WO2006125025A2 - Matrices extracellulaires de synthese regulant le comportement des cellules souches - Google Patents

Matrices extracellulaires de synthese regulant le comportement des cellules souches Download PDF

Info

Publication number
WO2006125025A2
WO2006125025A2 PCT/US2006/019130 US2006019130W WO2006125025A2 WO 2006125025 A2 WO2006125025 A2 WO 2006125025A2 US 2006019130 W US2006019130 W US 2006019130W WO 2006125025 A2 WO2006125025 A2 WO 2006125025A2
Authority
WO
WIPO (PCT)
Prior art keywords
collagen
composition
cells
matrix
solubilized
Prior art date
Application number
PCT/US2006/019130
Other languages
English (en)
Other versions
WO2006125025A3 (fr
Inventor
Sherry L. Voytik-Harbin
Beverly Z. Waisner
Original Assignee
Purdue Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Purdue Research Foundation filed Critical Purdue Research Foundation
Priority to CA2608422A priority Critical patent/CA2608422C/fr
Priority to AU2006247228A priority patent/AU2006247228B2/en
Priority to GB0724345A priority patent/GB2441268B/en
Publication of WO2006125025A2 publication Critical patent/WO2006125025A2/fr
Publication of WO2006125025A3 publication Critical patent/WO2006125025A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • A61L27/3834Cells able to produce different cell types, e.g. hematopoietic stem cells, mesenchymal stem cells, marrow stromal cells, embryonic stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/14Macromolecular materials
    • A61L27/22Polypeptides or derivatives thereof, e.g. degradation products
    • A61L27/24Collagen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3895Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells using specific culture conditions, e.g. stimulating differentiation of stem cells, pulsatile flow conditions

Definitions

  • This invention relates to the preparation of a collagen based matrix for culturing and differentiating stem cells and progenitor cells and the use of such compositions as tissue graft constructs.
  • ECM extracellular matrix
  • Tissue culture allows the study in vitro of animal cell behavior in an investigator-controlled physiochemical environment. Presumably cultured cells function best (i.e., proliferate and perform their natural in vivo functions) when cultured on substrates that closely mimic their natural environment. Currently, studies in vitro of cellular function are limited by the availability of cell growth substrates that present the appropriate physiological environment for proliferation and development of the cultured cells.
  • Complex scaffolds representing combinations of ECM components in a natural or processed form are commercially available, such as Human Extracellular Matrix (Becton Dickinson) and MATRIGEL ® .
  • modifying the conditions used to form a collagen based matrix from a solubilized collagen solution allows for the controlled alteration of the micro-structural and subsequent mechanical properties of the resulting ECM scaffold.
  • the micro-structural and mechanical properties of the ECM scaffold directly impact fundamental cell behavior including survival, adhesion, proliferation, migration and differentiation of cells cultured within the scaffold.
  • Basement membrane tissues and submucosal material harvested from warm blooded vertebrates have shown great promise as unique graft materials for inducing the repair of damaged or diseased tissues in vivo, and for supporting fundamental cell behavior (e.g., cell proliferation, growth, maturation, differentiation, migration, adhesion, gene expression, apoptosis and other cell behaviors) of cell populations in vitro.
  • Submucosal material can be extracted or fluidized to provide enriched extracts that can be utilized as additives for tissue culture media, or polymerized to form collagen based scaffolds, to promote in vitro cell growth and proliferation.
  • submucosal tissue undergoes remodeling and induces the growth of endogenous tissues upon implantation into a host.
  • Numerous studies have shown that submucosal tissue is capable of inducing host tissue proliferation, remodeling and regeneration of tissue structures following implantation in a number of in vivo environments, including the urinary tract, the body wall, tendons, ligaments, bone, cardiovascular tissues and other vascular tissues, and the central nervous system.
  • cellular infiltration and a rapid neovascularization are observed and the submucosa materials are remodeled into host replacement tissue with site-specific structural and functional properties.
  • submucosa tissue can be used as a tissue graft construct, for example, in its native form, in its fluidized form, in the form of an extract, or as components extracted from submucosa tissue and subsequently purified.
  • the fluidized forms of vertebrate submucosa tissue can be gelled to form a semi-solid composition that can be implanted as a tissue graft construct or utilized as a cell culture substrate.
  • the fluidized form can be injected, or delivered using other methods, to living tissues to enhance tissue remodeling.
  • the fluidized form can be modified, or can be combined with specific proteins, growth factors, drugs, plasmids, vectors, or other therapeutic agents for controlling the enhancement of tissue remodeling at the site of injection.
  • the fluidized, solubilized form can be combined with primary cells or cell lines prior to injection to further enhance the remodeling properties that result in the repair or replacement of diseased or damaged tissues.
  • Controlling the assembly of the constituting monomers into tertiary or quaternary multimeric arrangements is very hard to achieve under such conditions.
  • One embodiment of the present invention is directed to controlling the polymerization of a composition comprising solubilized collagen to form a collagen based scaffold that has the requisite microstructure and composition to allow for the expansion, differentiation and/or clonal isolation of stem cells in a highly reproducible and predictable manner.
  • the present invention relates to compositions comprising a three dimensional matrix that is formed to have the requisite composition and microstructure to enhance the proliferation and /or differentiation of stem cells or progenitor cells cultured within such a matrix.
  • an improved method for culturing stem cells comprises preparing a solubilized collagen composition from a source of collagen, adding cells to the solubilized collagen composition and polymerizing the collagen composition under controlled conditions to provide a matrix formed from collagen fibrils and having the desired microstructure.
  • cells are added to the collagen based matrix at a cell density within two orders of magnitude of the minimum cell number required to maintain cell viability, and the cells are cultured under conditions suitable for proliferation of the cells.
  • the three dimensional matrix has a fibril area fraction (defined as the percent area of the total area occupied by fibrils in a cross-sectional surface of the matrix; providing an estimate of fibril density) of about 8% to about 26% and an elastic or linear modulus (defined by the slope of the linear region of the stress-strain curve) of about 0.5 to about 40 kPa.
  • the three dimensional matrix is further provided with an exogenous source of glucose and calcium chloride.
  • stem cell seeded engineered purified collagen based matrices are used as novel compositions for inducing the repair of damaged or disease tissues in vivo.
  • the tissue graft construct comprises an engineered purified collagen based matrix, wherein the matrix is formed by contacting purified collagen with hydrochloric acid to produce a solubilized collagen composition and subsequently polymerizing the solubilized collagen composition under controlled conditions and in the presence of a population of cells to produce the engineered purified collagen based matrix containing cells entrapped within the matrix, hi one embodiment the population of cells comprises stem cells initially added to the composition at a density of less than 10 5 cells per milliliter, or the progeny of such stem cells, hi one embodiment the stem cell seeded engineered purified collagen based matrices are implanted into a host without culturing the seeded stem cells in vitro. In another embodiment the stem cell seeded engineered purified collagen based matrix is further incubated under conditions suitable for inducing the proliferation and/or differentiation of the seeded stem cells.
  • the stem cells are added to the engineered purified collagen based matrices at densities of less than 10 3 cells per milliliter and the cells are cultured under conditions that are minimally permissive for stem cell functionality. These conditions result in the production of localized populations of stem cells and thus allow for the isolation of clonal populations of stem cells. Accordingly, in one embodiment, a method of isolating clonal populations of individual stem cells is provided.
  • the method comprises the steps of contacting a collagen based matrix with a low density of stem cells wherein said collagen matrix is formed by contacting a source of collagen with HCl to prepare a solubilized collagen composition, polymerizing the solubilized collagen composition using a final collagen concentration of 1.0 to 3.0 mg/ml, at a pH of about 6.5 to about 7.0.
  • the initial seeded population of stem cells ranges from about 10 to about 10 3 cells per milliliter.
  • the seeded stem cells are cultured under conditions suitable for proliferation of the cells and individual populations of stem cells are isolated.
  • Figs. 1 A-IF present data showing the effect of various parameters on the stiffness (elastic or linear modulus) of the formed matrix.
  • Fig IA represents the effect of polymerization temperature on a matrix formed from a solubilized collagen composition comprising 1 mg/ml collagen in IX PBS at pH 7.4;
  • Fig. IB represents the effect of the buffer type on a matrix formed from a solubilized collagen composition comprising 1 mg/ml collagen, and about 0.15 M NaCl at 37 0 C;
  • Fig. 1C represents the effect of pH (using a phosphate buffer) on a matrix formed from a solubilized collagen composition comprising 1 mg/ml collagen, in IX PBS at pH 7.4;
  • ID represents the effect of pH (using a tris buffer) on a matrix formed from a solubilized collagen composition comprising 1 mg/ml collagen, in 5OmM tris, and about 0.15 M NaCl at 37 0 C.
  • Fig. IE represents the effect of ionic strength on a matrix formed from a solubilized collagen composition comprising 1 mg/ml collagen, no buffer, at 37 0 C;
  • Fig. IF represents the effect of phosphate concentration on a matrix formed from a solubilized collagen composition comprising 1 mg/ml collagen, and about 0.15 M NaCl at 37 0 C;
  • Fig. IG represents the effect of SIS component concentration on a matrix formed from a solubilized ECM collagen composition in IX PBS at 37 0 C.
  • Figs. 2 A & 2B represent a series of graphs showing the quantification of fibril area fraction (Fig. 2A) and fibril diameter distribution (Fig. 2B) based upon confocal and SEM images, respectively. All fibril area fraction relationships showing statistically significant differences (p ⁇ 0.05) are indicated with symbols (*, **,*,o).
  • Figs. 3A-3D represent a series of graphs showing cell length (Fig. 3A), length/width ratio (Fig. 3B), width (Fig. 3C), and surface area (Fig. 3D) determined and compared for neonatal human dermal fibroblasts (NHDFs) seeded within 3D ECMs prepared with 1.5 mg/ml type I collagen, and a type III collagen content that varied from 0 to 0.75 mg/ml. Results represent the means and standard deviations for 10 ⁇ n ⁇ 23 cells analyzed for each ECM formulation at a given time point. All groups showing statistically significant differences (p ⁇ 0.05) are marked with the same symbol. Figs.
  • FIGS. 4A-4D represent a series of images depicting cell contractility and matrix remodeling by individual NHDFs resident within type I collagen (1.5 mg/ml) ECMs prepared with type III collagen concentrations of 0.25 mg/ml (Figs. 4A and 4B) and 0.75 mg/ml (Figs. 4C and 4D).
  • Figs. 4A and 4C represent 2D projections of confocal reflection image stacks showing changes to NHDF morphology and collagen fibril microstructure observed 5 hours after polymerization.
  • Figs. 4B and 4D represent quantified levels of local volumetric strain (matrix deformation) within the 3D tissue construct.
  • Fig. 5 represents a graph depicting contractility and matrix remodeling within engineered ECMs.
  • NHDFs were grown within engineered ECMs in which the type I collagen concentration was kept constant at 1.5 mg/ml and the amount of type III collagen was either 0.25 mg/ml or 0.75 mg/ml.
  • Average local 3D principal strains for a single cell and its surrounding ECM were quantified 5 hours post- polymerization (5 ⁇ n ⁇ 6).
  • Negative strain values indicate compressive deformations. All relationships showing statistically significant differences (p ⁇ 0.05) are indicated with symbols (*, **,»,o,D,+).
  • Fig. 6 represents a graph showing that points of maximum local deformation or strain induced within a 3D tissue construct, by low passage neonatal human dermal fibroblasts, occurred at distances further from the cell than for engineered ECMS prepared with lower amounts of type III collagen.
  • NHDFs were grown within engineered ECMs in which the type I collagen concentration was kept constant at 1.5 mg/ml and the amount of type III collagen was either 0.25 mg/ml or 0.75 mg/ml.
  • Figs. 7A & 7B represent a series of graphs depicting data regarding the proliferation of low passage human dermal fibroblasts when grown within a 3D ECM format consisting of type I collagen ECMs prepared within increasing amounts of type III collagen (see Fig. 7A).
  • Fig. 8 represents a bar chart showing differences in the expression of select tissue-specific genes by multi-potential bone marrow derived mesenchymal cells grown on standard 2D plastic and within 3D ECM microenvironments of increased fibril density and stiffness (elastic or linear modulus). Gene expression patterns for mesenchymal cells cultured within a given 2D or 3D format was also modulated by changing the composition of the culture medium.
  • Fig. 9 is a schematic representation of general cell behavior of multi- potential bone marrow derived mesenchymal cells when cultured within 3D matrices that differ in collagen concentration to provide an ECM microenvironment characterized by increased fibril density and stiffness (elastic or linear modulus). Points of arrow indicate low frequency events and wide ends of arrows indicate high frequency events.
  • stem cell refers to an unspecialized cell from an embryo, fetus, or adult that is capable of self-replication or self-renewal and can develop into specialized cell types of a variety of tissues and organs.
  • the term as used herein encompasses totipotent cells (those cells having the capacity to differentiate into extra-embryonic membranes and tissues, the embryo, and all post-embryonic tissues and organs), pluripotent cells (those cells that can differentiate into cells derived from any of the three germ layers), and multipotent cells (those cells having the capacity to differentiate into a limited range of differentiated cell types).
  • progenitor cell refers to a stem cell with more specialization and less differentiation potential than a totipotent stem cell.
  • progenitor cells include unipotential cells (those cells having the capacity to differentiate along a single cell lineage).
  • lyophilized relates to the removal of water from a composition, typically by freeze-drying under a vacuum.
  • lyophilization can be performed by any method known to the skilled artisan and the method is not limited to freeze-drying under a vacuum.
  • the lyophilized tissue is lyophilized to dryness, and in one embodiment the water content of the lyophilized tissue is below detectable levels.
  • isolated collagen composition refers to a composition that comprises collagen in a predominantly soluble monomelic form (for example wherein less than 20% of the collagen is insoluble, denatured, or assembled in higher ordered structures).
  • “solubilized extracellular matrix composition” refers to a naturally occurring extracellular matrix that has been treated, for example, with an acid to reduce the molecular weight of at least some of the components of the extracellular matrix and to produce a composition wherein at least some of the components of the extracellular matrix have been solubilized from the extracellular matrix.
  • the "solubilized extracellular matrix composition” may include insoluble components of the extracellular matrix as well as solubilized components.
  • collagen-based matrix refers to extracellular matrices that comprise collagen.
  • An "engineered purified collagen based matrix” as used herein relates to a composition comprising a collagen fibril scaffold that has been formed under controlled conditions from a solubilized collagen composition, wherein the solubilized collagen composition is prepared from a composition consisting essentially of collagen.
