Cell systems using spheroids and methods of making and using them

文档序号:1145049 发布日期:2020-09-11 浏览:37次 中文

阅读说明:本技术 使用球状体的细胞系统以及制造和使用它们的方法 (Cell systems using spheroids and methods of making and using them ) 是由 M·J·穆尔 J·L·科尔里 A·D·沙尔玛 D·波泽尔 J·本恩 于 2018-12-04 设计创作,主要内容包括:本公开总体上涉及细胞培养系统,并且特别地涉及用于神经元细胞的三维细胞培养系统,所述三维细胞培养系统促进模拟体内神经纤维的结构与功能特征的结构与功能特征,包括细胞髓鞘形成。利用双重水凝胶构建体以及包含神经元细胞的球状体,本公开提供了用于允许细胞内和细胞外电生理学测量和记录的体外空间受控的三维模型的方法、装置和系统。所述三维水凝胶构建体允许掺入的细胞类型、几何制造以及电操纵的灵活性,从而提供了用于培养、干扰以及测试具有生理相关结果的仿生神经生长的可行的系统。(The present disclosure relates generally to cell culture systems, and in particular to three-dimensional cell culture systems for neuronal cells that facilitate structural and functional features that mimic structural and functional features of nerve fibers in vivo, including cell myelination. The present disclosure provides methods, devices and systems for spatially controlled three-dimensional models in vitro that allow for intracellular and extracellular electrophysiological measurements and recordings, using dual hydrogel constructs and spheroids comprising neuronal cells. The three-dimensional hydrogel constructs allow flexibility in the type of cell incorporated, geometric fabrication, and electrical manipulation, providing a viable system for culturing, perturbing, and testing biomimetic nerve growth with physiologically relevant results.)

1. A composition comprising spheroids of cells comprising one or a combination of cells and/or tissues selected from the group consisting of: neuronal cells, nervous system ganglia, stem cells, and immune cells.

2. The composition of claim 1, wherein the spheroids comprise a tissue selected from the group consisting of: dorsal root ganglia and trigeminal ganglia.

3. The composition of claim 1 or 2, wherein the spheroids comprise one or more cells selected from the group consisting of: glial cells, embryonic cells, mesenchymal stem cells, cells derived from induced pluripotent stem cells, sympathetic neurons, parasympathetic neurons, spinal cord motor neurons, central nervous system neurons, peripheral nervous system neurons, enteric nervous system neurons, motor neurons, sensory neurons, cholinergic neurons, GABAergic neurons, glutamatergic neurons, dopaminergic neurons, hydroxytryptamine neurons, interneurons, adrenergic neurons, trigeminal ganglia, astrocytes, oligodendrocytes, Schwann cells, microglia, ependymal cells, radial glial cells, satellite cells, enteric glial cells, pituitary cells, and combinations thereof.

4. The composition of any one of claims 1 to 3, wherein the spheroids comprise one or more immune cells selected from the group consisting of: t cells, B cells, macrophages, astrocytes and combinations thereof.

5. The composition of any one of claims 1 to 4, wherein the spheroids comprise one or more stem cells selected from the group consisting of: embryonic stem cells, mesenchymal stem cells, induced pluripotent stem cells, and combinations thereof.

6. The composition of any one of claims 1 to 5, wherein the neuronal cells are derived from stem cells selected from the group consisting of: embryonic stem cells, mesenchymal stem cells, and induced pluripotent stem cells.

7. The composition of any one of claims 1 to 6, wherein the spheroids have a diameter of from about 200 microns to about 700 microns.

8. The composition of any one of claims 1-7, wherein the spheroid comprises one or more neuronal cells and one or more Schwann cells, the ratio of cell types being equal to about 4 neuronal cells per 1 Schwann cell.

9. The composition of any one of claims 1-8, wherein the spheroid comprises one or more neuronal cells and one or more astrocytes at a ratio of about 4 neuronal cells per 1 astrocyte.

10. The composition of any one of claims 1-9, wherein the spheroid comprises one or more neuronal cells and one or more astrocytes at a ratio of about 1 neuronal cell per 1 astrocyte.

11. The composition of any one of claims 1-10, wherein the spheroid comprises one or more neuronal cells and one or more schwann cells at a ratio of about 10 neuronal cells per 1 schwann cell.

12. The composition of any one of claims 1 to 11, wherein the spheroid comprises one or more neuronal cells and one or more glial cells at a ratio of about 4 neuronal cells per 1 glial cell.

13. The composition of any one of claims 1 to 12, wherein any one or more cells are differentiated from induced pluripotent stem cells.

14. The composition of any one of claims 1 to 13, wherein the spheroids are free of induced pluripotent stem cells and/or immune cells.

15. The composition of any one of claims 1 to 14, wherein the spheroids are free of undifferentiated stem cells.

16. The composition of any one of claims 1-15, wherein the spheroid comprises no less than about 30,000, 35,000, 40,000, 45,000, 50,000, 55,000, 60,000, 65,000, 70,000, 75,000, 80,000, 90,000, 100,000, 150,000, 200,000, 225,000, or 250,000 cells.

17. The composition of any one of claims 1-16, wherein the spheroid comprises no less than 75,000 cells.

18. The composition of any one of claims 1 to 17, wherein the spheroids further comprise one or more magnetic particles.

19. A system, comprising:

(i) a cell culture vessel comprising a hydrogel;

(ii) one or more spheroids comprising one or more neuronal cells and/or isolated tissue explants;

(iii) an amplifier including a current generator;

(iv) a voltmeter and/or ammeter; and

(v) at least a first stimulating electrode and at least a first recording electrode;

wherein the amplifier, voltmeter and/or ammeter, and electrode are electrically connected to each other by an electrical circuit, wherein current is fed from the amplifier to the at least one stimulating electrode, and current is received at the recording electrode and fed to the voltmeter and/or ammeter; wherein the stimulating electrodes are positioned at or adjacent to one or more cells of the neuronal cells and/or isolated tissue explants and the recording electrodes are positioned at a predetermined distance distal to the cells such that an electric field is established across the cell culture container.

20. The system of claim 19, wherein the spheroid is a spheroid of any one of claims 1 to 18.

21. The system of any one of claims 19 or 20, wherein the culture vessel comprises 96, 192, 384 or more internal chambers.

22. The system of claim 21, wherein the 96, 192, 384 or more internal chambers comprise one or more isolated schwann cells and/or one or more oligodendrocytes sufficiently proximal to the one or more isolated tissue explants and/or the one or more neuronal cells such that the schwann cells or the oligodendrocytes deposit myelin to grow axons from the tissue explants and/or neuronal cells.

23. The system of any one of claims 19 to 22, further comprising a solid substrate having a hydrogel matrix crosslinked thereto, the solid substrate comprising at least one plastic surface having pores with a diameter of about 1 micron to about 5 microns.

24. The system of claim 23, wherein the solid substrate comprises a continuous outer surface and an inner surface, such solid substrate comprising at least one portion in a cylindrical or substantially cylindrical shape and at least one hollow interior defined at its edges by at least a portion of the inner surface, the inner surface comprising one or more pores having a diameter of about 0.1 microns to about 1.0 microns, wherein the hollow interior of the solid substrate is accessible from a point external to the solid substrate through at least one opening; wherein the hollow interior portion comprises a first portion adjacent the opening and at least a second portion distal to the opening; wherein the one or more neuronal cells and/or the one or more tissue explants are positioned at or adjacent to a first portion of a hollow interior and in physical contact with the hydrogel matrix, and wherein the second portion of the at least one hollow interior is in fluid communication with the first portion such that axons can grow from the one or more neuronal cells and/or the one or more tissue explants into the second interior portion of the hollow interior.

25. The system of any one of claims 19 to 24, wherein the composition is free of a sponge.

26. The system of any one of claims 19 to 25, wherein the hydrogel comprises at least a first cell-impenetrable polymer and a first cell-penetrable polymer.

27. The system of claim 26, wherein the at least one cell-impenetrable polymer comprises no more than about 15% PEG, and the at least one cell-penetrable polymer comprises about 0.05% to about 1.00% of one or a combination of self-assembling peptides selected from the group consisting of: RAD16-I, RAD16-II, EAK 16-I, EAK 16-II and dEAK 16.

28. The system of any one of claims 19-24, wherein the composition is free of polyethylene glycol (PEG).

29. The system of any one of claims 19 to 28, wherein the hydrogel comprises a first region and a second region, the first region is formed in the shape of a cylinder or rectangular prism oriented with its longitudinal axis through the top and bottom of the cell culture vessel, and each of the cylinder or rectangular prism comprises a space defined by the inner surface of the cylinder or rectangular prism, the space accessible through the top of the cell culture vessel via one or more openings; wherein the second region includes a space formed in the shape of an inner wall thereof, and an opening on a side surface thereof is adjacent to and in fluid communication with the first region.

30. The system of any one of claims 19 to 29, further comprising a cell culture medium comprising Nerve Growth Factor (NGF) at a concentration of about 5 to about 20 picograms per milliliter, and/or ascorbic acid at a concentration in the range of about 0.001% volume by volume to about 0.01% volume by volume.

31. The system of any one of claims 19 to 30, wherein the one or more spheroids comprise at least one or a combination of cells selected from: glial cells, embryonic cells, mesenchymal stem cells, cells derived from induced pluripotent stem cells, sympathetic neurons, parasympathetic neurons, spinal cord motor neurons, central nervous system neurons, peripheral nervous system neurons, enteric nervous system neurons, motor neurons, sensory neurons, cholinergic neurons, GABAergic neurons, glutamatergic neurons, dopaminergic neurons, hydroxytryptamine neurons, interneurons, adrenergic neurons, trigeminal neurons, astrocytes, oligodendrocytes, Schwann cells, microglia, ependymal cells, radial glial cells, satellite cells, enteric glial cells, and pituitary cells.

32. The system of any one of claims 19 to 31, further comprising one or more of stem cells, pluripotent cells, myoblasts, and osteoblasts.

33. The system of any one of claims 19-32, wherein the one or more neuronal cells comprise primary mammalian cells derived from the peripheral nervous system of the mammal.

34. The system of any one of claims 19-33, wherein the hydrogel comprises at least 1% polyethylene glycol (PEG).

35. The system of any one of claims 19-34, wherein the spheroids are in culture for no less than about 3, 30, 90, or 365 days.

36. The system of any one of claims 19 to 35, wherein at least a portion of the solid substrate is cylindrical or substantially cylindrical such that at least a portion of an inner surface of the solid substrate defines a cylindrical or substantially cylindrical hollow interior chamber in which the spheroid is positioned.

37. The system of any one of claims 19-36, wherein the one or more spheroids comprise one or more neuronal cells having axons that grow about 100 microns to about 500 microns in width and about 0.11 microns to about 10,000 microns in length.

38. The system of any one of claims 19 to 37, wherein the hydrogel comprises a series of two or more cavities in fluid communication with each other through a series of channels, at least one cavity comprising a spheroid, and at least a second cavity comprising a second spheroid, a suspension of cells, or a DRG; wherein the spheroid and the second spheroid, the suspension of cells, or the DRG are connected by a three-dimensional axon.

39. The system of claim 38, wherein the three-dimensional axon has a height at its lowest point of at least about 10 microns, or at least three monolayers of cells in height.

40. The system of claim 38, wherein the cavity is a well with a U-shaped well or circular well positioned in a horizontal or substantially horizontal plane of a solid substrate, wherein each channel comprises one or more axons connecting one or more spheroids.

41. The system of claim 40, wherein the system comprises a first spheroid comprising:

(i) one or more neuronal cells; and/or

(ii) One or more schwann cells or oligodendrocytes; and

a second spheroid comprising:

(i) one or more peripheral neurons;

wherein each spheroid is positioned in the cavity.

42. The system of claim 40, wherein the system comprises a first cavity, a second cavity, and a third cavity, each cavity configured to hold a spheroid and at least 50 microliters of cell culture medium, wherein the cavities are aligned such that the first cavity is positioned proximal to the second cavity and distal to the third cavity.

43. The system of claim 42, wherein the system comprises at least a fourth cavity, the cavities positioned therein in a pattern such that each cavity defines a corner of a square.

44. The system of claim 42, wherein the cavities are aligned in a line such that axons originating from the first spheroid in the first cavity extend to the second cavity and axons from the spheroid in the second cavity extend to axons in the third cavity.

45. A method of making a three-dimensional culture of one or more spheroids in a culture vessel comprising a solid substrate, the method comprising:

(a) contacting one or more neuronal cells with the solid substrate, the substrate comprising at least one outer surface, at least one inner surface, and at least one interior chamber defined by the at least one inner surface and accessible from a point external to the solid substrate through at least one opening;

(b) positioning one or more spheroids comprising neuronal cells into the at least one internal chamber; and

(c) applying cell culture medium to the culture vessel, wherein the volume of cell culture medium is sufficient to cover the at least one spheroid; wherein at least a portion of the inner surface comprises a first cell-impenetrable polymer and a first cell-penetrable polymer.

46. The method of claim 45, wherein step (b) comprises locating spheroids comprising tissue explants selected from one or a combination of: isolated dorsal root ganglia, spinal cord explants, retinal explants, and cortical explants.

47. The method of claim 45 or claim 46, wherein the spheroid is formed as a suspension of neuronal cells selected from one or a combination of: motor neurons, sensory neurons, sympathetic neurons, parasympathetic neurons, cortical neurons, spinal neurons, peripheral neurons, optionally derived from stem cells.

48. The method of any one of claims 45-47, wherein the spheroid further comprises isolated Schwann cells and/or oligodendrocytes.

49. The method of any one of claims 45 to 48, further comprising step (d): allowing the spheroids to grow neurites and/or axons after step (c) for a period of about 12 hours to about 1 year.

50. The method of any one of claims 45 to 49, further comprising the step of:

(i) isolating one or more neural cells from the sample prior to step (a); and/or

(ii) Isolating Dorsal Root Ganglia (DRGs) from one or more mammals prior to step (b) if said one or more spheroids comprise DRGs; and/or

(iii) Isolating one or more Schwann cells and/or one or more oligodendrocytes if the one or more spheroids comprise Schwann cells or oligodendrocytes.

51. The method of any one of claims 45 to 50, further comprising positioning at least one stimulating electrode at or adjacent to a cell body of the one or more neuronal cell or tissue explants and positioning at least one recording electrode at or adjacent to an axon at a point most distal to the cell body such that, upon introduction of an electrical current in the stimulating electrode, the recording electrode is capable of receiving a signal corresponding to one or more electrophysiological metrics capable of being measured at the recording electrode;

wherein the one or more electrophysiological metrics are one or a combination of: a conductance rate, an action potential, an amplitude of a wave associated with passage of an electrical pulse along a membrane of one or more neuronal cells, a width of the electrical pulse along the membrane of one or more neuronal cells, a latency of the electrical pulse along the membrane of one or more neuronal cells, and an envelope of the electrical pulse along the membrane of one or more neuronal cells.

52. A method of evaluating the toxicity and/or neuroprotective effect of an agent, the method comprising:

(a) culturing one or more spheroids in the composition of any one of claims 1 to 18;

(b) exposing at least one agent to the one or more spheroids; and

(c) measuring and/or observing one or more morphometric changes and/or one or more electrophysiological indicators of the one or more spheroids.

53. A method of measuring myelination or demyelination of one or more axons of one or more spheroids, the method comprising:

(a) culturing one or more spheroids in the composition of any one of claims 1 to 18 in the presence or absence of an agent for a time and under conditions sufficient for at least one axon to grow; and

(b) detecting an amount of myelination on one or more axons from the one or more spheroids; wherein the detection optionally comprises the steps of:

(i) measuring and/or observing one or more morphometric changes and/or one or more electrophysiological indicators of the one or more spheroids in the presence or absence of an agent; and

(ii) correlating one or more morphometric changes and/or one or more electrophysiological indicators of the one or more spheroids in the presence or absence of an agent with a quantitative or qualitative change in myelination of the spheroids.

54. A method of detecting and/or quantifying neuronal cell growth and/or axonal degeneration, the method comprising:

(a) quantifying the number or density of one or more spheroids and/or axons growing from the spheroids;

(b) culturing the one or more spheroids in the composition of any one of claims 1 to 18; and

(c) after culturing the spheroids for a period of time sufficient to allow growth of one or more axons or cell growth in the spheroids, calculating the number of cells within the spheroids and/or the number or density of axons growing from spheroids in the composition;

wherein step (b) optionally comprises contacting the one or more spheroids with one or more agents; wherein step (c) optionally comprises detecting internal and/or external recordings of one or more spheroids after culturing such spheroids, and correlating said recordings to measurements of the same recordings corresponding to a known or control number of cells; or

Wherein step (c) optionally comprises the additional steps of: (i) measuring intracellular and/or extracellular recordings and/or morphometric changes before and after the step of contacting the one or more spheroids with one or more agents; and (ii) correlating the difference in the recorded and/or morphometric change prior to contacting the one or more spheroids with the one or more agents and the recorded and/or morphometric change after contacting the one or more spheroids with the one or more agents to a change in the number of cells and/or the number or density of axons.

