ELECTROSPUN ELECTROACTIVE POLYMERS FOR REGENERATIVE MEDICINE APPLICATIONS

Due to the size and complexity of tissues such as the spinal cord and articular cartilage, specialized constructs incorporating cells as well as smart materials may be a promising strategy for achieving functional recovery. Aspects of the present invention describe the use of an electroactive, or piezoelectric, material that will act as a scaffold for stem cell induced tissue repair. Embodiments of the inventive material can also act alone as an electroactive scaffold for repairing tissues. The piezoelectric material of the present invention acts as a highly sensitive mechanoelectrical transducer that will generate charges in response to minute vibrational forces.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

The application claims the benefit of U.S. Provisional Patent Application Ser. No. 61/039, 214 filed Mar. 25, 2009, entitled “Electrospun Electroactive Polymer for Regenerative Medicine Applications”, the entire disclosure of which is incorporated herein by reference

FIELD OF THE INVENTION

The present invention relates to the use of a synthetic electroactive, or piezoelectric, biomaterial useful as an electroactive scaffold for repairing tissues. The scaffold may be used alone or in combination with cells as scaffold for tissue repair or regeneration.

BACKGROUND OF THE INVENTION

Tissue engineering is the application of principles and methods of engineering and life sciences toward a fundamental understanding and development of biological substitutes to restore, maintain and improve human tissue functions.

Orthopedic management of lesions to articular cartilage remains a persistent problem for the orthopedist and patient because articular cartilage has a limited intrinsic ability to heal. This has prompted the development of numerous procedures to treat these lesions and to halt or slow the progression to diffuse arthritic changes.

Tissue engineering may eliminate many of the problems associated with current surgical options. Current tissue engineering methods are aimed at filling the cartilage defects with cells with or without scaffolds to promote cartilage regeneration. Implantation of scaffolds alone leads to a poor quality reparative tissue. Chondrocytes implanted either alone or in combination with a scaffold have failed to restore a normal articular surface, and the hyaline cartilage formed early on in response to chondrocyte-containing scaffolds seems to deteriorate with time.

Damage to the spinal cord may result in autonomic dysfunction, a loss of sensation, or a loss of mobility. Such spinal cord injury (SCI) frequently is caused by trauma, tumors; ischemia, developmental disorders, neurodegenerative diseases, demyelinative diseases, transverse myelitis, vascular malformations, or other causes. The consequences of SCI depend on the specific nature of the injury and its location along the spinal cord. In addition because SCI is a dynamic process, the full extent of injury may not be apparent initially in all acute cord syndromes. Incomplete cord lesions may evolve into more complete lesions; more commonly, the injury level rises one or two spinal levels during the hours to days after the initial event. A complex cascade of pathophysiologic events accounts for this clinical deterioration.

The psychological and social impact of SCIs often is devastating. Some of the general disabling conditions associated with SCI are permanent paralysis of the limbs, chronic pain, muscular atrophy, loss of voluntary control over bladder and bowel, sexual dysfunction, and infertility.

Recent advances in neuroscience have drawn considerable attention to research into SCI and have made significantly better treatment and rehabilitation options available. Functional electrical stimulation (FES), for example, has shown the potential to enhance nerve regeneration and allow significant improvements in restoring and improving functional capacity after SCI. However, not all patients with spinal cord injury qualify for FES (a complete lesion of the spinal cord must be established); the patient must be in a neurologically stable condition; and the peripheral nerves must be intact to respond to exogenous electrical stimulations. Therefore, tissue engineering methods that could successfully restore, maintain, and improve the damage caused by spinal cord injury would eliminate many of the problems associated with current treatment options.

The development of improved tissue regeneration strategies will require a multi-disciplinary approach combining several technologies. Due to the size and complexity of tissues such as the spinal cord and articular cartilage, specialized constructs incorporating cells as well as smart materials may be a promising strategy for achieving functional recovery.

The use of stem cells for tissue engineering therapies is at the forefront of scientific investigation. Stem cells have the ability to differentiate into various cells types and thus promote the regeneration or repair of a diseased or damaged tissue of interest.

For example, mesenchymal stem cells (MSC) are multipotent cells that are capable of differentiating along several lineage pathways. In the body, adult stem cells often are localized to specific chemically and topologically complex microenvironments, or so-called “niches”. Increasing experimental evidence supports the notion that stem cells can adjust their properties according to their surroundings and select specific lineages according to cues they receive from their niche (Xie L, Spradling, A C, “A Niche Maintaining Germ Line Stem Cells In Drosophila Ovary,” Science 290:328 (2000); Fuchs E, Segre J, “Stem Cells: A New Lease On Life,” Cell 100: 143-155 (2000), Watt F M, Hogan B L M, “Out Of Eden: Stem Cells And Their Niches,” Science 287:1427 (2000)). It follows that in order for an MSC therapy to be successful in the repair of a specific tissue type, the microenvironment of the cells should be designed to relay the appropriate chemical and physical signals to them.

A viable approach may be to mimic characteristics of the microenvironment during cartilage and neurite development may be a viable approach. During cartilage development, for example, one of the earliest events is pre-cartilage mesenchymal cell aggregation and condensation resulting from cell-cell interaction, which is mediated by cell-cell and cell-matrix adhesion (fibronectin, proteoglycans, and collagens). (DeLise A. M., Fischer L, Tuan R S, “Cellular Interactions And Signaling In Cartilage Development. Osteoarthritis and Cartilage 8: 309-334 (2000)). Type I collagen, the predominant matrix protein present in the early stages of development, is later transformed to Type II collagen by increased cell synthesis during differentiation. (Safronova E E, Borisova N V, Mezentseva S V, Krasnopol'skaya K D, “Characteristics Of The Macromolecular Components Of The Extracellular Matrix In Human Hyaline Cartilage At Different Stages Of Ontogenesis.” Biomedical Science 2: 162-168 (1991)). Multiple growth factors and morphogens are also present contributing to the regulation of the differentiation process.