  • the conditions controlled during the polymerization reaction include one or more of the following: pH, phosphate concentration, temperature, buffer composition, ionic strength, and composition and concentration of purified collagen components.
  • an “engineered extracellular matrix” relates to a solubilized extracellular matrix composition that is polymerized to form a collagen fibril containing matrix under controlled conditions, wherein the controlled conditions include pH, phosphate concentration, temperature, buffer composition, ionic strength, and composition and concentration of the extracellular matrix components which includes both collagen and non-collagenous molecules.
  • a “bioactive engineered extracellular matrix” composition refers to an engineered extracellular matrix composition that can be polymerized to form a three dimensional scaffold that is capable of remodeling tissues in vivo.
  • naturally occurring extracellular matrix comprises any noncellular material naturally secreted by cells (such as intestinal submucosa) isolated in their native configuration with or without naturally associated cells.
  • submucosal matrices refers to natural extracellular matrices, known to be effective for tissue remodeling, that have been isolated in their native configuration, including submucosa derived from vertebrate intestinal tissue, stomach tissue, bladder tissue, alimentary tissue, respiratory tissue and genital tissue.
  • exogenous or “exogenously added” designates the addition of a new component to a composition, or the supplementation of an existing component already present in the composition, using material from a source external to the composition.
  • sterilization or “sterilize” or “sterilized” means removing unwanted contaminants including, but not limited to, endotoxins, nucleic acid contaminants, and infectious agents.
  • stiffness or elastic or linear modulus” refers to the fundamental material property defined by the slope linear portion of a stress-strain curve that results from conventional mechanical testing protocols.
  • the term "purified” and like terms relate to the isolation of a molecule or compound in a form that is substantially free from other components with which they are naturally associated (e.g., the total amount of nondesignated components present in the composition representing less than 5%, or more typically less than 1%, of total dry weight).
  • 3D matrix three dimensional purified collagen matrix
  • a "3D purified collagen matrix populated/seeded with cells” further comprises a viable population of cells contained within the matrix.
  • 3D ECM three dimensional extracellular matrix
  • three dimensional matrix (3D matrix) is a generic term that is intended to include both "three dimensional purified collagen matrices (3D purified collagen matrices)” as well as “three dimensional extracellular matrices (3D ECM).
  • collagen fibril refers to a quasi-crystalline, filamentous structure formed by the self-assembly of soluble trimeric collagen molecules. The collagen molecules in a collagen fibril typically pack in a quarter- staggered pattern giving the fibril a characteristic striated appearance or banding pattern along its axis. Solubilized collagen that is assembled in vitro to form collagen fibrils exhibit similarities to collagen structures found in vivo (Veis and George, 1994 Fundamental of interstitial collagen assembly.
  • collagen fibrils are organized as bundles in a hierarchical manner to form fibers. Collagen fibers are further organized in a tissue-specific fashion to provide tissues with specific structural-functional properties. Collagen fibrils are distinct from the amorphous aggregates or precipitates of insoluble collagen that can be formed by dehydrating (e.g., lyophilization) collagen suspensions to form porous network scaffolds. Collagen networks formed from amorphous aggregates, or precipitates of insoluble collagen, have characteristics that distinct from those formed from collagen fibrils as defined above. EMBODIMENTS
  • Cell culture scaffolds presenting a more biologically relevant microenvironment are disclosed. More particularly, these cell culture scaffolds comprise three-dimensional matrices/biomaterials that are created from solubilized collagen compositions.
  • the solubilized collagen compositions are prepared from biological sources, such as naturally occurring extracellular matrices, including for example submucosal matrices. More particularly, the soluble polymers suitable for use in the present invention can be isolated, to varying degrees of purity, from natural tissues and include, but are not limited to, type I collagen, type III collagen, growth factors and glycosaminoglycans.
  • the solubilized collagen composition comprises purified type I collagen or a mixture of purified type I and type III collagen.
  • the solubilized collagen composition undergoes polymerization/self assembly to form a three dimensional scaffold/biomaterial comprised of collagen fibrils.
  • the soluble polymers comprise type I collagen monomers, where upon polymerization the resulting scaffolds represent a composite material comprising insoluble collagen fibrils and an interfibrillar fluid component, referred to herein as a three dimensional matrix.
  • An array of scaffolds/biomaterials can be created by varying the composition of ECM molecules as well as the self-assembly/polymerization conditions.
  • applicants have discovered that upon seeding progenitor cells or stem cells within engineered purified collagen based matrices (scaffolds) representing different microstructural compositions (e.g., having different dimensioned and organizations of the collagen fibrils and filaments), distinct patterns of cell survival, growth, proliferation, and differentiation are obtained.
  • engineered purified collagen based matrices representing different microstructural compositions (e.g., varied fibril dimensions (length, diameter) and densities) will impact the rate of cell proliferation as well as the pattern of cellular condensation, aggregation, fusion, and cellular differentiation events and their associated time-line.
  • engineered purified collagen based matrices can be specifically designed to foster the proliferation of stem cells while maintaining their precursor or multi- potential status.
  • engineered purified collagen based matrices can be designed to direct differentiation of cells down a specific cell lineage (such as fat, bone, muscle, or cartilage) to form 3D organotypic tissues (that is pronounced of in vivo tissue structure and function).
  • stem cells and/or progenitor cells are seeded at relatively low densities on or within the various engineered purified collagen based matrices. It is known that, in general, cell behavior is determined by a combination of signal inputs arising from soluble factors, biophysical factors, the extracellular matrix substrate, and cell-cell interactions. Seeding cells at a relative low cell density on or within the collagen based matrices of the present invention allows ECM-based signaling to predominate over signals derived from cell-cell interactions. In accordance with one embodiment, cells are initially seeded on or within the engineered purified collagen based matrices at a minimal cell density that will allow for cell viability and replication (i.e., the minimal functionality density). This minimal functional density can be easily established for the particular cell type to be cultured and for the specific culture conditions utilized.
  • the stem cells or progenitor cells are seeded within the collagen based matrix at a cell density substantially higher than the minimal functionality density but at a relative low density compared to standard cell culture techniques.
  • the cells comprise stem cells, wherein the cells are seeded at a density within 3 orders of magnitude of the minimal functionality density, in another embodiment stem cells are seeded at a density within 2 orders of magnitude of the minimal functionality density, and in another embodiment the stem cells are seeded at a density within an order of magnitude of the minimal functionality density.
  • the stem cells can be seeded at a relatively high density of about IX 10 6 to about I X lO 8 cells/ml, or at a more typical density of about IX 10 3 to about I X lO 5 cells/ml. Seeding the cells at the relative high density of about IX 10 6 to about 1 X 10 cells/ml will promote cell to cell interactions over cell to matrix interactions. Accordingly, stem cells seeded at relatively high densities will develop into fat tissue even when the cells are cultured within 3D matrices of high collagen fibril density, hi one embodiment stem cells are seeded at a density of less than 5 X 10 4 cells/ml, more typically at a density of about 5 X 10 4 cells/ml.
  • stem cells are seeded at a density of less than I X lO 4 cells/ml, in another embodiment stem cells are seeded at a density selected from a range of about 1 X 10 2 to about 5 X 10 3 .
  • an improved method for culruring stem cells uses three dimensional purified collagen based matrices. The improved method allows for enhanced proliferation of stem cells as well as better control over the differentiation of the cultured cells, rn one embodiment the method comprises the steps of providing a solubilized collagen composition, adding cells to the collagen composition, and polymerizing the solubilized collagen composition to form collagen fibrils.
  • the solubilized collagen composition comprises collagen that has been isolated with or without additional components from natural tissues.
  • the solubilized collagen composition is prepared using purified type I collagen as a starting material.
  • collagen, and more particularly type I or type III collagen, that has been isolated from tissues is subjected to a final purification step that removes any reagents that were used during the isolation steps.
  • the final purification step comprises dialyzing the isolated collagen in an aqueous solution, and in one embodiment the isolated collagen is dialyzed against a dilute acid solution, including for example, hydrochloric acid.
  • the final purification step comprises dialyzing the isolated collagen against a 0.01 N HCl solution.
  • Isolated type I or isolated type III collagen preparations are commercially available, and these commercially available materials are subjected to a further purification step, including for example, dialyzing against a dilute (about 0.001 N to about 0.1 N) hydrochloric acid solution to produce purified collagen suitable for use for forming 3D purified collagen matrices.
  • the dialysate can optionally be filtered and/or centrifuged to remove particulate matter.
  • the collagen component of the solubilized collagen composition consists essentially of purified collagen, the majority of which are in monomelic form.
  • the composition is formed from purified collagen (the majority of which are in monomeric form) that is greater than 75% type I collagen, or greater than 90% type I collagen.
  • a composition consisting essentially of purified collagen is dissolved in an acid solution, such as hydrochloric acid to prepare a solubilized collagen composition of the desired concentration.
  • the purified collagen is dissolved in about 0.001 N to about 0.1 N, from about 0.005 N to about 0.1 N, from about 0.005 N to about 0.01 N, from about 0.01 N to about 0.1 N, from about 0.05 N to about 0.1 N, from about 0.001 N to about 0.05 N, about 0.001 N to about 0.01 N, or from about 0.01 N to about 0.05 N hydrochloric acid solution.
  • a three dimensional purified collagen matrix is provided, wherein the matrix is formed from a solubilized collagen composition wherein the collagen components of the solubilized collagen composition consist essentially of purified type I and type III collagen.
  • the component fibrils of such matrices have been found to have a greater degree of flexibility relative to the fibrils of engineered purified collagen matrices that are formed using only type I collagen.
  • the matrix comprises type I collagen and type III collagen in a ratio of 200: 1.
  • the method of forming matrices with fibrils that exhibit a higher degree of flexibility comprises the steps of combining in vitro at least 100 ug/ml of type I collagen with at least 0.5 ug/ml of type III collagen to obtain a total amount of collagen, and forming in vitro a three dimensional purified collagen matrix wherein the three dimensional matrix has decreased stiffness compared to a 3D matrix formed in vitro with type I collagen when the total amount of collagen in the two matrices is equivalent.
  • a method of preparing an extracellular matrix composition comprises the steps of combining in vitro at least 100 ug/ml of type I collagen with at least 0.5 ug/ml of type III collagen to obtain a total amount of collagen, and forming in vitro a three dimensional matrix.
  • the type I and type III collagen is dissolved in about 0.001 N to about 0.1 N, from about 0.005 N to about 0.1 N, from about 0.005 N to about 0.01 N, from about 0.01 N to about 0.1 N, from about 0.05 N to about 0.1 N, from about 0.001 N to about 0.05 N, about 0.001 N to about 0.01 N, or from about 0.01 N to about 0.05 N hydrochloric acid solution either before or after the combining step.
  • an extracellular matrix composition for use in repairing diseased or damaged tissues.
  • the extracellular matrix composition comprises at least 100 ug/ml of type I collagen and at least 0.5 ug/ml of type III collagen, wherein the type I collagen to type III collagen ratio is selected from the group consisting of 200:1, 100:1, 50:1, 15:1, 10:1, 8:1, 6:1, 5:1, 3:1, and 2:1, and a population of cells.
  • the matrix is formed by provided a solubilized collagen composition comprising type I and type III collagen, in a ratio selected from the group consisting of 200:1, 100:1, 50:1, 15:1, 10:1, 8:1, 6:1, 5:1, 3:1, and 2:1, polymerizing the solubilized collagen composition to form collagen fibrils, and adding cells to the collagen composition either before or after the polymerization step.
  • a composition comprising solubilized collagen and stem cells is injected into a host and the polymerization of the solubilized collagen composition occurs in vivo to form a cell entrapping matrix.
  • the solubilized collagen composition can be polymerized in vitro and the polymerized matrix, comprising the population of cells, can be subsequently injected or implanted in a host.
  • the population of cells entrapped within the 3D matrix can be cultured in vitro, for a predetermined length of time, to increase cell numbers and/or induce differentiation of the cell population prior to implantation into a host, hi a further embodiment, the population of cells can be cultured in vitro, for a predetermined length of time, to increase cell numbers and/or induce differentiation of the cell population and the cells can be separated from the matrix and implanted into the host in the absence of the polymerized matrix.
  • the engineered purified collagen based matrix comprises type III collagen in the range of about 0.5% to about 33% of total collagen in the matrix. In another illustrative embodiment, the engineered purified collagen based matrix comprises type I collagen in the range of about 66% to about 99.5% of total collagen in the matrix. In yet another illustrative embodiment, the type I collagen to type III collagen ratio is in the range of about 2:1 to about 200:1, wherein the type I collagen to type III collagen ratio may be selected from the group consisting of 200:1, 100:1, 50:1, 15:1, 10:1, 8:1, 6:1, 5:1, 3:1, and 2:1.
  • a method of enhancing cell proliferation within an extracellular matrix composition comprises the steps of combining in vitro an amount of type I collagen with an amount of type III collagen to obtain a total amount of collagen wherein the ratio of type III collagen to type I collagen is at least 1:6, and forming in vitro a three-dimensional extracellular matrix wherein the extracellular matrix enhances cell proliferation compared to an extracellular matrix formed in vitro with type I collagen wherein the amount of type I collagen is equivalent to the total amount of type I collagen in the combining step.
  • the method comprises the steps of combining in vitro at least 3 ug/ml of type I collagen with at least 0.5 ug/rnl of type III collagen to obtain a total amount of collagen wherein the ratio of type III collagen to type I collagen is at least 1 :6, and forming in vitro a three-dimensional extracellular matrix wherein the extracellular matrix enhances cell proliferation compared to an extracellular matrix formed in vitro with type I collagen, wherein the amount of type I collagen is equivalent to the total amount of type I collagen in the combining step.
  • the method of preparing an engineered purified collagen based matrix comprises combining type I and type III collagen wherein the type III collagen is added in the range of about 17% to about 33% of total collagen in the matrix.
  • the type I collagen is added in the range of about 66% to about 83% of total collagen in the matrix.
  • the type I collagen to type III collagen ratio is in the range of about 6:1 to about 1:1, wherein the type I collagen to type III collagen ratio may be selected from the group consisting of 6:1, 5:1, 4:1, 3:1, 2:1, and 1:1.
  • 3D matrices can be prepared from solubilized collagen compositions having purified collagen concentrations ranging from as little as 0.05 mg/ml to as much as 40mg/ml.
  • the 3D matrices are prepared from purified solubilized collagen compositions having a collagen concentration selected from a range of about 0.1mg/ml to about 5.0mg/ml, and in one embodiment about 1.5 mg/ml to about 3.0 mg/ml.
  • Table 1 summarizes the effect of total collagen concentration on the fibril structure of the matrix:
  • the 3D matrices formed in accordance with the present disclosure represent a matrix of collagen fibrils.