55. A method of measuring or quantifying the neuromodulatory effect of an agent, the method comprising:

(a) culturing one or more spheroids in the composition of any one of claims 1 to 18 in the presence and absence of the agent;

(b) applying a voltage potential across the one or more spheroids in the presence and absence of the agent;

(c) measuring one or more electrophysiological metrics from the one or more spheroids in the presence and absence of the agent; and

(d) correlating a difference in one or more electrophysiological metrics by the one or more spheroids to a neuromodulatory effect of the agent, such that a change in the electrophysiological metric in the presence of the agent as compared to the electrophysiological metric measured in the absence of the agent indicates a neuromodulatory effect, and no change in the electrophysiological metric in the presence of the agent as compared to the electrophysiological metric measured in the absence of the agent indicates that the agent does not confer a neuromodulatory effect;

or

(a) Culturing one or more spheroids in the composition of any one of claims 1 to 18 in the presence and absence of the agent;

(b) measuring and/or observing one or more morphometric changes of the one or more spheroids in the presence and absence of the agent; and

(c) correlating the one or more morphometric changes to a neuromodulatory effect of the agent, such that a change in the morphometric result in the presence of the agent as compared to the morphometric result measured and/or observed in the absence of the agent is indicative of a neuromodulatory effect, and no change in the morphometric result in the presence of the agent as compared to the morphometric result measured and/or observed in the absence of the agent is indicative of the agent not conferring a neuromodulatory effect.

56. A method of manufacturing the system of any one of claims 19 to 37, the method comprising:

(a) culturing neuronal cells in a cell culture medium for a period of time sufficient for the cells to form spheroids;

(b) positioning the spheroids within the hydrogel; and

(c) exposing the spheroids to a cell culture medium for a period of time sufficient to allow neurite or axon growth.

57. The method of claim 56, wherein step (a) comprises mixing the neuronal cells with one or more magnetic particles.

58. The method of claim 57, wherein step (b) comprises using magnetic forces to position the spheroids within the cavity of the hydrogel.

59. The method of claim 56, wherein step (b) comprises positioning the spheroids within the cavity of the hydrogel using acoustic, mechanical, or fluidic forces.

60. The method of claim 56, wherein the spheroid is a spheroid as disclosed in any of the compositions of claims 1-18.

61. The method of claim 52, further comprising correlating one or more morphometric changes and/or one or more electrophysiological indicators of the one or more spheroids to a toxicity of the agent, such that the agent is characterized as toxic if the morphometric changes and/or electrophysiological indicators indicate a decrease in cell viability and as non-toxic and/or neuroprotective if the morphometric changes and/or electrophysiological indicators indicate no change or increase in cell viability.

Technical Field

The present disclosure relates generally to cell culture systems, and in particular to a spheroid-directed three-dimensional cell culture system that facilitates structural and functional properties that mimic those of nerve fibers in vivo, including cell myelination and the propagation of complex action potentials.

Background

Replicating functional aspects of physiology for bench-top evaluation is particularly challenging for peripheral neuronal tissue, where long-range bioelectricity is one of the most relevant physiological outcomes. Thus, three-dimensional tissue models of peripheral nerves lag those of epithelial, metabolic, and tumor tissues, where soluble analytes may serve as appropriate indicators. The application of electrophysiological techniques has recently become possible for screening of environmental toxins as well as disease modeling and therapeutic testing through multi-electrode array technology. This application is a breakthrough in the study of Peripheral Nervous System (PNS) and Central Nervous System (CNS) applications, but the dissociative nature of the culture fails to replicate the population level environment and indicators critical to peripheral tissues. Instead, clinical methods of studying peripheral neuropathy and neuroprotection include conducting nerve conduction tests by measuring Complex Action Potentials (CAP) and Nerve Fiber Density (NFD) using morphometric analysis of skin biopsies.

Disclosure of Invention

The present disclosure relates to a microphysiological model of the nervous system that provides a3D construct and a specified tissue. Other model systems tend to allow only one of them. Organotypic tissue slices may provide 3D constructs and tissues specified by properties, but these models are not suitable for very high throughput analysis.

The present disclosure relates to compositions comprising cell spheroids comprising one or a combination of cells and/or tissues selected from the group consisting of: neuronal cells, nervous system ganglia, stem cells, and immune cells. In some embodiments, the spheroid comprises a tissue selected from the group consisting of: dorsal root ganglia and trigeminal ganglia. In some embodiments, the spheroid comprises one or more cells selected from the group consisting of: glial cells, embryonic cells, mesenchymal stem cells, cells derived from induced pluripotent stem cells, sympathetic neurons, parasympathetic neurons, spinal cord motor neurons, central nervous system neurons, peripheral nervous system neurons, enteric nervous system neurons, motor neurons, sensory neurons, cholinergic neurons, GABAergic neurons, glutamatergic neurons, dopaminergic neurons, hydroxytryptamine neurons, interneurons, adrenergic neurons, trigeminal ganglia, astrocytes, oligodendrocytes, Schwann (Schwann) cells, microglia, ependymal cells, radial glial cells, satellite cells, enteric glial cells, and pituitary cells. In some embodiments, the spheroid comprises one or more glial cells. In some embodiments, the spheroid comprises one or more embryonic cells. In some embodiments, the spheroid comprises one or more mesenchymal stem cells. In some embodiments, the spheroid comprises one or more cells derived from an induced pluripotent stem cell. In some embodiments, the spheroid comprises one or more parasympathetic neurons. In some embodiments, the spheroid comprises one or more spinal motor neurons. In some embodiments, the spheroid comprises one or more central nervous system neurons. In some embodiments, the spheroid comprises one or more peripheral nervous system neurons. In some embodiments, the spheroid comprises one or more enteric nervous system neurons. In some embodiments, the spheroid comprises one or more motor neurons. In some embodiments, the spheroid comprises one or more sensory neurons. In some embodiments, the spheroid comprises one or more interneurons. In some embodiments, the spheroid comprises one or more cholinergic neurons. In some embodiments, the spheroid comprises one or more gabaergic neurons. In some embodiments, the spheroid comprises one or more glutamatergic neurons. In some embodiments, the spheroid comprises one or more dopaminergic neurons. In some embodiments, the spheroid comprises one or more hydroxytryptamine neurons. In some embodiments, the spheroid comprises one or more trigeminal ganglion cells. In some embodiments, the spheroid comprises one or more astrocytes. In some embodiments, the spheroid comprises one or more oligodendrocytes. In some embodiments, the spheroid comprises one or more schwann cells. In some embodiments, the spheroid comprises one or more microglia. In some embodiments, the spheroid comprises one or more ependymal cells. In some embodiments, the spheroid comprises one or more radial glial cells. In some embodiments, the spheroid comprises one or more satellite cells. In some embodiments, the spheroid comprises one or more enterocytes. In some embodiments, the spheroid comprises one or more pituitary cells.

In some embodiments, the spheroid comprises one or more of one or a combination of immune cells selected from the group consisting of: t cells, B cells, macrophages, and astrocytes. In some embodiments, the spheroid comprises one or more of one or a combination of stem cells selected from the group consisting of: embryonic stem cells, mesenchymal stem cells, and induced pluripotent stem cells. In some embodiments, the neuronal cell is derived from a stem cell selected from the group consisting of: embryonic stem cells, mesenchymal stem cells, and induced pluripotent stem cells. Embodiments include each of the above cell types, independently of each other or in combination.

In some embodiments, the spheroid has a diameter of about 200 microns to about 700 microns. In some embodiments, the spheroid has a diameter of about 150 microns to about 800 microns. In some embodiments, the spheroid has a diameter of about 200 microns. In some embodiments, the spheroid has a diameter of about 300 microns. In some embodiments, the spheroid has a diameter of about 400 microns. In some embodiments, the spheroid has a diameter of about 500 microns. In some embodiments, the spheroid has a diameter of about 600 microns. In some embodiments, the spheroid has a diameter of about 700 microns. In some embodiments, the spheroid has a diameter of about 800 microns. In some embodiments, the spheroid has a diameter of about 900 microns. In some embodiments, the spheroid has a diameter of about 350 microns. In some embodiments, the spheroid has a diameter of about 450 microns. In some embodiments, the spheroid has a diameter of about 550 microns. In some embodiments, the spheroid has a diameter of about 650 microns.

In some embodiments, the spheroid comprises one or more neuronal cells and one or more schwann cells, the ratio of cell types being equal to about 4 neuronal cells per 1 schwann cell. In some embodiments, the spheroid comprises one or more neuronal cells and one or more astrocytes at a ratio equal to about 4 neuronal cells per 1 astrocyte. In some embodiments, the spheroid comprises one or more neuronal cells and one or more astrocytes at a ratio equal to about 1 neuronal cell per 1 astrocyte. In some embodiments, the spheroid comprises one or more neuronal cells and one or more schwann cells at a ratio equal to about 10 neuronal cells per 1 schwann cell. In some embodiments, the spheroid comprises one or more neuronal cells and one or more glial cells at a ratio equal to about 4 neuronal cells per 1 glial cell.

In some embodiments, any one or more of the cells described herein are differentiated from induced pluripotent stem cells. In some embodiments, the spheroid is free of induced pluripotent stem cells and/or immune cells. In some embodiments, the spheroid is free of undifferentiated stem cells.

In some embodiments, the spheroid comprises no less than about 30,000, 35,000, 40,000, 45,000, 50,000, 55,000, 60,000, 65,000, 70,000, or 75,000 cells. In some embodiments, the spheroid comprises no less than 75,000 cells. In some embodiments, the spheroid comprises no less than 65,000 cells. In some embodiments, the spheroid comprises no less than 60,000 cells. In some embodiments, the spheroid comprises no less than 100,000 cells. In some embodiments, the spheroid comprises no less than 125,000 cells. In some embodiments, the spheroid comprises no less than 150,000 cells. In some embodiments, the spheroid comprises no less than 175,000 cells. In some embodiments, the spheroid comprises no less than 200,000 cells.

In some embodiments, the spheroid comprises no less than 225,000 cells. In some embodiments, the spheroid comprises no less than 250,000 cells. In some embodiments, the spheroid comprises no less than 12,500 cells. In some embodiments, the spheroid comprises from about 12,500 cells to about 250,000 cells. In some embodiments, the spheroid comprises from about 12,500 cells to about 100,000 cells. In some embodiments, the spheroid comprises from about 12,500 cells to about 75,000 cells.

In some embodiments, the spheroid further comprises one or more magnetic particles. In some embodiments, the magnetic particle comprises one or more hollow interiors. In some embodiments, the magnetic particle comprises one or more layers of polymers on which the cells form spheroids.

The present disclosure also relates to a system comprising: (i) a cell culture vessel comprising a hydrogel; (ii) one or more spheroids comprising one or more neuronal cells and/or isolated tissue explants; (iii) an amplifier including a current generator; (iv) a voltmeter and/or ammeter; and (v) at least a first stimulating electrode and at least a first recording electrode; wherein the amplifier, voltmeter and/or ammeter and the electrodes are electrically connected to each other by an electrical circuit in which electrical current is fed from the amplifier to the at least one stimulating electrode and electrical current is received at the recording electrode and fed to the voltmeter and/or ammeter; wherein the stimulating electrodes are positioned at or adjacent to one or more cells of the neuronal cell and/or isolated tissue explant, and the recording electrodes are positioned at a predetermined distance distal to the cells such that an electric field is established across the cell culture vessel. In some embodiments, the spheroid is any one of the spheroids described herein.

In some embodiments, the culture vessel comprises 96, 192, 384 or more internal chambers. In some embodiments, the 96, 192, 384 or more internal chambers comprise one or more isolated schwann cells and/or one or more oligodendrocytes sufficiently proximal to the one or more isolated tissue explants and/or the one or more neuronal cells such that the schwann cells or oligodendrocytes deposit myelin to grow axons from the tissue explants and/or neuronal cells.

In some embodiments, the system further comprises a solid substrate having a hydrogel matrix crosslinked thereon, the solid substrate comprising at least one plastic surface having pores with a diameter of about 1 micron to about 5 microns. In some embodiments, the solid substrate comprises a continuous outer surface and an inner surface, such solid substrate comprising at least one portion in a cylindrical or substantially cylindrical shape and at least one hollow interior defined at its edges by at least a portion of said inner surface, said inner surface comprising one or more pores having a diameter of about 0.1 microns to about 1.0 microns, wherein the hollow interior of the solid substrate is accessible from a point external to the solid substrate through at least one opening; wherein the hollow interior portion comprises a first portion adjacent to the opening and at least a second portion distal to the opening; wherein the one or more neuronal cells and/or the one or more tissue explants are positioned at or adjacent to the first portion of the hollow interior and are in physical contact with the hydrogel matrix, and wherein the second portion of the at least one hollow interior is in fluid communication with the first portion such that axons can grow from the one or more neuronal cells and/or the one or more tissue explants into the second interior portion of the hollow interior.

In some embodiments, the system or composition is free of a sponge. In some embodiments, the hydrogel comprises at least a first cell-impermeable polymer and a first cell-permeable polymer. In some embodiments, the at least one cell impermeable polymer comprises no greater than about 15% PEG, and the at least one cell permeable polymer comprises from about 0.05% to about 1.00% of one or a combination of self-assembling peptides selected from the group consisting of: RAD16-I, RAD16-II, EAK 16-I, EAK 16-II and dEAK 16. In some embodiments, the composition does not contain polyethylene glycol (PEG). In some embodiments, the hydrogel comprises a first region and a second region, the first region being formed in the shape of a cylinder or a rectangular prism passing through the top and bottom of the cell culture container with its longitudinal axis, and each of the cylinder or the rectangular prism comprising a space defined by the inner surface of the cylinder or the rectangular prism, the space being accessible through the top of the cell culture container via one or more openings; wherein the second region includes a space formed in the shape of an inner wall thereof, and an opening on a side surface thereof is adjacent to and in fluid communication with the first region. In some embodiments, the hydrogel comprises at least 1% polyethylene glycol (PEG).

In some embodiments, the system further comprises a cell culture medium comprising Nerve Growth Factor (NGF) at a concentration of about 5 to about 20 picograms per milliliter, and/or ascorbic acid at a concentration in a range of about 0.001% by volume to about 0.01% by volume.

In some embodiments, the system comprises one or more spheroids comprising at least one or a combination of cells selected from the group consisting of: glial cells, embryonic cells, mesenchymal stem cells, cells derived from induced pluripotent stem cells, sympathetic neurons, parasympathetic neurons, spinal cord motor neurons, central nervous system neurons, peripheral nervous system neurons, enteric nervous system neurons, motor neurons, sensory neurons, cholinergic neurons, GABAergic neurons, glutamatergic neurons, dopaminergic neurons, hydroxytryptamine neurons, interneurons, adrenergic neurons, trigeminal neurons, astrocytes, oligodendrocytes, Schwann cells, microglia, ependymal cells, radial glial cells, satellite cells, enteric glial cells, and pituitary cells. In some embodiments, the system further comprises one or more of stem cells, pluripotent cells, myoblasts, and osteoblasts. In some embodiments, the one or more neuronal cells comprise primary mammalian cells derived from the peripheral nervous system of a mammal.

In some embodiments, the spheroid is cultured for no less than about 3, 30, 90, or 365 days.

In some embodiments, at least a portion of the solid substrate is cylindrical or substantially cylindrical such that at least a portion of the inner surface of the solid substrate defines a cylindrical or substantially cylindrical hollow interior chamber in which the spheroid is located. In some embodiments, the hydrogel comprises a series of two or more cavities in fluid communication with each other through a series of channels, at least one cavity comprises spheroids, and at least a second cavity comprises a second spheroid, a suspension of cells, or a DRG; wherein the spheroid and second spheroid, a suspension of cells, or a DRG are connected by a three-dimensional axon. In some embodiments, the cavity is a well with a U-shaped or circular well positioned in a horizontal or substantially horizontal plane of the solid substrate, wherein each channel comprises one or more axons connecting one or more spheroids.

In some embodiments, the one or more spheroids comprise one or more neuronal cells having axons that grow about 100 microns to about 500 microns in width and about 0.11 microns to about 10000 microns in length. In some embodiments, the three-dimensional axon is at least about 10 microns in height at its lowest point, or at least three monolayers of cells in height.

In some embodiments, the system comprises a first spheroid comprising: (i) one or more neuronal cells; and/or (ii) one or more schwann cells or oligodendrocytes; and the second spheroid comprises: (i) one or more peripheral neurons; wherein each spheroid is positioned in the cavity. In some embodiments, the system comprises a first cavity, a second cavity, and a third cavity, each configured to hold a spheroid and at least 50 microliters of cell culture medium, wherein the cavities are aligned such that the first cavity is positioned proximal to the second cavity and distal to the third cavity. In some embodiments, the system includes at least a fourth cavity, the cavities positioned therein in a pattern such that each cavity defines a corner of a square. In some embodiments, the cavities are aligned in a line such that axons originating from a first spheroid in a first cavity extend to a second cavity and axons from a spheroid in the second cavity extend to axons in a third cavity.