A few studies have demonstrated the use of MSCs for cartilage repair through intra-articular injection and have shown promise. (Murphy M, Fink D J, Hunziker E B, Barry F P, “Stem Cell Therapy In A Caprine Model Of Osteoarthritis,” Arthritis Rheumatism 48: 3464-3474 (2003); Ponticello M S, Schinagel R M, Kadiyala, S, Barry F P, “Gelatin-Based Resorbable Spone As A Carrier Matrix For Human Mesenchymal Stem Cells In Cartilage Regeneration Therapy,” J Biomed Materials Res 52: 246-255 (2000)). The MSCs are injected at a high cell density either alone (in saline) or in combination with a gelatinous/hydrogel matrix in order to promote cell-cell aggregation. However, the use of MSCs in combination with biomaterials of varying architectures that may closely mimic the physical architecture of the native extracellular matrix during development to direct chondrogenic differentiation has yet to be investigated.

Schwann cell-laden grafts and nerve conduits have shown promise for repairing nervous tissue (Brook G A, Lawrence J M, Raisman G. Columns of Schwann cells extruded into the CNS induce in-growth of astrocytes to form organized new glial pathways. Glia 2001; 33:118-130) and optic nerves (Negishi H. Optic nerve regeneration within artificial Schwann cell graft in the adult rat. Brain Research Bulletin 2001; 55:409-419), as have injections of adult stem cells (Desawa M. Specific induction of neuronal cells from bone marrow stromal cells and application for autologous transplantation. Journal of Clinical Investigation 2004; 113:1701-1710), but the size and complexity of the spinal cord warrants the development of specialized constructs.

The present invention addresses these problems. From a biological viewpoint, almost all human tissues and organs are characterized by well-organized hierarchical fibrous structures through the assembly of nanoscale elements. It is believed that converting biopolymers into fibers and networks that mimic native structures will ultimately enhance the utility of these materials as scaffolds. Nanoscale fibrous scaffolds may provide an optimal template for stem cell growth, differentiation, and host integration.

It is known that cells will attach to synthetic polymer scaffolds leading to the formation of tissue. (Sachlos, E. and Czernuszka, Eur. Cells & Materials 5: 29-40 (2003)). Using fetal bovine chondrocytes maintained in vitro, Li et al. have shown that scaffolds constructed from electrospun three-dimensional nanofibrous poly(∈-capro-lactone) act as a biologically preferred scaffold/substrate for proliferation and maintenance of the chondrocyte phenotype. (Wan-Ju Li, et al., J. Biomed. Mater. Res. 67A: 1105-1114 (2003)).

Because electric polarization can influence cell growth and behavior, e.g., growth of different cell types (Yang, X. L. et al., J. Mater. Sci.:Mater. Med. 17: 767 (2006)), enhancement of nerve regeneration (Kotwal, A. et al., Biomaterials, 22: 1055 (2001)), and cell adhesion and morphology (Kapurr., et al., J. Biomed Mater. Res. 27: 133 (1996)), it was hypothesized that a three-dimensional, electrically charged polymer scaffold would be a promising approach for a number of tissue engineering applications (e.g., nerve, bone, cartilage regeneration).

SUMMARY OF THE INVENTION

In one aspect, the present invention provides an electroactive structure for growing isolated differentiable cells that comprises a three dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material wherein the matrix of fibers is seeded with the isolated differentiable cells and forms a supporting scaffold for growing the isolated differentiable cells, and wherein the matrix of fibers stimulates differentiation of the isolated differentiable cells into a mature cell phenotype on the structure. In one embodiment, the biocompatible synthetic piezoelectric polymeric material is poly(vinylidene fluoride trifluoroethylene) copolymer. In another embodiment, the matrix fibers is a non-woven mesh of nanofibers. In another embodiment, the three dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material is formed by electrospinning. In another embodiment, the isolated differentiable cells are multipotent human mesenchymal cells. In another embodiment, the human mesenchymal stem cells are isolated from human bone marrow. In another embodiment, the isolated differentiable human mesenchymal cells have a CD44+CD34− CD45− phenotype. In another embodiment, the mature cell phenotype comprises a chondrogenic cell phenotype. In another embodiment, the chonodrogenic cell phenotype on the structure produces at least one glycosaminoglycan. In another embodiment, the mature cell phenotype comprises a neuronal cell phenotype.

In another aspect, the present invention provides a composition for use in tissue engineering that comprises (a) isolated differentiable cells, and (b) a supporting electroactive scaffold for growing the isolated differentiable cells, the supporting scaffold comprising a three dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material, wherein the matrix of fibers is seeded with the isolated differentiable cells and forms a supporting scaffold for growing the isolated differentiable cells, and wherein the matrix of fibers stimulates differentiation of the isolated differentiable cells into a mature cell phenotype on the structure. In one embodiment, the biocompatible synthetic piezoelectric polymeric material is poly(vinylidene fluoride trifluoroethylene) copolymer. In another embodiment, the three dimensional matrix of fibers is a non-woven mesh of nanofibers. In another embodiment, the three dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material is formed by electrospinning. In another embodiment, the isolated differentiable cells are multipotent human mesenchymal cells. In another embodiment, the human mesenchymal stem cells are isolated from human bone marrow. In another embodiment, the isolated differentiable human mesenchymal cells have a CD44+ CD34− CD45− phenotype. In another embodiment, the mature cell phenotype comprises a chondrogenic cell phenotype. In another embodiment, the chonodrogenic cell phenotype on the structure produces at least one glycosaminoglycan. In another embodiment, the mature cell phenotype comprises a neuronal cell phenotype.

