal cord NSC. Some embodiments comprise an ex vivo near-homogeneous population of spinal cord NSC (in the sense that the population is highly enriched for spinal cord NSC, but not that the NSC are all of the same kind). Some embodiments consist essentially of an ex vivo spinal cord NSC and spinal cord neuronal progenitor cells. Some embodiments predominantly comprise an ex vivo spinal cord neuronal progenitor cells. In aspects of these embodiments, the spinal cord neuronal progenitor cells comprise neural, astrocytic, and oligodendrocytic spinal cord progenitor cells. In further aspects, the neural spinal cord progenitor cells can comprise motor neuron and/or p3 progenitor cells, in further aspects the neural spinal cord progenitor cells can comprise dorsal and ventral spinal cord progenitor cells.
To be suitable for implantation with survival and engraftment, the various cellular compositions herein disclosed should comprise at least 50-200 million cells. Thus various embodiments are cell cultures and populations comprising at least 50 million, or 100 million, or 150 million, or 200 million cells. In some embodiments, the culture or population does not comprise more than 500 million or 750 million, or one billion cells.
The present technology includes methods for generating a clinically translatable spinal cord neural stem cell. Compared to previous methods, the present methods generate cells that are typically karyotypically stable and safe for human translation. A set of pre-clinical in vitro and in vivo studies optimize the determination of cells for clinical use.
Recent findings reveal that appropriately patterned neural stem cells (NSCs), which can provide tissue-specific developing microenvironments, have superior potential to reconstitute damaged neural circuitry and enable axonal regeneration. For treating disorders of the spinal cord, including spinal cord injury (SCI), amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), and other spinal cord-specific disorders, a source of cells of regional spinal cord identity that is capable of generating multiple neuronal and glial derivatives is needed. However, protocols to derive and maintain regionalized spinal cord NSCs from human pluripotent stem cells (hPSCs) have not existed.
The importance of generating NSCs of regional spinal cord identity is illustrated by the fact that corticospinal axons, the most important system for human voluntary motor function, only regenerate into neural grafts of spinal cord identity. This homologous reconstitution of the lesioned adult spinal cord with spinal cord multipotent neural progenitor cells (NPCs) was achieved using developmental primary spinal cord tissue, and these grafted cells formed synaptic relays across lesion sites to significantly improve functional outcomes. In contrast, corticospinal axons did not regenerate into rostrally-fated (brain) neural progenitors. Human embryonic stem cell (hESC) lines are readily available as a source for human clinical trials and are more broadly accepted ethically than primary human neural tissue, highlighting the importance of generating spinal cord NSCs from such cell lines.
Recent progress in spinal cord developmental research has revealed that SOX2+/Brachyury(T)+ neuromesodermal progenitors (NMPs) reside in a caudal stem zone and are the endogenous cellular source of the spinal cord. Early epiblasts acquire neural fates either in the anterior neural plate, which contributes brain progenitors, or via the induction of primitive streak-associated neuromesodermal progenitors, which contribute spinal cord progenitors. The brain and spinal cord therefore have independent origins early in development. Although retinoic acid has been used to generate caudalized cells, retinoic acid activates only rostral homeobox (HOX) genes (HOX1-5 paralog), which pattern brain neuroepithelial cells (NEPs) with a broad brainstem-to-rostral cervical spinal cord identity. In contrast, synergistic wingless-type MMTV integration site protein family (WNT) and fibroblast growth factor (FGF) signals are necessary and sufficient to induce more caudal neuraxis spinal HOX gene expression (HOX6-9 paralog) and specify cervical and thoracic spinal cord identity.
The herein disclosed methods induce and maintain functional human spinal cord NSCs. Importantly, these NSCs include all six dorsal spinal cord neuronal progenitor cell types (pd1-6), and all of the ventral spinal cord neuronal progenitor cell types (pV0-3 and pMN) and can yield a broad range of identified spinal cord neuronal phenotypes (D11-6, V0-3, and Motor neurons), but not rostral identity cells. The ability to create spinal cord motor neurons and interneurons can facilitate disease modeling and drug screening for several disorders with extensive spinal cord pathology, including ALS, SMA, progressive muscular atrophy, hereditary spastic paraplegia, Friedreich's ataxia, tabes dorsalis, and others.
Stem cells are cells that can differentiate into other cell types, but that can also self-renew indefinitely, to replace themselves. Totipotent and pluripotent stem cells can differentiate into all, or a wide array, of cell types influenced by signals present in the environment. In an early stage in differentiation, stem cells retain their “stemness”, but become restricted to giving rise to a particular lineage of cell types, for example, hematopoietic stem cells and NSC. However, these cells still give rise to many cell types and are termed multipotent. Stem cells can also acquire patterning, giving the a positional identity within the body of an organism, such as spinal cord NSC, as distinct from other types of NSC that, for example give rise to neuronal cells in various regions of the brain. In a further stage of differentiation, the ability to self-renew becomes finite and the cells give rise to only one or a few cell types. These are unipotent and oligopotent progenitor cells.
Of further importance, human embryonic stem cell-derived NSCs are an appropriate cell type for clinical translation for spinal cord “replacement” strategies in spinal cord injury and other disorders. Only spinal cord-fated cells enable regeneration of corticospinal axons, but an hPSC-derived spinal cord NSC cell line has not been previously derived. It is demonstrated herein that spinal cord NSCs enable host corticospinal regeneration and synapse formation in the injury site, and in turn, grafted human NSCs extended very large numbers of axons into the host spinal cord over long distances. These extending human axons form synapses with the host spinal cord below the lesion, establishing a potential neural relay across the lesion site. By using a retrograde monosynaptic rabies tracing system, it is shown that human spinal cord NSC grafts establish connectivity with all of the classic brainstem and cortical host neurons that influence motor function, together with a diverse population of intrinsic spinal cord neuronal populations as well as primary sensory neurons.
Described herein are spinal cord NSCs from hESCs or other hPSC and methods for generating the same. In some embodiments, methods are described to determine whether these cells can reconstitute damaged spinal neural circuitry and support corticospinal regeneration. In some embodiments, described is a derivation of spinal cord NSCs from hESCs by activation of WNT and FGF signaling, together with dual inhibition of SMAD signaling, that is, inhibition of both BMP and TGF-β. These human spinal cord NSCs generated motor neurons and a diversity of spinal interneurons comprising multiple positions in the spinal cord dorso-ventral axis, which could be maintained in vitro for prolonged time periods, and survived in lesioned rat spinal cords where they enabled robust corticospinal regeneration. Furthermore, grafts interconnected with multiple intraspinal and supraspinal systems, as assessed by glycoprotein-deleted rabies trans-synaptic virus tracing. Together, hPSC-derived spinal NSCs could represent an optimal cell type for spinal cord study and therapeutic transplantation for several spinal cord disorders.
Access to cells providing “spinal cord neurons” enable a broad range of biomedical applications in vitro and these newly generated hPSC-derived spinal cord NSCs thus constitute the optimal cell type for clinical translation for spinal cord “replacement” strategies in several spinal cord disorders.