The present disclosure also relates to methods of making a three-dimensional culture of one or more spheroids in a culture vessel. In some embodiments, the method comprises: (a) contacting one or more neuronal cells with a solid substrate comprising at least one external surface, at least one internal surface, and at least one internal chamber defined by at least one internal standard and accessible from a point external to the solid substrate through at least one opening; (b) positioning one or more spheroids comprising neuronal cells into at least one internal chamber; and (c) applying a cell culture medium to the culture vessel, the volume of cell culture medium being sufficient to cover the at least one spheroid; wherein at least a portion of the inner surface comprises a first cell-impenetrable polymer and a first cell-penetrable polymer. In some embodiments, step (b) comprises locating spheroids comprising tissue explants selected from one or a combination of: isolated dorsal root ganglia, spinal cord explants, retinal explants, and cortical explants.

In some embodiments, the spheroid is formed as a suspension of neuronal cells selected from one or a combination of: motor neurons, sensory neurons, sympathetic neurons, parasympathetic neurons, cortical neurons, spinal neurons, peripheral neurons, optionally derived from stem cells. In some embodiments, the spheroid is formed from a suspension of neuronal cells selected from one or a combination of: motor neurons, sensory neurons, sympathetic neurons, parasympathetic neurons, cortical neurons, spinal neurons, peripheral neurons, optionally derived from stem cells. In some embodiments, the spheroid further comprises isolated schwann cells and/or oligodendrocytes.

In some embodiments, the method further comprises step (d): allowing the spheroids to grow neurites and/or axons for a period of about 12 hours to about 1 year after step (c). In some embodiments, the method further comprises the steps of: isolating one or more neural cells from the sample prior to step (a); and/or if the one or more spheroids comprise a DRG, isolating a Dorsal Root Ganglion (DRG) from one or more mammals prior to step (b); and/or isolating one or more schwann cells and/or one or more oligodendrocytes if the one or more spheroids comprise schwann cells or oligodendrocytes.

In some embodiments, the method further comprises positioning at least one stimulation electrode at or adjacent to a cell body of one or more neuronal cell or tissue explants, and positioning at least one recording electrode at or adjacent to an axon at a point furthest from the cell body, such that upon introduction of a current into the stimulation electrode, the recording electrode is capable of receiving a signal corresponding to one or more electrophysiological metrics capable of being measured at the recording electrode; wherein the one or more electrophysiological metrics are one or a combination of: a conductance rate, an action potential, an amplitude of a wave associated with passage of an electrical pulse along a membrane of one or more neuronal cells, a width of the electrical pulse along the membrane of one or more neuronal cells, a latency of the electrical pulse along the membrane of one or more neuronal cells, and an envelope of the electrical pulse along the membrane of one or more neuronal cells.

The present disclosure also relates to methods of evaluating the toxicity and/or neuroprotective effects of an agent comprising: (a) culturing one or more spheroids in any of the compositions disclosed herein; (b) exposing at least one agent to the one or more spheroids; (c) measuring and/or observing one or more morphometric changes and/or one or more electrophysiological metrics of the one or more spheroids; and (d) correlating the one or more morphometric changes and/or one or more electrophysiological indicators of the one or more spheroids to the toxicity of the agent, such that if the morphometric changes and/or electrophysiological indicators indicate a decrease in cell viability, the agent is characterized as toxic, and if the morphometric changes and/or electrophysiological indicators indicate no change or increase in cell viability, the agent is characterized as non-toxic and/or neuroprotective.

The present disclosure also relates to a method of measuring myelination or demyelination of one or more axons of one or more spheroids, comprising: (a) culturing one or more spheroids in any composition disclosed herein, in the presence or absence of an agent, for a time and under conditions sufficient for at least one axon to grow; and (b) detecting the amount of myelination on one or more axons from one or more spheroids; wherein the detection optionally comprises the steps of: (i) measuring and/or observing one or more morphometric changes and/or one or more electrophysiological indicators of the one or more spheroids in the presence or absence of an agent; and (ii) correlating one or more morphometric changes and/or one or more electrophysiological indicators of the one or more spheroids in the presence or absence of the agent with a quantitative or qualitative change in myelination of the spheroid.

The present disclosure also relates to methods of detecting and/or quantifying neuronal cell growth and/or axonal degeneration comprising: (a) quantifying the number or density of one or more spheroids and/or axons growing from the spheroids; (b) culturing the one or more spheroids in any of the compositions disclosed herein; and (c) after culturing the spheroid for a period of time sufficient to allow growth of one or more axons or cell growth in the spheroid, calculating the number of cells within the spheroid and/or the number or density of axons growing from spheroids in the composition. In some embodiments, step (b) optionally comprises contacting the one or more spheroids with one or more agents. In some embodiments, step (c) optionally comprises detecting internal and/or external recordings of such one or more spheroids after culturing the spheroids, and correlating the recordings to measurements of the same recordings corresponding to a known or control number of cells. In some embodiments, step (c) optionally includes the following additional steps: (i) measuring intracellular and/or extracellular recordings and/or morphometric changes before and after the step of contacting the one or more spheroids with one or more agents; and (ii) correlating the difference in the recorded and/or morphometric change prior to contacting the one or more spheroids with the one or more agents and the recorded and/or morphometric change after contacting the one or more spheroids with the one or more agents to a change in the number of cells and/or the number or density of axons.

The present disclosure also relates to methods of measuring or quantifying the neuromodulatory effect of an agent, comprising: (a) culturing one or more spheroids in any of the compositions disclosed herein, in the presence and absence of the agent; (b) applying a voltage potential across one or more spheroids in the presence and absence of the agent; (c) measuring one or more electrophysiological metrics from the one or more spheroids in the presence and absence of the agent; and (d) correlating the difference in the one or more electrophysiological metrics for the one or more spheroids to the neuromodulatory effect of the agent, such that a change in the electrophysiological metric in the presence of the agent as compared to the electrophysiological metric measured in the absence of the agent indicates a neuromodulatory effect, and no change in the electrophysiological metric in the presence of the agent as compared to the electrophysiological metric measured in the absence of the agent indicates that the agent does not confer a neuromodulatory effect.

The present disclosure also relates to methods of measuring or quantifying the neuromodulatory effect of an agent, comprising: (a) culturing one or more spheroids in any of the compositions disclosed herein in the presence and absence of the agent; (b) measuring and/or observing one or more morphometric changes of the one or more spheroids in the presence and absence of the agent; and (c) correlating one or more morphometric changes to the neuromodulatory effect of the agent, such that a change in the morphometric result in the presence of the agent as compared to the morphometric result measured and/or observed in the absence of the agent is indicative of neuromodulatory effect, and no change in the morphometric result in the presence of the agent as compared to the morphometric result measured and/or observed in the absence of the agent is indicative of the agent not conferring neuromodulatory effect.

Drawings

Fig. 1A-1B show a fabricated 3D hydrogel scaffold (scaffold) for use in the design of a neuro-on-a-chip.

Figure 2 shows representative spheroid and axon growth within the hydrogel. The hydrogel construct was able to direct and limit 3D axonal growth and cellular localization in order to mimic the nerve fiber bundle.

Fig. 3 shows a list of morphological and physiological measurements that may be taken at the ganglion, at the proximal bundle, at the midpoint of the bundle, and at the distal bundle of the dorsal root ganglion.

Fig. 4 shows a confocal image stack of unmyelinated nerve fiber bundles proximal, midpoint, and distal to the dorsal root ganglion: staining with β -III tubulin to show neurites, with DAPI to show nuclei, and with S100 to show schwann cells.

Fig. 5 shows a confocal depth map showing 3D neurite density.

Fig. 6A to 6C show Transmission Electron Microscopy (TEM) of nerve culture cross sections. Figure 6A shows a high density of parallel, bundled unmyelinated neurites 1.875mm from the nerve in the tunnel. Figure 6B shows the focus centered on Schwann Cell (SC) -encapsulated axons (Ax) that are 1mm from the nerve sparing. Figure 6C shows myelin sheath around individual nerve fibers in 25 day cultures.

Fig. 7A to 7B illustrate three-dimensional rendering of confocal images. Figure 7A shows immunohistochemistry for MBP protein. Figure 7B shows immunohistochemistry for MAG. Both cultures were 190 μm thick, confirming the three-dimensional myelin forming ability of the in vitro system.

Figure 8 shows neurite outgrowth from spheroids formed by human neurons derived from induced pluripotent stem cells. Neurites were extended on a 2D surface from human motor neuron spheroids co-cultured with astrocytes (left) and Schwann (Schwann) cells (right).

Figure 9 shows spheroids of motor neurons and astrocytes grown in a3D hydrogel system. These spheroids showed robust 3D neurite outgrowth (about 5 mm).

Figure 10 shows neurites extending in 3D from human motor neuron/schwann cell spheroids (top panel) and human sensory neurons (bottom panel).

Fig. 11 shows a 96-well sphere printing drive. A 96-well plate is positioned on top of the driver, effectively placing a magnet in the center of each well. The magnetized cells are then attracted by a magnet, causing aggregation and allowing spheroids to form.

Figure 12 shows a protocol for making rat spinal cord spheroids. Spheroids can be formed by adding magnetic nanoparticles to cultured cells and culturing in a non-adherent plate over a magnet. (not shown: spheroids can also be formed by rotation in a non-adherent circular base plate in the presence of magnetic nanoparticles.) one or more magnetic cell spheroids can be held in place with a magnet while the hydrogel growth matrix is added.

Figure 13 shows a device for placing magnetic spheroids into hydrogel voids. The outer part of this design has a magnet in the center. The dark gray section allows movement in the y-direction and the inner/uppermost part receives the glass slide, aligns the insert, and allows movement in the x-direction. A single magnet at the center allows for control of placement of constructs requiring a single magnet regardless of the shape of the construct. Other device designs incorporate multiple magnets for placing the spheroid in the connected hole.

Fig. 14A-14B illustrate placement of spheroids in a hydrogel construct. Neurite outgrowth from rat embryonic spinal cord spheroids as indicated by β -III tubulin staining followed the hydrogel morphology of the extramold. Figure 14A shows the misplacement of a spheroid when no magnet is used. Figure 14B shows the correct placement when using the device shown in figure 13.

FIG. 15 shows a bar graph showing reproducibility of spheroid production. The same spheroid formation method achieves batch-to-batch consistency. Spheroids of 27,000 cells had an average diameter of 0.47. + -. 0.03mm and a circularity of 0.72. + -. 0.15 in the xy direction.

Fig. 16A-16B show representative phase diagrams and viability of cell spheroids. These figures show an example of reproducible, well-formed spheroid formation from primary embryonic rat spinal cord cells, which show consistent size and shape.

Figure 17 shows neurite outgrowth from spheroids in matrigel at 1:20 dilution. The neurites are confined to growth-permissive channels in a3D environment. Neurites were grown to a thickness of about 100-150 μm.

Figure 18 shows the defined circuits created using Dorsal Root Ganglion (DRG) explants in combination with schwann cell spheroids in a gelatin methacrylate hydrogel. This configuration allows for unidirectional growth of DRG neurites.

Figure 19 shows defined loops arranged in a rectangular form created with multiple schwann cell spheroids in a 1:20 dilution of matrigel.

Fig. 20 shows a schematic of a method of producing a neural micro-physiological system using magnetic spheroids. Digital projection lithography can be used to solidify the hydrogel "mold" in which the growth-allowed hydrogel is contained.

Fig. 21 shows 3D spheroid seeding using nanocapsules to place spheroids within micropatterned hydrogels. Spheroids can be formed by adding magnetic nanoparticles to cultured cells and culturing in a non-adherent plate over a magnet. When the hydrogel growth matrix is added, one or more magnetic cell spheroids may be held in place with a magnet.

Figure 22 shows a bar graph showing that the number of cells seeded affects the diameter of spheroids.

Fig. 23A-23B show the motility and 3D structure of spheroids. FIG. 23A: calcein staining indicated high viability of cells in spheroids formed from primary rat embryonic spinal cord tissue. FIG. 23B: cross-sectional views of β -III tubulin stained spheroids in the obtained hydrogel constructs were examined with a fluorescent slide microscope to demonstrate the 3D structure of the spheroids.

Figure 24 shows spheroids incorporating different cell types. As indicated by antibody staining, neurons (β -III), oligodendrocyte lineage cells (Olig2), astrocytes (GFAP), and microglia and macrophages (CDIIb) were present in the spheroids.

Figure 25 shows 3D neural network formation using spheroids and micropatterned hydrogel. As shown by calcein staining, spheroids formed by rat embryonic DRG cells (left) extended neurites to spinal cord spheroids in methacrylated gelatin (right), but not vice versa.

Fig. 26 illustrates the placement of a plurality of spinal cord spheroids in a fixed position and neural network consistent with the outer mold geometry.

Figure 27 shows confocal images (10x on top and 20x on bottom) of cocultured iPSC-derived motor neurons grown on top of human muscle myocytes and myotubes. Muscle cells were allowed to differentiate in growth medium for 3 days, then motor neurons were added and the medium was switched to motor neuron medium.

Fig. 28 depicts confocal images (10x) of 2D cultures of mature myotubes forming 3D sheaths expressing heavy chain myosin. Myotubes were cultured for 3 weeks: cultured in growth medium for 3 days, in differentiation medium for 7 days, and in growth medium for the rest of the time.

Figure 29 shows 10x phase contrast images of 3D muscle cells encapsulated in 5% GelMA after 3 weeks in culture.

FIG. 30 shows the 255 μ M10 x maximum intensity projection Z-stack of human skeletal myotubes expressing desmin after 3 weeks in culture.

Figure 31 shows the 96 μ M10 x maximum intensity projection Z-stack of human skeletal myotubes expressing heavy chain myosin after 3 weeks in GelMA culture.

Figure 32 shows induced 3D spheroid generation by neurons in pendant droplets at different apparent densities (seeing diversity).

Figure 33 shows induced 3D spheroid generation by neurons in U-bottom wells at different apparent densities.

Fig. 34 shows 3D spheroid generation by motor neurons in hanging droplets at different apparent densities.

Fig. 35 shows 3D spheroid generation by motor neurons in U-bottom wells at different apparent densities.

Figure 36 shows 3D spheroid generation by motor neurons in pendant drops at different apparent densities within 24 or 48 hours.

Figure 37 shows 3D spheroid generation by motor neurons in U-bottomed wells at different apparent densities within 24 or 48 hours.

Figure 38 shows the growth of pendant droplet spheroids seeded with 25,000 iPSC-derived motor neurons and seeded with 25,000 astrocytes after 4 days.

Figure 39 shows the growth of U-bottom-well spheroids seeded with 25,000 iPSC-derived motor neurons and seeded with 25,000 astrocytes after 4 days.

Figure 40 shows the growth of pendant droplet spheroids seeded with 40,000 iPSC-derived motor neurons and seeded with 10,000 astrocytes after 4 days.

Figure 41 shows different combinations of neurons and glia within 24 hours.

Fig. 42 shows spheroids of oligodendrocyte precursor cells at 4X (top panel) and 10X (bottom panel).

Fig. 43A to 43J show the preparation of spheroids consisting of human neurons (hN) and/or human schwann cells (hSC) after 2 days in vitro. The presence of hscs promoted spheroid formation in the co-culture system, whereas the hN-only condition did not form spheroids within 2 days. hSC spheroids were prepared at three different cell densities: 25,000(a), 50,000(b) and 75,000 (c). At constant hN density (75,000) but varying hSC density: 25,000(d), 50,000(e) and 75,000(f) create co-cultured spheroids. In parallel, at three different densities: hN spheroids were prepared at 50,000(g), 75,000(h) and 100,000 (i). (j) Comparison of the diameters of the different spheroids revealed that the co-cultured spheroids were more dense (compact) compared to the single culture spheroids, indicating that the affinity of hSC for hN leads to a more tightly packed cell cluster. K (thousand fold). N-4, error bars represent standard error of the mean (SEM). Scale bar: 100 μm. And (p value is less than or equal to 0.0001), and (p value is less than or equal to 0.001), and (p value is less than or equal to 0.01), and (p value is less than or equal to 0.05).

Figure 44 shows the formation of spheroids composed of individual human neurons (hN). Qualitative examination at >2 days after starting hN only culture showed a consistent significant increase in spheroid size as the total number of cells increased (25K, 50K, 75K and 100K). A chart comparing individual spheroid diameters supports this qualitative check. K (thousand fold). N-4, error bars represent standard error of the mean (SEM). Scale bar: 100 μm. (p-value is less than or equal to 0.0001) and (p-value is less than or equal to 0.01).