In another aspect, the present invention provides a method of making an implantable electroactive scaffold, the method comprising the steps: (a) isolating differentiable human cells from a human donor; (b) preparing a three-dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material to form a cell scaffold; (c) seeding the cell scaffold with the isolated differentiable human cells; and (d) growing the differentiable human cells on the cell scaffold so that the differentiable human cells differentiate into a mature cell phenotype on the scaffold. In one embodiment, the differentiable human cells in step (a) are multipotent human mesenchymal cells. In another embodiment, step (a) further comprises the step of obtaining the differentiable human mesenchymal cells from bone marrow. In another embodiment, the differentiable human mesenchymal cells in step (a) have a CD44+ CD34− CD45− phenotype. In another embodiment, the biocompatible synthetic piezoelectric polymeric material in step (b) is poly(vinylidene fluoride trifluoroethylene) copolymer. In another embodiment, the three dimensional matrix of fibers in step (b) is a non-woven mesh of nanofibers. In another embodiment, the three dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material is formed by electrospinning. In another embodiment, the mature cell phenotype in step (d) comprises a chondrogenic cell phenotype. In another embodiment, the chonodrogenic cell phenotype in step (d) produces at least one glycosaminoglycan. In another embodiment, the mature cell phenotype in step (d) comprises a neuronal cell phenotype.

BRIEF DESCRIPTION OF THE FIGURES

FIG. 1 shows a scanning electron micrograph (1600×) of PVDF-TrFE nanofiber mesh, average fiber diameter 970±480 nm.

FIG. 2 shows TSC analysis of PVDF-TrFE powder and electrospun PVDF-TrFE mat (externally applied field: 100V).

FIG. 3 shows data resulting from X-ray diffraction analysis of PVDF-TrFE (A) electrospun nanofibers; (B) raw/unprocessed PVDF-TrFE powder.

FIG. 4 shows FTIR analysis of PVDF-TrFE powder (gray line) and electrospun PVDF-TrFE mat (black line).

FIG. 5 shows confocal images of PC-12 cells cultured on (a) PVDF-TrFE meshes in induction media; (b) in standard growth media; and (c) PC-12 cells on PLLA meshes in induction media (60× objective D); and metabolic activity of PC-12 cells at 10 days in culture *P<0.05 for PVDF-TrFE versus collagen.

FIG. 6 shows chondroadherin and focal adhesion kinase (FAK) gene expression in human mesenchymal stem cells (hMSCs) cultured for 28 days on PLLA and PVDF-TrFE scaffolds. Cell pellet cultures serve as controls.

FIG. 7 shows glycosaminoglycan production (sGAG) for human mesenchymal stem cells cultured in chondrogenic induction media on PLLA and PVDF-TrFE meshes at 28 days. Pellet cultures served as a positive control. *p<0.05.

FIG. 8 shows (a) viability and growth of human skin fibroblasts on electrospun PVDF-TrFE fiber scaffold compared to tissue culture polystyrene (TCPS); (b) SEM image of electrospun PVDF-TrFE fibers.

FIG. 9A/B shows confocal scanning laser microscopy images of human skin fibroblasts attached to PVDF-TrFE fibers after 1 day and after 7 days of cell culture.

DETAILED DESCRIPTION OF THE INVENTION

The present invention provides an electroactive, or piezoelectric, biomaterial as an electroactive scaffold for repairing tissues. The piezoelectric material acts as a highly sensitive mechanoelectrical transducer that will generate charges in response to minute vibrational forces. It further provides piezoelectric compositions comprising a three-dimensional matrix of nanofibers of piezoelectric synthetic or biological polymers used as an implantable scaffolding for delivery of differentiable human mesenchymal cells in tissue engineering applications and methods of preparing them. The piezoelectric scaffolds, which demonstrate electrical activity in response to minute mechanical deformation, allow the achievement of local electric fields characteristic of the natural extracellular matrix observed during development and regeneration.

As used herein, the term “stem cells” refers to undifferentiated cells having high proliferative potential with the ability to self-renew that can migrate to areas of injury and can generate daughter cells that can undergo terminal differentiation into more than one distinct cell phenotype. These cells have the ability to differentiate into various cells types and thus promote the regeneration or repair of a diseased or damaged tissue of interest.

Specialized protein receptors that have the capability of selectively binding or adhering to other signaling molecules coat the surface of every cell in the body. Cells use these receptors and the molecules that bind to them as a way of communicating with other cells and to carry out their proper functions in the body. Each cell type has a certain combination of receptors, or markers, on their surface that makes them distinguishable from other kinds of cells.

Stem cell markers are given short-hand names based on the molecules that bind to the corresponding stem cell surface receptors. In many cases, a combination of multiple markers is used to identify a particular stem cell type. Researchers often identify stem cells in shorthand by a combination of marker names reflecting the presence (+) or absence (−) of them. For example, a special type of hematopoietic stem cell from blood and bone marrow called “side population” or “SP” is described as (CD34−/low, c-Kit+, Sca-1+).

The following markers commonly are used by skilled artisans to identify stem cells and to characterize differentiated cell types (http://stemcells.nih.gov/info/scireport/appendixE.asp, (visited Dec. 28, 2007)):

Marker Cell Type Significance CD34 Hematopoietic stem cell a highly glycosylated type I transmembrane protein (HSC), muscle satellite, expressed on 1-4% of bone marrow cells, endothelial progenitor CD38 immature T and B cells a type II transmembrane protein found on immature T and B cells but not most mature peripheral lymphocytes CD41 platelets and megakaryocytes; the integrin αIIb subunit CD45 WBC progenitor the leukocyte common antigen found on all cells of hematopoietic origin CD105 Endothelial cells a disulfide-linked homodimer found on endothelial cells but absent from most T and B cells; CD133 primitive hematopoietic a pentaspan transmembrane glycoprotein progenitors CD3 T cells a member of the T cell receptor complex; CD4, CD8 Mature T cells Cell-surface protein markers specific for mature T lymphocyte (WBC subtype) CD7 Early T cells An early T cell lineage marker, CD10 early T and B a type II membrane metalloprotease cell precursors; CD13 granulocytes, monocytes a type II membrane metalloprotease, and their precursors CD14 myelomonocytic lineage a GPI-linked protein expressed mainly on myelomonocytic lineage cells; CD19 B cells a component of the B cell antigen signaling complex; CD33 Myelomonocytic precursors a sialic acid binding protein absent from pluripotent stem cells that appears on myelomonocytic precursors after CD34; CD38 WBC lineages A Cell-surface molecule that identifies WBC lineages. Selection of CD34+/CD38− cells allows for purification of HSC populations CD44 Mesenchymal A type of cell-adhesion molecule used to identify specific types of mesenchymal cells CD56 NK cells an isoform of the neural adhesion molecule found exclusively on natural killer (NK) cells; CD127 lymphocytes the high affinity interleukin 7 receptor expressed on lymphocytes; CD138 Immature B cells an extracellular matrix receptor found on immature B cells and plasma cells and plasma cells; Glycophorin A RBCs, embryoid a sialoprotein present on human RBCs and embryoid precursors precursors; CD90 prothymocytes a GPI-cell anchored molecule found on prothymocyte cells in humans. c-kit HSC, MSC Cell-surface receptor on BM cell types that identifies HSC and MSC; binding by fetal calf serum (FCS) enhances proliferation of ES cells, HSCs, MSCs, and hematopoietic progenitor cells Fetal liver endothelial Cell-surface receptor protein that identifies endothelial cell kinase-1 (Flk-1) progenitor; marker of cell-cell contacts