Provided herein are methods for generating spinal cord neural stem cells. Such methods include contacting human pluripotent stem cells in a stem cell-appropriate medium with a combination of appropriate factors as described in detail below. In certain embodiments, the methods disclosed herein are used to generate spinal cord neural stem cells that are karyotypically stable. In addition such methods generate spinal cord neural stem cells that are capable of engraftment in vivo, survival in vivo, and undergoing neuronal and glial differentiation in vivo.
According to the embodiments disclosed herein, pluripotent stem cells (for example, hESC or induced pluripotent stem cells (iPSC)) are grown to about 70% confluence in an appropriate medium (such as an hESC growth medium, for example, mTeSR medium, but any complete media for human ESC or iPSC could be used). In some embodiments the culture plates are coated with MATRIGEL® or vitronectin. No feeder cells are required and in some embodiments none are used. The culture medium is then switched to an induction medium containing inhibitors of bone morphogenetic protein (BMP), activin, and transforming growth factor β (TGF-β); as well as one or more caudalizing morphogens. Some embodiments further include an inhibitor of Notch signaling. In some embodiments the base medium for the induction medium is N2B27 medium. (N2B27 medium is knockout Dulbecco's Modified Eagle's Medium (DMEM)/F12: Neurobasal (1:1), 1×N2, 1×B27, 1× penicillin/streptomycin, 1× Glutamax). In some embodiments, the BMP signaling inhibitor is LDN-193189. In some embodiments, the activin and TGF-β signaling inhibitor is SB-431542. In some embodiments, the one or more caudalizing morphogens are fibroblast growth factor 2 and fibroblast growth factor 8 (FGF2/8), and an activator of WNT signaling. In some embodiments, the activator of WNT signaling is a GSK3β inhibitor, for example, CHIR99021. In some embodiments, the inhibitor of Notch signaling is a γ-secretase inhibitor, for example, DAPT. In some embodiments, the cells are cultured in induction medium for 10 days, the cells are split and fresh medium is added at regular intervals, for example daily. This culture protocol induces the hPSC to develop into neural stem cells and, ultimately, into spinal cord neural stem cells.
In some embodiments, the methods include contacting human pluripotent stem cells in a stem cell-appropriate medium with a combination of appropriate factors. In certain embodiments, the combination of appropriate factors includes a combination of SB-431542, CHIR99021, FGF2, and FGF8b. In other embodiments, the combination of appropriate factors includes a combination of SB-431542, CHIR99021, FGF2, FGF8b, and LDN-193189. In other embodiments, the combination of appropriate factors includes a combination of SB-431542, CHIR99021, FGF2, FGF8b, LDN-193189, and DAPT.
In some embodiments, SB-431542 is used at a concentration of 5-10 μM, or any integral value therein. In some embodiments, CHIR99021 is used at a concentration of 3-4 μM, or any integral value therein. In some embodiments, FGF2 and FGF8 are used at a concentration of 25-100 ng/ml each, or any integral value therein. In some embodiments an inhibitor of Notch signaling, such as DAPT, is used. Other components may be used at the concentrations described in the Examples below. Generally, use of the various components at the lower end of the above concentration ranges leads to greater genetic and phenotypic stability, including stability of the spinal cord NSC phenotype and lower potential for tumorigenicity. This consideration also applies in the maintenance and expansion phase.
In some embodiments where the method includes contacting a human pluripotent stem cell with a combination of appropriate factors that includes CHIR99021, the CHIR99021 is used at a concentration of less than about 4 μM CHIR99021 or no more than about 4 μM CHIR99021. In other embodiments, the CHIR99021 is used at a concentration of between about 0.5 μM and 4 μM. In some embodiments, the CHIR99021 is used at a concentration of about 0.5 μM, about 1 μM, about 2 μM, about 3 μM, or about 4 μM. In certain embodiments, the CHIR99021 is not part of the combination of appropriate factors.
In some embodiments where the method includes contacting a human pluripotent stem cell with a combination of appropriate factors that includes SB431542, the SB431542 is used at a concentration of less than about 10 μM SB431542 or no more than about 10 μM SB431542. In other embodiments, the SB431542 is used at a concentration of between about 1 μM and 10 μM. In some embodiments, the SB431542 is used at a concentration of about 1 μM, about 2.5 μM, about 5 μM, about 7.5 μM, or about 10 μM. In certain embodiments, the SB431542 is not part of the combination of appropriate factors.
In some embodiments where the method includes contacting a human pluripotent stem cell with a combination of appropriate factors that includes FGF-2, the FGF-2 is used at a concentration of less than about 25 ng/ml FGF-2 or no more than about 25 ng/ml FGF-2. In other embodiments, the FGF-2 is used at a concentration of between about 25 ng/ml and 200 ng/ml. In some embodiments, the FGF-2 is used at a concentration of about 25 ng/ml, about 100 ng/ml, about 200 ng/ml.
In some embodiments where the method includes contacting a human pluripotent stem cell with a combination of appropriate factors that includes FGF-8b, the FGF-8b is used at a concentration of less than about 25 ng/ml FGF-8b or no more than about 25 ng/ml FGF-8b. In other embodiments, the FGF-8b is used at a concentration of between about 25 ng/ml and 100 ng/ml. In some embodiments, the FGF-8b is used at a concentration of about 25 ng/ml, or about 100 ng/ml. In certain embodiments, the FGF-8b is not part of the combination of appropriate factors.
In embodiments where the combination of appropriate factors includes LDN-193189, the LDN-193189 is used at a concentration of about 100 nM or any other suitable concentration.
In some embodiments, the method comprises contacting a human pluripotent stem cell with a set of factors having any of the combinations of concentrations shown in Table 1 or Table 2. In some particular embodiments, the combination of appropriate factors is SB-431542, CHIR99021, FGF-2, and FGF-8b, where the concentration of SB-431542 is 5 μM, the concentration of CHIR99021 is 3 μM, the concentrations of FGF2 and FGF8 are 25 ng/ml, and no DAPT is used. In other particular embodiments, the combination of appropriate factors is SB-431542, CHIR99021, FGF-2, FGF-8b, and LDN-193189, where the concentration of SB-431542 is about 5 μM, the concentration of CHIR99021 is about 3 μM, the concentrations of FGF2 and FGF8 are about 25 ng/ml, the concentration of LDN-193189 is about 100 nM, and no DAPT is used. In a further aspect of these embodiments, no inhibitor of Notch signaling is used. In some embodiments the plate coating is changed to CELLSTART® (ThermoFisher Scientific) at passage 4 and to poly-L-ornithine at passage 6.
In some embodiments, the spinal cord neural stem cells generated from the methods disclosed herein are karyotypically stable. A spinal cord neural stem cell is “karyotypically stable” if the spinal cord neural stem cells maintain chromosome stability over several (e.g., 5, 10, 15) passages without generating significant abnormal cells. An abnormal cell is a cell containing any karyotype different from normal 46 chromosomes. In certain embodiments, a population of kayotypically stable spinal cord neural stem cells generated from the methods disclosed herein is one that demonstrates fewer abnormal cells than from other methods. In some aspects, a spinal cord neural stem cell is karyotopically stable if the spinal cord neural stem cell has fewer than 1 karyotypic abnormalities per preparation, or between 0 to 1 karyotypic abnormalities per preparation.