Fig. 45A and 45B show schwann cells migrating out of the spheroid and elongating along the axon. (45A) The figure shows how human schwann cells (hSC) stained for the hSC marker S100 (light grey) migrate out of the spheroid over a period of 4 weeks with growing axons (grey) stained for β III-tubulin. Nuclei were labeled with DAPI (dark grey). Scale bar: 1000 μm. (45B) The high magnification image of the inset in panel a. Scale bar: 25 μm.

Fig. 46A-46C show recordings of axons growing from spheroids in culture. Fig. 46B' and 46B "show graphical representations of collected records. These graphs show the Nerve Conduction Velocity (NCV) of two spheroids measured in meters per second over time. Figure 46C shows the same NCV experiment, with bar graphs showing at two time points: the starting value of the current and the NCV at the peak.

Figures 47A to 47f various stages of myelin formation observed in vitro reconstituted human nerves. (47A) Non-dense myelin. (47B) Dense myelin. (47C) The myelin during compaction (compaction). (47D) Myelin formation in the absence of axons. (47E) Intracytoplasmic lamina. (47F) Naked (unmyelinated) axons.

Fig. 48A-48 d. dorsal horn spheroid characterization. (48A) Micrographs show typical DRG to DH synapse cultures at 14 DIV. (48B) The fluorescence images show β 3-tubulin staining of synaptic cultures at 28 DIV. (48C) Micrographs show synaptic cultures on E-phys rig (rig). The blue arrow indicates the distance between the stimulating electrode and the recording electrode, which is 3.1 mm. This image shows the stimulating electrodes at the DRG axons and the recording electrodes at the DH spheroids. (48D) Electrical activity was recorded at the DH spheroids in response to 20V stimulation at the DRG axons. The X-axis is in milliseconds; the y-axis is in millivolts.

Detailed Description

Various terms relating to the methods and other aspects of the present disclosure are used throughout the specification and claims. Such terms are to be given their ordinary meaning in the art unless otherwise indicated. Other specifically defined terms are to be construed in a manner consistent with the definitions provided herein.

As used in this specification and the appended claims, the singular forms "a", "an" and "the" include plural referents unless the content clearly dictates otherwise.

The term "greater than 2" as used herein is defined as any integer greater than the number 2, such as 3, 4, or 5.

The term "about" as used herein when referring to measurable values such as amounts, time intervals, and the like, is intended to encompass variations of ± 20%, ± 10%, ± 5%, ± 1%, ± 0.9%, ± 0.8%, ± 0.7%, ± 0.6%, ± 0.5%, ± 0.4%, ± 0.3%, ± 0.2%, or ± 0.1% from the specified values, as such variations are suitable for performing the disclosed methods.

As used herein in the specification and in the claims, the phrase "and/or" should be understood to mean "either or both" of the elements so combined (i.e., elements that are present in combination in some instances and are present in isolation in other instances). In addition to the elements specifically identified by "and/or" sentence pattern, other elements may optionally be present, whether related or unrelated to those elements specifically identified, unless clearly stated to the contrary. Thus, as a non-limiting example, a reference to "a and/or B," when used in conjunction with an open-ended term such as "comprising," can refer in one embodiment to a and B is not present (optionally including elements other than B); in another embodiment, B may be referred to and a is absent (optionally including elements other than a); may refer to a and B (optionally including other elements) in yet another embodiment; and so on.

As used herein in the specification and in the claims, "or" should be understood to have the same meaning as "and/or" as defined above. For example, where items are separated in a list, "or" and/or "should be interpreted as being inclusive, i.e., including at least one of the plurality of elements or list of elements, but also including more than one of the plurality of elements or list of elements, and optionally including additional unlisted items. Terms such as "only one of," or "exactly one of," or "consisting of … …, when used in the claims, are intended to mean that it includes a plurality of elements or exactly one of the elements in the list of elements. In general, as used herein, the term "or" preceded by an exclusive wording only, "either of", "one of", "only one of", or "exactly one of" is to be interpreted as indicating an exclusive alternative (i.e., "one or the other but not both"), "consisting essentially of … …" shall have the ordinary meaning as used in the patent law field when used in the claims.

As used herein, the terms "comprising" (and any form comprising "(such as" comprises "and" comprising) "," having "(and any form having" (such as "having" and "having)", "including" (and any form comprising (including), such as "comprising" and "including)", or "containing" (and any form containing (such as "containing" and "containing)", are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.

As used herein, the phrase "integer from X to Y" means any integer including the endpoints. That is, if a range is disclosed, each integer within the range, including the endpoints, is disclosed. For example, the phrase "integer from X to Y" discloses 1, 2, 3, 4, or 5 and a range of 1 to 5.

The term "plurality" as used herein is defined as any quantity or number greater than or exceeding 1.

As used herein, "substantially equivalent" may be, for example, within a range known to correlate with an abnormal or normal range at a given measurement index. For example, if the control sample is from a diseased patient, then essentially equivalent is within the abnormal range. If the control sample is from a patient known not to have the condition to be tested, then substantially equivalent is within the normal range for that given index.

The present disclosure relates generally to systems capable of receiving and culturing one or more spheroids in a three-dimensional culture. In some embodiments, the system uses a solid substrate, such as plastic or similar polymer, that includes pores on which hydrogels of any shape or size can reside. The hydrogel of the system in some embodiments serves as a support for the cells of the present disclosure to grow and proliferate neurites and/or to form axons under conditions sufficient for mature cells of the nervous system to grow, divide and/or proliferate axons (either in spheroid form or in suspension). In some embodiments, the system includes a hydrogel that forms a cavity having at least two regions: a first region resembling a well having a flat or curved bottom and a diameter spanning the longitudinal plane of the solid support, the first region further comprising an opening located at the top of the region and accessible from the exterior of the system, and an opening located on at least one side of the first region and in fluid communication with the second region. In some embodiments, the diameter of the first region is about 1mm or less. The second region is in the form of a channel extending laterally from the first region, the sides of which define the height of the channel. In some embodiments, the channel has a width of about 10 to about 750 microns. In some embodiments, the channel has a length of from about 100 to about 10000 microns. After growing a spheroid from any one or combination of cells identified in the present disclosure, the spheroid can be placed into a first region containing cell culture medium. After placement, the neurites may grow spontaneously or be promoted to grow by exposure to one or more growth-stimulating molecules. Neurite and/or axon growth may occur in a second region, originating from the first region of the system and passing through at least one opening on the side of the hydrogel and into the second region. After the neurite or axon has grown to a desired length, the agent can be exposed to cell culture to determine how the agent affects the growth, morphology, or action potential of the axon or neurite.

In some embodiments, the cavities or apertures that receive the spheroids and define the first regions may be in a pattern or network connected by respective second regions such that the spheroids may be connected by channels of axons that grow from one or more spheroids. In some embodiments, the spheroids are in a square or rectangular pattern connected by channels positioned between each spheroid. In some embodiments, the pattern is an "L" shape, wherein the spheroids define the ends and corners of the "L" configuration. In some embodiments, the spheroids may be positioned in a triangular or angular pattern with three channels between each of the three cavities comprising the spheroids. At one end of the hydrogel network, the first cavity may comprise a spheroid having central nervous system characteristics. In these embodiments, the cells that normally fill the central nervous system constitute spheroids. Such cells may be selected from any combination or composition comprising a single neuronal cell, and may also include astrocytes or immune cells. In the same embodiment, the cavity located furthest from the first cavity may accommodate spheroids having sensory characteristics, such as those including sensory neurons. Thus, the axonal connection between the first spheroid and the distal-most spheroid of the first spheroid mimics a sensory nerve fiber where the axon extends from a spheroid characteristic of the central nervous system to a spheroid that includes peripheral sensory neurons. Electrophysiological measurements between such spheroids can be made by placing electrodes at either end of the circuit and measuring the recordings.

In some embodiments, the spheroid comprises a mixture of neuronal cells and non-neuronal cells. Non-neuronal cells include skeletal muscle cells, cardiac muscle cells, and smooth muscle cells. Non-neuronal cells also include cells from organ tissues, such as kidney cells, liver cells, and pancreatic cells. Examples of non-neuronal cells also include endothelial cells, epithelial cells of the skin, and corneal cells of the eye. In some embodiments, the cell is a mammalian cell, a non-human animal cell, or a human cell. In some embodiments, any one or more cells of the spheroid are primary human cells. In some embodiments, the cell is taken from a human subject. In some embodiments, the cell is a rat or murine cell. In some embodiments, the cell is a non-human primate cell, a porcine cell, a canine cell, or a bovine cell. In some embodiments, any of the disclosed systems can include a spheroid of neuronal cells mixed with or without non-neuronal cells.

The methods of the present disclosure include methods of culturing spheroids disclosed herein, and methods of measuring the toxicity or biological effects of a toxin, drug, therapeutic agent, biomolecule, or contaminant when such molecule, drug, or therapeutic agent is exposed to spheroids in a system and cultures of axons or neurites that germinate from such spheroids. In some embodiments, the method comprises a method of causing axons and/or neurites to grow unidirectionally in a culture from a first spheroid to a second spheroid. In some embodiments, any of the disclosed systems comprise an agent that stimulates, accelerates, slows, or halts neurite and/or axon growth in culture. In some embodiments, any of the methods of the present disclosure comprise stimulating directional growth of axons in culture. In certain embodiments, the agent is used to attract guidance of axons and/or neurites or to prevent growth of axons and/or neurons. In some embodiments, the attractive targeting protein is added to a system selected from the group consisting of: axon-directing factor, neurotrophic factor, adhesive extracellular matrix protein, cell adhesion receptor (e.g., cadherin, Ig-CAM, or integrin); one can also use peptides that mimic the putative binding sites of these proteins. In some embodiments, the protein that prevents axonal and/or neurite growth is a component of the system. Rejection proteins include: ephrins (sometimes), Semaphorins (most of the time), Slits; chondroitin sulfate proteoglycan, and the like.

Methods of making spheroids with and without magnetic particles or beads are also disclosed. If the magnetic particles are components of spheroids, any device including a magnet may be used to place one or more spheroids at a location within one of the disclosed cavities formed by the hydrogel walls. The present disclosure generally relates to an apparatus including a movable frame that is movable in any lateral direction parallel to a horizontal direction in which the apparatus operates. The frame is attached to one or more magnets having a magnetic force sufficient to attract the spheroids containing the magnetic particles. In some embodiments, a frame movable in the x and y directions of the longitudinal plane of the device is mechanically attached to the magnets by glue, polymer, or fasteners, such that movement of the frame causes the magnets to move, the magnetic force being sufficient to move the spheroids in any direction if the spheroids are within the magnetic field of the magnets. In some embodiments, the device comprises a first frame and a second frame, at least one of the first or second frames being movable in a lateral direction parallel to a longitudinal plane of the device and a horizontal or substantially horizontal direction on the device.

The term "bioreactor" refers to an enclosure or partial enclosure in which cells are cultured, optionally in suspension. In some embodiments, a bioreactor refers to an enclosure or partial enclosure in which cells are cultured, wherein the cells may be in a liquid suspension or, alternatively, may be in contact with, grown on, or within another non-liquid substrate (including but not limited to a solid growth support material). In some embodiments, the solid growth support material or solid substrate comprises at least one or a combination of: silica, plastic, metal, hydrocarbon, or gel. The present disclosure relates to systems comprising a bioreactor comprising one or more culture vessels in which neuronal cells can be cultured in the presence or absence of a cell growth medium.

The term "culture vessel" as used herein may be any vessel suitable for growing cells, culturing cells, incubating cells, proliferating cells, propagating cells, or otherwise similarly manipulating cells. The culture vessel may also be referred to herein as a "culture insert". In some embodiments, the culture vessel is made of biocompatible plastic and/or glass. In some embodiments, the plastic is a thin layer of plastic comprising one or more pores that allow diffusion of proteins, nucleic acids, nutrient (e.g., heavy metals and hormones) antibiotics, and other cell culture medium components through the pores. In some embodiments, the width of the aperture is no greater than about 0.1, 0.5, 1.0, 2, 3, 4,5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, 50 microns. In some embodiments, the culture vessel is in a hydrogel matrix and is free of a substrate or any other structure. In some embodiments, the culture vessel is designed to contain a hydrogel or hydrogel matrix and various culture media. In some embodiments, the culture vessel consists of or consists essentially of a hydrogel or hydrogel matrix. In some embodiments, the only plastic component of the culture container is the component that constitutes the sidewall and/or bottom of the culture container, which separates the volume of the well or area in which the cells grow from the point outside the culture container. In some embodiments, the culture vessel comprises a hydrogel and one or more isolated glial cells. In some embodiments, the culture vessel comprises a hydrogel and one or more isolated glial cells seeded with one or more neuronal cells.

The term "electrical stimulation" refers to the process by which cells are exposed to an Alternating Current (AC) or Direct Current (DC) current. The current may be introduced into a solid substrate or applied through the cell culture medium or other suitable component of the cell culture system. In some embodiments, electrical stimulation is provided to the device or system by placing one or more electrodes at different locations within the device or system to generate a voltage potential across the cell culture container. The electrodes are in operative connection with one or more amplifiers, voltage meters, current meters, and/or electrochemical systems (e.g., batteries or generators) via one or more wires. Such devices and wires form an electrical circuit through which current is generated and through which an electrical potential is generated across the tissue culture system.

The term "hydrogel" as used herein may be any water-insoluble, crosslinked, three-dimensional network of, for example, polymer chains, wherein the interstices between the polymer chains are filled or capable of being filled with water. The term "hydrogel matrix" as used herein refers to, for example, any three-dimensional hydrogel construct, system, device, or similar structure. Hydrogel and hydrogel matrices are known in the art and include, for example, U.S. patent nos. 5,700,289 and 6,129,761 and currley and Moore, 2011; curley et al, 2011; various types of hydrogels and hydrogel matrices are described in Irons et al, 2008 and Tibbitt & Anseth, 2009; each of these patents and documents is incorporated by reference in its entirety. In some embodiments, the hydrogel or hydrogel matrix may be cured by subjecting the liquefied pre-gel solution to ultraviolet, visible, or ay light having a wavelength greater than about 300nm, 400nm, 450nm, or 500 nm. In some embodiments, the hydrogel or hydrogel matrix may be cured into various shapes, for example, designed to mimic the bifurcated shape of a nerve bundle. In some embodiments, the hydrogel or hydrogel matrix comprises poly (ethylene glycol) dimethacrylate (PEG). In some embodiments, the hydrogel or hydrogel matrix comprises a peptide fragment hydrogel (Puramatrix). In some embodiments, the hydrogel or hydrogel matrix comprises glycidyl methacrylate-dextran (menex). In some embodiments, the neuronal cells are incorporated into a hydrogel or hydrogel matrix. In some embodiments, cells from the nervous system are incorporated into the hydrogel or hydrogel matrix. In some embodiments, the cells from the nervous system are schwann cells and/or oligodendrocytes. In some embodiments, the hydrogel or hydrogel matrix comprises a tissue explant from the nervous system of an animal (e.g., a mammal) and a population of supplemental cells that are derived from the nervous system but are isolated and cultured to enrich its population in culture. In some embodiments, the hydrogel or hydrogel matrix comprises a tissue explant, such as a retinal tissue explant, DRG, or spinal cord tissue explant, and isolated and cultured schwann cells, oligodendrocytes, and/or microglia. In some embodiments, two or more hydrogel or hydrogel matrices are used simultaneously in a cell culture vessel. In some embodiments, two or more hydrogels or hydrogel matrices are used simultaneously in the same cell culture vessel, but the hydrogels are separated by walls, forming independently addressable microenvironments in the tissue culture vessel, e.g., well. In a multiplexed tissue culture vessel, for some embodiments, any number of the aforementioned wells or independently addressable locations may be included within a cell culture vessel such that the hydrogel matrix in one well or location is different from or the same as the hydrogel matrix in another well or location of the cell culture vessel.