The term “cellular differentiation” as used herein refers to the process by which cells acquire a cell type.

The term “chondrocytes” as used herein refers to cells found in cartilage that produce and maintain the cartilaginous matrix. From least to terminally differentiated, the chondrocytic lineage is (i) Colony-forming unit-fibroblast (CFU-F); (ii) mesenchymal stem cell/marrow stromal cell (MSC); (iii) chondrocyte. The term “chondrogenesis” refers to the formation of new cartilage from cartilage forming or chondrocompetent cells.

The term “ΔHf” refers to Heat of Fusion.

The term “nanoscale fiber” generally refers to fibers whose diameter ranges from about 1 to about 1000 nanometers.

The term “progenitor cell” as used herein refers to an immature cell in the bone marrow that can be isolated by growing suspensions of marrow cells in culture dishes with added growth factors. Progenitor cells are referred to as colony-forming units (CFU) or colony-forming cells (CFC). The specific lineage of a progenitor cell is indicated by a suffix, such as, but not limited to, CFU-F (fibroblastic).

As used herein, the terms “osteoprogenitor cells,” “mesenchymal cells,” “mesenchymal stem cells (MSC),” or “marrow stromal cells” are used interchangeably to refer to multipotent stem cells that differentiate from CFU-F cells capable of differentiating along several lineage pathways into osteoblasts, chondrocytes, myocytes and adipocytes. When referring to bone or cartilage, MSCs commonly are known as osteochondrogenic, osteogenic, chondrogenic, or osteoprogenitor cells, since a single MSC has shown the ability to differentiate into chondrocytes or osteoblasts, depending on the medium.

The term “piezoelectric material” as used herein refers to any material that exhibits piezoelectric properties or effects. The terms “piezoelectric properties” or “piezoelectric effects” are used interchangeably to refer to the property exhibited by piezoelectric materials of becoming electrically polarized when mechanically strained and of becoming mechanically strained when an electric field is applied.

The term PLLA as used herein refers to biodegradable aliphatic polyester homopolymer poly L-lactic acid (PLLA) obtained from Alkermes, Inc.

The present invention described hereinabove has both human and veterinary utility. The term “subject” as used herein therefore includes animals of mammalian origin, including humans.

The term “Tm” refers to melting point.

The terms “Transforming Growth Factor”, “tumor growth factor” or “TGF” are used interchangeably to describe two classes of polypeptide growth factors, TGFα and TGFβ. TGFα, which is upregulated in some human cancers, is produced in macrophages, brain cells, and keratinocytes, and induces epithelial development. TGFβ exists in three known subtypes in humans, TGFβ1, TGFβ2, and TGFβ3 that are upregulated in some human cancers, and play crucial roles in tissue regeneration, cell differentiation, embryonic development, and regulation of the immune system. TGFβ receptors are single pass serine/threonine kinase receptors.

Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges which may independently be included in the smaller ranges is also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either both of those included limits are also included in the invention.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and described the methods and/or materials in connection with which the publications are cited.

It must be noted that as used herein and in the appended claims, the singular forms “a”, “and”, and “the” include plural references unless the context clearly dictates otherwise. All technical and scientific terms used herein have the same meaning.

The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates which may need to be independently confirmed.

EXAMPLES

The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are neither intended to limit the scope of what the inventors regard as their invention nor they intended to represent that the experiments below are all or the only experiments performed.

Example 1 Fabrication Of Piezoelectric Tissue Engineering Scaffolds

The present invention makes use of fibers formed from a permanently piezoelectric poly(vinylidene fluoride trifluoroethylene) (PVDF-TrFE) copolymer. The PVDF-TrFE copolymer was fabricated into a nanofibrous scaffold using an electrospinning technique.

The electrospinning process is affected by varying the electric potential, flow rate, solution concentration, capillary-collector distance, diameter of the needle, and ambient parameters like temperature. PVDF-TrFE and PLLA were electrospun into fibers according to commonly used optimization procedures whereby porosity, surface area, fineness and uniformity, diameter of fibers, and the pattern thickness of the sheet could be manipulated. See, e.g., Greiner, A. et al Angew Chem. Int. Ed. Engl. 46: 5670 (2007).

The electrospinning setup used herein is described in U.S. patent application Ser. No. 11/291,701, which is incorporated herein by reference. It is comprised a syringe pump containing a 13-20 gauge needle mounted on a robotic arm in order to control the splaying of fibers on the collector. An electrically grounded stainless steel plate of dimensions 15×30 cm is used as the collector.

PVDF-TrFE copolymer (65/35) purchased from Solvay Solexis, Inc. (NJ, USA) was dissolved in Methylethylketone (MEK). For the successful formation of fibers, a 15% w/v solution concentration of the polymer in MEK was used. The syringe pump was filled with the polymer solution, and a constant flow rate of 0.035 ml/min was maintained using the syringe pump. The positive output lead of a high voltage power supply (Gamma High Voltage, Inc.) was attached to a 20 gauge needle, and a 25 kvolt voltage was applied to the solution. The collector-to-needle distance was 18.5 cm. The electrospinning process was performed in about 12% to about 13% humidity at 25 degrees C. When the charge of the polymer at increasing voltage exceeded the surface tension at the tip of the needle, the polymer splayed randomly as fibers. These were collected as nonwoven mats on the grounded plate.