  • the fibrils of the matrices are formed at a fibril area fraction (density) of about 7.7% to about 25% total volume.
  • the 3D matrices have a fibril area fraction of about 12.8% to about 18.3% total volume.
  • the 3D matrices have a fibril area fraction of about 18.5% to about 25% total volume.
  • the 3D matrix has a fibril area fraction of about 12.8% to about 18.3% total volume and the fibrils have a hydrated diameter of about 350 to about 475nm.
  • the 3D matrix has a fibril area fraction of about 18.5% to about 25% total volume and the fibrils have a hydrated diameter of about 375 to about 500nm.
  • 3D matrices having low fibril density and low stiffness enhance stem cell proliferation with decreased differentiation of the cells. Accordingly, 3D matrices formed from solubilized collagen compositions having about 0.1 mg/ml to about 3 mg/ml collagen, and more typically about 0.5 mg/ml to about 2.5 mg/ml collagen are utilized to stimulate stem cell proliferation.
  • the 3D matrices so formed will have a fibril predicted fibril area fraction (density) of about 7.7% to about 18.3% total volume and about 9.2% to about 16.5% total volume, respectively, hi one embodiment the 3D matrices are formed from solubilized collagen compositions having about 3 mg/ml to about 1.5 mg/ml collagen and in one embodiment the solubilized collagen compositions have about 2.5, 2.0, 1.5, or 1.0 mg/ml of collagen. Alternatively, higher concentrations of total collagen present in the three dimensional matrix leads to differentiation of stem cells.
  • 3D matrices (having a fibril area fraction of at least about 18% total volume) formed from solubilized collagen compositions having more than about 3 mg/ml are utilized to stimulate differentiation of stem cells cultured within the matrix.
  • the 3D matrices are formed from solubilized collagen compositions having about 3.2, 3.4, 3.6, 3.8, 4.0, 4.5 or 5.0 mg/ml of collagen, resulting in 3D matrices having a fibril area fraction of about 19%, 19.7%, 20.5%, 21.2%, 22%, 23.8% and 25.6% total volume, respectively.
  • 3D matrix can be modified by controlling the relative proportion of type I to type III collagen, the fibril area fraction (density), or the fibril diameter of the collagen fibrils in the 3D matrix.
  • 3D matrices are prepared having a relatively low stiffness (elastic or linear modulus) of about 0.48 to about 24.0 IcPa. In one embodiment these matrices are used to propagate stem cells and progenitor cells without further differentiation of the cells and/or their progeny. In another embodiment 3D matrices are prepared having a relatively high stiffness of about 25 to about 40 kPa. In one embodiment these relatively stiffer matrices are used to induce the differentiation of stem cells and progenitor cells and/or their progeny.
  • a 3D matrix having a relatively low stiffness of about 0.48 to about 24.0 kPa and a relatively low fibril area fraction (density) of about 7% to about 18% total volume.
  • a 3D matrix is provided having a relatively high stiffness of about 25 to about 40 kPa and a relatively high fibril area fraction (density) of about 19% to about 26% total volume.
  • the solubilized collagen composition comprises collagen monomers isolated from natural tissues, and includes additional components that are naturally associated with the native tissues and/or exogenously added components. In one embodiment various exogenous materials, such as growth factors are added to the collagen based matrices of the present invention.
  • the solubilized collagen composition represents a solubilized fraction of a naturally occurring extracellular matrix, and in one embodiment the naturally occurring extracellular matrix is a vertebrate submucosal matrix. In one embodiment the solubilized collagen composition represents a solubilized fraction of vertebrate intestinal submucosa.
  • acetic acid formic acid, lactic acid, citric acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid can be used to solubilize the naturally occurring extracellular matrix (or a purified lyophilized collagen composition) to produce a solubilized collagen composition.
  • the solubilized collagen composition derived from a naturally occurring extracellular matrix, such as vertebrate intestinal submucosa, can then be polymerized to form an engineered extracellular matrix.
  • the invention also relates to methods of preparation and compositions comprising solubilized extracellular matrix components polymerized in vitro where the extracellular matrix components are solubilized by other methods known in the art.
  • the polymerizing step can be performed under conditions that are systematically varied where the conditions are selected from the group consisting of pH, phosphate concentration, temperature, buffer composition, ionic strength, the extracellular matrix components in the solubilized extracellular matrix composition, and the concentration of the extracellular matrix components in the solubilized extracellular matrix composition.
  • a method of forming a 3D matrix comprising stem cells is provided. The method comprises the steps of providing an acid solubilized purified type I collagen composition, hi one embodiment the collagen composition further comprises type III collagen.
  • the purified collagen represents a commercially available isolated preparation of collagen that is further subjected to purification, including for example dialyzing against an solution of about 0.005 N to about 0.1 N HCl, more typically about 0.01 N HCl.
  • the solubilized collagen composition comprises purified collagen that is suspended in about 0.005 N to about 0.1 N HCl solution, and in one embodiment suspended in 0.0 IN HCl.
  • the solubilized collagen composition is also typically sterilized using standard techniques including for example contact with chloroform or peracetic acid. Stem cells are then added to the solubilized collagen composition at a specific density, typically ranging from about 1 X 10 to about 1 X 10 .
  • the stem cells are added to the solubilized collagen composition at a concentration of less than 5 X 10 4 cells per milliliter, and in one embodiment the cells are added at a density of about 10 to about 10 3 per milliliter, hi accordance with one embodiment the collagen/cell suspension is then pipetted into a well plate and allowed to polymerize in a humidified environment at 37 0 C for approximately 30 minutes. In an alternative embodiment the collagen/cell suspension is injected into a host and the composition is polymerized in vivo.
  • solubilized collagen compositions can be prepared from vertebrate submucosal matrices wherein the collagen compositions comprise additional components besides collagen.
  • Vertebrate submucosal matrices can be obtained from various sources, including intestinal tissue harvested from animals raised for meat production, including, for example, pigs, cattle and sheep or other warm-blooded vertebrates.
  • the solubilized collagen composition is derived from one or more sources selected from the group consisting of intestinal submucosa, stomach submucosa, urinary bladder submucosa, uterine submucosa, and. any other submucosal material that can be used to remodel endogenous tissue.
  • the submucosa comprises the tunica submucosa delaminated from both the tunica muscularis and at least the luminal portion of the tunica mucosa of a warm-blooded vertebrate.
  • Such constructs can be prepared by mechanically removing the luminal portion of the mucosa and the external muscle layers and lysing resident cells with hypotonic washes.
  • compositions comprising the tunica submucosa delaminated from both the tunica muscularis and at least the luminal portion of the tunica mucosa of the submucosal tissue of warm-blooded vertebrates can be used as tissue graft materials (see, for example, U.S. Patents Nos. 4,902,508 and 5,281,422 incorporated herein by reference).
  • Such submucosal tissue preparations are characterized by excellent mechanical properties, including high compliance, high tensile strength, a high burst pressure point, and tear-resistance.
  • Submucosa-derived matrices are collagen based biodegradable matrices comprising highly conserved collagens, glycoproteins, proteoglycans, and glycosaminoglycans in their natural configuration and natural concentration.
  • Such submucosal material serves as a matrix for the regrowth of endogenous tissues at the implantation site (e.g., biological remodeling).
  • the submucosal material serves as a rapidly vascularized matrix for support and growth of new endogenous connective tissue.
  • submucosa matrices have been found to be trophic for host cells of tissues to which it is attached or otherwise associated in its implanted environment, hi multiple experiments submucosal tissue has been found to be remodeled (resorbed and replaced with autogenous differentiated tissue) to assume the characterizing features of the tissue(s) with which it is associated at the site of implantation or insertion.
  • Small intestinal submucosa tissue is an illustrative source of submucosal tissue for use in this invention.
  • Submucosal tissue can be obtained from various sources, for example, intestinal tissue can be harvested from animals raised for meat production, including, pigs, cattle and sheep or other warm-blooded vertebrates.
  • Small intestinal submucosal tissue is a plentiful by-product of commercial meat production operations and is, thus, a low cost material.
  • Suitable intestinal submucosal tissue typically comprises the tunica submucosa delaminated from both the tunica muscularis and at least the luminal portion of the tunica mucosa, but other tissue constructs can also be used.
  • the intestinal submucosal tissue comprises the tunica submucosa and basilar portions of the tunica mucosa including the lamina muscularis mucosa and the stratum compactum which layers are known to vary in thickness and in definition dependent on the source vertebrate species.
  • submucosal tissue is described in U.S. Patent. No. 4,902,508, the disclosure of which is expressly incorporated herein by reference.
  • a segment of vertebrate intestine for example, preferably harvested from porcine, ovine or bovine species, but not excluding other species, is subjected to abrasion using a longitudinal wiping motion to remove the outer layers, comprising smooth muscle tissues, and the innermost layer, i.e., the luminal portion of the tunica mucosa.
  • the submucosal tissue is rinsed under hypotonic conditions, such as with water or with saline under hypotonic conditions, and is optionally sterilized.
  • the submucosal tissue can be sterilized using conventional sterilization techniques including glutaraldehyde tanning, formaldehyde tanning at acidic pH, propylene oxide or ethylene oxide treatment, gas plasma sterilization, gamma radiation, electron beam, and/or peracetic acid sterilization. Sterilization techniques which do not adversely affect the structure and biotropic properties of the submucosal tissue can be used.
  • An illustrative sterilization technique is exposing the submucosal tissue to peracetic acid, 1-4 Mrads gamma irradiation (or 1-2.5 Mrads of gamma irradiation), ethylene oxide treatment, exposure to chloroform, or gas plasma sterilization.
  • the submucosal tissue can be subjected to one or more sterilization processes.
  • the intact extracellular matrix material can be sterilized with peracetic acid or the solubilized collagen composition can be sterilized.
  • the submucosal tissue can be subjected to one or more sterilization processes.
  • the submucosal tissue can be stored in a hydrated or dehydrated state prior to solubilization in accordance with the invention.
  • Extracellular matrix-derived tissues other than intestinal submucosa tissue may be used in accordance with the methods described herein and used as a source for preparing solubilized collagen compositions. Methods of preparing and treating other extracellular matrix-derived tissues are known to those skilled in the art and may be similar to the methods described above.
  • the extracellular matrix material is solubilized with an acid and the solubilized fraction is recovered for polymerization to form the collagen based matrices of the present invention.
  • the source extracellular matrix material is comminuted by tearing, cutting, grinding, or shearing the harvested extracellular matrix material.
  • the extracellular matrix material can be comminuted by shearing in a high-speed blender, or by grinding the extracellular matrix material in a frozen or freeze-dried state, and then lyophilizing the material to produce a powder having particles ranging in size from about 0.1 mm 2 to about 1.0 mm 2 .
  • the extracellular matrix material powder can thereafter be hydrated with, for example, water or buffered saline to form a fluid or liquid or paste-like consistency.
  • the extracellular matrix tissue is comminuted by freezing and pulverizing under liquid nitrogen in an industrial blender.
  • the preparation of fluidized forms of the source extracellular matrix material, such as submucosa tissue, is described in U.S. Patent No. 5,275,826, the disclosure of which is expressly incorporated herein by reference.
  • an acid such as hydrochloric acid, acetic acid, formic acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid, is used to solubilize the source extracellular matrix material.
  • the acidic conditions for solubilization can include solubilization at about O 0 C to about 6O 0 C, and incubation periods of about 5 minutes to about 96 hours.
  • the concentration of the acid can be from about 0.001 N to about 0.1 N, from about 0.005 N to about 0.1 N, from about 0.01 N to about 0.1 N, from about 0.05 N to about 0.1 N, from about 0.001 N to about 0.05 N, about 0.001 N to about 0.01 N, or from about 0.01 N to about 0.05 M.
  • the solubilization can be conducted at any temperature, for any length of time, and at any concentration of acid.
  • the solubilization step can be performed in the presence of an acid or in the presence of an acid and an enzyme.
  • the acid solubilization step results in a solubilized extracellular matrix composition that remains bioactive (i.e., is capable of polymerizing and remodeling tissues in vivo) after lyophilization.
  • the extracellular matrix material is treated with one or more enzymes before, during, or after the acid solubilization step.
  • the extracellular matrix material is treated with the enzyme before the acid solubilization step or after the acid solubilization step, but under conditions that are not acidic.
  • Enzymatic digestion of the extracellular matrix material is conducted under conditions that are optimal for the specific enzyme used and under conditions that retain the ability of the solubilized components of the extracellular matrix material to polymerize.
  • the concentration of the enzyme depends on the specific enzyme used, the amount of extracellular matrix material to be digested, the desired time of digestion, and the desired temperature of the reaction. In various illustrative embodiments, about 0.01% to about 0.5% (weight per volume, such that 0.01% is equivalent to 0.01 g/100 ml) of enzyme is used.
  • Exemplary enzymes include pepsin, bromelain, cathepsins, chymotrypsin, elastase, papain, plasmin, subtilisin, thrombin, trypsin, matrix metalloproteinases (e.g., stromelysin, elastase), glycosaminoglycan-specific enzymes (e.g., chondroitinase, hyaluronidase, heparinase)and the like, or combinations thereof.
  • the source extracellular matrix material can be treated with one or more enzymes.
  • the enzyme digestion can be performed at about 2 0 C to about 37 0 C. However, the digestion can be conducted at any temperature, for any length of time (e.g., about 5 minutes to about 96 hours), and at any enzyme concentration.
  • the ratio of the extracellular matrix material (hydrated) to total enzyme (weight/weight) ranges from about 25 to about 2500. If an enzyme is used, it should be removed (e.g., by fractionation) or inactivated after the desired incubation period for the digestion so as to not compromise stability of the components in the solubilized extracellular matrix composition. Enzymes, such as pepsin for example, can be inactivated with protease inhibitors, a shift to neutral pH, a drop in temperature below O 0 C, or heat inactivation, or a combination of these methods.
  • the source extracellular matrix material can be extracted in addition to being solubilized with hydrochloric acid.
  • the solubilized collagen composition comprises soluble and insoluble components, and at least a portion of the insoluble components of the solubilized collagen composition can be separated from the soluble components.
  • the insoluble components can be separated from the soluble components by centrifugation (e.g., at 12,000 rpm for 20 minutes at 4 0 C).
  • solubilized extracellular matrix composition prepared with or without the above-described separation step, is fractionated prior to polymerization.
  • solubilized extracellular matrix composition can be fractionated by dialysis.
  • Exemplary molecular weight cut-offs for the dialysis tubing or membrane are from about 3,500 to about 12,000 or about 3,500 to about 5,000.