The term "immune cell" as used herein may be, for example, any cell involved in the immunological activity of a subject, including protecting the subject from infection or symptoms of infection, or attacking, clearing, or otherwise eliminating dysfunctional cells or pathogens from cells in the subject, or ameliorating symptoms of a disease caused by a pathogen. In some embodiments, the immune cells include one or more B cells, T cells, antigen presenting cells (such as astrocytes, dendritic cells, and macrophages), stellate cells, granulocytes, monocytes, basophils, eosinophils, and/or mast cells. In some embodiments, the immune cell expresses CD4 or CD8 and one or more immunomodulatory molecules. In some embodiments, the immunomodulatory molecule is selected from one of: IL-28, MHC, CD80, CD86, IL-1, IL-2, IL-4, IL-5, IL-6, IL-10, IL-18, MCP-1, Mp-a, Mp- Ρ - β, IL-8, L-selectin, P-selectin, E-selectin, CD34, GlyCAM-1, MadCAM-1, LFA-1, VLA-1, Mac-1, pl50.95, PECAM, ICAM-1, ICAM-2, ICAM-3, CD2, LFA-3, M-CSF, G-CSF, mutant of IL-18, CD40, CD40L, vascular growth factor, fibroblast growth factor, IL-7, nerve growth factor, vascular endothelial growth factor, Fas, TNF receptor, Fit, Apo-1, P55, L-1, IL-7, nerve growth factor, WS, TNF receptor, Fit, Apo-1, P55, IL-1, and MPA-1 DR3, TRAMP, Apo-3, AIR, LARD, NGRF, DR4, DR5, KILLER, TRAIL-R2, TRICK2, DR6, caspase ICE, Fos, c-jun, Sp-1, Ap-2, p38, p65Rel, MyD88, IRAK, TRAF6, IkB, inactive NIK, SAP K, SAP-1, JNK, interferon-responsive genes, NFkB, Bax, TRAIL, TRAILrec, TRAILrecDRC5, TRAIL-R3, TRAIL-R4, RANK, RANK LIGAND, Ox40, Ox40 LIGAND, NKG2D, MICA, MICB, NKG2A, NKG2B, NKG2C, NKG2, 2E 3, TA 2F, TAR 2, and functional fragments thereof, or combinations thereof. Immunomodulatory proteins are exemplified in U.S. patent No. 8008265.

The term "immunomodulator" refers to a substance that has a modulating effect on the immune system. Such substances can be conveniently identified using standard assays that indicate various aspects of the immune response, such as cytokine secretion, antibody production, NK cell activation, and T cell proliferation. See, for example, WO 97/28259; WO 98/16247; WO 99/11275; krieg et al (1995) Nature374: 546-549; yamamoto et al (1992) J.Immunol.148: 4072-76; ballas et al (1996) J.Immunol.157: 1840-45; klinman et al (1997) J.Immunol.158: 3635-39; sato et al (1996) Science 273: 352-; pisetsky (1996) J.Immunol.156: 421-; shimada et al (1986) Jpn.J.cancer Res.77: 808-; cowdery et al (1996) J.Immunol.156: 4570-75; roman et al (1997) nat. Med.3: 849-854; lipford et al (1997a) Eur.J.Immunol.27: 2340-44; WO 98/55495 and WO 00/61151. Thus, these and other methods can be used to identify, test and/or confirm immunostimulatory substances, such as immunostimulatory nucleotides, immunostimulatory isolated nucleic acids.

In some embodiments, the two or more hydrogels may comprise different amounts of PEG and/or peptide fragment hydrogels. In some embodiments, the two or more hydrogels may have various densities. In some embodiments, the two or more hydrogels may have various permeabilities that are capable of allowing cells to grow in the hydrogel. In some embodiments, the two or more hydrogels may have various flexibilities. In some embodiments, a bioreactor, cell culture device, or composition disclosed herein comprises a hydrogel comprising two layers of polymers: cell permeable polymers and cell impermeable polymers. In some embodiments, the cell permeable layer is layered in at least a region on top of the cell impermeable layer.

The term "cell permeable polymer" refers to a hydrophilic polymer having identical or mixed monomer subunits at a concentration and/or density sufficient to form spaces upon crosslinking on a solid substrate in a solid or semi-solid state, such spaces being sufficiently biocompatible to allow cells or portions of cells to grow in culture.

The term "cell impermeable polymer" refers to a hydrophilic polymer having identical or mixed monomeric subunits at a concentration and/or density sufficient to not form biocompatible spaces or compartments upon cross-linking in a solid or semi-solid state on a solid substrate. In other words, a cell-impenetrable polymer is a polymer that cannot support the growth of cells or parts of cells in culture after cross-linking at a particular concentration and/or density.

The term "functional fragment" can be any portion of a polypeptide or nucleic acid sequence to which a corresponding full-length polypeptide or nucleic acid is directed that is of sufficient length and has sufficient structure to confer a biological effect that is at least similar or substantially similar to the full-length polypeptide or nucleic acid on which the fragment is based. In some embodiments, a functional fragment is a portion of a full-length or wild-type nucleic acid sequence encoding any of the nucleic acid sequences disclosed herein, and the portion encodes a polypeptide having a length and/or structure that is less than full-length but encodes a domain that is still biologically functional as compared to the full-length or wild-type protein. In some embodiments, a functional fragment may have reduced, about equivalent, or enhanced biological activity as compared to the wild-type or full-length polypeptide sequence on which the fragment is based. In some embodiments, the functional fragment is derived from a sequence of an organism (such as a human). In such embodiments, the functional fragment may retain 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, or 90% sequence identity to the wild-type human sequence from which the sequence is derived. In some embodiments, the functional fragment may retain 87%, 85%, 80%, 75%, 70%, 65%, or 60% sequence homology to the wild-type sequence from which the sequence is derived.

It will be appreciated by the skilled person that the cell-impermeable polymer and the cell-permeable polymer may comprise the same or substantially the same polymer, but that the difference in concentration or density after cross-linking forms a hydrogel matrix, some portion of which facilitates growth of the cells or a portion of the cells in culture.

In some embodiments, the hydrogel or hydrogel matrix may have various thicknesses. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 150 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 200 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 250 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 300 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 350 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 400 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 450 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 500 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 550 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 600 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 650 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 700 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 750 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 750 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 700 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 650 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 600 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 550 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 500 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 450 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 400 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 350 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 300 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 250 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 200 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 150 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 300 μm to about 600 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 400 μm to about 500 μm.

In some embodiments, the hydrogel or hydrogel matrix may have various thicknesses. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 10 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 150 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 200 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 250 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 300 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 350 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 400 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 450 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 500 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 550 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 600 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 650 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 700 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 750 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 800 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 850 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 900 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 950 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 1000 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 1500 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 2000 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 2500 μm to about 3000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 2500 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 2000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 1500 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 1000 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 950 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 900 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 850 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 800 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 750 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 700 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 650 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 600 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 550 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 500 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 450 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 400 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 350 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 300 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 250 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 200 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 100 μm to about 150 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 300 μm to about 600 μm. In some embodiments, the hydrogel or hydrogel matrix has a thickness of about 400 μm to about 500 μm.

In some embodiments, the hydrogel or hydrogel matrix comprises one or more synthetic polymers. In some embodiments, the hydrogel or hydrogel matrix comprises one or more of the following synthetic polymers: polyethylene glycol (polyethylene oxide), polyvinyl alcohol, poly 2-hydroxyethyl methacrylate, polyacrylamide, silicone, and any derivative or combination thereof.

In some embodiments, the hydrogel or hydrogel matrix comprises one or more synthetic and/or natural polysaccharides. In some embodiments, the hydrogel or hydrogel matrix comprises one or more of the following polysaccharides: hyaluronic acid (hyaluronic acid), heparin sulfate, heparin, dextran, agarose, chitosan, alginate, and any derivative or combination thereof.

In some embodiments, the hydrogel or hydrogel matrix comprises one or more proteins and/or glycoproteins. In some embodiments, the hydrogel or hydrogel matrix comprises one or more of the following proteins: collagen, gelatin, elastin, titin, laminin, fibronectin, fibrin, keratin, silk fibroin and any derivative or combination thereof.

In some embodiments, the hydrogel or hydrogel matrix comprises one or more synthetic and/or natural polypeptides. In some embodiments, the hydrogel or hydrogel matrix comprises one or more of the following polypeptides: polylysine, polyglutamate or polyglycine.

In some embodiments, the hydrogel comprises one or a combination of polymers selected from the group consisting of polymers disclosed in the following references: khoshakhllagh et al, "Photocurable interactive network of hydrophilic and Puramatrix as a selectively curable scaffold for neural growth", Acta biomaterials 2015, 1, 21 days.

Any hydrogel suitable for cell growth can be formed by placing any one or combination of the polymers disclosed herein at a concentration and under conditions and for a sufficient period of time sufficient to form two different densities of crosslinked polymers: a cell permeable polymer, a cell impermeable polymer. The polymer may be a synthetic polymer, a polysaccharide, a natural protein or glycoprotein, and/or a polypeptide, such as those selected from the group consisting of.

Synthetic polymers

Such as polyethylene glycol (polyethylene oxide), polyvinyl alcohol, poly-2-hydroxyethyl methacrylate, polyacrylamide, silicone, combinations thereof, and derivatives thereof.

Polysaccharides(whether synthetic or derived from natural sources)

Such as hyaluronic acid, heparan sulfate, heparin, dextran, agarose, chitosan, alginate, combinations thereof, and derivatives thereof.

Natural protein or glycoprotein

Such as collagen, gelatin, elastin, titin, laminin, fibronectin, fibrin, keratin, silk fibroin, combinations thereof, and derivatives thereof.

Polypeptides(whether of synthetic or natural origin)

Such as polylysine and all of the listed RAD and EAK peptides.

The term "three-dimensional" or "3D" as used herein means, for example, the thickness of a culture of cells such that at least three layers of cells grow adjacent to each other. In some embodiments, the term three-dimensional means that the thickness or height of the neurites and/or axons is from about 10 to about 1000 microns in the context of the disclosed systems. In some embodiments, the term three-dimensional means that the thickness or height of the neurites and/or axons is from about 10 to about 100 microns in the context of the disclosed systems.

The term "isolated neurons" refers to neuronal cells that have been removed or dissociated from the organism or culture from which they were originally grown. In some embodiments, the isolated neuron is a neuron in suspension. In some embodiments, the isolated neuron is a component of a larger mixture of cells, including a tissue sample or a suspension containing non-neuronal cells. In some embodiments, the neuronal cells have become isolated when they are removed from the animal from which they were derived, for example in the case of tissue explants. In some embodiments, the isolated neurons are those in DRGs excised from an animal. In some embodiments, the isolated neuron comprises at least one or more cell from a species or combination of species selected from the group consisting of: sheep cells, goat cells, horse cells, cow cells, human cells, monkey cells, mouse cells, rat cells, rabbit cells, dog cells, cat cells, pig cells, or other non-human mammals. In some embodiments, the isolated neuron is a human cell. In some embodiments, the isolated neuron is a stem cell that has been pre-treated to have a similar or substantially similar differentiation phenotype as a human neuronal cell. In some embodiments, the isolated neuron is a human cell. In some embodiments, the isolated neuron is a stem cell that has been pre-treated to have a similar or substantially similar differentiation phenotype as a non-human neuronal cell. In some embodiments, the stem cell is selected from the group consisting of: mesenchymal stem cells, induced pluripotent stem cells, embryonic stem cells, hematopoietic stem cells, epidermal stem cells, stem cells isolated from the umbilical cord of a mammal, or endodermal stem cells.

The term "neurodegenerative disease" is used throughout the specification to describe a disease caused by damage to the central nervous system and/or the peripheral nervous system. Exemplary neurodegenerative diseases that can be examples of diseases that can be studied using the disclosed models, systems, or devices include, for example, parkinson's disease, huntington's disease, amyotrophic lateral sclerosis (Lou Gehrig's disease), alzheimer's disease, lysosomal storage disorders (such as, for example, Folkerth, j. neuropath, exp. neuro, 58, 9, sep., 1999, "leukopathies" or gliosis/demyelinating diseases), Tay-Sachs disease (β -hexosaminidase deficiency), other genetic diseases, multiple sclerosis, brain injury or trauma resulting from ischemia, accident, environmental injury, spinal cord injury, ataxia, and alcoholism. In addition, the invention can be used to test the efficacy, toxicity or neurodegenerative effects of agents on neuronal cells in culture to investigate treatments for neurodegenerative diseases. The term neurodegenerative disease also includes neurodegenerative diseases, including, for example, autism and related neurological diseases, such as schizophrenia and the like.

As used herein, the term "neuronal cell" refers to a cell, e.g., comprising at least one or a combination of dendrites, axons, and somatic cells, or any cell or group of cells isolated from nervous system tissue. In some embodiments, the neuronal cell is any cell that comprises or is capable of forming an axon. In some embodiments, the neuronal cell is a schwann cell, a glial, a cortical neuron, an embryonic cell isolated or obtained from neuronal tissue or that has differentiated into a cell having a neuronal phenotype or a phenotype substantially similar to that of a neuronal cell, an induced pluripotent stem cell (iPS) that has differentiated into a neuronal phenotype, or a mesenchymal stem cell derived from neuronal tissue or differentiated into a neuronal phenotype. In some embodiments, the neuronal cell is a neuron from Dorsal Root Ganglion (DRG) tissue, retinal tissue, spinal cord tissue, or brain tissue of an adult, juvenile, or fetal subject. In some embodiments, the neuronal cell is any one or more cells isolated from neuronal tissue of a subject. In some embodiments, the neuronal cell is a mammalian cell. In some embodiments, the cell is a human cell and/or a rat cell. In some embodiments, the cell is a non-human mammalian cell or is derived from a cell isolated from a non-human mammal. Neuronal cells may comprise isolated neurons from more than one species if isolated or dissociated from the original animal from which the cell originated. In some embodiments, the spheroid is free of DRG tissue.

In some embodiments, the neuronal cell is one or more of: central nervous system neurons, peripheral nervous system neurons, sympathetic neurons, parasympathetic neurons, enteric nervous system neurons, spinal cord motor neurons, sensory neurons, autonomic neurons, somatic neurons, dorsal root ganglia, cholinergic neurons, GABAergic neurons, glutamatergic neurons, dopaminergic neurons, hydroxytryptamine neurons, interneurons, adrenergic neurons, and trigeminal ganglia. In some embodiments, the glial cells are one or more of: astrocytes, oligodendrocytes, schwann cells, microglia, ependymal cells, radial glial cells, satellite cells, enteroglial cells, and pituitary cells. In some embodiments, the immune cell is one or more of: macrophages, T cells, B cells, leukocytes, lymphocytes, monocytes, mast cells, neutrophils, natural killer cells, and basophils. In some embodiments, the stem cell is one or more of: hematopoietic stem cells, neural stem cells, embryonic stem cells, adipose-derived stem cells, bone marrow-derived stem cells, induced pluripotent stem cells, astrocyte-derived induced pluripotent stem cells, fibroblast-derived induced pluripotent stem cells, renal epithelium-derived induced pluripotent stem cells, keratinocyte-derived induced pluripotent stem cells, peripheral blood-derived induced pluripotent stem cells, hepatocyte-derived induced pluripotent stem cells, mesenchymal-derived induced pluripotent stem cells, neural stem cell-derived induced pluripotent stem cells, adipose-derived induced pluripotent stem cells, preadipocyte-derived induced pluripotent stem cells, chondrocyte-derived induced pluripotent stem cells, and skeletal muscle-derived induced pluripotent stem cells. In some embodiments, spheroids may also include other cell types, such as keratinocytes or endothelial cells.

The term "neuronal cell culture medium" or simply "culture medium" as used herein may be any nutrient suitable for supporting growth, culturing, growing, proliferating, propagating or otherwise manipulating neuronal cells. In some embodiments, the culture medium comprises a neural basal medium supplemented with Nerve Growth Factor (NGF). In some embodiments, the medium comprises Fetal Bovine Serum (FBS). In some embodiments, the medium comprises L-glutamine. In some embodiments, the medium comprises ascorbic acid at a concentration in the range of about 0.001% by volume to about 0.01% by volume. In some embodiments, the medium comprises ascorbic acid at a concentration in the range of about 0.001% by volume to about 0.008% by volume. In some embodiments, the medium comprises ascorbic acid at a concentration in the range of about 0.001% volume by volume to about 0.006% volume by weight. In some embodiments, the medium comprises ascorbic acid at a concentration in the range of about 0.001% by volume to about 0.004% by volume. In some embodiments, the medium comprises ascorbic acid at a concentration in the range of about 0.002% by volume to about 0.01% by volume. In some embodiments, the medium comprises ascorbic acid at a concentration in the range of about 0.003% by volume to about 0.01% by volume. In some embodiments, the medium comprises ascorbic acid at a concentration in the range of about 0.004% by volume to about 0.01% by volume. In some embodiments, the medium comprises ascorbic acid at a concentration in the range of about 0.006% volume to about 0.01% volume weight. In some embodiments, the medium comprises ascorbic acid at a concentration in the range of about 0.008% volume to about 0.01% volume. In some embodiments, the culture medium comprises ascorbic acid at a concentration in the range of about 0.002% by volume to about 0.006% by volume. In some embodiments, the medium comprises ascorbic acid at a concentration in the range of about 0.003% by volume to about 0.005% by volume.