Example 2 Characterization of the Electrospun PVDF-TrFE Fibers

Structure and piezoelectric activity were examined by differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), thermally stimulated current (TSC) spectroscopy, X-ray diffraction (XRD) and fourier transform infrared spectroscopy (FTIR). Comparisons were made between PVDF-TrFE polymer powder and electrospun PVDF-TrFE fibers.

The fiber diameter of electrospun PVDF-TrFE fibers was characterized using Scanning Electron Microscopy (SEM) according to established methods and compared to poly L-lactic acid (PLLA) meshes. FIG. 1 shows that the resulting fibrous meshes had an average fiber diameter of 970±480 nm, with uniform fiber morphologies having no beading, as characterized by scanning electron microscopy. The fiber mats were free of droplets.

Thermally stimulated current (TSC) spectroscopy is widely used to understand dielectric relaxation in complex solid systems. TSC is based on the ability of polar molecules to be moved by an electric static field. At a temperature Tp, an electric field is applied during a time tp long enough to let the dipoles orient themselves. This configuration is fixed by a rapid decrease in temperature to reach a temperature T0. At T0, the sample is short-circuited during a time t0 to remove the space charges and to equilibrate the temperature. The progressive and sequential release of the entities oriented previously can be observed during a linear rise in temperature. The depolarization current is then recorded as a function of the temperature.

TSC measurements confirmed that the electrospun PVDF-TrFE fiber scaffolds have internal charges comparable to the original piezoelectric polymer powder. The electrospun and powder forms were heated from −60° C. to 140° C. (7 C per min) and were subjected to an externally applied field of 100 V. FIG. 2 shows the data resulting from TSC analysis of the electrospun PVDF-TrFE mat and the non-processed powder form. It shows that for both the powder and electrospun forms, there was polarization due to the applied electric field followed by a spontaneous relaxation.

Thermal Gravimetric Analysis (TGA) was performed to detect any remaining solvent in the nanofiber mat using a Thermal Gravimetric Analyzer (TA Instrument model Q50). The analyzer measures weight changes in materials with regard to temperature, which allows for the effective quantitative analysis of thermal reactions that are accompanied by mass changes resulting from dehydration, decomposition and oxidation of a sample.

The nanofiber mat was subjected to vacuum prior to the analysis. A sample of the test material was placed into a high alumina cup supported on, or suspended from, an analytical balance located outside the furnace chamber. The balance was zeroed, and the sample cup heated according to a predetermined thermal cycle. The balance sends the weight signal to the computer for storage, along with the sample temperature and the elapsed time. The TGA curve plots the TGA signal, converted to percent weight change, on the Y-axis against the reference material temperature on the X-axis.

The results showed that fibrous meshes with vacuum treatment had a 0.5% solvent content as demonstrated by a loss of 0.5 weight percent as compared to the unprocessed/raw polymer.

Results obtained by DSC, XRD and FTIR showed that the electrospinning process did not alter significantly the polymer structure compared to the original piezoelectric polymer powder.

Differential scanning calorimetry (DSC) is used to study the thermal behavior of polymers. In this technique, separate chambers for the sample and reference are heated equally. Transformations taking place in the sample are detected by the instrument, which compensates by changing the heat input so that there is a zero temperature difference between the reference and sample. The amount of electrical energy supplied to the heating elements is then proportional to the heat released by the sample. Thermal analysis was performed with a TA Model Q100 Differential Scanning Calorimeter.

Fourier-Transform Infrared Spectroscopy (FTIR) is used to observe vibrational changes in chemical bonds. Here, infrared radiation in the range from 4000 to 600 cm-1, the mid-infrared region, was used. The presence and intensity of specific vibrational frequencies allows for determination of functional groups in organic molecules. The class of material (proteinaceous, cellulosic, and so forth) then can be identified from these functional groups.

A micro x-ray diffractometer capable of focusing a collimated x-ray beam (20 to 800 micron diameter range) onto areas of interest within the sample was used to generate an x-ray diffraction (XRD) pattern characteristic for the crystalline phases contained within the sample. X-rays diffracted by the sample strike a detector and are converted to an electronic signal that is then further processed by software. Search-match software then was used to match the unknown diffraction pattern to a database of diffraction patterns collected from reference compounds.

The degree of crystallinity was determined, and the piezoelectric crystal form of the copolymer present in the electrospun PVDF-TrFE mats was confirmed, by DSC. Comparisons of PVDF-TrFE mats with the piezoelectric unprocessed powder and solvent-cast film as well as with nonpiezoelectric-unpoled PVDF pellets were made.

TABLE 1 Comparison of DSC data with literature values PVDF-TrFE PVDF-TrFE PVDF-TrFE PVDF (65/35) (65/35) (65/35) Physical form Pellet Powder Solvent-cast film Electrospun fiber Tm(C) 171 (161*) 107 (1 peak) 115 (1 peak) 115 (1 peak) 147 (154.55**) (2 peak) 147 (2 peak) 149 (2 peak) ΔHf(J/g)  45 (50*)  13 (1 peak)  13 (1 peak)  15 (1 peak)  23 (30**) (2 peak)  34 (2 peak)  28 (2 peak) *Zhao, Z. et al., J. Appl. Polym. Sci. 97: 466-74 (2005); **Data provided by supplier (Solvay Solexis, Inc.)

Table 1, which compares the experimental DSC data with literature values for test polymers (in parentheses), shows that low and high temperature peaks were observed in the PVDF-TrFE polymer during the first and second heating cycle. The differences in the first heating cycle between the test polymers were not detectable in the second heating cycle, which suggests that there is no chemical degradation or changes in the chemical structure due to the fabrication process. The melting points and heats of fusion for PVDF-TrFE materials are distinct from values obtained for the unpoled PVDF pellet, indicating that the piezoelectric beta-phase crystal form is present in the electrospun mat.