  • the solubilized extracellular matrix composition is dialyzed against an acidic solution having a low ionic strength.
  • the solubilized extracellular matrix composition can be dialyzed against a hydrochloric acid solution, however any other acids can be used, including acetic acid, formic acid, citric acid, lactic acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid.
  • the extracellular matrix composition can be dialyzed against water as long as the pH is approximately 6 or below.
  • the fractionation for example by dialysis, can be performed at about 2 0 C to about 37 0 C for about 1 hour to about 96 hours.
  • the concentration of the acid such as acetic acid, hydrochloric acid, formic acid, citric acid, lactic acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid, against which the solubilized extracellular matrix composition is dialyzed, can be from about 0.001 N to about 0.1 N, from about 0.005 N to about 0.1 N, from about 0.01 N to about 0.1 N, from about 0.05 N to about 0.1 N, from about 0.001 N to about 0.05 N, about 0.001 N to about 0.01 N, or from about 0.01 N to about 0.05 N.
  • the solubilized extracellular matrix composition is dialyzed against 0.01 N HCl.
  • the fractionation can be performed at any temperature
  • the 3D matrix used for culturing stem cells comprises a lyophilized, solubilized collagen composition that is rehydrated prior to contact with the cells.
  • lyophilized means that water is removed from the composition, typically by freeze-drying under a vacuum (typically to dryness).
  • a solubilized extracellular matrix composition is lyophilized after solubilization.
  • the solubilized extracellular matrix composition is lyophilized after the solubilized portions have been separated from the insoluble portions, hi yet another illustrative aspect, the solubilized extracellular matrix composition is lyophilized after a fractionation step but prior to polymerization, hi another illustrative embodiment, the polymerized matrix is lyophilized.
  • the solubilized extracellular matrix composition is first frozen, and then placed under a vacuum.
  • the solubilized extracellular matrix composition is freeze-dried under a vacuum. Any method of lyophilization known to the skilled artisan can be used.
  • the solubilized collagen composition is sterilized before polymerization, m one embodiment the source of the solubilized collagen (e.g., a naturally occurring extracellular matrix, or a lyophilized purified collagen composition) is sterilized prior to the solubilization step. Sterilization of the extracellular matrix material can be performed, for example, as described in U.S. Patents Nos. 4,902,508 and 6,206,931, incorporated herein by reference. In another embodiment, the solubilized collagen composition is directly sterilized, for example, with peracetic acid.
  • the source of the solubilized collagen e.g., a naturally occurring extracellular matrix, or a lyophilized purified collagen composition
  • Sterilization of the extracellular matrix material can be performed, for example, as described in U.S. Patents Nos. 4,902,508 and 6,206,931, incorporated herein by reference.
  • the solubilized collagen composition is directly sterilized, for example, with peracetic acid.
  • sterilization can be carried out either before or after the fractionation step.
  • the lyophilized composition itself is sterilized before rehydration, for example using an e-beam sterilization technique.
  • the polymerized matrix formed from the components of the solubilized collagen matrix composition is sterilized.
  • the solubilized extracellular matrix composition is directly sterilized before the fractionation/separation step, for example, with peracetic acid or with peracetic acid and ethanol (e.g., by the addition of 0.18% peracetic acid and 4.8% ethanol to the solubilized extracellular matrix composition before the separation step).
  • sterilization can be carried out during the fractionation step.
  • the solubilized extracellular matrix composition can be dialyzed against chloroform, peracetic acid, or a solution of peracetic acid and ethanol to disinfect or sterilize the solubilized extracellular matrix composition.
  • the solubilized extracellular matrix composition can be sterilized by dialysis against a solution of peracetic acid and ethanol (e.g., 0.18% peracetic acid and 4.8% ethanol).
  • peracetic acid and ethanol e.g., 0.18% peracetic acid and 4.8% ethanol.
  • the chloroform, peracetic acid, or peracetic acid/ethanol can be removed prior to polymerization of the solubilized collagen composition, for example by dialysis against an acid, such as 0.01 N HCl.
  • the lyophilized collagen matrix composition can be stored frozen or at room temperature (for example, at about -8O 0 C to about 25 0 C). Storage temperatures are selected to stabilize the components of the solubilized collagen matrix composition.
  • the compositions can be stored for about 1-26 weeks, or longer, hi one illustrative embodiment, the storage solvent is hydrochloric acid.
  • storage solvent means the solvent that the solubilized collagen matrix composition is in prior to and during lyophilization.
  • hydrochloric acid, or other acids at concentrations of from about 0.001 N to about 0.1 N, from about 0.005 N to about 0.1 N, from about 0.01 N to about 0.1 N, from about 0.05 N to about 0.1 N, from about 0.001 N to about 0.05 N, from about 0.001 N to about 0.01 N, or from about 0.01 N to about 0.05 N can be used as the storage solvent for the lyophilized, solubilized collagen matrix composition.
  • Other acids can be used as the storage solvent including acetic acid, formic acid, citric acid, lactic acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid, and these acids can be used at any of the above-described concentrations.
  • the lyophilizate can be stored (i.e., lyophilized in) an acid, such as acetic acid, at a concentration of from about 0.001 M to about 0.5 M or from about 0.01 M to about 0.5 M.
  • the lyophilizate can be stored in water with a pH of about 6 or below.
  • lyoprotectants, cryoprotectants, lyophilization accelerators, or crystallizing excipients e.g., ethanol, isopropanol, mannitol, trehalose, maltose, sucrose, tert-butanol, and Tween 20
  • crystallizing excipients e.g., ethanol, isopropanol, mannitol, trehalose, maltose, sucrose, tert-butanol, and Tween 20
  • the sterilized, solubilized collagen composition can be dialyzed against 0.01 N HCl, for example, prior to lyophilization to remove the sterilization solution and so that the solubilized extracellular matrix composition is in a 0.01 N HCl solution as a storage solvent.
  • the solubilized extracellular matrix composition can be dialyzed against acetic acid as the storage solvent, for example, prior to lyophilization and can be lyophilized in acetic acid and
  • the resulting lyopliilizate can be redissolved in any solution, but may be redissolved in an acidic solution or water.
  • the lyophilizate can be redissolved in, for example, acetic acid, hydrochloric acid, formic acid, citric acid, lactic acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid, at any of the above-described concentrations, or can be redissolved in water, hi one illustrative embodiment the lyophilizate is redissolved in 0.01 N HCl.
  • the redissolved lyophilizate can be subjected to varying conditions (e.g., pH, phosphate concentration, temperature, buffer composition, ionic strength, and composition and concentration of solubilized extracellular matrix composition components (dry weight/ml)) that result in polymerization to form engineered extracellular matrices for specific tissue graft applications.
  • varying conditions e.g., pH, phosphate concentration, temperature, buffer composition, ionic strength, and composition and concentration of solubilized extracellular matrix composition components (dry weight/ml)
  • a solubilized collagen composition is prepared by enzymatically treating the source extracellular matrix material with 0.1% (w/v) pepsin in 0.01 N HCl to initially solubilized the extracellular matrix material, centrifuging the enzymatically treated composition at 12,000 rpm for 20 minutes at 4 0 C to remove insoluble components, fractionating the soluble fraction by dialysis against a 0.01 N HCl solution, and then polymerizing the dialyzed fraction.
  • the method does not involve a fractionation step.
  • the source extracellular matrix material is enzymatically treated with 0.1% (w/v) pepsin in a 0.01 N hydrochloric acid solution to produce a solubilized collagen composition, the solubilized composition is then centrifuged to remove insoluble components, and then the solubilized fraction is polymerized.
  • a solubilized collagen composition is prepared by grinding source vertebrate submucosa into a powder and enzymatically digesting the powderized submucosa with 0.1% w/v pepsin and solubilizing in 0.01 N HCl for one to three days at 4 0 C.
  • solubilized components of the solubilized submucosa composition are separated from the insoluble components by centrifugation at 12,000 rpm at 4 0 C for 20 minutes.
  • the supernatant, comprising the soluble components, is recovered and the pellet containing insoluble components is discarded.
  • the supernatant is then fractionated by dialyzing the solubilized submucosa composition against 0.01 N HCl.
  • the solubilized submucosa composition is dialyzed against several changes of 0.01 N hydrochloric acid at 4 0 C using dialysis membranes having a molecular weight cut-off of 3500.
  • the solubilized submucosa composition is fractionated to remove components having a molecular weight of less than about 3500.
  • dialysis tubing or membranes having a different molecular weight cut-off can be used.
  • the fractionated solubilized submucosa composition is then polymerized to produce the collagen based matrices of the present invention.
  • a solubilized collagen composition is prepared by grinding vertebrate submucosa into a powder and digesting the powderized submucosa composition with 0.1% w/v pepsin and solubilizing in 0.01 N hydrochloric acid for one to three days at 4 0 C.
  • the solubilized components are then separated from the insoluble components, for example, by centrifugation at 12,000 rpm at 4 0 C for 20 minutes.
  • the supernatant, comprising the soluble components is recovered and the pellet containing insoluble components is discarded.
  • the non-fractionated solubilized submucosa composition is then polymerized.
  • the present invention encompasses the formation of a solubilized collagen composition from a complex extracellular matrix material without purification of the matrix components.
  • the components of the naturally occurring extracellular matrices can be partially purified and the partially purified composition can be used in accordance with the methods described herein to prepare a solubilized collagen composition.
  • the solubilized collagen composition includes purified type I collagen or type I and type III collagen as the only protein constituents of the composition.
  • the solubilized collagen composition can be polymerized under different conditions to produce a collagen based matrix having the desired microstrutures and mechanical properties. Polymerization of purified type I collagen solutions at different concentrations of collagen affected fibril density while maintaining a relatively constant fibril diameter. In addition, both fibril length and diameter are affected by altering the pH of the polymerization reaction.
  • Additional conditions can be varied during the polymerization reaction to provide engineered purified collagen matrices that have the desired properties
  • the conditions that can be varied include pH, phosphate concentration, temperature, buffer composition, ionic strength, the extracellular matrix components in the solubilized extracellular matrix composition, and the concentration of solubilized extracellular matrix composition components (dry weight/ml). These conditions result in polymerization of the extracellular matrix components to form engineered extracellular matrices with desired compositional, microstructural, and mechanical characteristics.
  • these compositional, microstractural, and mechanical characteristics can include fibril length, fibril diameter, number of fibril-fibril connections, fibril density, fibril organization, matrix composition, 3-dimensional shape or form, viscoelastic, tensile, or compressive behavior, shear (e.g., failure stress, failure strain, and modulus), permeability, swelling, hydration properties (e.g., rate and swelling), and in vivo tissue remodeling and bulking properties, and desired in vitro cell responses.
  • the matrices described herein have desirable biocompatibility, vascularization, remodeling, and bulking properties, among other desirable properties.
  • qualitative and quantitative microstructural characteristics of the engineered matrices can be determined by environmental or cryostage scanning electron microscopy, transmission electron microscopy, confocal microscopy, second harmonic generation multi-photon microscopy.
  • polymerization kinetics may be determined by spectrophotometry or time-lapse confocal reflection microscopy.
  • tensile, compressive and viscoelastic properties can be determined by rheometry or uniaxial tensile testing.
  • a rat subcutaneous injection model can be used to determine remodeling and bulking properties.
  • the solubilized collagen composition is polymerized at a final total collagen concentration of about 1 to about 40 mg/ml, and in one embodiment about 1 to about 30 mg/ml, in another embodiment about 2 to about 25 mg/ml and in another embodiment about 5 to about 15 mg/ml.
  • the final total collagen is selected from a range of about 0.25 to about 5.0 mg/ml, or in another embodiment the final total collagen concentration is selected from the range of about 0.5 to about 4.0 mg/ml, and in another embodiment the final total collagen concentration is selected from the range of about 1.0 to about 3.0 mg/ml, and in another embodiment the final total collagen concentration is about 0.3, 0.5, 1.0, 2.0 or 3.0 mg/ml.
  • the components of the solubilized extracellular matrix composition are polymerized at final concentrations (dry weight/ml) of about 0.25 to about 10 mg/ml, about 0.25 to about 20 mg/ml, about 0.25 to about 30 mg/ml, about 0.25 to about 40 mg/ml, about 0.25 to about 50 mg/ml, about 0.25 to about 60 mg/ml, or about 0.25 to about 80 mg/ml.
  • the total collagen comprising the solubilized collagen composition comprises type I and type III collagen, wherein the percent range of the type III collagen and type I collagen is selected from about 17- 33% and about 66-83%, respectively, to achieve various collagen type VUl ratios.
  • percentage ranges of type III collagen and type I collagen, respectively that may be used in the matrices include 17% and 83%; 20% and 80%; 25% and 75%; 30% and 70%; and 33% and 66%, respectively.
  • the type I collagen to type III collagen ratio may be in the range of about 6: 1 to about 1:1.
  • Examples of the type I collagen to type III collagen ratios that may be used in the matrices include 6:1, 5:1, 4:1, 3:1, 2:1, 1.5:1, and 1:1.
  • At least 3 ug/ml of type I collagen is combined with at least 0.5 ug/ml of type III collagen to obtain a total amount of collagen.
  • Examples of the amount of type I collagen combined with type III collagen, respectively, that may be used in the matrices include 3 ug/ml and 0.5 ug/ml; 1500 ug/ml and 250 ug/ml; 1500 ug/ml and 500 ug/ml; 1500 ug/ml and 750 ug/ml; and 1500 ug/ml and 1500 ug/ml.
  • I collagen and type III collagen can be the same as those described above for the method of decreasing stiffness of an extracellular matrix composition.
  • the matrix compositions produced by the methods described herein can be combined, prior to, during, or after polymerization, with stem cells or progenitor cells, to further enhance the repair or replacement of diseased or damaged tissues.
  • progenitor cells include those that give rise to blood cells, fibroblasts, endothelial cells, epithelial cells, smooth muscle cells, skeletal muscle cells, cardiac muscle cells, multi-potential progenitor cells, pericytes, and osteogenic cells.
  • the population of progenitor cells can be selected based on the cell type of the intended tissue to be repaired.
  • the progenitor cells can produce cardiac muscle cells.
  • the matrix composition can also be seeded with autogenous cells isolated from the patient to be treated, hi an alternative embodiment the cells may be xenogeneic or allogeneic in nature. hi any of the embodiments described above using purified collagen, the purified collagen can be sterilized after purification, hi yet other embodiments, the collagen that is purified can be sterilized before or during the purification process, hi other embodiments, purified collagen can be sterilized before polymerization or the matrix can be sterilized after polymerization.