In some embodiments, the hydrogel, hydrogel matrix, and/or neuronal cell culture medium comprises any one or more of the following components: artemisinin, ascorbic acid, ATP, beta-endorphin, BDNF, calf serum (bovine calfserum), bovine serum albumin, calcitonin gene-related peptide, capsaicin, carrageenan, CCL2, ciliary neurotrophic factor, CX3CL1, CXCL1, CXCL2, D-serine, fetal bovine serum, fluorocitrate (fluoroform), formalin, glial cell line-derived neurotrophic factor, glial fibrillary protein, glutamate, IL-1 alpha, IL-1 beta, IL-6, IL-10, IL-12, IL-17, IL-18, insulin, laminin, lipoxin, mac-1-saporin, methionine sulfoximine, minocycline, neuregulin-1, neuroprotectin, rank neurotropin, NGF, nitric oxide, NT-3, NT-4, cyclophilin, NT-4, and/or dihydrocarb, persephin, platelet lysate, PMX53, poly-D-lysine (PLL), poly-L-lysine (PLL), propentofylline, resolvin (resolvins), S100 calbindin B, selenium, substance P, TNF- α, collagen type I-V, and zymosan.

As used herein, the term "optogenetics" refers to a biotechnology involving the use of light to control cells in living tissue (typically neurons) that have been genetically modified to express photosensitive ion channels. It is a neuromodulation method used in neuroscience that uses a combination of optical and genetic techniques to control and monitor the activity of individual neurons in living tissue-even in freely moving animals-and accurately measures the effects of those manipulations in real time. The key reagent used in optogenetics is the light sensitive protein. Spatially accurate neuronal control is achieved using optogenetic actuators such as channelrhodopsin, halorhodopsin (halorhodopsin) and archaerhodopsin (archaerhodopsin), while temporally accurate recordings can be made with the aid of optogenetic sensors for calcium ions (aequorin, carmelon (Cameleon), GCaMP), chloride ions (clomereon) or membrane voltages (Mermaid). In some embodiments, neural cells modified with optogenetic actuators and/or sensors are used in the culture systems described herein.

The term "plastic" refers to a biocompatible polymer comprising hydrocarbons. In some embodiments, the plastic is selected from the group consisting of: polystyrene (PS), Polyacrylonitrile (PAN), Polycarbonate (PC), polyvinylpyrrolidone, polybutadiene (PVP), polyvinyl butyral (PVB), polyvinyl chloride (PVC), Polyvinylmethylether (PVME), polylactic-co-glycolic acid (PLGA), poly (l-lactic acid), polyesters, Polycaprolactone (PCL), polyethylene oxide (PEO), Polyaniline (PANI), polyfluorene, polypyrrole (PPY), Polyethylenedioxythiophene (PEDOT) and mixtures of two or any two or more of the foregoing polymers. In some embodiments, the plastic is a mixture of three, four, five, six, seven, eight, or more polymers.

The term "seeding" as used herein refers to, for example, transferring a quantity of cells into a new culture vessel. The amount may be defined and the volume or number of cells may be used as a basis for defining the amount. The cell may be part of a suspension.

The term "sequence identity" as used herein refers to a particular percentage of residues that are identical within a particular region in the context of two or more nucleic acid or polypeptide sequences. The term is synonymous with "sequence homology" or "homology" of a sequence to another sequence. The percentage can be calculated by the following method: optimally aligning two sequences, comparing the two sequences over a specified region, determining the number of positions at which identical residues occur in the two sequences to produce a number of matched positions, dividing the number of matched positions by the total number of positions in the specified region, and multiplying the result by 100 to yield the percentage of sequence identity. Where the two sequences are of different lengths or the alignment results in one or more staggered ends and the specified region of comparison comprises only a single sequence, the residues of the single sequence are included in the denominator of the calculation rather than the numerator. When comparing DNA and RNA, thymine (T) and uracil (U) can be considered equivalent. Identity can be calculated manually or by using a computer sequence algorithm such as BLAST or BLAST 2.0.

The term "solid substrate" as used herein refers to any substance that is a solid support that is free or substantially free of cytotoxins. In some embodiments, the solid substrate comprises one or a combination of silicon dioxide, plastic, and metal. In some embodiments, the solid substrate comprises pores of sufficient size and shape to allow diffusion or inactive transport of proteins, nutrients, and gases through the solid substrate in the presence of cell culture media. In some embodiments, the pore size is no more than about 10, 9, 8, 7, 6,5, 4, 3, 2, or 1 microns in diameter. One of ordinary skill can determine what pore size is necessary based on the contents of the cell culture medium and the exposure of the cells growing on the solid substrate in a particular microenvironment. For example, one of ordinary skill in the art can observe whether any cultured cells in a system or device are viable under conditions using a solid substrate comprising pores of various diameters. In some embodiments, the solid substrate comprises a base having a predetermined shape that defines the shape of the outer surface and the inner surface. In some embodiments, the substrate comprises one or a combination of silica, plastic, ceramic, or metal, and wherein the substrate is cylindrical in shape or substantially similar to a cylinder such that the first cell-impermeable polymer and the first cell-permeable polymer coat an inner surface of the substrate and define a cylindrical or substantially cylindrical interior chamber; and wherein the opening is positioned at one end of the cylinder. In some embodiments, the base comprises one or more pores of sufficient size and shape to allow diffusion of proteins, nutrients, and oxygen through the solid substrate in the presence of the cell culture medium. In some embodiments, the solid substrate comprises a plastic base having a pore size of no greater than 1 micron in diameter, and comprises at least one layer of a hydrogel matrix; wherein the hydrogel matrix comprises at least a first cell-impenetrable polymer and at least a first cell-penetrable polymer; the substrate comprises a predetermined shape around which the first cell-impenetrable polymer and the at least first cell-penetrable polymer are physically adhered or chemically bonded; wherein the solid substrate comprises at least one compartment at least partially defined by the shape of the inner surface of the solid substrate and accessible from a point external to the solid substrate through an opening optionally positioned at one end of the solid substrate. In some embodiments, where the solid substrate comprises a hollow interior portion defined by at least one interior surface, cells in a suspension or tissue explant may be seeded by placing the cells at or near the opening such that the cells may adhere to at least a portion of the interior surface of the solid substrate prior to growth. The at least one compartment or hollow interior of the solid substrate allows for containment of cells in a particular three-dimensional shape defined by the shape of the interior surface of the solid substrate and promotes directed growth of cells away from the opening. In the case of neuronal cells, the degree and shape of containment of the at least one compartment facilitates axonal growth from a cell body positioned in the at least one compartment and at or near the opening. In some embodiments, the solid substrate is cylindrical, tubular, or substantially tubular or cylindrical such that the shape of the interior compartment is cylindrical or partially cylindrical. In some embodiments, the solid substrate comprises one or more branched tubular interior compartments. In some embodiments, the bifurcated or multi-bifurcated shape of the hollow interior portion of the solid is configured to or allows axons to grow in a multi-branch pattern. When and if electrodes are placed at the distal end of the axon to its vicinity and at or near the neuronal cell body, electrophysiological metrics, such as intracellular action potentials, can be measured within the device or system. In some embodiments, the electrodes are operably connected to a voltmeter, an ammeter, and/or a device capable of generating an electric current on a length of wire physically connecting the electrodes to the voltmeter, ammeter, and/or device.

The present disclosure relates to suitably filled hydrogels comprising a mixture of a cell permeable polymer and a cell impermeable polymer. In some embodiments, the hydrogel comprises about 10% to about 20% PEG, and has a total modulus of about 0.1 to about 200 Pa. In some embodiments, the hydrogel has a modulus of about 0.5 Pa. In some embodiments, the hydrogel has a modulus of about 10 Pa. In some embodiments, the hydrogel has a modulus of about 50 Pa. In some embodiments, the hydrogel has a modulus of about 75 Pa. In some embodiments, the hydrogel has a modulus of about 90 Pa. In some embodiments, the hydrogel has a modulus of about 100 Pa. In some embodiments, the hydrogel has a modulus of about 125 Pa. In some embodiments, the hydrogel has a modulus of about 150 Pa. In some embodiments, the hydrogel has a modulus of about 175 Pa. In some embodiments, the hydrogel has a modulus of about 200 Pa. In some embodiments, the hydrogel has a modulus of no greater than 230 Pa.

Spherical body

As used herein, a "spheroid" or "cell spheroid" may be, for example, any grouping of cells of a three-dimensional shape that generally corresponds to an ellipse or circle or convex or concave arc rotated about one of its major axes (major or minor) and includes three-dimensional ovoid, oblate and prolate spheroids, lenslets, or substantially equivalent shapes.

The spheroids of the present invention may have any suitable width, length, thickness and/or diameter. In some embodiments, the spheroid may have a width, a thickness, a width, a thickness, or a diameter in a range of about 10 μm to about 50,000 μm or any range therein (such as, but not limited to, about 10 μm to about 900 μm, about 100 μm to about 700 μm, about 300 μm to about 600 μm, about 400 μm to about 500 μm, about 500 μm to about 1,000 μm, about 600 μm to about 1,000 μm, about 700 μm to about 1,000 μm, about 800 μm to about 1,000 μm, about 900 μm to about 1,000 μm, about 750 μm to about 1,500 μm, about 1,000 μm to about 5,000 μm, about 1,000 μm to about 10,000 μm, about 2,000 to about 50,000 μm, about 25,000 μm to about 40,000 μm, or about 3,000 μm to about 15,000 μm). In some embodiments, the spheroid can have a width, length, thickness, and/or diameter of about 50 μm, 100 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, 1,000 μm, 5,000 μm, 10,000 μm, 20,000 μm, 30,000 μm, 40,000 μm, or 50,000 μm. In some embodiments, a plurality of spheroids are generated, and each spheroid of the plurality of spheroids may have a width, length, thickness, and/or diameter that varies by less than about 20%, such as, for example, less than about 15%, 10%, or 5%. In some embodiments, each spheroid of the plurality of spheroids may have a different width, length, thickness, and/or diameter within any of the ranges described above.

The cells in the spheroid may have a particular orientation. In some embodiments, the spheroid may include an inner core and an outer surface. In some embodiments, the spheroid may be hollow (i.e., may not contain cells inside). In some embodiments, the inner core cell and the outer surface cell are different types of cells. In some embodiments, the core comprises a magnetic nanoparticle.

The stiffness of the spheroid may vary, for example, as measured by the modulus of elasticity (pascal; Pa). In certain embodiments, the spheroids have an elastic modulus in the range of about 100Pa to about 10,000Pa, e.g., about 100Pa to about 12,000Pa or about 100Pa to about 4800 Pa. In some embodiments, the spheroid may have an elastic modulus of about 1200 Pa. As another example, the spheroid modulus may vary from about at least 10Pa, at least about 100Pa, at least about 150Pa, at least about 200Pa, or at least about 450 Pa. In some embodiments, a composition or device of the present disclosure comprises one or more wells, and each well comprises one or more different spheroids, a first, second, third, fourth, or fifth or more spheroid population. In one embodiment, the first spheroid comprises an elastic modulus of about 100Pa to about 300Pa, and the second spheroid comprises an elastic modulus of about 400Pa to about 800 Pa. In another example, the first spheroid is characterized by an elastic modulus of about 50 to about 200Pa and the second spheroid is characterized by an elastic modulus of about 250Pa to about 500 Pa.

In some embodiments, the spheroid may be composed of one, two, three, or more different cell types (including one or more neuronal cell types and/or one or more stem cell types). In some embodiments, the inner core cell can be composed of one, two, three, or more different cell types. In some embodiments, the outer surface cells may consist of one, two, three, or more different cell types.

In some embodiments, the spheroid comprises at least two types of cells. In some embodiments, the spheroid comprises neuronal cells and non-neuronal cells. In some embodiments, the spheroid comprises neuronal cells and astrocytes in a ratio of about 5:1, 4:1, 3:1, 2:1, or 1: 1. In some embodiments, the spheroid comprises neuronal cells and non-neuronal cells in a ratio of about 5:1, 4:1, 3:1, 2:1, or 1: 1. In some embodiments, the spheroid comprises neuronal cells and non-neuronal cells in a ratio of about 1:5, 1:4, 1:3, or 1: 2. Any combination of cell types disclosed herein can be used in the spheroids of the present disclosure at the ratios identified above.

According to particular embodiments, the groups of cells may be placed in any suitable shape, geometry, and/or pattern. In some embodiments, the cells are arranged in spheres across the surface area of a bead or nanoparticle having a solid or hollow core. For example, individual groups of cells may be deposited as spheroids, and the spheroids may be arranged within a three-dimensional grid or any other suitable three-dimensional pattern. The individual spheroids may all comprise about the same number of cells and be of about the same size, or alternatively, different spheroids may have different numbers of cells and different sizes. In some embodiments, a plurality of spheroids may be arranged radially from a single point or multiple points in a shape such as an L or T shape, sequential spheroids may be arranged in a single line or parallel lines, a tube, a cylinder, a torus (toroid), a network of hierarchically branched vessels, a high aspect ratio object, a thin closed shell, an organoid, or other complex shape that may correspond to the geometry of a tissue, vessel, or other biological structure.

Any suitable physiological response of the spheroid can be determined, evaluated, measured, and/or identified in the methods of the present disclosure. In some embodiments, 1, 2, 3, 4, or more physiological responses of the spheroid can be determined, evaluated, measured, and/or identified in the methods of the present disclosure. In some embodiments, the physiological response of the spheroid may be a change in morphology of the spheroid. The method may include determining a change in morphology of the spheroid, which may include estimating at least one morphological parameter prior to contacting the spheroid with an agent (such as a chemical and/or biological compound), estimating the at least one morphological parameter after contacting the spheroid with the agent, and calculating a difference between the at least one morphological parameter before and after contacting the spheroid with the agent to provide a change in morphology of the spheroid. In some embodiments, the physiological response of the spheroid may be the contraction or swelling of the spheroid in response to contact with an agent. The morphology of the spheroid may be determined using any method known to those skilled in the art, such as, but not limited to, quantifying eccentricity and/or cross-sectional area.

In some embodiments, the physiological response of the spheroid may be a change in volume of the spheroid. The method may include determining a change in volume of a spheroid, which may include estimating a first volume before contacting the spheroid with an agent, estimating a second volume after contacting the spheroid with the agent, and calculating a difference between the first volume and the second volume to provide the change in volume of the spheroid. In some embodiments, the physiological response of the spheroid may be the contraction or swelling of the spheroid in response to contact with an agent.

The agent can be any suitable compound, such as an organic compound, a small molecule compound (e.g., a small molecule organic compound), a protein, an antibody, an oligonucleotide (e.g., DNA and/or RNA), a gene therapy vehicle (e.g., a viral vector), and any combination thereof. One or more (e.g., 1, 2, 3, 4,5, or more) agents can be used in the methods of the invention. For example, the methods of the invention can comprise contacting the spheroids of the invention with two or more different agents. In some embodiments, the methods of the invention can modulate activity in the spheroids indirectly, such as, for example, by contacting the spheroids of the invention with a gene therapy vehicle (e.g., a viral vector).

The methods of the invention may comprise culturing cells and/or spheroids. The cultivation can be carried out using methods known to the person skilled in the art. In some embodiments, the cells and/or spheroids may be cultured for any desired period of time, such as, but not limited to, hours, days, weeks, or months. In some embodiments, the cells and/or spheroids may be cultured for about 1, 2, 3, 4,5, 6, or 7 days, or for about 1, 2, 3, 4,5, 6, 7, 8, 9, 10, or 11 or more weeks.

Cell culture media suitable for use in the methods of the invention are known in the art and include, but are not limited to, BEGMTMBronchial epithelial cell growth medium, Dulbecco's Modified Eagle's Medium (DMEM), high glucose Dulbecco's modified Eagle's medium (DMEM-H), McCoy's 5A modified medium, RPMI, Ham medium, medium 199, mTeSR, and the like. The cell culture medium may be supplemented with additional components such as, but not limited to, vitamins, minerals, salts, growth factors, carbohydrates, proteins, serum, amino acids, attachment factors, cytokines, growth factors, hormones, antibiotics, therapeutics, buffers, and the like. Cell culture components and/or conditions may be selected and/or altered during performance of the methods of the invention to enhance and/or mimic certain cellular characteristics and/or properties. Examples of inoculation methods and cell culture methods are described in U.S. Pat. Nos. 5,266,480, 5,770,417, 6,537,567 and 6,962,814 and in "De novo recording of a functional amino acid black by tissue engineering" Nature Biotechnology17:149-155(1999), Oberpenning et alAre incorporated by reference herein in their entirety. In some embodiments, the cell culture medium is altered in a stepwise manner to promote myelination of axons in the culture. Pre-myelination (Pre-myelination) and myelination media include the following components:

TABLE 1 components of myelination induction medium protocol.

Figure BDA0002610186270000421

In some embodiments, the solid substrate, cell culture device, or nanoparticle comprises a spheroid comprising one or more cell types disclosed herein. Any particle may comprise any one or combination of 1, 2, 3, 4,5, 6, 7, 8, or more cell types herein.