Moreover, the electrospun electroactive PVDF-TrFE fibers of the present invention do not require poling to show a piezoelectric effect. The term “poling” as used herein refers to the adjustment of the polarity of a substance. For example, electric dipoles may be aligned (meaning arranged, positioned or synchronized in a manner that allows for proper or optimal functioning) by utilizing an electric field. The term “polarity” refers to the property, state or condition of having or manifesting two opposite or opposing charges within the same body.

FIG. 3 shows X-ray diffractograms of (A) electrospun; and (B) non-processed powder form of PVDF-TrFE. FIG. 4 shows FTIR analysis of PVDF-TrFE powder (gray line) and electrospun PVDF-TrFE mat (black line). Together, XRD and FTIR analysis confirmed the presence of the poled, piezoelectric phase (beta-crystal phase) in the electrospun PVDF-TrFE.

FIG. 4 shows that unprocessed and electrospun mats have similar FTIR spectra.

Example 3 PVDF-TrFE Fiber Mats Support Stem Cells

Three studies were conducted to establish that the PVDF-TrFE fiber mesh can be used as a scaffold to support stem cells or other cell types

Materials and Methods

1. Cells

(a) Cell line model for neuronal differentiation. When treated with nerve growth factor (NGF), PC12 cells, a cell line derived from a pheochromocytoma of the rat adrenal medulla, stop dividing, grow long neurites, and undergo terminal differentiation, which makes this cell line a useful model system for neuronal differentiation.

PC12 cells (ATCC number CRL-1721) were seeded at 3×103 cells per cm2 culture dish and maintained in ATCC formulated F-12K medium containing 1.5% fetal bovine serum and 15% horse serum. Cultures were maintained at 37 C, 95% air, 5% CO2 atmosphere. For induction of the neuronal phenotype, 50 ng/ml NGF (Chemicon) was added to the medium at the start of the culture and maintained throughout the duration of the culture. The term “induction media” refers to the medium containing NGF.

(b) Fibroblasts. Normal human skin fibroblasts (ATCC number SCRC-1041) were seeded at 5×103 cells per cm2 culture dish and maintained in Dulbecco's modified Eagle's medium containing 15% fetal bovine serum.

(c) Mesenchymal stem cells. Human mesenchymal stem cells (hMSCs) were prepared as described in Livingston, et al., J. Materials Science: Materials in Med. 14: 211-218 (2003) and in U.S. Pat. No. 5,486,359, which are incorporated herein by reference. In brief, bone marrow aspirates of 30-50 mL were obtained from healthy human donors. Marrow samples were washed with saline and centrifuged over a density cushion of ficoll. The interface layer was removed, washed, and the cells counted. Nucleated cells recovered from the density separation were washed and plated in tissue culture flasks in Dulbecco's Modified Eagle's Medium (DMEM) containing 10% fetal bovine serum (“FBS”, HyClone Laboratories, Inc.). Non-adherent cells were washed from the culture during biweekly feedings. Colony formation was monitored for a 14-17 day period. MSC's were passaged when the tissue culture flasks were near confluent. At the end of the first passage, MSCs were enzymatically removed from the culture flask using trypsin-EDTA and replated at a lower density for further expansion. At the end of the second passage, MSC's were either seeded onto scaffolds or cryopreserved until future use.

The hMSC cells were identified as multipotent stem cells based on surface marker characterization, which distinguishes the stem cells from other cell types in the bone marrow, for example white blood cells. Cells expressing CD44 (CD44+) and the absence of CD45 (CD45−) and CD34 (CD34−) surface antigens were verified by fluorescence-activated-cell-sorter.

Chondrogenic differentiation of hMSCs was performed according to published procedures. See Barry, F. et al., Exp. Cell Res. 268, 189 (2001), which is incorporated herein by reference. 2×105 cells were seeded on PVDF-TrFE scaffolds in 24-well plates using three different culture media: (i) the chondrogenic culture media containing TGFb3, or induction media, (CCM+), consisted of 1 mM sodium pyruvate (Sigma), 0.1 mM ascorbic acid-2-phosphate (Wako), 1×10-7 M dexamethasone (Sigma), 1% ITS 1 (Collaborative Biomedical Products), and 10 ng/ml recombinant human TGF-β3 (Oncogene Sciences) dissolved in Dulbecco's Modified Eagle's Medium containing 4-5 g/L glucose (DMEM-LG), (ii) chondrogenic culture media (CCM) without TGFβ3 (CCM−); (iii) mesenchymal stem cell growth media (MSCGM), the standard growth media for hMSCs, consisting of DMEM-LG with 10% fetal bovine serum and 1% antibiotic-antimycotic. Cells were harvested after 1, 14, and 28 days of culture.

Cell pellet cultures served as controls for these experiments. A single cell pellet was produced by centrifuging 2.5×105 cells in a 15 mL centrifuge tube and culturing the pelleted cells in the tube.

Cell viability: Metabolic activity and cell growth were measured using the XTT kit (Biotium, USA). XTT is a tetrazolium derivative that measures cell viability based on the activity of mitochondria enzymes in live cells that reduce XTT and are inactivated shortly after cell death. XTT is reduced to a highly water-soluble orange colored product, the amount of which is proportional to the number of living cells in the sample, and can be quantified by measuring absorbance at wavelength of 475 nm.

Cells were plated onto scaffolds in 96-well tissue culture plates at 10,000 cells per well for up to 7 days. Reagents were added such that the final volume of tissue culture medium (containing 10% FBS) in each well was 0.1 mL. For one 96-well plate, 25 μL Activation Reagent was mixed with 5 mL XTT Solution to derive activated XTT solution. 25 μL or 50 μL of the activated XTT solution was added to each well and the plate incubated in an incubator for 4 hours. The plate was shaken gently to evenly distribute the dye in the wells. The absorbance of the samples was measured spectrophotometrically at a wavelength of 450-500 nm. Reference absorbance is measured at a wavelength of 630-690 nm.