  • progenitor or stem cells to treat damaged tissues (including for example treating myocardial infarction followed by heart failure) has demonstrated early evidence of potential utility.
  • recent data has revealed three key issues that significantly limit successful delivery of reparative cells to tissues.
  • a novel cell delivery strategy involves the suspension of cells in a liquid-phase, injectable solubilized collagen composition that polymerizes in situ to form a three-dimensional (3D) matrix.
  • the 3D matrix is designed to both entrap cells and provide them with an "instructive" microenvironment which promotes cell survival and modulates their fate. It is anticipated that the introduction of cells in the presence of a comparatively viscous medium (i.e., the solubilized collagen composition, which will subsequently assemble in situ shortly after post-injection) will enhance the cells local retention.
  • the components of the 3D matrix and their microstructural organization play an important role in determining cell fate with respect to survival, proliferation, and differentiation.
  • the biophysical signals provided by a 3D self- assembled collagen microenvironment can be used to direct the proliferation and differentiation capacity of multi-potential, bone marrow-derived stem cells.
  • 3D purified collagen matrices characterized by a relatively high fibril density and stiffness supported an increase in clonal growth and enhanced osteogenesis (bone formation).
  • these results demonstrate the ability to engineer injectable, self-assembling 3D purified collagen matrices in which the composition, microstructure, and mechanical properties are defined and systematically varied with discrete outcomes.
  • the biophysical features of the 3D matrix in addition to cellular signaling modalities consisting of soluble factors and cell-cell interactions, are determinants of cell fate and represent a new target for therapeutic manipulation.
  • a method of enhancing the repair of damaged, diseased or congenital defective tissues comprises the steps of suspending a population of cells within a solubilized collagen composition, inducing the polymerization of the solubilized collagen composition, and injecting the composition into warm blooded species.
  • the composition is injected into a mammalian species, including a human for example, and in one embodiment the cells represent autologous, hi an alternative embodiment the cells may be xenogeneic or allogeneic cells.
  • the injected solubilized collagen composition polymerizes in vivo to form a 3D matrix with the population of cells embedded within the collagen matrix.
  • the population of cells comprise stem cells, hi one embodiment the soluble collagen composition comprises purified type I collagen, glucose, and calcium chloride.
  • a 3D purified collagen matrix is provided comprising collagen fibrils at a fibril area fraction of about 12% to about 25% (area of fibril to total area) that comprises glucose and CaCl 2 .
  • the solubilized collagen composition comprises about 0.05 mg/ml to about 5 mg/ml total purified collagen (either type I alone or a combination of type I and type III collagen) about 1.1 ImM to about 277.5mM glucose and about 0.2 mM to about 4.0 mM CaCl 2 .
  • the solubilized collagen composition comprises about 0.1 mg/ml to about 3 mg/ml total purified collagen (either type I alone or a combination of type I and type III collagen) in about 0.05 to about 0.005N HCl (and in one embodiment about 0.01N HCl), about 0.07M to about 0.28M NaCl (and in one embodiment about 0.137M NaCl), about 1.3 to about 4.5mM KCl (and in one embodiment about 2.7mM KCl) 5 about 4.0 to about 16mM Na 2 HPO 4 (and in one embodiment about 8.ImM Na 2 HPO 4 ), about 0.7 to about 3.OmM KH 2 PO 4 (and in one embodiment about 1.5mM KH 2 PO 4 ), about 0.25 to about 1.OmM MgCl 2 (and in one embodiment about 0.5mM M
  • Polymerization of the solubilized collagen composition is induced by the addition of a neutralizing solution such as NaOH.
  • a neutralizing solution such as NaOH.
  • a NaOH solution can be added to a final concentration of 0.0 IN NaOH.
  • the cells are then added to the composition after the addition of neutralizing solution, hi accordance with one embodiment a calcium chloride solution is also added to the solubilized collagen composition, hi this embodiment, calcium chloride is added to bring the final concentration OfCaCl 2 in the solubilized collagen composition to about 0.4 rnM to about 2.0 mM CaCl 2 (and in one embodiment about 0.9 mM CaCl 2 ).
  • composition is then allowed to polymerize either in vitro or in vivo to form a 3D matrix comprised of collagen fibrils wherein the cells are embedded within the 3D matrix.
  • polymerization reaction is conducted in a buffered solution using any biologically compatible buffer system known to those skilled in the art.
  • the buffer may be selected from the group consisting of phosphate buffer saline (PBS), Tris (hydroxymethyl) aminomethane Hydrochloride (Tris-HCl), 3-(N-Morpholino) Propanesulfonic Acid (MOPS), piperaziiie-n,n'-bis (2-ethanesulfonic acid) (PIPES), [n-(2-Acetamido)]-2-Aminoethanesulfonic Acid (ACES), N-[2-hydroxyethyl] piperazine-N 1 - [2-ethanesulfonic acid] (HEPES) and 1,3- bis[tris(Hydroxymethyl)methylamino]propane (Bis Tris Propane).
  • PBS phosphate buffer saline
  • Tris-HCl Tris (hydroxymethyl) aminomethane Hydrochloride
  • MOPS 3-(N-Morpholino) Propanesulfonic Acid
  • PPES piperazi
  • KH 2 PO 4 is varied. Ionic strength may be adjusted as an independent variable by varying the molarity of NaCl only.
  • the polymerization of the solubilized collagen composition is conducted at a pH selected from the range of about 6.0 to about 9.0, and in one embodiment polymerization is conducted at apH selected from the range of about 5.0 to about 11.0 and in one embodiment about 6.0 to about 9.0, and in one embodiment polymerization is conducted at a pH selected from the range of about 6.5 to about 8.5, in another embodiment polymerization of the solubilized collagen composition is conducted at a pH selected from the range of about 7.0 to about 8.0, and in another embodiment polymerization of the solubilized collagen composition is conducted at a pH selected from the range of about 7.3 to about 7.4.
  • the ionic strength of the buffered solution is also regulated.
  • the ionic strength of the solubilized collagen composition is selected from a range of about 0.05 to about 1.5 M, in another embodiment the ionic strength is selected from a range of about 0.10 to about 0.90 M, in another embodiment the ionic strength is selected from a range of about 0.14 to about 0.30 M and in another embodiment the ionic strength is selected from a range of about 0.14 to about 0.17 M.
  • the polymerization is conducted at temperatures selected from the range of about O 0 C to about 6O 0 C.
  • polymerization is conducted at temperatures above 2O 0 C, and typically the polymerization is conducted at a temperature selected from the range of about 2O 0 C to about 4O 0 C, and more typically the temperature is selected from the range of about 3O 0 C to about 4O 0 C. In one embodiment the polymerization is conducted at about 37 0 C.
  • the phosphate concentration is varied.
  • the phosphate concentration is selected from a range of about .005 M to about 0.5 M.
  • the phosphate concentration is selected from a range of about 0.01 M to about 0.2 M.
  • the phosphate concentration is selected from a range of about 0.01 M to about 0.1 M.
  • the phosphate concentration is selected from a range of about 0.01 M to about 0.03 M.
  • the solubilized collagen composition can be polymerized by, for example, dialysis against a solution under any of the above-described conditions (e.g., dialysis against PBS at pH 7.4), extrusion or co-extrusion of submucosa formulations into a desired buffer, including the buffers described above, or wet-spinning to form strands of extracellular matrix material.
  • the strands can be formed by extrusion of a solubilized collagen composition through a needle and can be air-dried to form threads.
  • the strands can be formed by extrusion through a needle and can be air-dried to form fibers or threads of various dimensions.
  • the syringe can be adapted with needles or tubing to control the dimensions (e.g., diameter) of the fibers or threads.
  • the extrusion process involves polymerization of the solubilized extracellular matrix composition followed by extrusion into a bath containing water, a buffer, or an organic solvent (e.g., ethanol).
  • the extrusion process involves coextrusion of the solubilized extracellular matrix composition with a polymerization buffer (e.g., the buffer such as Tris or phosphate buffers at various concentrations can be varied to control pH and ionic strength).
  • a polymerization buffer e.g., the buffer such as Tris or phosphate buffers at various concentrations can be varied to control pH and ionic strength.
  • the extrusion process involves extrusion of the solubilized extracellular matrix composition into a polymerization bath (e.g., the buffer such as Tris or phosphate buffers at various concentrations can be varied to control pH and ionic strength).
  • a polymerization bath e.g., the buffer such as Tris or phosphate buffers at various concentrations can be varied to control pH and ionic strength.
  • the bath conditions affect polymerization time and properties of the fibers or threads, such as mechanical integrity of the fibers or threads, fiber dimensions, and the like.
  • the extrusion of a solubilized collagen composition through a needle is used a method to control orientation of polymerized fibrils within the fibers.
  • the fibers can be air-dried to create materials that can be crosslinked or woven into three dimensional meshes or mats that can serve as a substrate, or a component of a substrate, for culturing cells.
  • engineered extracellular matrices can be polymerized from the solubilized extracellular matrix composition at any step in the above-described methods.
  • the engineered matrices can be polymerized from the solubilized extracellular matrix composition after the solubilization step or after the separation step, the filtration step, or the lyophilization and rehydration steps, if the separation step, the filtration step, and/or the lyophilization and rehydration steps are performed.
  • the engineered matrices can be combined, prior to, during, or after polymerization, with nutrients, including minerals, amino acids, pharmaceutical agents, sugars, peptides, proteins, vitamins (such as ascorbic acid), or glycoproteins that facilitate cellular proliferation, such as laminin and fibronectin, or growth factors such as epidermal growth factor, platelet-derived growth factor, transforming growth factor beta, or fibroblast growth factor, and glucocorticoids such as dexamethasone.
  • fibrillogenesis modulators such as alcohols, glycerol, glucose, or polyhydroxylated compounds can be added prior to or during polymerization.
  • cells can be added to the solubilized extracellular matrix composition as the last step prior to the polymerization or after polymerization of the matrix.
  • particulate extracellular matrix compositions can be added to the solubilized extracellular matrix composition and can enhance in vivo bulking capacity.
  • cross-linking agents such as carbodiimides, aldehydes, lysl-oxidase, N-hydroxysuccinimide esters, imidoesters, hydrazides, and maleimides, and the like can be added before, during, or after polymerization.
  • Hyaluronic acid is a glycosaminoglycan found naturally within the extracellular matrix. This mucopolysaccharide is made up of a repetitive sequence of two modified simple sugars, glucuronic acid and N-acetyl glucosamine. HA molecules are negatively charged and typically high in molecular weight (long in size). The size and charged nature of this molecule allow it to bind water to produce a high viscosity gel. When hyaluronic acid is added to soluble collagen compositions and the solubilized collagen compositions are allowed to polymerize, it appears that only subtle changes occur to the fibrillar microstructure of the resultant 3D matrix.
  • HA content represents a further variable of the present engineered 3D matrices that can be manipulated to provide an optimal microenvironment for cells cultured within the matrices.
  • the engineered purified collagen based matrices of the present invention can be used as cell culture substrates that more accurately mimic the substrates that various cells contact in vivo. Accordingly, collagenous based matrices can be designed for specific cell types to mimic their native environment.
  • stem cells or progenitor cells can be cultured in vitro without altering the fundamental cell behavior (e.g., cell proliferation, growth, maturation, differentiation, migration, adhesion, gene expression, apoptosis and other cell behaviors) of the cells
  • the engineered purified collagen based matrices of the present invention can be used to expand or differentiate a cell population, such a stem cell population (including pluripotent or unipotent cells), primary cells, progenitor cells or other eukaryotic cells by seeding the cells on, or within, the collagen based matrix and culturing the cells in vitro for a predetermined length of time under conditions conducive for that cell type's proliferation (i.e., appropriate nutrients, temperature, pH, etc.).
  • cells are added to the solubilized collagen composition as the last step prior to the polymerization of the solubilized collagen composition.
  • the engineered purified collagen based matrices of the present invention can be combined with nutrients, including minerals, pharmaceutical agents, amino acids, sugars, peptides, proteins, vitamins (such as ascorbic acid), or glycoproteins that facilitate cellular proliferation, such as laminin and fibronectin and growth factors such as epidermal growth factor, platelet-derived growth factor, transforming growth factor beta, or fibroblast growth factor, and glucocorticoids such as dexamethasone.
  • a sterilized engineered purified collagen based matrix may be seeded with living cells and packaged in an appropriate medium for the cell type used.
  • a cell culture medium comprising Dulbecco's Modified Eagles Medium (DMEM) can be used with standard additives such as non-essential amino acids, glucose, ascorbic acid, sodium pyruvate, fungicides, antibiotics, etc., in concentrations deemed appropriate for cell type, shipping conditions, etc.
  • DMEM Dulbecco's Modified Eagles Medium
  • the cell seeded engineered purified collagen based matrices of the present invention can be used simply for culturing cells in vitro, or the composition can be implanted or injected as a tissue graft construct to enhance the repair of damaged or diseased tissue.
  • an improved tissue graft construct is provided wherein the construct comprises a 3D purified collagen based matrix and a population of cells.
  • the 3D purified collagen based matrix is formed from a solubilized collagen composition wherein the solubilized composition is formed by contacting a source of purified collagen with an acid selected from the group consisting of hydrochloric acid, acetic acid, formic acid, sulfuric acid, ethanoic acid, carbonic acid, nitric acid, or phosphoric acid.
  • the solubilized collagen composition is then polymerized as described above to form the 3D purified collagen based matrix.
  • Cells and in one embodiment stem cells, are combined with the collagen based matrix at a low density and can be either added to the solubilized collagen composition prior to polymerization, or after formation of the collagen based matrix.
  • This initial seeded population of cells can be expanded by incubating the composition under conditions suitable for replication of the seeded cells.
  • cell seeded 3D purified collagen based matrices of the present invention comprise a population of cells that consists of, or are the progeny of, eukaryotic stem cells initially added to the composition at a low density, m one embodiment a tissue graft construct is prepared comprising the 3D purified collagen based matrices of the present invention that have been seeded with a low density of cells, wherein the cells are cultured within the matrix to expand and/or differentiate the seeded population of cells prior to implantation of the graft construct in a host, hi one embodiment the cells, and more particularly stem cells, are initially seeded within the 3D purified collagen matrix at a final concentration of about 10 to about 10 8 cells per milliliter, and in one embodiment at a final concentration of less than 10 5 cells per milliliter.
  • culturing conditions can be selected wherein a decreased seeding density of viable pluripotent or multipotent stem cells within an engineered purified collagen based matrix leads to clonal growth of cells representing a single cell lineage.
  • Such cells can be isolated and transferred to a second engineered purified collagen based matrix and conditions can be altered to enhance the proliferation of the isolated clonal population of cells.