Once the spinal cord neural stem cell phenotype is obtained, the cells can be further expanded and the phenotype maintained in culture by switching to a neural maintenance medium (a neurobasal medium; that is, a basal medium that meets the cell culture requirements of neuronal cells) containing inhibitors of activin and TGF-β signaling, and activators of WNT and SHH signaling. In some embodiments the base medium of the neural maintenance medium is N2B27 medium. In some embodiments the activin and TGF-β signaling inhibitor is SB-431542. In some embodiments the activator of WNT signaling is a GSK3β inhibitor, for example, CHIR99021. In some embodiments the activator of SHH is Hh-Ag1.5.
In some embodiments, the concentration of SB-431542 is 2 μM. In some embodiments, the concentration of CHIR99021 is reduced to 2 μM from the amount used during induction. In some embodiments the concentration of Hh-Ag1.5 is 100 nM. In some embodiments, components may be used independently at the concentrations described in the Examples below.
One of the major impediments to the development of practical biomedical uses of neural stem cells for humans has been the rapidity with which they undergo terminal differentiation in culture. This has made it impractical to generate the number of cells needed, and have them available, for transplant. While neural stem cells and neural progenitor cells can survive implantation and engraftment, cells that have already differentiated past the progenitor cell stage do not survive. Although spinal cord NSC cultured and expanded in the disclosed maintenance medium do not retain a stem cell phenotype indefinitely, eventually differentiating into neural, astrocyte, and oligodendrocyte progenitor cells, they do not differentiate past the progenitor cell stage. Additionally they retain their spinal cord positional identity. Thus it becomes possible to generate substantial numbers of cells, sufficient for implantation and capable of survival and engraftment.
It is important that the differentiation into progenitor cells produces progenitors for all three types of neuronal cell types: neurons, astrocytes, and oligodendrocytes. In repairing neuronal lesions, whether due to injury, disease or disorder all three cell types play a role. Neurons replace neurons that have been destroyed or damaged beyond repair. Astrocytes support the engraftment and survival of the neurons and can also reconstitute the blood-brain barrier. Oligodendrocytes mediate myelination of the axons of the new neurons and remyelination of damaged neurons.
Generally, any source of hPSC can be used in the induction of spinal cord NSC, but spinal cord NSC (including progenitor cells) generated from different sources offer different advantages. Established hESC lines are well-characterized and readily obtainable. They are suited to a wide variety of laboratory application. However, if used in biomedical applications (that is, for implantation into a patient), they will generally not provide an immunological, major histocompatibility complex (MHC) match (that is, they will be allogenic) to any particular patient, so treatment with allogeneic hESCs will typically need to include immune suppression.
iPSC can also be used and avoid ethical issues related to the acquisition of human embryonic tissue. They are similarly suitable for laboratory applications. In biomedical use, iPSC offer the possibility of autologous sourcing, ensuring an MHC match and thus obviating immune suppression. iPSC can be used for allogeneic donation as well. However, the risk of tumorigenicity is greater than with hESC-derived cells.
Another approach to obtaining MHC-matched hPSC for individual patients is to establish a bank of MHC-typed hPSC lines of diverse types. Both hESC and iPSC, or a mixture thereof, could be used to establish the bank.
The induction culture results in a population of cells with a spinal cord neural stem cell phenotype. This positional identity or patterning sets these cells apart from previously obtained neural stem cell cultures, which all had forebrain or hindbrain positional identities (patterning). Spinal cord neural stem cells exist in nature, but only as integrated into a complete (if possibly still developing) organism. Nor has it been feasible to isolate and culture them. The presently disclosed spinal cord NSC cultures represent more highly-enriched populations of spinal cord NSC than has been previously achieved. In some embodiments the cells are >95% spinal cord NSC. However, any population will typically comprise multiple kinds of NSC, such as, SOX1+/SOX2+ NSCs, SOX1−/SOX2+ NSCs, SOX2+/PAX6+ NSCs, SOX2+/NKX6.1+ NSCs, and SOX2+/OLIG2+ NSCs. Thus, enriched or homogenous populations of spinal cord neural stem cells, and in vitro cultures thereof, constitute embodiments disclosed herein.
Unlike earlier efforts, the present spinal cord neural stem cells can be maintained in a non-terminally differentiated state for an extended period of time of at least several months, and the number of cells can be expanded to enable laboratory and clinical uses. The maintenance of the stem cell aspect of the phenotype is not indefinite, with differentiation into the various neuronal progenitor cell types. However, because all three progenitor types (neuronal, astrocyte, and oligodendrocyte) are produced, this population of cells continue to enable uses including implantation into a patient where the cell engraft, survive, and make functional connections with the endogenous tissue. Importantly, the patterning of spinal cord positional identity is maintained. Thus, some embodiments are ex vivo populations of cells predominantly comprising (or consisting essentially of) neural stem cells and neural progenitor cells with spinal cord positional identity patterning. Other embodiments are ex vivo populations of cells predominantly comprising (or consisting essentially of) neural progenitor cells with spinal cord positional identity patterning. In aspects of these embodiments, the neural progenitor cells are a mixture of neuronal progenitor cells, astrocyte progenitor cells, and oligodendrocyte progenitor cells. In further aspects the neural progenitor cells include motor neuron progenitor cells. In further aspects the neural progenitor cells can comprise dorsal and ventral spinal cord progenitors.
In various embodiments the cell compositions comprise pharmaceutically acceptable diluents, excipients, or carriers. These can include culture media and buffered saline solutions. In still further embodiments the cell compositions comprise a cryopreservative so that they may be stored frozen. In some embodiments the cryopreservative (or cryoprotectant) comprises glycerol or dimethyl sulfoxide.
Terminology such as enriched, near-homogeneous, consisting essentially of, and predominantly comprising, acknowledge that cultures and cell populations may not consist exactly of only the indicated cells types. It is used to indicate that the indicated cell type(s) make up a much larger proportion of the culture or population than would be found in any tissue sample directly obtained from a living organism, or cultured therefrom (or that exists in a living organism), and to indicate that other cell types are present in negligible amounts, whether from contamination or non-synchrony of differentiation. While these terms are used to convey that the overwhelming majority of the cells are spinal cord NSC and/or spinal cord neuronal progenitor cells, it should not be interpreted to indicate that these cells are all of the same kind or subtype that exist within the rubrics of spinal cord NSC and spinal cord neuronal progenitor cells, unless explicitly stated.
These cells, cultured neural stem cells and/or neural progenitor cells, are capable of being used for implantation into a patient for therapeutic effect, as they will survive, engraft, and form functional connections with cells of the recipient. In various embodiments survival and engraftment in a human requires implantation of at least 50 to 200 million cells. In one embodiment survival and engraftment in a human requires implantation of at least 100 million cells. They are also useful in modeling diseases and disorders impacting the spinal cord and in drug screening.
The present technology includes spinal cord neural stem cells generated according to the methods of the present technology. The present technology includes methods of engrafting a spinal cord neural stem cell into a subject in need thereof. The method of engrafting a spinal cord neural stem cell may comprising implanting the spinal cord neural stem cell of the present technology into a subject in need thereof. In some aspects, the subject has a spinal cord injury. In some aspects the subject has a neurodegenerative disease.