Real time reverse transcriptase-polymerase chain reaction (RT-PCR): RNA was isolated using a Qiagen Mini kit (Qiagen). Samples were lysed and then homogenized using QiaShredder columns (Qiagen). Ethanol was added to the lysate and the lysate was loaded onto the RNeasy silica-gel membrane. Pure, concentrated RNA then was eluted from the membrane in water.

Relative gene expression analysis (QuantiTect SYBR Green RT-PCR kit, Qiagen) for chondrogenic markers (chondroadherin, type II collagen), and focal adhesion kinase (FAK) was performed using the MX4000 detection system (Stratagene). Ribosomal protein, large, PO (“RPLPO”) was used as housekeeping gene.

Qiagen PCR kit: 2× QuantiTect SYBR Green RT-PCR Master Mix (stored at −20° C.), template RNA, primers, and RNase-free water were thawed, mixed individually and placed on ice. A reaction components master mix was prepared as follows:

Component Volume/reaction Final concentration 2x QuantiTect SYBR Green 12.5 μl 1x RT-PCR Master Mix Primer A Variable 0.5-2.0 μM Primer B Variable 0.5-2.0 μM QuantiTect RT Mix 0.25 μl 0.25 μl RNAse-free water Variable Optional: Uracil-N-glycolase, Variable 1-2 units/reaction heat labile Template RNA Variable ≦500 ng/reaction Total volume   25 μl

Where final reaction volumes other than 25 μl were used, the volumes of 2× Quanti-Tect SYBR Green RT-PCR Master Mix and Quanti Tect RT Mix used were adjusted so that the ratio between them remained constant.

The master mix was mixed thoroughly and appropriate volumes dispensed into PCR tubes. Template RNA (≦500 ng/reaction) was added to the individual PCR tubes and incubated on ice for less than 30 min. The MX4000 was programmed and data acquisition performed during the extension step. A melting curve analysis of the RT-PCR product(s) between 55 C and 95 C was performed to verify specificity and identify of the RT-PCR products.

A standard curve was generated using various RNA concentrations, which contain substantial levels of chondrogenic markers (chondroadherin, type II collagen) and focal adhesion kinase (FAK). Two optical channels, one for SYBR Green and one for a reference dye (ROX), were used to correct for volume and plate location differences. Each template was analyzed in triplicate. Stratagene reaction tubes (Cat. No. 41002) and caps (Cat. No. 410024) were used, and fluorscence data was collected for SYBR Green. A typical thermal profile used was the following:

50 C for 30 min (reverse transcriptase step)

95° C. for 15 min (to activate the DNA polymerase)

40 cycles of:

    • 94° C. for 15 sec
    • 55° C. for 30 sec
    • 72° C. for 30 sec (triplicate readings of fluorescence were taken during this phase of the cycle.)

A dissociation curve was generated after the amplification cycles were completed. For the amplification plots, fluorescence was analyzed as “dRn” to generate Ct values for all of the samples simultaneously. Gene expression levels were analyzed according to Mueller (Mueller, P. Y., Janoviak, H., Miserez, A. R., Dobbie, Z., Biotechniques 32, 1372-74 (2002)), which is incorporated herein by reference, and expressed as “mean normalized expression.”

Confocal fluorescence microscopy was used to obtain fluorescence images of cells cultured on fiber scaffolds. A fluorescent stain, which visualizes nuclear DNA (4′,6-diamidino-2-phenylindole, DAPI, Invitrogen, USA) and the actin cytoskeleton (Alexa Fluor 488 phalloidin; Invitrogen, USA) in fixed cells was used. Fluorescence images of cells cultured on fiber scaffolds were taken with a confocal fluorescence microscope (Clsi, Nikon, Japan).

Cell proliferation. Cell number over time was measured using the PicoGreen assay (Invitrogen).

sGAG synthesis: Absorbance at 656 nm was used to measure total sulfated proteoglycan content (“sGAG”) using the Blycan assay (Biodyne Science, UK).

Results

The results show that PDVF-TrFE fiber piezoelectric scaffolds are biocompatible and stimulate differentiation of hMSCs into chondrocytes, PC-12 neuronal cells into neurites; and stimulate attachment and growth of fibroblasts on the PVDF-TrFE scaffold as compared to growth of these cells under normal culture conditions.

FIGS. 5a-5c shows that at 10 days in culture, extensive neurite extension on PVDF-TrFE meshes was seen with or without media containing Nerve Growth Factor (NGF). Neurite extension of cells grown on electrospun poly-L-lactic acid [PLLA] (average fiber diameter of 1.0+/−0.4 μm) scaffolds appeared less extensive and only occurred in the presence of NGF. As shown in FIG. 5d, cell growth, as measured by metabolic activity using the XTT kit (Biotium, USA), was significantly lower on PVDF-TrFE meshes for both growth and induction media as compared to tissue culture polystyrene and PLLA scaffolds, suggesting that PVDF-TrFE downregulates proliferation and facilitates differentiation.

FIGS. 6 and 7 shows that for human mesenchymal stem cell chondrogenesis, glycosaminoglycan production by cells on PVDF-TrFE meshes/mats was significantly higher than for cells on PLLA or in pellet culture (positive control) in inductive media. It is known that transforming growth factor β (TGF-β) induces chondrogenesis in hMSCs and involves deposition of a cartilage-specific extracellular matrix. Barry, F. et al., Exp. Cell Res. 268, 189 (2001). Initial studies showed that chondrogenic markers and sGAG synthesis was significantly induced by CCM+ media. As shown in FIG. 6, the sGAG concentrations and chondroadherin/FAK gene expression was significantly higher on PVDF-TrFE as compared to PLLA scaffolds (p<0.01). However, no significant differences between PVDF-TrFE and PLLA scaffolds could be seen using CCM- and MSCGM media (chondroadherin, type II collagen, and FAK gene expression; sGAG synthesis).