  • Estimates of optimal cell densities for clonal growth range from about 10 cells/ml to about 10 3 cells/ml and depend upon the specific seeding efficiencies.
  • a method of isolating clonal populations of individual stem cells comprises the steps of contacting a source of collagen with hydrochloric acid to prepare a solubilized collagen composition. The solubilized collagen composition is then polymerized to form an engineered purified collagen based matrix.
  • the stem cells are seeded on or within the engineered purified collagen based matrix at a low density that maintains the functionality of the stem cells but allows for the isolation of clonal populations of cells.
  • the solubilized collagen composition is prepared having a type I collagen concentration selected from the range of about 1.0 to 3.0 mg/ml, and a pH of about 6.5 to about 7.0, wherein the solubilized collagen composition further comprises glucose and calcium chloride.
  • stem cells are seeded at a concentration selected from the range of from about 10 to about 10 3 cells per milliliter.
  • the source of collagen used to prepare the solubilized collagen composition comprises a purified preparation of type I collagen that has been dissolved in a hydrochloric acid solution.
  • the source of collagen comprises a hydrochloric acid solubilized fraction of a naturally occurring extracellular matrix, such as a submucosal matrix.
  • the solubilized collagen composition is prepared from vertebrate intestinal submucosa.
  • the hydrochloric acid solution used to prepared the solubilized collagen composition can be from about 0.005 N to about 0.1 N, from about 0.01 N to about 0.1 N, from about 0.05 N to about 0.1 N, from about 0.001 N to about 0.05 N, about 0.001 N to about 0.01 N, or from about 0.01 N to about 0.05 N HCl.
  • the solubilized collagen composition (purified collagen or extracellular matrix components) can be polymerized at final concentrations of collagen (dry weight/ml) of about 5 to about 10 mg/ml, about 5 to about 30 mg/ml, about 5 to about 50 mg/ml, about 5 to about 100 mg/ml, about 20 to about 50 mg/ml, about 20 to about 60 mg/ml, or about 20 to about 100 mg/ml.
  • the three-dimensional matrices may contain fibrils with specific characteristics, including, but not limited to, a fibril area fraction (defined as the percent area of the total area occupied by fibrils in a cross-sectional surface of the matrix; i.e., fibril density) of about 7% to about 26%, about 20% to about 30%, about 20% to about 50%, about 20% to about 70%, about 20% to about 100%, about 30% to about 50%, about 30% to about 70%, or about 30% to about 100%.
  • a fibril area fraction defined as the percent area of the total area occupied by fibrils in a cross-sectional surface of the matrix; i.e., fibril density
  • the three-dimensional matrices have an elastic or linear modulus (defined by the slope of the linear region of the stress-strain curve obtained using conventional mechanical testing protocols; i.e., stiffness) of about 0.5 kPa to about 40 kPa, about 30 IcPa to 100 kPa, about 30 IcPa to about 1000 kPa, about 30 kPa to about 10000 IcPa, about 30 IcPa to about 70000 kPa, about 100 IcPa to 1000 IcPa, about 100 IcPa to about 10000 IcPa, or about 100 IcPa to about 70000 IcPa.
  • an elastic or linear modulus defined by the slope of the linear region of the stress-strain curve obtained using conventional mechanical testing protocols; i.e., stiffness
  • a kit for preparing 3D matrices that have been optimized for a particular cell that is to be seeded within the formed 3D matrix.
  • the kit is provided with purified individual components that can be combined to form a solubilized collagen composition that upon polymerization forms a 3D matrix comprised of collagen fibrils that presents an optimal microenvironment for a population of cells.
  • the population of cells represent cells provided separately from the kit, but in one embodiment the cells may also constitute a component of the kit.
  • the cells are mammalian cells, including human cells, and in a further embodiment the cells are stem or progenitor cells.
  • a kit is provided comprising a solubilized collagen composition and a polymerization composition.
  • the solubilized collagen composition comprises purified type I collagen as the sole collagen component.
  • the solubilized collagen composition comprises purified type I collagen and type III collagen as the sole collagen components.
  • the kit comprises separate vessels, each containing one of the following components: purified type I collagen, a phosphate buffer solution, a glucose solution, a calcium chloride solution and a basic neutralizing solution.
  • the purified type I collagen of the kit is provided in a lyophilized form and the kit is further provided with a solution of HCl (or other dilute acid including for example, acetic acid, formic acid, lactic acid, citric acid, sulfuric acid, ethaiioic acid, carbonic acid, nitric acid, or phosphoric acid) for resuspending the lyophilized collagen.
  • HCl or other dilute acid including for example, acetic acid, formic acid, lactic acid, citric acid, sulfuric acid, ethaiioic acid, carbonic acid, nitric acid, or phosphoric acid
  • the kit is provided with a solution comprising a solubilized collagen composition, and in a further embodiment the solubilized collagen composition comprises a solubilized extracellular matrix composition.
  • the kit comprises a phosphate buffer solution, a glucose solution, a calcium chloride solution, and acid solution, a basic neutralizing solution, a vessel comprising purified type I collagen, and a vessel comprising purified type III collagen.
  • the polymerization composition comprises a phosphate buffer that has a pH of about 7.2 to about 7.6 and the acid solution is an HCl solution comprising about 0.05N to about 0.005N HCl, and in one embodiment the acid solution is about 0.0 IN HCl.
  • the glucose solution has a concentration selected from the range of about 0.2% to about 5% w/v glucose, or about 0.5% to about 3% w/v glucose, and in one embodiment the glucose solution is about 1% w/v glucose.
  • the CaCl 2 solution has a concentration selected from the range of about 2 mM to about 40.0 mM CaCl 2 or about 0.2 mM to about 4.0 mM CaCl 2 , or about 0.2 to about 2mM CaCl 2 .
  • the kit is provided with a 1OX PBS buffer having a pH of about pH 7.4, and comprising about 1.37M NaCl, about 0.027M KCl, about 0.08 IM Na 2 HPO 4 , about 0.015M KH 2 PO 4 , about 5mM MgCl 2 and about 1% w/v glucose.
  • the kit can further be provided with instructional materials describing methods for mixing the kit reagents to prepare 3D matrices.
  • the instructions materials provide information regarding the final concentrations and relative proportions of the matrix components that give optimal microenvironmental conditions including fibril microstructure and mechanical properties for a particular cell type or for a particular desired result (i.e., clonal expansion of cells, differentiation, etc.).
  • Small intestinal submucosa is harvested and prepared from freshly euthanized pigs as previously disclosed in U.S. Patent. No. 4,956,178.
  • Intestinal submucosa is powderized under liquid nitrogen and stored at -8O 0 C prior to use. Digestion and solubilization of the material is performed by adding 5 grams of powdered tissue to each 100 ml of solution containing 0.1% w/v pepsin in 0.01 N hydrochloric acid and incubating for 72 hours at 4 0 C. Following the incubation period, the resulting solubilized composition is centrifuged at 12,000 rpm for 20 minutes at 4 0 C and the insoluble pellet is discarded.
  • the supernatant is dialyzed against at least ten changes of 0.01 N hydrochloric acid at 4 0 C (MWCO 3500) over a period of at least four days.
  • the solubilized fractionated composition is then sterilized by dialyzing against 0.18% peracetic acid/4.8% ethyl alcohol for about two hours. Dialysis of the composition is continued for at least two more hours, with additional changes of sterile 0.01 N hydrochloric acid per day, to eliminate the peracetic acid.
  • the contents of the dialysis bags are then lyophilized to dryness and stored.
  • Small intestinal submucosa was harvested and prepared from freshly euthanized pigs as previously disclosed in U.S. Patent No. 4,956,178. Intestinal submucosa was powderized under liquid nitrogen and stored at -8O 0 C prior to use. Partial digestion of the material was performed by adding 5 g powdered tissue to each 100 ml solution containing 0.1% w/v pepsin in 0.01M hydrochloric acid and digesting for 72 hours at 4 0 C. Following partial digestion, the suspension was centrifuged at 12,000 rpm for 20 minutes at 4 0 C and the insoluble pellet discarded. The supernatant was lyophilized to dryness. EXAMPLE 3
  • lyophilized material from Example 2 consisting of a mixture of extracellular matrix components, was reconstituted in 0.01 N HCl.
  • reconstituted extracellular matrix solutions were diluted and brought to a particular pH, ionic strength, and phosphate concentration by the addition of a phosphate buffer and concentrated HCl and NaOH solutions. Polymerization of neutralized solutions was then induced by raising the temperature from 4 0 C to 37 0 C.
  • Various polymerization buffers including, e.g., phosphate buffers
  • the pH of the polymerization reaction was controlled by varying the ratios of mono- and dibasic phosphate salts.
  • Type I collagen prepared from calfskin was obtained from Sigma- Aldrich Corporation, St. Louis, MO, and dissolved in and dialyzed extensively against 0.01 M hydrochloric acid (HCl) to achieve desired concentrations.
  • Interstitial ECM was prepared from porcine small intestinal submucosa (SIS). SIS was powdered under liquid nitrogen and the powder stirred (5% w/v) into 0.01 N hydrochloric acid containing 0.1% (w/v) pepsin for 72 h at 4 0 C. The suspension was centrifuged at 12,000 xg for 20 min at 4 0 C to remove undissolved tissue particulate and lyophilized to dryness.
  • the lyophilized material was redissolved in 0.01 N HCl to achieve desired collagen concentrations.
  • each solution was diluted and brought to the specified pH, ionic strength, and phosphate concentration by the addition of a polymerization composition and concentrated HCl and NaOH solutions. Polymerization of neutralized solutions was induced by raising the temperature from 4 0 C to 37°C.
  • Various polymerization compositions were used to make final solutions with the properties shown in Table 2.
  • ECMs representing purified type I collagen or a complex mixture of interstitial ECM components (SIS) were prepared at varied pH (series 1), ionic strength (series 2), and phosphate concentration (series 3).
  • [C] represents collagen concentration in mg/ml
  • [Pj] represents phosphate concentration in M
  • I represents ionic strength in M.
  • Figs. 1 A-IG Representative data showing the results of varying the polymerization temperature, buffer system, pH (using either a phosphate or tris buffer), ionic strength, phosphate concentration or concentration of ECM material, on stiffness (elastic modulus) of the formed 3D matrix is presented in Figs. 1 A-IG.
  • the polymerization temperature is increased from 4 0 C up to 37 0 C, the polymerization rate and the stiffness of the formed 3D matrix increases.
  • the effect of a temperature gradient profile on the microstructural composition of the 3D matrix was also investigated.
  • EXAMPLE 5 Quantification of Fibril Properties from Three Dimensional Images Quantification of the fibril diameter distribution within engineered extracellular matrices was conducted based upon two- and three- dimensional image sets obtained via electron and confocal microscopy techniques using methods described within Brightman et al., Biopolymers 54:222-234, 2000. More recently, a Matlab program with a graphical user interface was written for measurement of fibril diameters from these images. For three-dimensional confocal images, depth attenuation was corrected by normalizing against a fitted logarithmic curve, after which images were binarized into white and black pixels using a threshold value. Three rectangles were outlined in the x-y plane across each fibril, with one axis aligned with the fibril.
  • Average fibril diameter in each rectangle was calculated as the total white area divided by the rectangle's length.
  • the average diameter of each fibril was taken to be the average of the three measurements, and the average diameter in a given matrix was calculated as an average of all measurements.
  • Length of fibril per volume was estimated by dividing the total white volume of an image by the average cross-sectional area of fibrils in that image. Due to distortion in the z-plane, the fibril cross-sections in the image could not be assumed circular and calculated from diameter. Rather, the average cross-sectional area was found by expanding the rectangles described above into three-dimensional boxes. The cross-sectional area of a fibril in was found by dividing the total white volume contained in the box by the length of the box's axis aligned with the fibril.
  • a Matlab program has also been developed to determine fibril density from two- and three- dimensional images. This method involves thresholding and binarizing the image data to discriminate fibrils from the background. The surface area or volume representing fibrils is then quantified and normalized to the surface area or volume of the image.
  • Small intestinal submucosa was harvested and prepared from freshly euthanized pigs as previously disclosed in U.S. Patent No. 4,956,178. Intestinal submucosa was powderized under liquid nitrogen and stored at -8O 0 C prior to use. Digestion and solubilization of the material was performed by adding 5 grams of powdered tissue to each 100 ml of solution containing 0.1% pepsin in 0.01 N hydrochloric acid and incubating with stirring for 72 hours at 4 0 C. Following the incubation period, the solubilized composition was centrifuged at 12,000 rpm for 20 minutes at 4 0 C and the insoluble pellet was discarded.
  • the supernatant was dialyzed extensively against 0.01 N HCl at 4 0 C in dialysis tubing with a 3500 MWCO (Spectrum Medical Industries). Polymerization of the solubilized extracellular matrix composition was achieved by dialysis against PBS, pH 7.4, at 4 0 C for about 48 hours. The polymerized construct was then dialyzed against several changes of water at room temperature and was then lyophilized to dryness.
  • the polymerized construct had significant mechanical integrity and, upon rehydration, had tissue-like consistency and properties.
  • glycerol was added prior to polymerization by dialysis and matrices with increased mechanical integrity and increased fibril length resulted.
  • EXAMPLE 9 Preparation of Extracellular Matrix Threads
  • Small intestinal submucosa was harvested and prepared from freshly euthanized pigs as previously disclosed in U.S. Patent No. 4,956,178.
  • Intestinal submucosa was powderized under liquid nitrogen and stored at -8O 0 C prior to use. Digestion and solubilization of the material was performed by adding 5 grams of powdered tissue to each 100 ml of solution containing 0.1% w/v pepsin in 0.01 N hydrochloric acid and incubating for 72 hours at 4 0 C. Following the incubation period, the solubilized composition was centrifuged at 12,000 rpm for 20 minutes at 4 0 C and the insoluble pellet was discarded.
  • the solubilized extracellular matrix composition (at 4 0 C) was placed in a syringe with a needle and was slowly injected into a PBS solution at 4O 0 C.
  • the solubilized extracellular matrix composition immediately formed a filament with the diameter of the needle. If a blunt-tipped needle is used, straight filaments can be formed while coiled filaments can be formed with a bevel-tipped needle. Such filaments can be used as resorbable sutures.
  • solubilized extracellular matrix compositions were dialyzed (MWCO 3500) extensively against water and 0.01 M acetic acid to determine the effects of these media on the lyophilization product. Aliquots of the solubilized extracellular matrix composition in 0.01 M acetic acid were created and glacial acetic acid (17.4 M) was added to create a range of concentrations from 0.01 to 0.5 M acetic acid.