In some embodiments, the engrafted spinal cord neural stem cell demonstrates increased engraftment compared to a control spinal cord neural stem cell. In some embodiments, the engrafted spinal cord neural cell survives at least 3-months post engraftment. In some embodiments, the engrafted spinal cord neural cell undergoes both neuronal and glial differentiation in vivo. In some embodiments, the engrafted spinal cord neural stem cell demonstrates increased survival and undergoes neuronal and glial differentiation in vivo.
To model diseases and disorders pluripotent stem cells can be obtained from a subject that is genetically determined or predisposed to develop the condition. Induced pluripotent stem cells will typically be the more readily obtained source tissue in this instance. Alternatively, pluripotent stem cells from a healthy donor can be genetically engineered to carry a genetic lesion associated with a disease or disorder, if the genetic basis of the condition is known. In this instance both iPSC and hESC are similarly suitable sources of pluripotent stem cells. In either case, these pluripotent stem cells are then cultured in the neural stem cell induction conditions. As they differentiate, their development can be compared to normal cells to identify markers of the disease process. Chemical and biological agents that might contribute to development of disease can be added to the culture. Chemical and biologic agents (including gene therapeutic agents) can then be tested for their ability to prevent, interrupt or reverse development of the disease, or at least its correlate in tissue culture.
Human H9 ESCs cultured under feeder-free conditions were converted into NSCs by switching from hESC growth media to N2B27 media supplemented with LDN-193189 (LDN; a BMP signaling inhibitor), SB-431542 (SB; an activin and TGF-β signaling inhibitor), DAPT (a γ-secretase inhibitor, an inhibitor of Notch signaling), CHIR99021 (CHIR; a GSK3β inhibitor, an activator of WNT signaling), and FGF2+FGF8 (FGF2/8) (
Any differentiation protocol faces the challenge of maintaining cells for extended time periods in vitro, which is important to the generation of sufficient quantities of cells to fill human-sized spinal cord lesion sites. WNT and Sonic hedgehog (SHH) are potent mitogens, whereas bone morphogenetic protein (BMP) directs cells to differentiate into the neural crest lineage in the developing spinal cord. Activation of WNT/SHH and suppression of BMP could specify cells to become spinal cord NSCs and maintain their stemness for prolonged time periods. The neural induction media as described above was changed to N2B27 media supplemented with CHIR, SB, and Hh-Ag1.5 (a potent SHH agonist) ten days after neural induction. In this chemically defined condition, SOX1+/SOX2+/Nestin+ NSCs could be expanded over four months in vitro, through passage 30 (
To further confirm these results, the gene expression profile of human H9 ESC-derived spinal cord NSCs (H9-derived spinal cord NSCs) was examined two months after neural induction. When compared to expression profiles of human fetal brain and spinal cord, cultured H9-derived spinal cord NSCs expressed high levels of caudal HOX genes and low levels of the telencephalic markers FOXG1, SIX3, and OTX2 (
To further examine the regional identity of H9-derived spinal cord NSCs along the rostrocaudal axis, their gene expression profiles were compared to human fetal central nervous system (CNS) tissues, fetal spinal cord-derived NPCs, and to “default” H9-NSCs that were induced by conventional methods that lead to a default brain identity (see Example 7) using RNA-Sequencing. HOX cluster (HOX A-D) expression was observed in H9-derived spinal cord NSCs, but not in default H9-NSCs (
To assess the robustness of the differentiation protocol, it was repeated in another hESC line, UCSF4-ESCs, and in two human episomal induced pluripotent cell (iPSC) lines: RNA induced PSCs (RiPSCs) and IPS11 cells (see Example 7). hPSC-derived NSCs from these three additional cell lines acquired a spinal cord positional identity and maintained their regional identity and neurogenic potential over a long time period (>2 months;
A major challenge to the field of NSC transplantation is the development of NSC types that are optimized to treat a specific disease indication. In the case of spinal cord injury (SCI), driving grafted cells to spinal cord identities has been a key limiting factor to advancement of transplantation therapies. To examine whether the disclosed human spinal cord NSCs exhibit properties in this context, they were grafted into models of rat SCI. Adult athymic rats underwent cervical level 4 (C4) lesions of the dorsal spinal cord, which interrupt corticospinal projections, the most important voluntary motor system in humans. H9-derived spinal cord NSCs expressing green fluorescent protein (GFP) were grafted into lesion sites two weeks following injury (see Spinal Cord Surgeries in Example 7). When examined up to three months post-transplantation, grafted cells survived and extended very large numbers of axons (labeled with NF70) into the injured host spinal cord (
The specific neuronal lineages adopted by H9-derived spinal cord NSCs were identified. H9-derived spinal cord NSCs generated a variety of spinal interneuronal subtypes (
GFP-expressing human axons emerged from the lesion site in very large numbers and over very long distances (more than 10 spinal segments, a distance of 45 mm). Axons co-expressed human TAU, confirming their identity as axons, and grew in organized, linear rostro-caudal trajectories in the host dorsal column white matter (
To examine whether synapses were functional between host and the human neural graft, and the pattern of connectivity with host, mono-trans-synaptic rabies virus technology was utilized. A critical feature of this retrograde tracing system is exclusive transfer through functional synaptic connections, permitting unambiguous identification of interconnected neuronal circuitry. An H9-derived spinal cord NSC line was established that stably expressed: 1) GFP, 2) the TVA receptor, and 3) rabies glycoprotein. This vector allows for retrograde trans-synaptic spread of rabies expressing mCherry across one synapse following injection of glycoprotein deleted rabies virus (RVdG); moreover, rabies is exclusively transmitted by grafted human neural cells because only they possess the rabies TVA and glycoprotein to allow primary viral uptake and synaptic transport. This H9-derived spinal cord NSC line was grafted into sites of SCI two weeks after C4 spinal cord dorsal column lesions (n=4). Control animals were grafted with H9-derived spinal cord NSCs lacking the rabies glycoprotein and TVA (n=2). Animals received injections of RVdG into grafts four months post-transplantation, and were perfused a week later. In control animals, there were no mCherry+ cells in the host spinal cord, brainstem, or brain (
Graft connectivity was further assessed with host supraspinal (brain and brainstem) axonal systems. Of substantial importance was the observation that numerous mCherry-labeled neurons were observed in layer V of the motor cortex (
Corticospinal axons are of critical importance in controlling human voluntary movement, and spinal cord, but not forebrain, neural progenitor cells support robust corticospinal regeneration. To determine whether H9-ESCs driven to spinal cord identities support corticospinal regeneration, these cells were grafted to sites of SCI and compared their effects to H9-ESCs driven to rostral neuraxis identities. In the absence of the caudalizing morphogens CHIR and FGF2/8, H9-ESC-derived NSCs adopted a forebrain identity. Omission of only FGF2/8 resulted in NSCs of hindbrain identity. These results were confirmed by immunolabeling for the brain-specific marker OTX2 and the spinal cord-specific marker CDX2 (
hES cells (WA-09 (H9) provided from WiCell Research Institute; passages 33-41, UCSF4 provided by Dr. Susan Fisher (UCSF); passages P13-P23) were cultured in mTeSR medium (Stem Cell Technologies) on MATRIGEL® (Corning Inc.)—coated plates. Cells were passaged using Versene (Thermo Fisher Scientific), washed and replated at a dilution of 1:10. Two human episomal IPSC lines were used in this study. IPS11 was purchased from ALSTEM and RIPSCs was provided from Allele Biotechnology. IPS11 (passages; 1*-10*) were cultured under the same conditions to hES cells. RiPSC line (passages 23-30) was cultured in E8 TeSR media (Stem Cell Technologies) on vitronectin (Stem Cell Technologies)—coated plates and passaged at a dilution of 1:6 using Gentle Cell Dissociation Reagent (Stem Cell Technologies).