The term "nanoparticle" or "nanoshuttle" when each term is used interchangeably is a particle that includes at least one region. Magnetic particles in the range of about 0.7 microns to about 1.5 microns have been described in patent documents including, for example, U.S. patent nos. 3,970,518, 4,018,886, 4,230,685, 4,267,234, 4,452,773, 4,554,088, 4,659,678, 6,623982, 6,645,731 and U.S. application No. 20110250146, each of which is hereby incorporated by reference in its entirety. The nanoparticles can be used to magnetize, i.e., make responsive to a magnetic field, any of the cells or spheroids described herein. Some compositions and/or systems of the present disclosure include a cell in contact with a magnetically-responsive element or a spheroid containing a magnetically-responsive element. In some embodiments, the compositions and/or systems of the present disclosure comprise a cell or spheroid comprising one or more magnetic nanoparticles in contact with one or more magnetic nanoparticles. As used herein, a "magnetically-responsive element" can be any element or molecule that will respond to a magnetic field. The one or more nanoparticles must contain or be a magnetically responsive element. In some embodiments, the nanoparticle can be taken up or adsorbed by any cell described herein. In some embodiments, the magnetic field may be used to manipulate the position, shape, pattern, or motion of the cell or spheroid.

In some embodiments, the charged nanoparticles have nanoscale dimensions. In some embodiments, the nanoparticles of the present disclosure have a size from about 5nm to about 1000nm, or any range therein. In some embodiments, the nanoparticles have a diameter size of about 5nm to about 250nm, about 25nm to about 225nm, about 50nm to about 200nm, about 75nm to about 150 nm. In some embodiments, the nanoparticle has a diameter of about 5nm, about 10nm, about 20nm, about 40nm, about 60nm, about 80nm, about 100nm, about 120nm, about 140nm, about 160nm, about 180nm, about 200nm, about 250nm, about 300nm, about 400nm, about 500nm, about 600nm, about 700nm, about 800nm, about 900nm, or about 1000 nm. In some embodiments, the nanoparticle has a diameter of no greater than about 5nm, about 10nm, about 20nm, about 40nm, about 60nm, about 80nm, about 100nm, about 120nm, about 140nm, about 160nm, about 180nm, about 200nm, about 250nm, about 300nm, about 400nm, about 500nm, about 600nm, about 700nm, about 800nm, about 900nm, or about 1000 nm. In some embodiments, the nanoparticles are of substantially uniform size. In some embodiments, the nanoparticles are of different sizes. In some embodiments, the size of the nanoparticle will depend on what type of cell is being used.

The magnetically-responsive element may be any element or molecule that will respond to a magnetic field. In some embodiments, the magnetically-responsive element is a rare earth magnet, such as, for example, samarium cobalt (SmCo) or neodymium iron boron (NdFeB). In some embodiments, the magnetically-responsive element is a ceramic magnet material, such as, for example, strontium ferrite. In some embodiments, the magnetically-responsive element is a magnetic element, such as, for example, iron, cobalt, nickel, or any alloy or oxide thereof. In some embodiments, the magnetically-responsive element comprises gold. In some embodiments, the magnetically-responsive element is a paramagnetic material that reacts to a magnetic field, and is not a magnet per se, as this makes assembly of the material easier.

At one endIn some embodiments, the nanoparticles comprise one or more iron oxides, including, for example, iron (III) oxide, α -Fe2O3、γ-Fe2O3、β-Fe2O3、-Fe2O3Iron (II) oxide, or iron (II, III) oxide. In some embodiments, the nanoparticle comprises one or more of gold, iron oxide, and polylysine.

In some aspects of the present disclosure, coated magnetic particles are provided that comprise a nanoparticle core of a magnetic material and a substrate coating material on the magnetic core in an amount sufficient to impede non-specific binding of a biomacromolecule to the magnetic core. These magnetic particles are characterized by extremely low non-specific binding and efficient target capture, which is essential to achieve the level of enrichment required to effectively isolate very rare cells (such as neurons or other cell types disclosed herein). In an alternative embodiment, a coated magnetic particle is provided comprising the structure: i. a nanoparticle core of a magnetic material; a substrate coating material that forms a discontinuous coating on the magnetic core, thereby providing at least one discontinuous region that, if accessible, promotes non-specific binding of the substrate-coated particles to the biomacromolecule; additional coating material that blocks the entrance of biological macromolecules into the discrete regions. The magnetic core material of the particles described immediately above may comprise at least one transition metal oxide and a suitable substrate coating material comprises a protein. Proteins suitable for coating magnetic particles include, but are not limited to, bovine serum albumin and casein. The additional coating material may be the original coating protein or one member of a specific binding pair coupled to the substrate material on the magnetic core. Exemplary specific binding pairs include biotin-streptavidin, antigen-antibody, receptor-hormone, receptor-ligand, agonist-antagonist, lectin-carbohydrate, protein a-antibody Fc, and avidin-biotin. In one embodiment, the member of the specific binding pair is coupled to the substrate coating material via a bifunctional linking compound. Exemplary bifunctional linking compounds include succinimidyl-propionyl-dithiopyridine (SPDP) and sulfosuccinimidyl-4- [ maleimidomethyl ] cyclohexane-1-carboxylate (SMCC), but various other such heterobifunctional linker compounds are available from Pierce, Rockford, III.

The coated magnetic particles of the present invention preferably have a magnetic mass of 70-90%. In some embodiments, the majority of the magnetic particles have a particle size in the range of from about 90 to about 150 nm. The particles may be synthesized such that they are more monodisperse, for example, in the range of from about 90 to about 120nm or from about 120 to about 150 nm. The particles of the invention are typically suspended in a biocompatible medium.

In some embodiments, the nanoparticles may be combined with a support molecule. A "support molecule" is generally a polymer or other long molecule used to intimately mix the nanoparticle and the cell together. The support molecule may be positively charged, negatively charged or mixed charge or neutral, and may be a combination of more than one support molecule. In some embodiments, the support molecule is a natural polymer or a cell-derived polymer. Non-limiting examples of such polymers include peptides, polysaccharides, and nucleic acids. In other embodiments, the support molecule is a synthetic polymer. In some embodiments, the polymer is polylysine. In some embodiments, the support molecule may be one or more of the following: polylysine, fibronectin, collagen, laminin, BSA, hyaluronic acid (hyaluronan), glycosaminoglycans, anionic non-sulfated glycosaminoglycans, gelatin, nucleic acids, extracellular matrix protein mixtures, matrigel, antibodies, and mixtures and derivatives thereof. In some embodiments, the nanoparticle comprises Ferridex, a material consisting of dextran-coated superparamagnetic iron oxide nanoparticles (SPIONs).

The nanoparticles may be positively or negatively charged. In some embodiments, the negatively charged nanoparticles contain a charge-stabilized metal (e.g., silver, copper, platinum, palladium, gold). In some embodiments, the negatively charged nanoparticles comprise gold.

In some embodiments, the positively charged nanoparticles contain surfactant or polymer stabilized or coated alloys and/or oxides (e.g., elemental iron, iron-cobalt, nickel oxide, iron oxide). In some embodiments, the positively charged nanoparticles comprise iron oxide.

The present disclosure also relates to a system, comprising:

(i) a hydrogel matrix;

(ii) one or more spheroids;

(iii) a current generator;

(iv) a voltmeter and/or ammeter;

(v) at least a first stimulating electrode and at least a first recording electrode;

wherein the generator, the voltmeter and/or ammeter and the electrodes are electrically connected to each other by an electrical circuit, wherein an electrical current is fed from the generator to the at least one stimulation electrode and an electrical current is received at the recording electrode and fed to the voltmeter and/or ammeter; wherein the stimulating electrode is positioned at or adjacent to one or more cell bodies of the neuronal cell and the recording electrode is positioned at a predetermined distance distal to the cell bodies such that an electrical potential is established across the cell culture container.

In some embodiments, the solid substrate is comprised of a hydrogel or hydrogel matrix. In some embodiments, the solid substrate is comprised of a hydrogel or hydrogel matrix and is free of glass, metal, or ceramic. In some embodiments, the solid substrate is shaped into a form or mold that is predetermined to seed cells of a particular size suitable for axonal growth. In some embodiments, the solid substrate or at least one substrate portion is shaped to have an inner tubular structure of at least one branch, wherein the tube is optionally tapered in diameter the further away from the location of seeding with the tissue explant or neuronal cell. For example, the present disclosure contemplates a center point at one end of a semi-cylindrical or cylindrical portion of a solid substrate that is accessible through an opening or hole at an outer surface at a point external to the solid substrate. The opening or hole can be used to place or seed a cell (any one or more of any one or combination of cells of the disclosed cells) at the aforementioned center point. When the cells are allowed to grow in culture for several days, the cells are exposed to a medium containing any of the components disclosed herein at a concentration and for a period of time sufficient to allow axons to grow from the neuronal cells. If cells are to be myelinated or myelination is desired for study, glial cells may be introduced through the same hole and seeded before neuronal cells or explants are added. The axonal process growth may occur at increasing distances from the center point as the axon grows in a semi-cylindrical or tubular structure. The entry point or opening at a point in the solid substrate that is further and further away from the central point (or seeding point) may be used to locate or observe axonal growth in axonal status. The present disclosure contemplates that the structure of the solid substrate takes any form that promotes axonal growth. In some embodiments, the interior chamber or compartment housing the axon projections comprises a semi-circular or substantially cylindrical diameter. In some embodiments, the solid substrate branches in two or more internal compartments at a point distal from the central point. In some embodiments, such branches may resemble a keyhole shape or tree, where there are 2, 3, 4,5, 6, 7, or 8 or more tubular or substantially cylindrical interior chambers in fluid communication with each other, such that axonal growth originates at the inoculation point of one or more cells, and extends longitudinally along the interior chambers and into one or more branches. In some embodiments, one or more electrodes may be placed at or adjacent to one or more openings so that recordings may be taken across one or more locations along the length of the axon. This can also be used to probe one or more locations along the length of the axon.

The present disclosure relates to a system for accurately measuring recordings between artificial central nervous system nodes and peripheral nervous system nodes, the system comprising at least a first spheroid and a second spheroid, the first spheroid comprising a dorsal root ganglion or a neuronal cell or a mammalian embryonic cell from the central nervous system; and the second spheroid comprises at least one neuronal cell or primary mammalian stem cell from the peripheral nervous system. The present disclosure relates to manufacturing any of the systems disclosed herein by: at least a first or second spheroid containing a magnetic substance is positioned in a hole or channel defined by the hydrogel, and the first or second spheroid is moved by aligning a magnet at or adjacent to the hole or channel. If the system mimics an axon traveling between a central nervous system node or group of cells and a peripheral nervous system node or group of cells, in some embodiments, the first spheroid is positioned at or near a first pore or channel and the second spheroid is positioned at or near a second pore or channel at a distance sufficient to allow axonal growth between the two spheroids upon exposure to cell culture media. The present disclosure relates to measuring recordings between a central neural node or group of cells and a peripheral node or group of cells, the method comprising placing an electrode at or adjacent a first spheroid, placing an electrode at or near a second spheroid, and electrically stimulating the system using an amplifier or generator comprising a generator. In some embodiments, the method further comprises measuring the electrophysiological response.

The term "recording" as used herein refers to, for example, measuring the response of one or more neuronal cells. Such a response may be an electrophysiological response, e.g. patch clamp electrophysiological recording or field potential recording.

The present disclosure discloses methods and apparatus for obtaining physiological measurements of micro-scale organotypic models of neural tissue in vitro that simulate clinical nerve conduction and NFD testing. The results obtained by using these methods and devices may better predict clinical outcomes, thereby enabling a more cost-effective approach for selecting promising lead compounds with a higher likelihood of late-stage success. The present disclosure includes the fabrication and utilization of a three-dimensional micro-engineered system that enables exceptionally dense, highly parallel nerve fiber bundle growth. Due to the limited nature of the beam, this in vitro model is able to measure both CAP and intracellular patch clamp recordings. In addition, subsequent confocal and Transmission Electron Microscopy (TEM) analysis allows quantitative structural analysis, including NFD. In summary, this in vitro model system has a new capability to evaluate histomorphometry and population electrophysiology, similar to clinical histopathology and nerve conduction tests.

The present disclosure also provides methods for measuring axonal myelination created using the in vitro models described herein. Similar to the structure of human afferent peripheral nerves, Dorsal Root Ganglion (DRG) neurons in these in vitro constructs project long, parallel, fasciculated axons towards the periphery. In natural tissues, axons of different diameters and myelination degrees conduct sensory information back to the central nervous system at different rates. Schwann cells support sensory relays by myelinating axons and providing isolation for faster conduction. Similarly, the three-dimensional growth induced by this in vitro construct comprises axes of various diameters spanning distances up to 3mm in a closely parallel orientation. Schwann cell presence and ensheathing (sheathong) was observed in confocal and TEM imaging.

Although neuronal morphology is a useful indicator of phenotypic maturity, a more definitive hallmark of healthy neurons is their ability to conduct action potentials. Apoptosis alone is not a comprehensive measure of neuronal health, as many pathological changes may occur before cell death manifests. Electrophysiological studies of action potential generation can determine whether the observed structure supports predicted function, and the ability to measure clinically relevant endpoints produces more predictive results. Similarly, information gathered from imaging can determine a quantitative indicator of the degree of myelination, while CAP measurements indicate the overall health of myelin and help to gain further insight into the toxicity and neuroprotective mechanisms of various agents or compounds of interest.

In some embodiments, the at least one agent comprises a small chemical compound. In some embodiments, the at least one agent comprises at least one environmental or industrial pollutant. In some embodiments, the at least one agent comprises one or a combination of small chemical compounds selected from the group consisting of: chemotherapeutic agents, analgesics, cardiovascular modulators, cholesterol, neuroprotective agents, neuromodulatory agents, immunomodulators, anti-inflammatory agents and antimicrobial agents.

In some embodiments, the at least one agent comprises one or a combination of chemotherapeutic agents selected from the group consisting of: actinomycin, alitretinol, all-trans retinoic acid, azacitidine, azathioprine, bexarotene, bleomycin, bortezomib, capecitabine, carboplatin, chlorambucil, cisplatin, cyclophosphamide, cytarabine, Dacarbazine (DTIC), daunorubicin, docetaxel, doxifluridine, doxorubicin, epirubicin, Epothilone (Epothilone), erlotinib, etoposide, fluorouracil, gefitinib, gemcitabine, hydroxyurea, idarubicin, imatinib, irinotecan, nitrogen mustard, melphalan, mercaptopurine, methotrexate, mitoxantrone, nitrosourea, oxaliplatin, paclitaxel, pemetrexed, Romidepsin, taclofluposide (Tafluposide), temozolomide (oral dacarbazine), teniposide, thioguanine (formerly thioguanine), topotecan, valrubic acid, pentan (Valbixin) Vemurafenib, vinblastine, vincristine, vindesine, vinorelbine, Vismodegib (Vismodegib), and Vorinostat (Vorinostat).

In some embodiments, the at least one agent comprises one or a combination of analgesics selected from the group consisting of: paracetamol (Paracetamol), non-steroidal anti-inflammatory drugs (NSAIDs), COX-2 inhibitors, opioids, flupirtine, tricyclic antidepressants, carbamazepine, gabapentin and pregabalin.

In some embodiments, the at least one agent comprises one or a combination of cardiovascular modulators selected from the group consisting of: nepicastat (nepicastat), cholesterol, niacin, scutellaria baicalensis (scutellaria), prenylamine (prenylamine), dehydroepiandrosterone, monatepile (monatepil), esketamine, niguldipine (niguldipine), asenapine, atomoxetine, flunarizine, milnacipran, mexiletine, amphetamine, thiopentasodium, flavonoids, bromobenzylamine, oxazepam, and magnolol (honokiol).

In some embodiments, the at least one agent comprises one or a combination of neuroprotective and/or neuromodulatory agents selected from the group consisting of: tryptamine, galanin receptor 2, phenylalanine, phenylethylamine, N-methylphenethylamine, adenosine, kyphotide (kyptorphin), substance P, 3-methoxytyramine, catecholamine, dopamine, GABA, calcium, acetylcholine, epinephrine, norepinephrine, and hydroxytryptamine.

In some embodiments, the at least one agent comprises one or a combination of immunomodulatory agents selected from the group consisting of: clinumomab (clenolizimab), epritumumab (enotizumab), rigolizumab (ligelizumab), cetuzumab (simtuzumab), vatuzumab (vatelizumab), parsuzumab (parsatuzumab), imatuzumab (Imgatuzumab), trastuzumab (tregalizumab), pertuzumab (patelizumab), naloxonumab (namulumab), perrazumab (perazimab), framumab (patrimumab), attitumumab (atib), urituximab (urtitumumab), rituximab (ubuliximab), trastuzumab (futuximab) and duritumumab (duligitumab).

In some embodiments, the at least one agent comprises one or a combination of anti-inflammatory agents selected from the group consisting of: ibuprofen, aspirin, ketoprofen, sulindac, naproxen, etodolac, fenoprofen, diclofenac, flurbiprofen, ketorolac, piroxicam, indomethacin, mefenamic acid, meloxicam, nabumetone, oxaprozin, ketoprofen, famotidine, meclofenamate (meclofenamate), tolmetin, and salsalate (salsalate).