Human skin fibroblasts (ATCC number SCRC-1041) were cultured on PVDF-TrFE fiber scaffolds over a 7-day period. Tissue culture polystyrene (TCPS) served as the control). FIG. 8 and FIG. 9 show that fibroblasts grew and were well-spread on PVDF-TrFE meshes. This was comparable to growth on tissue culture plastic (positive control).

Confocal fluorescence microscopy verified the attachment and proliferation of the cells on the PVDF-TrFE fiber scaffolds. FIG. 9 shows confocal scanning laser microscopy images of human skin fibroblasts attached to PVDF-TrFE fibers after 1 day and after 7 days of cell culture. The cell morphologies of one day cultures on the fiber scaffolds are distinctly different from those of 7-day cultures. On day 1, the cells are not fully spread out. When grown on the scaffolds for a longer time (7 days) cells exhibit a more elongated and spread-out morphology.

While the present invention has been described with reference to the specific embodiments thereof it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adopt a particular situation, material, composition of matter, process, process step or steps, to the objective spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims appended hereto.

Claims

1. An electroactive structure for growing isolated differentiable cells comprising a three dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material

wherein the matrix of fibers is seeded with the isolated differentiable cells and forms a supporting scaffold for growing the isolated differentiable cells, and
wherein the matrix of fibers stimulates differentiation of the isolated differentiable cells into a mature cell phenotype on the structure.

2. The electroactive structure according to claim 1, wherein the biocompatible synthetic piezoelectric polymeric material is poly(vinylidene fluoride trifluoroethylene) copolymer.

3. The electroactive structure according to claim 1, wherein the matrix fibers is a non-woven mesh of nanofibers.

4. The electroactive structure according to claim 1, wherein the three dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material is formed by electrospinning.

5. The electroactive structure according to claim 1, wherein the isolated differentiable cells are multipotent human mesenchymal cells.

6. The electroactive structure according to claim 5, wherein the human mesenchymal stem cells are isolated from human bone marrow.

7. The electroactive structure according to claim 5, wherein the isolated differentiable human mesenchymal cells have a CD44+ CD34− CD45− phenotype.

8. The electroactive structure according to claim 5, wherein the mature cell phenotype comprises a chondrogenic cell phenotype.

9. The electroactive structure according to claim 5, wherein the chonodrogenic cell phenotype on the structure produces at least one glycosaminoglycan.

10. The electroactive structure according to claim 1, wherein the mature cell phenotype comprises a neuronal cell phenotype.

11. A composition for use in tissue engineering, comprising

(a) Isolated differentiable cells, and
(b) a supporting electroactive scaffold for growing the isolated differentiable cells, the supporting scaffold comprising a three dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material
wherein the matrix of fibers is seeded with the isolated differentiable cells and forms a supporting scaffold for growing the isolated differentiable cells, and
wherein the matrix of fibers stimulates differentiation of the isolated differentiable cells into a mature cell phenotype on the structure.

12. The composition according to claim 11, wherein the biocompatible synthetic piezoelectric polymeric material is poly(vinylidene fluoride trifluoroethylene) copolymer.

13. The composition according to claim 11, wherein the three dimensional matrix of fibers is a non-woven mesh of nanofibers.

14. The composition according to claim 11, wherein the three dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material is formed by electrospinning.

15. The composition according to claim 11, wherein the isolated differentiable cells are multipotent human mesenchymal cells.

16. The composition according to claim 15, wherein the human mesenchymal stem cells are isolated from human bone marrow.

17. The composition according to claim 15, wherein the isolated differentiable human mesenchymal cells have a CD44+ CD34− CD45− phenotype.

18. The composition according to claim 15, wherein the mature cell phenotype comprises a chondrogenic cell phenotype.

19. The composition according to claim 15, wherein the chonodrogenic cell phenotype on the structure produces at least one glycosaminoglycan.

20. The composition according to claim 11, wherein the mature cell phenotype comprises a neuronal cell phenotype.

21. A method of making an implantable electroactive scaffold, the method comprising the steps:

(a) isolating differentiable human cells from a human donor;
(b) preparing a three-dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material to form a cell scaffold;
(c) seeding the cell scaffold with the isolated differentiable human cells; and
(d) growing the differentiable human cells on the cell scaffold so that the differentiable human cells differentiate into a mature cell phenotype on the scaffold.

22. The method according to claim 21, wherein the differentiable human cells in step (a) are multipotent human mesenchymal cells.

23. The method according to claim 22, wherein step (a) further comprises the step of obtaining the differentiable human mesenchymal cells from bone marrow.

24. The method according to claim 22, wherein the differentiable human mesenchymal cells in step (a) have a CD44+ CD34− CD45− phenotype.

25. The method according to claim 21, wherein the biocompatible synthetic piezoelectric polymeric material in step (b) is poly(vinylidene fluoride trifluoroethylene) copolymer.

26. The method according to claim 21, wherein the three dimensional matrix of fibers in step (b) is a non-woven mesh of nanofibers.

27. The method according to claim 21, wherein the three dimensional matrix of fibers formed of a biocompatible synthetic piezoelectric polymeric material is formed by electrospinning.

28. The method according to claim 21, wherein the mature cell phenotype in step (d) comprises a chondrogenic cell phenotype.

29. The method according to claim 28, wherein the chonodrogenic cell phenotype in step (d) produces at least one glycosaminoglycan.

30. The method according to claim 21, wherein the mature cell phenotype in step (d) comprises a neuronal cell phenotype.

Patent History
Publication number: 20090325296
Type: Application
Filed: Mar 25, 2009
Publication Date: Dec 31, 2009
Applicant: New Jersey Institute of Technology (Newark, NJ)
Inventors: Treena L. Arinzeh (West Orange, NJ), Norbert Weber (Piscataway, NJ), Michael Jaffe (Maplewood, NJ)
Application Number: 12/411,320
Classifications
Current U.S. Class: Support Is A Resin (435/396)
International Classification: C12N 5/08 (20060101);