For spinal cord stem cell induction, at about 70% confluence, mTeSR medium was changed to N2B27 medium (knockout Dulbecco's Modified Eagle's Medium (DMEM)/F12: Neurobasal (1:1), 1×N2, 1×B27, 1× penicillin/streptomycin, 1× Glutamax) supplemented with 100 nM LDN-193189 (Stemgent), 10 μM SB431542 (Stemgent), 4 μM CHIR99021 (Stemgent), 1 μM DAPT (Stemgent), 100 ng/ml fibroblast growth factor (FGF) 2 (Peprotech, Rocky Hill, NJ) and 100 ng/ml FGF8 (Peprotech). The medium was replaced the following day with fresh neural induction media every day and cells were split 1:3 with Accutase (Innovative Cell Technologies Inc.). 10 μM Y-27632 (ROCK inhibitor; Stemgent) was used to enhance cell survival. 10 days after neural induction, neural induction media was switched to neural maintenance media (N2B27 medium supplemented with 2 μM SB431542, 3 μM CHIR99021, and 200 nM Hh-Ag 1.5). At passage 4, plate coating was changed from MATRIGEL® or vitronectin to CELLSTART® (Thermo Fisher Scientific), changed again at passage 6 to poly-L-ornithine (PLO; Sigma-Aldrich)/laminin (LAM; Sigma-Aldrich). The change in coating will accelerate neural differentiation, as MATRIGEL® and vitronectin tend to retain the immature stage while CELLSTART® and PLO favor differentiation into NSC. Neuronal differentiation was performed in N2B27 medium supplemented with 300 ng/mL cAMP (Sigma-Aldrich) and 0.2 mM vitamin C (Sigma-Aldrich) from passage 10 to passage 15 spinal cord NSCs. While cAMP and vitamin C can accelerate neuronal differentiation, these agents are not required for differentiation to occur.
The human spinal cord NPC line UCSD1113 (Passages; 4-18) generated from a 9-week-old fetal spinal cord cultured on plates coated with CELLSTART, using N2B27 medium supplemented with 2% of STEMPRO Neural Supplement (all from Life Technologies), 20 ng/ml FGF2, 20 ng/ml epidermal growth factor (EGF), and 10 ng/ml leukemia inhibitory factor (LIF). Default H9-NSCs (Passages; 1*-8*) purchased from Life Technologies were cultured with 20 ng/ml FGF2 and 20 ng/ml EGF on PLO/LAM coated plates.
To induce H9-forebrain NSCs, mTeSR medium was changed to N2B27 medium supplemented with 100 nM LDN-193189 and 10 μM SB431542. Neural induction media was changed every day and cells were split 1:3 with Accutase. Seven days after neural induction, induction media was switched to N2B27 medium supplemented with 20 ng/ml FGF 2, 20 ng/ml EGF, and 10 ng/ml LIF. At passage 4, plate coating was changed from MATRIGEL® to poly-D-lysine/laminin. To induce H9-forebrain NSCs, mTeSR medium was changed into N2B27 medium supplemented with LDN-193189, 10 μM SB431542, 4 μM CHIR99021, and 1 μM DAPT. From 10 days after neural induction, cells were cultured with exactly the same condition to H9-spinal cord NSCs.
Cultures were fixed for 30 minutes in 4% paraformaldehyde (PFA) in 0.1M PB at room temperature and post-fixed in methanol for 15 minutes. After being washed three times with Tris-buffered saline (TBS), fixed samples were permeabilized with 0.25% Triton X-100 with 5% normal horse serum in TBS for 1 h. Primary antibodies in the blocking solution were applied overnight. The primary antibodies used for immunolabeling are listed in
Total RNA was isolated from cultures using the RNeasy Mini kit (Qiagen) following the manufacturer's protocol. Human fetal brain and spinal Cord Poly A+ RNA (Clontech Laboratories, Inc.) were used for control. Total RNA was quantified with NanoDrop® (Thermo-Fischer). For cDNA synthesis, the reverse transcription reaction was performed using the PrimeScript™ RT Master Mix (Perfect Real Time, Clontech) and quantitative PCR was performed using primers specific for the genes of interest (see
H9-derived spinal cord NSCs were cultured on glass coverslips coated with POL/LAM and infected with lentivirus expressing GCaMP5 under control of the MAP2 promoter (generous gift from Russell C. Addis, UPenn). Cells on coverslips transferred into a recording chamber. Recordings were made in a submersion-type recording chamber and perfused with oxygenated ACSF containing (in mM) 119 NaCl, 2.5 KCl, 2 MgCl2, 2.5 CaCl2), 1.3 NaH2PO4, 26.0 NaHCO3, 20 glucose (˜295 mOsml) at 23° C. at a rate of 2-3 ml/minute. Whole-cell patch clamp recordings were obtained using Multiclamp 700B patch amplifiers (Molecular Devices) and data was analyzed using pClamp 10 software (Molecular Devices). Data were low-pass filtered at 2 kHz, and digitized at 10 kHz. Whole-cell voltage and current clamp recordings were made at room temperature using pulled patch pipettes (5-6 MO) filled with internal solution containing (150 mM K-gluconate, 1.5 mM MgCl2, 5.0 mM HEPES, 1 mM EGTA, 10 mM phosphocreatine, 2.0 mM ATP, and 0.3 mM GTP).
Total RNA was collected using the RNeasy Mini kit and stored at −70° until needed. Total RNA integrity was examined using the Agilent Bioanalyzer 2000. TrueSeq stranded mRNA-seq libraries were prepared from 5 μg of total RNA (Illumina mRNA-seq kit, RS-122-2103) and sequenced using Illumina HiSeq 2500 PE-100 (sequences publically available from GEO, accession number: GSE83107). TrueSeq stranded mRNA-seq libraries were prepared and sequenced using Illumina HiSeq 2500 at the IGM Genomics Center, University of California, San Diego. Unsupervised hierarchical clustering was performed in Cluster 3.0. Samples were clustered using uncentered Pearson correlation and average linkage. RNA-Seq data was downloaded from GEO, accession number GSM1944034 (fetal brain-derived NPCs) and GSM1381228-1381231 (default H9 NSCs) for unsupervised hierarchical clustering.