  • the solubilized extracellular matrix compositions were frozen using a dry ice/ethanol bath and lyophilized to dryness. The lyophilized preparations were observed, weighed, and dissolved at 5 mg/ml in either 0.01 N HCl or water. The dissolution and polymerization properties were then evaluated. The results are shown in Tables 2-6. Table 3. Gross appearance of solubilized extracellular matrix compositions following lyophilization at various hydrochloric acid concentrations.
  • the SIS/pepsin solution When removed from stirring, the SIS/pepsin solution should appear viscous and somewhat uniform. Pour SIS/pepsin solution into centrifuge jars. Balance jars as necessary. 2.2. This mixture should be centrifuged at 16,000 x g for 30 minutes at 4°C.
  • the tube should be full and taut.
  • dialysis tubing filled with SIS should be removed from the HCl.
  • SIS should be refrigerated until use.
  • the condenser is the metal cylinder which opens on the front of the lyophilizer.) Ensure that the black rubber collection tubing attached to the bottom of the condenser is plugged. This can be accessed by opening the grate on the front of the lyophilizer.
  • the jar may be placed in a freezer until all material is solid. In a -8O 0 C freezer, this takes about 30 minutes.
  • More jars may be added to freeze-dry simultaneously, but add jars one or two at a time. Wait until the vacuum pressure falls to a reasonable range (e.g., 200 millitorr) to ensure that the last jar is sealed before adding subsequent jars.
  • a reasonable range e.g. 200 millitorr
  • Llyophilized material is not immediately used, it should be stored in a dry environment. Use a large, sealable container with Dri-Rite or another desiccant, and place containers of lyophilized material therein.
  • Mixing may be accelerated by shaking, stirring, etc. Store container under refrigeration until dissolution of SIS is complete.
  • dialysis tubing filled with solubilized SIS should be removed from the dialysis tank aseptically. 4. Remove dialysis clips and pour or pipette solubilized SIS into a sterile jar.
  • the disinfected solubilized SIS should be stored at 4°C until use.
  • dialysis tubing filled with solubilized SIS should be removed from the dialysis tank aseptically.
  • the disinfected solubilized SIS should be stored at 4°C until use.
  • ECM extracellular matrix
  • the three-dimensional (3D) extracellular matrix (ECM) of tissues in vivo represents a complex array of macromolecules that serves to provide biochemical and biophysical microenvironmental cues to resident cells.
  • ECM extracellular matrix
  • the exact role of any one biophysical feature or molecular component within the ECM in regulating cellular behavior has been difficult to elucidate due to the inherent interdependence of ECM compositional, structural, and mechanical properties.
  • the 3D microstructural composition of fibrils within engineered ECMs created from purified type I collagen regulates cell-matrix adhesion, matrix remodeling, and proliferation properties of fibroblasts. It is further anticipated that altering the ratios of collagen types I and III within engineered ECMs would affect the hierarchical assembly of fibrils, and therefore the ECM signaling capacity.
  • a multipotential mesenchymal stem cell line (Dl) derived from mouse bone-marrow stroma was obtained from American Type Culture Collection (ATCC). Dl cells were propagated in Dulbecco's modified Eagle medium containing 4.5 g/L glucose, 110 mg/L sodium pyruvate, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin, and 10% fetal bovine serum (FBS) within a humidified atmosphere of 5% carbon dioxide at 37 0 C. Three-dimensional collagen ECMs were prepared by dissolving native, acid-solubilized type I collagen from calf skin (Sigma Chemical Co, St. Louis, MO) in 0.01 N hydrochloric acid to achieve desired concentrations.
  • the isolated collagen obtained from Sigma Chemical was dialyzed against an acidic solution having a low ionic strength (0.01 N HCl) for 1-2 days, using dialysis tubing or a membrane having a molecular weight cut-off selected from a range of about 3,500 to about 12,000 daltons.
  • the purified collagen solution was layered onto a volume of chloroform. After incubation for 18 hours at 4 0 C, the collagen solution layer was carefully removed so as not to include the collagen-chloroform interface layer.
  • collagen solutions were polymerized under different conditions. Specifically, to create collagen matrices consisting of collagen fibrils at increasing densities, collagen solutions were polymerized at final collagen concentrations of 1.0 to 3.0 mg/ml.
  • the polymerization composition comprised a 1OX phosphate buffered saline (PBS) with an ionic strength of 0.14 N and a pH of 7.4.
  • PBS 1OX phosphate buffered saline
  • the specific formulation of the 1OX phosphate buffer is as follows:
  • solubilized collagen e.g., type I collagen
  • the composition is mixed well after each additional component is added.
  • the composition is then combined with a cell pellet of known cell number to create desired cell density; mixed well; and allowed to polymerize.
  • the resulting polymerized 3D matrix has a final concentration of glucose and CaCl 2 of about 5.55mM glucose and about 0.9046mM CaCl 2 .
  • collagen solutions were polymerized at a pH selected from the range of 6.5-8.5.
  • Dl cells were harvested in complete medium, collected by centrifugation, and added as the last component before polymerization.
  • Tissue constructs were prepared at a relatively low cell density of 5 * 10 4 cells/ml. Previous studies by applicants have shown that this cell density is suitable for maintaining cell viability, minimizing cell-cell interaction, and allowing the study of the dynamic relationship between an individual cell and its surrounding ECM.
  • tissue constructs were cultured for 48 hours at 37 0 C in a humidified environment consisting of 5% CO 2 in air. After 48 hours, each of the constructs comprising Dl cells seeded within a specific ECM microstructure were cultured under 3 different conditions: 1) complete medium no supplements
  • the results of this experiment revealed the following: 1) multipotential stem cells seeded within engineered ECMs proliferated at rates that were dependent upon microstructural composition of the engineered ECM and the media composition; 2) time-dependent patterns of cellular condensation and aggregation exhibited by multipotential stem cells were dependent upon microstructural composition of the engineered ECM and the media composition;
  • Ll A unipotential stem (precursor) cell line (Ll) derived from mouse and representing pre-adipocytes was obtained from American Type Culture Collection (ATCC). Ll cells were propagated in Dulbecco's modified Eagle medium containing 4.5 g/L glucose, 110 mg/L sodium pyruvate, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin, and 10% fetal bovine serum (FBS) within a humidified atmosphere of 5% carbon dioxide at 37 0 C. To enhance cell viability, cells representing passage numbers greater than 5 were maintained in complete media in which the penicillin and streptomycin were reduced to 25 U/ml and 25 ⁇ g/ml, respectively.
  • ATCC American Type Culture Collection
  • tissue constructs representing Ll cells seeded within 3D engineered ECMs of different microstructural compositions were carried out as described in Example 13. Immediately after polymerization (20 minutes or less), complete medium was added and the tissue constructs were cultured for 48 hours at 37 0 C in a humidified environment consisting of 5% CO 2 in air. After 48 hours, each of the constructs comprising Ll cells seeded within a specific ECM microstructure were cultured under 3 different conditions:
  • the differentiation medium consists of DMEM supplemented with 10% FBS, 25 U/ml penicillin, 25 ⁇ g/ml streptomycin, 115 ⁇ g/ml methyl-isobutyl xanthine, 10 ⁇ g/ml insulin, and 5 x 10 "7 M dexamethasone.
  • the post differentiation medium consisted of DMEM supplemented with 10% FBS, 25 U/ml penicillin, 25 ⁇ g/ml streptomycin, and 10 ⁇ g/ml insulin.
  • parallel experiments were conducted on Ll cells grown in a standard 2D format on tissue- culture plastic. Cell behavior and morphology were monitored throughout the duration of the experiment using standard brightfield microscopy.
  • Multi-potential stem cells derived from the bone marrow of mice were suspended at 5 x 10 4 cells/ml within purified type I collagen solutions (Sigma Chemical Co.) at varying collagen concentrations ranging from 1.5-3.6 mg/ml using the procedures described in Example 13.
  • Tissue constructs consisting of Dl cells entrapped within a 3D ECM were formed by inducing self-assembly (polymerization) at pH 7.4, 137 mM NaCl, and 37 0 C.
  • an increase in collagen concentration as a self-assembly parameter was used to generate a 3D ECM microenvironment in which the density of the resultant fibrils and stiffness (linear or elastic modulus) of the matrix were systematically increased.
  • the 3D constructs and resident cells were maintained in one of three different media formulations (Table 8) at 37 0 C in a humidified environment consisting of 5% CO 2 in oxygen for periods of time up to 4 weeks.
  • Basal medium consisted of Dulbecco's modified Eagle's medium supplemented with 4 mM L-glutamine, 4.5g/L glucose, 1.5 g/L sodium bicarbonate, 1 mM sodium pyruvate, 10% fetal bovine serum, 100 U/ml > penicillin, and 100 ⁇ g/ml streptomycin.
  • Dl cells also were cultured in a parallel fashion in the standard 2D format on the surface of tissue culture plastic.
  • the proliferative and differentiation status of the cells were determined qualitatively or quantitatively. Qualitative evaluation of cell number and morphology was conducted several times a week using light microscopy. Real-time RT-PCR was used to quantify and compare the expression levels of CFBAl (runx2), LPL (lipoprotein lipase), and procollagen II as indicators of osteogenesis (bone formation), adipogenesis (fat formation), and chondro genesis (cartilage formation), respectively. Histochemical stains, including alkaline phosphatase and oil red O, were applied to whole mount or cryosectioned samples to detect osteogenic and adipogenic activity, respectively, in some cases immunohistochemical staining was used to corroborate results.
  • medium formulation A demonstrated a high frequency of adipogenesis on plastic and within 3D ECMs of low fibril density and stiffness (1.5 mg/ml).
  • fibril density and stiffness of the 3D ECM microenvironment increased, adipogenesis events decreased and osteogenesis increased.
  • Medium formulation B appeared to support differentiation of Dl cells into fat (adipogenesis) and (bone) osteogenesis on plastic. Limited areas of osteogenesis and adipogenesis were noted amongst a large number of spindle-shaped cells for Dl cells grown within ECMs of low stiffness under these same medium conditions.
  • stiffness of the 3D ECM increased, cells more uniformly developed regional areas of osteogenesis and myo genesis-like events.
  • a 2D projection of one confocal image revealed cells organized or fused to form a multi-cellular structure reminiscent of a myotube. These events were limited to 3D ECM microenvironments of high stiffness (3.4 mg/ml and greater). While these myotube-like events were noted in all three medium formulations, they appeared to occur more frequently in medium formulations B and C. The cells of the myotube-like structure were stained immunohistochemically for F-actin to demonstrate the fusion of and connectivity of the actin cytoskeleton between individual cells.
  • Real-time RT-PCR confirmed that biophysical features of the 3D ECM microenvironment (e.g., fibril density and ECM stiffness) could be modulated to regulate stem cell growth and differentiation.
  • the tissue specific genes CBFAl (runx2), LPL (lipoprotein lipase), and Pro Col II (procollagen II) were selected as indicators of osteogenesis, adipogenesis, and chondrogenesis, respectively.
  • Results showed that cells grown for 2 weeks on 2D plastic in the basal medium (no additives) remain relatively undifferentiated, more specifically, limited expression of the osteogenic, adipogenic, and chondrogenic indicators.
  • Dl cells show an increase in LPL (adipogenesis) when cultured on plastic in the presence of Medium A or Medium B.
  • the expression of LPL correlates well with the observed fat cell morphology developed within the cultures.
  • the gene expression patterns developed by cells grown within a 3D ECM microenvironment were dramatically different from those observed for cells grown on plastic.
  • the expression of CBFAl, indicative of osteogenesis could be enhanced by growing the cells within 3D ECMs of increased stiffness or Medium B.
  • the increased expression of CFBAl correlated well with cell morphologies and histochemical staining.
  • chondrogenesis events as indicated by high procollagen II expression appeared to be enhanced within Dl cells cultured in 3D ECMs of high stiffness.
  • the starting cell density was also a critical determinant of the stem cell fate within the 3D culture formats studied.
  • NHDFs human dermal fibroblasts
  • FBS fetal bovine serum
  • 3D Engineered ECMs and 3D Tissue Constructs Purified type I and type III collagens, that were solubilized from bovine dermis and human placenta, respectively, were obtained from Sigma Chemical Company (St. Louis, MO). Three-dimensional engineered ECMs were prepared at a constant collagen type I concentration of 1.5 mg/ml and type III collagen concentrations of either 0, 0.25, 0.50, and 0.75 mg/ml (Table 9) using the general procedures described in Example 13.
  • the polymerization buffer consisted of 1OX phosphate buffered saline (PBS) with an ionic strength of 0.14 M and a pH of 7.4.
  • AU 3D engineered ECMs and tissue constructs were polymerized in vitro within a humidified environment at 37 0 C.
  • 3D tissue constructs were formed by first harvesting NHDFs in complete media and then adding the cells as the last component to the collagen solutions prior to polymerization. Tissue constructs were prepared at a relatively low cell density of 5xlO 4 cells/ml in order to minimize cell-cell interactions. Immediately after polymerization (20 minutes or less), complete medium was added and the tissue constructs were maintained at 37 0 C in a humidified environment consisting of 5% CO 2 in air.
  • Table 9 Summary of formulations for 3D engineered ECMs prepared with varied ratios of collagen types I and III.
  • 3D ECM Microstructural Composition Two quantitative parameters describing the 3D ECM microstructural composition, fibril area fraction (a 2D approximation of 3D fibril density) and fibril diameter, were determined based upon confocal reflection and scanning electron microscopy (SEM) images. Prior to microstructural analysis, engineered 3D ECM constructs were polymerized within four-well Lab-Tek coverglass chambers (Nalge Nunc International, Rochester, NY) and placed within a humidified environment at 37 0 C where they were maintained for approximately 15 hours.
  • the confocal microscope was used to obtain high resolution, 3D, reflection images of the component collagen fibrils within each ECM (Brightman at al., Biopolymers 54: 222-234, 2000; Voytik-Harbin et al., Methods Cell Biol 63: 583- 597, 2001). Three images (at least 10 ⁇ m in thickness) were taken at random locations within each of 2 specimens representing a given 3D ECM composition. The confocal image stacks were then read into Matlab (The Mathworks, Natick, MA), and 2D projections, representing 21 z-sections, of each image were created and a threshold chosen for binarization.
  • Matlab The Mathworks, Natick, MA
  • engineered ECM constructs were fixed in 3% glutaraldehyde in 0.1M cacodylate at pH 7.4, dehydrated with ethanol, and critical point dried. Samples were sputter-coated with gold/palladium prior to imaging. Duplicate samples were imaged in a JEOL (Peabody, MA) JSM-840 SEM using 5 IcV accelerating voltage and a magnification of 3,00OX. Digital images were captured using 1280x960 resolution and 160 second dwell time. From each image obtained from duplicate samples, forty fibrils were chosen at random (10 fibrils per quadrant).