In some embodiments, the at least one agent comprises one or a combination of antimicrobial agents selected from the group consisting of: antibacterial, antifungal, antiviral, antiparasitic, heat, radiation, and ozone.

The present disclosure additionally discloses methods of measuring both intracellular and extracellular recordings of biomimetic neural tissue in a three-dimensional culture platform. Previously, electrophysiological experiments were performed in dissociated surface-seeded cultures or organotypic slice preparations with limitations inherent to each approach. Studies in dissociated cell cultures are generally limited to single cell recordings due to the lack of organized multicellular neurite architecture, as will be seen in organotypic preparations. Organotypic preparations have intact neural circuits and allow both intracellular and extracellular studies. However, acute brain slices present a complex series of simultaneous variables without a means of controlling a single factor and are therefore inherently limited in flux potential.

Intracellular recordings in three-dimensional cultures in vitro have been previously demonstrated. However, neuronal outgrowth is not spatially limited to anatomically relevant structures that support studies of extracellular populations. A more biomimetic three-dimensional neural culture is needed to allow examination of electrophysiological behavior at the population level. The present disclosure supports the technique of whole-cell patch clamping and synchronized population-level events in extracellular field recordings generated by restricted neurite outgrowth in three-dimensional geometry. Prior to the present disclosure, the measurement of these endpoints was directly analogous to clinical nerve conduction testing, and yet to be validated for pure cell in vitro studies.

Using the methods and devices disclosed herein, field recording is used to measure the combined extracellular changes in potential caused by signaling in all recruited fibers. The population response elicited by electrical stimulation is CAP. The electrically induced population peak potentials are graded in nature, including the combined effect of action potentials in slow fibers and fast fibers. Spike potential is a single coherent event with a fast onset phase and short duration, which is characteristic of the response of CAP or action potentials that contain only rapid signaling in the absence of synaptic input. The three-dimensional neural constructs disclosed in this disclosure also support CAP stimulation from greater distances along the neurite tracts or channels, which demonstrates the ability of neural cultures to be able to rapidly transmit signals from distant stimuli, much like afferent peripheral nerves. The three-dimensional neural cultures of the present disclosure support proximal and distal stimulation techniques that can be used to measure conduction properties.

The present disclosure may be used with one or more growth factors that induce recruitment of many fiber types typical of a bundle of nerve fibers. In particular, Nerve Growth Factor (NGF) preferentially recruits small diameter fibers, which are often associated with pain signaling, as demonstrated in the data provided herein. It has been demonstrated that brain-derived neurotrophic factor (BDNF) and neurotrophic factor 3(NT-3) preferentially support the growth of larger diameter proprioceptive fibers. Growth-affecting factors such as bioactive molecules and pharmacological agents can be incorporated with electrophysiological studies to allow systematic manipulation of conditions for mechanistic studies.

Three-dimensional neural cultures formed using the present disclosure can be used as a platform to study the mechanisms underlying myelin-damaging disease and peripheral neuropathy by studying the effects of known dysmyelination agents, neuropathy-inducing culture conditions, and toxic neuropathy-inducing compounds on the neural cultures. The present disclosure allows for the use of conduction velocity as a functional measure of myelin and nerve fiber integrity under toxic and therapeutic conditions, thereby facilitating studies on drug safety and efficacy. The incorporation of genetic mutations and drugs into neural cultures produced using the techniques disclosed herein may enable the reproduction of disease phenomena in a controlled manner, leading to a better understanding of neurodegeneration and possible therapeutic therapies.

The present disclosure provides devices, methods, and systems related to generating, maintaining, and physiologically exploring micro-engineered neural cells and neural networks designed to mimic the native neural tissue anatomy. In some embodiments, the devices and systems include one or more cultured or isolated schwann cells and/or one or more cultured or isolated oligodendrocytes that are in contact with one or more neuronal cells in a cell culture container comprising a solid substrate comprising at least one outer surface, at least one inner surface, and at least one interior chamber; the shape of the interior chamber is defined at least in part by the at least one interior surface and is accessible from a point exterior to the solid substrate through at least one opening in the exterior surface; wherein a cell body of the one or more neuronal cells is positioned at one end of the internal chamber and an axon is capable of growing within the internal chamber along at least one length of the internal chamber such that a location of an axon tip extends distally from the cell body. In some embodiments, the inner surface of the solid substrate is cylindrical in shape or is substantially cylindrical such that a cell body from a neuronal cell is positioned at an open proximal end at one end of the cylindrical or substantially cylindrical inner surface, and an axon of the neuronal cell comprises a length of cellular material extending along the length of the inner surface from a point at the edge of the cell body to a point distal to the cell body. In some embodiments, the inner surface of the solid substrate is cylindrical in shape or is substantially cylindrical such that a cell body from a neuronal cell is positioned at an open proximal end at one end of the cylindrical or substantially cylindrical inner surface, and an axon of the neuronal cell comprises a length of cellular material extending along the length of the inner surface from a point at the edge of the cell body to a point distal to the cell body. In some embodiments, the inner surface of the solid substrate is cylindrical in shape or is substantially cylindrical such that a cell body from a neuronal cell is positioned at an open proximal end at one end of the cylindrical or substantially cylindrical inner surface, and an axon of the neuronal cell comprises a length of cellular material extending along the length of the inner surface from a point at the edge of the cell body to a point distal to the cell body; wherein, if the cell culture vessel contains a plurality of neuronal cells, the plurality of axons extend from the plurality of cells (or cell bodies) such that the plurality of axons define a bundle of axons capable of growing distally from the cell bodies along the length of the inner surface. In some embodiments, the neuronal cells are grown on and within a penetrable polymer. In some embodiments, one or more electrodes are positioned at or adjacent to the tip of at least one axon and one or more electrodes are positioned at or adjacent to the cell body such that a voltage potential is established across the length of one or more neuronal cells.

It is another object of the present disclosure to provide medium to high throughput assays of neurological function for screening pharmacological and/or toxicological properties of chemical and biological agents. In some embodiments, the agent is a cell (such as any type of cell disclosed herein) or an antibody (such as an antibody for treating a clinical disease). In some embodiments, the agent is any drug or agent used to treat human diseases such that toxicity, impact, or neuromodulation can be compared between new agents as proposed mammalian treatments and existing treatments for human diseases. In some embodiments, the novel agent for treating a human disease is for treatment of a neurodegenerative disease and is compared to existing treatments for neurodegenerative diseases. In the case of multiple sclerosis, as a non-limiting example, the effects of a new agent (modified cells, antibodies, or small chemical compounds) can be compared and compared to the same effects of existing treatments for multiple sclerosis such as Copaxone (Copaxone), riti (Rebif), other interferon therapies, tysbri (Tysabri), dimethyl fumarate, fingolimod, teriflunomide (teriflunomide), mitoxantrone, prednisone, tizanidine, and baclofen.

It is another object of the present disclosure to use a unique set of techniques such as two-dimensional and three-dimensional micro-engineering of nerve bundles in conjunction with electrophysiological stimulation and recording of populations of nerve cells.

It is another object of the present disclosure to provide a novel approach for assessing neurophysiologic function in vitro that uses the Compound Action Potential (CAP) as a clinically similar indicator to obtain results that are more sensitive and predictive of human physiology than those provided by current methods.

It is another object of the present disclosure to provide a micro-engineered neural tissue that mimics natural anatomical and physiological features and is easily evaluated using high-throughput electrophysiological stimulation and recording methods.

It is another object of the present disclosure to provide methods for replicating, manipulating, modifying and evaluating the mechanisms underlying myelin-damaging diseases and peripheral neuropathies.

Another object of the present disclosure is to allow medium to high throughput assays of neuromodulation of human neural cells for screening pharmacological and/or toxicological activity of chemical and biological agents.

It is another object of the present disclosure to use a unique set of technologies such as 2D and 3D micro-engineering nerve bundles in conjunction with optical and electrochemical stimulation and recording of human neural cell populations.

Another object of the present disclosure is to quantify postsynaptic potentials induced in biomimetic engineered thalamocortical circuits. We observed population spikes generated retrograde in the nerve bundle, suggesting that they are able to perform population-level physiological functions such as conduction of complex action potentials and postsynaptic potentials.

It is another object of the present disclosure to enable non-invasive stimulation and recording of multi-unit physiological responses to evoked potentials in neural circuits using optogenetic methods, hardware and software control of illumination, and fluorescence imaging.

It is another object of the present disclosure to use a micro-engineered loop to test selective 5-H T reuptake inhibitors (SSRIs) and secondary antipsychotics to see if they alter their developmental maturation.

In one embodiment, the combination of polyethylene glycol dimethacrylate and Puramatrix hydrogel was micropatterned using projection lithography using a Digital Micromirror Device (DMD), as shown in fig. 1. This approach enables rapid micropatterning of one or more hydrogels directly onto conventional cell culture materials. Since the photomask never comes into contact with the gel material, multiple hydrogels can be rapidly cured in series, enabling the production of many gel constructs within 1 hour without automation. This approach enables the use of polyethylene glycol (PEG), a mechanically robust cell growth-limiting gel, to confine neurite growth within a biomimetic growth-promoting gel. In some embodiments, such growth-promoting gels may be Puramatrix, agarose, or methacrylated dextran. When embryonic Dorsal Root Ganglion (DRG) explants were grown in this constrained three-dimensional environment, axons grew out of the ganglia at high density and fasciation as shown in fig. 5 and 6. Most axons appear as small diameter, unmyelinated fibers that grow to a length of approximately 1mm in 2 to 4 weeks. The structure of this culture model with dense, highly parallel, three-dimensional bundles of nerve fibers extending from the ganglia is roughly similar to the peripheral nerve architecture. Its morphology can be assessed using a neuromorphic assay, allowing clinically similar assessments to be made that are not available for traditional cellular assays.

In some embodiments, the culture model provides the ability to record electrically evoked population field potentials generated by Compound Action Potentials (CAP). The example traces show characteristic uniform, rapid, short latency, population spike responses that remain consistent under high frequency (100Hz) stimulation, as seen in fig. 8B. This CAP is reversibly eliminated by tetrodotoxin (TTX), as shown in fig. 8E and 8F, confirming that the drug can be administered and confirmed to have an effect. There is a measurable extension of the onset phase delay time associated with distal nerve bundle stimulation, as seen in fig. 8C and 8D. The response was insensitive to neurotransmitter blockers, indicating that the evoked response was primarily CAP and not synaptic potential, as shown in fig. 10. Embryonic DRG cultures have been used effectively as models of peripheral neurobiology for decades. While very useful as a model system, conventional DRG cultures are known to be poorly predictive of clinical toxicity when evaluated with traditional cell viability assays. Although single cell patch clamp recordings in DRG cultures were possible, no reports of CAP were recorded due to the lack of tissue architecture. In preferred embodiments, the present disclosure provides the ability to evaluate histomorphometric assays and population electrophysiology similar to clinical histopathology and nerve conduction tests.

In some embodiments, the present disclosure uses human neural cells to grow neural tissue in a three-dimensional environment, where neuronal cell bodies are bundled together and located at different locations from axonal fiber bundles, thereby simulating natural neural architecture and allowing measurement of morphometric and electrophysiological data, including CAP. In some embodiments, the present disclosure uses neuronal cells and glial cells derived from primary human tissue. In other embodiments, the neuronal and glial cells may be derived from human stem cells, including induced pluripotent stem cells.

In another embodiment, the present disclosure uses conduction velocity as a functional measure of the condition of neural tissue under toxic and therapeutic conditions. Information about the extent of myelination, myelin health, axonal transport, mRNA transcription, and neuronal damage can be determined by electrophysiological analysis. In conjunction with morphometric analysis of nerve density, percentage of myelination, and type of nerve fiber, the mechanism of action of the compound of interest can be determined. In some embodiments, the devices, methods, and systems disclosed herein can incorporate genetic mutations and drugs to reproduce disease phenomena in a controlled manner, thereby allowing a better understanding of neurodegeneration and possible therapeutic therapies.

The present disclosure relates to systems comprising any of the disclosed compositions, and methods of using the systems to capture involved data that is more physiologically relevant compared to data collected using a two-dimensional tissue culture system or a system that does not use multiple cell types. In some embodiments, the systems or compositions disclosed herein comprise one or more cells comprising any mutation. In some embodiments, at least 1, 100, 500, 1,000, or more cells comprise a mutation associated with a particular model of human disease. Any of the disclosed systems can include cells having the mutations disclosed in table B below. In some embodiments, cells of the present disclosure include or express endogenous muteins disclosed in table B or at least about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% of those muteins, and thus those model cells can be used to test the efficacy or toxicity of certain drugs, biomolecules, or other therapeutic agents added to the system. The model can also be used to understand basic biology in the context of environmental contaminants, pathogens, or endogenously expressed proteins, and how such molecules affect the nervous system based on such information as responses to agents and axonal growth, myelination, and demyelination or morphological changes of the cells themselves.

TABLE B mutations

Figure BDA0002610186270000571

Figure BDA0002610186270000591

Figure BDA0002610186270000601

Figure BDA0002610186270000631

Figure BDA0002610186270000651

Figure BDA0002610186270000731

Figure BDA0002610186270000811

Figure BDA0002610186270000821

Figure BDA0002610186270000831

Figure BDA0002610186270000841

Figure BDA0002610186270000871

Figure BDA0002610186270000891

Figure BDA0002610186270000921

Figure BDA0002610186270000931

Figure BDA0002610186270000941

Figure BDA0002610186270000961

Figure BDA0002610186270000971

Figure BDA0002610186270000991

Figure BDA0002610186270001001

Figure BDA0002610186270001041

Figure BDA0002610186270001051

Figure BDA0002610186270001061

Figure BDA0002610186270001081

In some embodiments, at least one cell in the disclosed system comprises any one or more of the mutations at the loci identified in table B. If Table B discloses mRNA sequences, one or more mutations of the cell may be located in the complementary endogenous DNA sequences disclosed at the GenBank sequences above. In some embodiments, the cell comprises a mutation of one or more of the sequences identified in table B above, or a sequence having at least about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to any of the sequences disclosed in table B, or, if the sequence is an mRNA sequence, the cell may comprise a mutation of one or more complementary DNA sequences of those identified in table B, or a mutation that is a sequence comprising or complementary to a sequence having at least about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any of the sequences disclosed in table B.

In some embodiments, the spheroids disclosed herein comprise 2, 3, 4, or 5 or more mutations in the genes identified in table B. If spheroids containing the specific mutations identified in table B are used within the system disclosed herein, the corresponding system can be used as an in vitro model of the corresponding disease states identified above.

In some embodiments, any composition, system or method as described in PCT/US2015/050061 can be used in embodiments of the disclosure.

In some embodiments, the method relates to a method of making a system, culture plate or device for culturing cells, the method comprising obtaining stem cells such as induced pluripotent stem cells, exposing the cells to one or more cell growth factors, differentiating the stem cells into neuronal cells, and seeding the cells onto a solid substrate comprising a first and/or second cavity or well. In some embodiments, the first and/or second cavities are U-shaped bottom holes, curved bottom holes, or flat bottom holes. In some embodiments, the method comprises seeding about 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 90, 100, 125, 150, 175, 200, 225, or 250 thousand cells. In some embodiments, the step of seeding the cells comprises seeding the one or more f-cells in a series of cavities or wells spaced within a solid substrate, and each cavity or well comprises a cell culture medium. In some embodiments, the step of seeding in the cavities or wells comprises seeding the cells in a pattern positioned within a solid substrate such that each well contains a spheroid of cells and each spheroid grows in suspension or hanging drops. In some embodiments, a method of making a system, culture plate, or device for culturing cells comprises allowing cells to be cultured undisturbed for a sufficient time to allow them to spontaneously form one or more spheroids.

The present disclosure also relates to methods of testing the toxicity of an agent by exposing the agent to one or more spheroids within a cavity or well within a solid substrate. In some embodiments, the method further comprises exposing the agent to the one or more spheroids for a time sufficient for the agent to be adsorbed by one or more cells of the one or more spheroids, and then measuring viability of the cells by recording, observing morphological changes, or a combination of both.

The disclosure also relates to methods of forming spheroids derived from cells derived from stem cells or cells from the nervous system of a subject. In some embodiments, a method of forming spheroids comprises: (i) differentiating the cell from the stem cell into one or more cell types that are one or a combination of neuronal cells, astrocytes, schwann cells, or any other cell disclosed herein, and then (ii) mixing the one or more cells for a time sufficient to form spheroids. In some embodiments, the method does not include the step of differentiating any cells after spheroids are formed. In some embodiments, the method does not expose the spheroids or any cells to one or more DRGs.

The following examples are intended as non-limiting examples of how embodiments disclosed in this application may be made and used. Any publications, patents, or patent applications disclosed in the examples or the subject matter of the specification are herein incorporated by reference in their entirety.

158页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:分泌IL-36的免疫应答细胞及其用途

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!

技术分类