A total of 32 athymic nude rats were subjects of this study (150-180 g, The Jackson Laboratory). NIH guidelines for laboratory animal care and safety were strictly followed. Animals had free access to food and water throughout the study. All surgery was done under deep anesthesia using a combination (2 ml/kg) of ketamine (25 mg/ml), xylazine (1.3 g/ml), and acepromazine (0.25 mg/ml).
Rat C4 dorsal column lesions were made using a tungsten wire knife to transect all of dorsal column axons as described previously (Kadoya, K., et al. Spinal cord reconstitution with homologous neural grafts enables robust corticospinal regeneration. Nat Med (2016)). Briefly, the tungsten wire knife was inserted 1 mm from the dorsal surface of spinal cord and raised to transect the corticospinal tract, leaving a fragment of the most dorsal aspect of the ascending dorsal column sensory axons intact to create a closed lesion cavity with intact dura. Two weeks after C4 corticospinal lesions, a 2 μl suspension of 106 NSCs (viability >90%) were injected into the lesion site using a four growth factor cocktail consisting of 50 μg/ml BDNF (Peprotech), 10 μg/ml FGF2, 10 μg/ml VEGF (Peprotech), and 50 μM MDL28170. The following groups and subject numbers were studied.
For the generation of NSCs expressing EnvA-pseudotyped rabies helper proteins, H9 NSCs previously transduced with lentivirus expressing CAG-GFP were transduced with a polycistronic lentivirus expressing TVA, SAD-B19 rabies G-protein, and GFP, under control of the CAG promoter (Salk Viral Vector Core Facility). After several passages (at least 3) and caudalization, H9-derived GFP+ NSCs expressing CAG-TVA-G-protein were grafted into lesion sites After 4-5 months of graft maturation, grafts were stereotactically injected with a total of 2.5 μL EnvA pseudotyped G-deleted (SADAG) rabies expressing mCherry (1 E7vg/mL in PBS) into the graft core and along the graft/host interface evenly across 15 sites, with three injection depths at each site (1, 0.7, and 0.5 mm), using pulled glass micropipettes and a PicoSpritzer, as guided by visualization of the graft with a GFP-excitation flashlight. Rats were perfused 7 days later.
Animals were perfused with 4% paraformaldehyde in 0.1 M phosphate buffer (pH 7.2). Spinal cord were removed, post-fixed and sectioned on a vibratome set at 30 μm intervals. Sections were incubated with primary antibodies overnight (see
For the quantification of neural differentiation in human cell grafts, Hu, human GFAP, NG2, CaMKII, ChAT, GABA, GlyT2, FOXG1, or LHX3 and NuNu were used. By using microscopy, cells were sampled in images (100× or 200× magnification) from randomly selected regions of grafts. The proportion of neural or neuronal marker expressing cells to total number of HuNu was then calculated and averaged among groups. The number of corticospinal axons regenerating into grafts in lesion sites was quantified using images taken by BZ-9000 digital microscope system as previously described (Kadoya, K., et al. Spinal cord reconstitution with homologous neural grafts enables robust corticospinal regeneration. Nat Med (2016)). Briefly, 1-in-6 sections were labeled for rCOMET-traced corticospinal axons. Dorsal-to-ventral virtual lines were placed and then examined under 400× magnification. RCOMET-labeled axons that intersected the line were marked and counted. Two sagittal sections containing the corticospinal main track were quantified. In all cases, observers were blinded to group identity.
Multiple group comparisons were made using one-way analysis of variance (ANOVA; JMP software) at a designated significance level of 95%. Non-parametric data were assessed using Mann-Whitney. Data are presented as mean±SEM. As stated above, all quantifications were performed by observers blinded to group identity.
Neural stem cells have exhibited efficacy in pre-clinical models of spinal cord injury (SCI) and are on a translational path to human testing. Neural stem cells must be driven to a spinal cord fate to optimize host axonal regeneration into sites of implantation in the injured spinal cord, where they subsequently form neural relays across the lesion that support significant functional improvement. Methods of deriving and culturing human spinal cord neural stem cells derived from embryonic stem cells that can be sustained over serial high passage numbers in vitro have been developed (as discussed above), providing a potentially optimized cell source for human clinical trials.
However, some studies of the spinal cord neural stem cells have revealed that spinal cord neural stem cells occasionally exhibited karyotypic abnormalities, and that some of these abnormalities were clonal in nature. Accordingly, the present technology includes modified methods for generating spinal cord neural stem cells to reduce rates of cell division, minimizing the risk that errors would occur during cell division that resulted in chromosomal aberrations. This development improves prospects for safe human translation.
A range of concentrations of reagents were tested to induce neural differentiation to reducing the extent of cell proliferation while retaining the intended differentiation of cells into a spinal cord neural stem cell identity. Table 1 and Table 2 lists the range of concentrations of FGF2, FGF-8b, CHIR99021, LDN-193189, SB431542, and DAPT tested over numerous rounds of induction of H9 embryonic stem cells to a spinal cord neural stem cell fate.
H9 ESCs were obtained from WiCell (Madison, WI) and expanded as colonies in mTesR1 media (Stem Cell Technology) on MATRIGEL® (Corning)-coated plates. Three days prior to neural induction, ESCs were split at a 1:10 ratio and plated in mTesR media on MATRIGEL®. On day 0, ESCs were changed to neural induction media composed of knockout DMEM/F12, Neurobasal (1:1 ratio), 2 mM GlutaMAX®, 1×Pen/Strep, 1×N2, 1×B27 minus vitamin A, 100 nM LDN193189, and a range of concentrations of CHIR99021 (0-4 μM), SB431542 (0-10 μM), FGF-2 (25-200 ng/ml), and FGF-8b (25-100 ng/ml) (Table 1 and Table 2). Media was changed daily and cells split at a ratio of 1:3 on day 4 onto MATRIGEL®-coated plates. On day 7, LDN was removed from the neural induction media. On day 10, media was changed to neural maintenance media consisting of knockout DMEM/F12, Neurobasal (1:1 ratio), 2 mM GlutaMAX, 1×Pen/Strep, 1×N2, 1×B27 minus vitamin A, 2 μM SB431542, 2 μM CHIR, 200 nM Hh-Ag 1.5. On day 12, cells were split at a ratio of 1:3 onto plated coated with poly D-lysine (Sigma, 100 μg/ml, O/N at 37° C.) and mouse laminin (Sigma, 10 μg/ml, 2 hr at 37° C.).
Side-by-Side Comparison of Original (Method 1) with Modified (Method 2) Protocol
H9-ESCs cultured in mTeSR1 (Stem Cell Technology) were split at a ratio of 1:6 and plated onto Matrigel-coated 6-well plates (Corning). The next day media was changed to Method 1 or Method 2 (Table 3, below) neural induction media; media was subsequently changed daily. Cells were split on days 2 and 6 with TrypLE and re-plated at 200,000 cells/cm2 onto Matrigel-coated 6-well plates. On day 10, media was replaced with neural maintenance media containing knockout DMEM/F12:Neurobasal (1:1), 2 mM GlutaMAX, 1×Pen/Strep, 2 μM CHIR99021, 2 μM SB431542, 200 nM Hh-Ag1.5. Additionally, on day 10, RNA was isolated from approximately 1×106 cells with the RNAeasy miniprep kit (Qiagen) resulting in 1.2±0.2 μg total RNA per sample. 1 μg RNA was used to generate cDNA with the Qiagen first-strand cDNA synthesis kit and 5 ng of cDNA for used for each PCR reaction. On day 12, H9-scNSCs were re-plated at 200,000 cells/cm2 onto PDL/laminin (Sigma)-coated flasks. Cells were expanded until day 30 with daily media changes and split every 4 days. On day 30, cells were analyzed for RNA, as described above for day 10.
On day 30, spinal cord NSCs were dissociated into single cells with Accutase and washed with cold DPBS. For each flow cytometry labeling, 1×106 cells were transferred to protein low-bind Eppendorf tubes and incubated with the live/dead dye, zombie green (1:500, Biolegend) for 15 minutes. Cells were washed with cold DPBS and centrifuged at 500 rcf for 5 min.
For in vitro neuronal differentiation, dissociated cells were plated on chamber slides coated with poly l-lysine (100 μg/ml) and mouse laminin (25 μg/ml). The next day media was changed to neuronal differentiation media consisting of neurobasal plus, B27-plus and culture-1 supplements, 200 μM ascorbic acid, 1 μM db-cAMP and 10 μg/ml mouse laminin. Half media changes were performed every 2-3 days for 14 days. At 14-days, cells were fixed with 4% paraformaldehyde and immunolabeled with antibodies to chicken anti-MAP2 (0.5 μg/ml from Millipore) and mouse anti-TAU (1 μg/ml from Biolegend) and imaged on an FV3000 confocal microscope.
For in vitro astrocyte differentiation, dissociated cells were plated on chamber slides coated with poly l-lysine (100 μg/ml) and mouse laminin (25 μg/ml). The next day media was changed to astrocyte differentiation media (Stem Cell Technologies). Half media changes were performed every 2-3 days for 14 days. At 14-days, cells were fixed with 4% paraformaldehyde and immunolabeled with antibodies to mouse anti-GFAP (1 μg/ml from Millipore) and goat anti-SOX9 (1 μg/ml from R&D Systems) and imaged on an FV3000 confocal microscope.
For cell surface screening, BD Lyoplates (BD Sciences) were used containing an array of 242 purified monoclonal antibodies to cell surface markers. Cells were labeled according to manufacturer's instructions and analyzed with a Acea Novocyte flow cytometry machine equipped with a high throughput 96-well adapter.
For single cell flow cytometry, dissociated cells were incubated with live/dead stain zombie green (1:500 in DPBS, Biolegend) for 15 min at room temperature in the dark. DPBS was added to cells and centrifuged 500 rcf for 5 min at 4° C. Cells were then re-suspended in flow cytometry staining buffer (BD Sciences) containing fluorescent conjugated antibodies: VioBlue-SSEA-4, APC-CD24, APC-PSA-NCAM, APC-CD133, Pacific Blue-A2B5 (all 1:100 from Miltenyi Biotech). Cells were labeled for 30 min on ice in the dark, then washed with cold DPBS. Following centrifugation, cells were re-suspended in 300 μl staining buffer and sorted with an Acea Novocyte 3000 flow cytometer. 30,000 events were collected with the “medium” setting at a 35 μl/min flow rate with a 12.2 μm diameter nozzle. Based on previous work, the markers listed in Table 4 represented the intended spinal cord neural stem cell fate.
Karyotype analyses (G-banding) were performed by Cell Line Genetics LLC (Madison, WI) or UCSD Cytogenetics Lab (La Jolla, CA) from live cell cultures.
Having identified a putative optimized regimen for generating spinal cord NSCs in vitro, these cells were grafted in vivo in models of spinal cord injury (SCI) to determine whether they exhibited the predicted properties of: 1) successful engraftment, with neural stem cell fill of the lesion site; 2) extension of graft axons in large numbers and over distances into the distal host spinal cord; and 3) regeneration of host axons into the grafts. Athymic nude female rats underwent T10 moderate spinal cord contusions. The Infinite Horizon device (PSI Systems) equipped with a 2.5 mm round impactor tip was used to deliver a force of 150 kDyns. Animals received 0.5 mg/kg ampicillin and 1 mg/kg banamine daily for 3 days post-injury and bladders were manually expressed twice per day for 7-10 days. Two weeks later, the candidate neural stem cells were grafted into the lesion site. H9-spinal cord NSCs at day 30 in vitro (passage 5) were suspended at a concentration of 2×106 cells/ml, and two aliquots of 1×106 cells were transferred to Eppendorf tubes and centrifuged 3,500 rpm for 5 min. For grafting, the pellet was resuspended in 3 μl each of human fibrinogen (Sigma, 20 μg/ml in PBS) or human thrombin (Sigma, 20 U/ml in 10 mM CaCl2) with supplemental growth factor to improve cell survival after grafting: 50 μg/ml BDNF, 10 μg/ml FGF-2, 10 μg/ml VEGF-165 (all from Peprotech) and 50 μM MDL-28170 (Aobious). Three microliters of the cell slurry was injected into the lesion cavity. Animals received 0.5 mg/kg ampicillin and 1 mg/kg banamine daily for 3 days afterward. For anterograde tracing of the corticospinal tract, AAV9 vectors were used that expressed a codon-optimized membrane-targeted tdTOMATO (Charles River, formally Vigene Biosciences). AAV9-tdTOMATO was injected into both motor cortices with a pulled glass pipette attached to a PicosSpritzer II (General Valve). AAV9-tdTOMATO (5×1012 genome copies/ml) was injected into 8 sites per hemisphere (stereotaxic coordinates relative to bregma: Antero-posterior −0.5, −0.1, −0.15, −0.2 cm; Medio-lateral 0.18 or 0.25 cm; Dorso-ventral depth 0.12 cm) at a volume of 0.5 μl per site. Rats were sacrificed 1 month later.
Rats were perfused with cold 4% paraformaldehyde in 0.1M phosphate buffer and spinal cord were post-fixed overnight in 4% PFA at 4° C. After three days in 30% sucrose solution, spinal cords were sectioned in the transverse plane on a microtome set at 35 μm intervals. A series of 1-in-12 sections were immunolabeled with antibodies directed against mouse human-specific nuclei (Millipore, 2.5 μg/ml), guinea pig NeuN (to identify neurons; Millipore, 1 μg/ml), rabbit KI-67 (to identify dividing cells; Abcam, 2 μg/ml), mouse STEM121 (to identify human cytoplasm; Takara, 1 μg/ml), human-specific TAU (to identify graft-derived axons; Biolegend 2 μg/ml), rabbit GFAP (to identify all species of astrocytes; Dako, 1 μg/ml), and mouse STEM123 (to identify human astrocytes; Takara, 1 μg/ml). Confocal images of labeled sections containing the lesion/graft site were acquired on an Olympus FV3000 microscope with a 20× air objective and a 2× zoom. Two random regions within the graft from 2 sections (approximately 400 μm apart) were imaged for quantification. The total number of hNuc, NeuN and KI67 cells were quantified using ImageJ software. Images were imported into ImageJ and split into individual channels. Individual channels were thresholded, then processed using the watershed function. The analyze particles function was used to count the number of cells with the following parameters: Particle size: 100-infinity, 0.3-1 circularity, exclude edges, include holes.
Different protocols were assessed for inducing human spinal cord neural stem cells from starter pluripotent embryonic stem cells. The different protocols varied the concentrations as shown in Table 1 and Table 2. Ultimately a set of parameters was identified that consistently generated the intended cells with stable karyotype. This method consisted of the reagents at the concentrations listed in Table 4.
Using these methods, production of neural stem cells with a spinal cord identity were confirmed using flow cytometry and quantitative PCR at 5 and 15 days post-neural induction. At 5 days post-neural induction, cells exhibited a rapid loss of OCT4 protein mRNA expression. CDX2, a marker of a neuro-mesodermal intermediate cell, was present in ˜50% of cells, while >80% of cells immunolabeled for neural stem cell markers SOX1, SOX2 and Nestin (
Karyotypic analyses of spinal cord neural stem cells generated with the modified methods are depicted in
Three independent cell lines were generated comparing the protocol of the present technology (“modified protocol” or “Method 2,”
To demonstrate reproducibility of the optimized protocol for producing human spinal cord neural stem cells, five separate cell batches were repeatedly generated using the same modified protocol. Thirty days post-neural induction, the cells were immunolabeled for Nestin, SOX1 and SOX2 (
Flow cytometry was also used to examine consistency in expression of neural stem cell markers among the five different NSC batches (
The neuronal differentiation potential of the modified spinal cord NSC protocol was confirmed by subjecting cells to differentiation conditions for 14 days after completion of neural induction. Flow cytometry after differentiation indicated that greater than 95% of cells immunolabeled for the neuron-specific marker B3-tubulin (
Next, it was examined whether human spinal cord NSCs generated with the modified methods would successfully engraft, survive, and undergo neuronal and glial differentiation in vivo. All five batches of spinal cord NSCs generated above were evaluated in immunodeficient rats that underwent T10 contusion injuries two weeks earlier. Each sample cell batch was grafted into four animals (total N=20 rats). All cell batches survived engraftment (
To assess in vivo neuronal differentiation of each NSC batch 12 weeks after grafting, sections were immunolabeled for NeuN, a marker of mature neurons, and the human cell-specific marker HuNu (
The proportion of grafted cells undergoing division was also quantified using the marker Ki67 (
The extent of astrocytic differentiation was characterized by immunolabeling with human-specific GFAP antibodies. All batches showed differentiation into astrocytes (
A hallmark of neural stem cells implanted into sites of SCI is the extraordinary outgrowth of axons from grafts into the host spinal cord (Cell 2012; Nat Med 2016; Sci Transl Med 2017). To assess whether the modified methods for generating human spinal cord NSCs also supported extensive axonal outgrowth into the host, spinal cord sections were immunolabeled for the human-specific axonal marker Human Tau. Very dense numbers of human Tau+ axons extended outward from grafts placed into the T10 spinal cord contusion cavity (
Another hallmark of spinal cord neural stem cells that are grafted into lesion cavities is that they support regeneration of injured host axons. To assess host axonal regeneration into the graft, host serotonergic axons (5HT) were immunolabeled. Indeed, host serotonergic axons penetrated grafts (
To determine the safety and tolerability of the H9-spinal cord NSC batches over three months, a functional observational battery (FOB) was performed weekly and compared outcomes to animals that received lesions alone. On this scale, there were no significant differences among groups over weeks (ANOVA P=0.6;
Embryonic stem cells driven to a spinal cord identity represent a potential therapy for spinal cord injury. Improved methods have now been identified for generating these cells from the parent H9 ES cell line that generates cells that are sustainable in vitro over serial passages and, importantly, that exhibit improved karyotypic stability compared to previous methods. This improved protocol supports a path to clinical translation.
The newly generated cell lines exhibit considerable batch-to-batch consistency in vitro, as demonstrated by immunolabeling, PCR and a panel of flow cytometry markers. Karyotypic analysis also demonstrates considerable consistency of the different batches, with only occasional karyotypic abnormalities that were not sustained over higher passage numbers. In future clinical manufacturing of these cells, cellular identify markers and karyotype in all master cell banks could be sampled, and eliminate those that did not meet criteria for cell identity or karyotype. For example, if a master cell bank exhibited expanding karyotypic abnormalities over higher passage numbers or clonal abnormalities, that bank would be discarded.
The newly generated cell lines also exhibit good batch-to-batch consistency on in vivo grafting in terms of survival, graft axon outgrowth and host axon ingrowth. One of five spinal cord NSC batches was inferior to the other batches in measures of in vivo survival and fill of the lesion cavity. Based on this observation, candidate master cell banks generated for clinical use undergo in vivo testing, and any banks that exhibit inferior survival be discarded. The present technology suggest that this might occur in 20% of batches.
All five grafted batches of spinal cord neural stem cells exhibited good in vivo safety at the three-month time point.
Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term “about.” As used herein the terms “about” and “approximately” means within 10 to 15%, preferably within 5 to 10%. Accordingly, unless indicated to the contrary, the numerical parameters set forth in the specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by the present invention. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value, however, inherently contains certain errors necessarily resulting from the standard deviation found in their respective testing measurements.
The terms “a,” “an,” “the” and similar referents used in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention otherwise claimed. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the invention.
Groupings of alternative elements or embodiments of the invention disclosed herein are not to be construed as limitations. Each group member may be referred to and claimed individually or in any combination with other members of the group or other elements found herein. It is anticipated that one or more members of a group may be included in, or deleted from, a group for reasons of convenience and/or patentability. When any such inclusion or deletion occurs, the specification is deemed to contain the group as modified thus fulfilling the written description of all Markush groups used in the appended claims.
Certain embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Of course, variations on these described embodiments will become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventor expects skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.
Specific embodiments disclosed herein may be further limited in the claims using consisting of or consisting essentially of language. When used in the claims, whether as filed or added per amendment, the transition term “consisting of” excludes any element, step, or ingredient not specified in the claims. The transition term “consisting essentially of” limits the scope of a claim to the specified materials or steps and those that do not materially affect the basic and novel characteristic(s). Embodiments of the invention so claimed are inherently or expressly described and enabled herein.
Furthermore, numerous references have been made to patents and printed publications throughout this specification. Each of the above-cited references and printed publications are individually incorporated herein by reference in their entirety.
In closing, it is to be understood that the embodiments of the invention disclosed herein are illustrative of the principles of the present invention. Other modifications that may be employed are within the scope of the invention. Thus, by way of example, but not of limitation, alternative configurations of the present invention may be utilized in accordance with the teachings herein. Accordingly, the present invention is not limited to that precisely as shown and described.
This application claims the benefit of U.S. Provisional Application No. 63/520,841, filed Aug. 21, 2023. This application is also related to U.S. patent application Ser. No. 16/530,777 filed Aug. 2, 2019, which claims the benefit of U.S. Provisional Application No. 62/714,590 filed on Aug. 3, 2018. The entire contents of the referenced applications are incorporated herein by reference.
Number | Date | Country | |
---|---|---|---|
63520841 | Aug 2023 | US |