  • ECM constructs were loaded uniaxially at an extension rate of 1 mm/min (corresponding to a strain rate of ⁇ - 0.04 /min) until failure. Images were collected at a rate of 0.1 frames/sec to provide sequential images at 0.64% strain intervals.
  • F was the force recorded by the Minimat and A 0 was the initial cross-sectional area (width x thickness) within the center of the specimen (Callister et al., Materials Science and Engineering: An Introduction. 3rd edition. New York, NY: John Wiley & Sons, 1994).
  • true stress the actual cross-sectional area of each specimen at a specific load was imaged, quantified, and substituted for A 0 in the engineering stress equation above. From the resulting stress-strain relationships ultimate strength (maximum stress achieved during tensile loading), failure strain (strain at which specimen fails), and linear or elastic modulus (stiffness; slope of linear region of stress-strain curve) were determined.
  • the confocal microscope was used in a reflection (back-scattered light) mode to obtain image stacks of an individual cell and the component collagen fibrils of its surrounding ECM as described previously (Brightman et al., Biopolymers 54: 222-234, 2000; Voytik- Harbin et al., Methods Cell Biol 63: 583-597, 2001). Images were collected at 30- minute intervals and a z-step of 0.5 ⁇ m to minimize exposure of the tissue constructs to radiation from the confocal microscope laser.
  • the processed image stack was used to determine fundamental morphological parameters including number of cytoplasmic projections, cell volume, 3D cell surface area, length, width, and height as described previously (Pizzo et al., JAppl Physiol 98: 1909-1921, 2005). Since each cell had a relatively unique orientation within the 3D matrix, these morphological parameters were defined based on a cellular coordinate system. Morphological evaluation was conducted on a total of 10 to 23 cells for each of the 3D ECM compositions studied. EXAMPLE 21
  • Equation (3) where ⁇ j j are average strains in the confocal coordinate system directions.
  • This average strain matrix in Equation (3) was then solved using eigenvector analysis (Strang et al., Linear Algebra and Its Applications. 3rd edition. San Diego, CA: Academic Press, 1988) to determine 3 average principal strains (E / , E 2 , Es) and associated directions such that,
  • [V] is a 3 x 3 matrix such that the column vectors (Vj, V 2 , Vi) are the directions of the principal strains given by
  • each cell had a unique set of average principal strains and directions in 3D.
  • Another analysis was performed to determine on a finer scale where the maximum local principal strains within the 3D ECM occurred in relationship to the cell. This analysis involved determination of local principal strains E 1 , E 2 , and E 3 , each with unique principal direction, at each of the 15 x 15 x 3 grid points. The maximum compressive E 1 , E 2 , and E 3 were then identified within the image volume. The location of each maximum compressive principal strain was known in terms of its IDVC grid location and also in ⁇ m. The distance from these three-maximum principal strain locations to the center of the cell body in 3D could then be determined using simple vector relationships. The locations of the maximum compressive principal strains did not necessarily occur at the same grid locations for each cell.
  • Tissue constructs formed by seeding NHDFs within specific 3D ECM formulations during polymerization were prepared in four-well Lab-Tek coverglass chambers (Nalge Nunc International, Rochester, NY) for visualization of the F- actin cytoskeleton.
  • constructs were fixed and permeabilized with a solution containing 0.1% Triton IOOX and 3% paraformaldehyde, post-fixed in 3% paraformaldehyde, and treated with 1% bovine serum albumin to minimize non- specific binding.
  • the constructs were then stained overnight at 4 0 C with Alexa Fluor 488 Phalloidin (Molecular Probes, Eugene, OR) and rinsed.
  • each well and tissue construct was examined microscopically to observe the viability, number, and morphology of the cells.
  • the medium from each well then was replaced with fresh medium containing the metabolic indicator dye alamarBlue (10% v/v; BioSource International, Inc., Camarillo, CA).
  • dye reduction was monitored spectrofiuorometrically using a FluoroCount Microplate Fluorometer (Packard Instruments, Meriden, CT) with excitation and emission wavelengths of 560 nm and 590 nm, respectively. Background fluorescence measurements were determined from wells containing only dye reagent in culture medium.
  • fibril diameter measurements made from confocal reflection images corroborated SEM results; however, fibril diameter values obtained from confocal images were greater than those obtained using SEM (Table 10) since confocal imaging was conducted on unprocessed, fully hydrated specimens. It should be noted that fibril diameter measurements made using confocal reflection imaging were considered somewhat less accurate and less precise since fibril diameters were near the limit of resolution for this imaging technique.
  • Table 10 Collagen fibril diameter measurement data for 3D engineered ECMs prepared from type I collagen in the absence and presence of type III collagen as determined from scanning electron (SEM) and confocal reflection (CRM) images.
  • ECMs engineered from type I collagen in the presence of type III collagen over the range of 0 to 0.75 mg/ml showed biphasic responses in terms of true stress calculated parameters ultimate strength and stiffness.
  • the mean ultimate strength obtained for ECMs prepared from 1.5 mg/ml type I collagen alone was 136.7 ⁇ 49.9 kPa.
  • ECMs prepared at 3 mg/ml type I collagen had ultimate strength and stiffness values that were 2.2 and 3.5 times, respectively, those obtained for ECM prepared at 1.5 mg/ml type I collagen.
  • the ability of cells to sense and respond to changes in the 3D ECM microenvironment that resulted from the addition of type III collagen initially was assessed by determining and comparing 3D cell morphology and cell-induced ECM remodeling (deformation and reorganization of component collagen fibrils).
  • Three- dimensional morphometric analyses for cells seeded within the different ECM microenvironments were conducted at 6 and 12 hours following tissue construct formation. ECM remodeling by individual cells was repeatedly monitored during a 5 to 6 hour time window shortly after construct formation.
  • the collagen type I/III ratio also affected the ability of individual cells to deform and reorganize the component collagen fibrils of their surrounding ECM.
  • Repeated monitoring of interactions between a cell and its surrounding collagen fibrils within a live tissue construct by confocal reflection microscopy provided a means of visualizing and quantifying this response over a 5 to 6 hour time window.
  • An IDVC algorithm (Roeder et al., JBiomech Eng 126: 699-708, 2004) was applied to consecutive confocal image stacks and used to determine 3D displacements and strains as they occurred locally to a given cell and adjacent collagen fibrils.
  • Data generated from this algorithm provided the basis for 1) quantification of volumetric strain induced by a single cell within a tissue construct; 2) a detailed analysis of average local principal strains for each imaged volume; and 3) determination of magnitudes and locations for points within the image volume where maximum principal strains, Ei, E 2 , and E 3 , occurred. This data was then compiled and used to compare the mechanical status of a large number of individual cells grown within the different ECM formulations.
  • Results showed that cells grown in ECMs containing type III collagen were less able to contract and remodel the surrounding matrix as the type III collagen content increased or type I/III ratio decreased.
  • Qualitative perspectives and corresponding volumetric strain data obtained for representative cells grown within type I collagen ECMs prepared with low (0.25 mg/ml) and high (0.75 mg/ml) type III collagen concentrations are shown (Fig. 4).
  • type III collagen containing ECMs with total collagen contents of 1.75 mg/ml to 2.25 mg/ml were characterized by 3D average local principal strain levels that were about 2 to 3 times greater than ECMs prepared of type I collagen alone and a total collagen content of 1 mg/ml.
  • ECMs prepared with a type III collagen content of 0.75 mg/ml generated maximum principal strains at distances of only 15-25 ⁇ m from the center of the cell.
  • type III collagen enabled cells to induce large principal strains within their ECMs, the distance at which maximum principal strain occurred was considerably less than that found for ECMs prepared at low levels of type I collagen alone (Fig. 6). More specifically, ECMs prepared at 1 mg/ml type I collagen yielded, on average, points of maximum principal strain for 1-, 2-, and 3- directions at distances of 48 ⁇ m, 45 ⁇ m, and 52 ⁇ m from the center of the cell, respectively.
  • ECMs containing a few scattered actin filaments were observed in ECMs prepared from type I collagen alone, but only at low collagen concentrations of 1.5 mg/ml and below. Cells with diffuse actin staining patterns were noted within ECMs prepared at collagen I levels greater than 1.5 mg/ml. Diffuse actin staining patterns were observed for cells grown in engineered ECMs representing type I collagen ECMs prepared at concentrations of 1 mg/ml and 3 mg/ml.
  • Fibroblast proliferation was enhanced in ECMs with increased type III collagen content (Fig. 7A). Since the type I collagen content was kept constant, increasing the amount of type III collagen also increased the overall collagen content. Although the total number of cells within all ECM formulations increased between 24 and 48 hours, the total number of fibroblasts was greatest for ECMs prepared with the highest type III collagen concentration for both time points. When type III collagen was added at levels below 0.25 mg/ml, in the range of 0.02 mg/ml to 0.10 mg/ml, the proliferative capacity of the resident cells was lower than that obtained for 1.5 mg/ml type I collagen alone.
  • type III collagen affected not only microstructural-mechanical properties but also the macromolecular composition of the engineered ECMs, it was uncertain if changes in NHDF proliferation were a result of differences in biophysical or biochemical signals (cues) inherent in the 3D ECM microenvironments.
  • traditional experimental methods involving creation of 2D ECM surface coatings consisting of varied collagen I/III ratios to evaluate cell-ECM interactions were applied.
  • NHDF were seeded onto the ECM-coated surfaces and proliferation monitored. No significant difference was observed in cell proliferation due to type III collagen content at either the 24- or 48-hour time points (Fig. 7B).
  • AU coatings showed a significant increase (p ⁇ 0.05) in cell number between the 24- and 48-hour time points.
  • cells seeded on plastic showed significantly greater proliferation than those seeded on any of the ECM coated surfaces (p ⁇ 0.05).
  • ASC human adipose-derived stem cells
  • GM-CSF granulocyte- macrophage colony stimulating factor
  • VEGF vascular endothelial growth factor
  • HGF hepatocyte growth factor
  • bFGF transforming growth factor- ⁇
  • TGF- ⁇ transforming growth factor- ⁇
  • the survival, proliferation, and differentiation properties of human APC and EPC cells implanted within three dimensional matrices will be investigated using both standard cell culture media or by suspension in any of the formulations of "ready-to-assemble" components of self-assembling 3D matrix microenvironments, in which the microstructure, composition, and mechanical properties are quantified and systematically varied.
  • the delivery efficiency and subsequent engraftment (cell survival and differentiation) of human ASC or endothelial progenitor cells derived from human cord blood (EPC) implanted within an animal model of hindlimb muscle ischemia will also be investigated. More particularly, cells will be delivered with or without injectable 3D matrix microenvironments in which the "instructive" or signaling properties are controlled and systematically varied .
  • ASC adipose-derived stem cells
  • EPC human cord blood
  • 3D ECM microenvironments in which specific biophysical features including fibril density, length, and width and stiffness are systematically varied will be created from purified collagen as described previously [Pizzo et al., 2005, J. App. Phys. 98:1909-1921; Roeder et al., 2002 J. Biomech Eng. 124: 214- 22213,17].
  • composition is systematically varied by including ECM molecules such as type III collagen, hyaluronic acid, VEGF, bFGF will also be investigated. These molecules were chosen based upon their known role in neovascularization and cardiac muscle development.
  • cells will be added as the last component of the solubilized collagen matrix and the suspension will be injected over 30 seconds through a 25Ga needle (paralleling intramuscular injection for in vivo systems) into a well plate and polymerized at 37°C. Immediately following polymerization (less than 30 minutes), complete medium will be added to all constructs.
  • cell seeding densities ranging between 1x10 5 to 1x10 7 cells/ml will be evaluated.
  • Fundamental cell behaviors including survival, morphology, proliferation, and differentiation will be determined using techniques established previously [Pizzo et al., 2005, J. App. Phys. 98:1909-1921].
  • cells will be prelabeled with CellTracker dyes or transfected with GFP and analyzed in 3- or 4-dimensions using confocal microscopy in a combination reflection- fluorescence mode. Outcomes will be compared to those from control "deliveries" in which cells are injected into media within culture plates, parallel to the situation for cell injection into a tissue environment in the absence of a solubilized, self-assembling matrix.
  • the 3D cell containing matrices will be implanted via injection into either normal or ischemic muscle in vivo, using the hindlimb model of muscle ischemia that the March lab has established and published in the preliminary findings concerning adipose stem cells [Rehman et al., 2004, Circulation 109: 1292-8]. Briefly, nude mice are employed so that cells of human origin can be studied in the absence of xenogeneic barriers. The ilio-femoral artery is surgically ligated and excised as described previously, in the left hindlimb only. The right hindlimb thus serves as a non-ischemic control.
  • the musculature of the distal legs (e.g., tibialis anterior) then can be used as a well-demarcated delivery site for lOO ⁇ l injections into normal (right) and ischemic (left) muscle, that are performed under direct visualization.
  • Injections of precisely defined numbers of ASC or EPC will be conducted 1 day following the surgical induction of ischemia in mice, with groups of 5 animals for each condition to be evaluated.
  • the conditions will include control injections in saline (the previous standard) or in soluble self-assembling matrices.
  • the cells will be labeled with GFP to permit enumeration by subsequent flow cytometry following muscle dissociation, as well as microscopic evaluation of the anatomy of engraftment and differentiation in selected mice.
  • mice injected will be sacrificed at either 3 hours post-injection, to quantify the number of cells retained acutely following delivery; and at 2 weeks post-injection, to determine precisely the cell survival over time following the injection. Cells will be counted by flow cytometry with the addition of fluorescent particles to permit precise volumetric enumeration. A total of 60 mice will be used in this study (e.g., 2 cell types x 3 ECMs x 5 animals/group x 2 timepoints). The key endpoints will be quantitation of cell retention, and subsequent survival and engraftment into muscle or vasculature in the normal and ischemic muscles.
  • EXAMPLE 31 EXAMPLE 31
  • NHDFs low passage neonatal human dermal fibroblasts
  • growth media and passing solutions were obtained from Cambrex Bioproducts (Walkersville, MD).
  • NHDF were propagated in fibroblast basal medium supplemented with human recombinant fibroblast growth factor, insulin, gentamicin, amphotericin B, and FBS according to manufacturer's recommendation.
  • Cells were maintained in a humidified atmosphere of 5% CO 2 at 37 0 C and cell passage numbers representing 15 or less were used for all experiments.
  • type I collagen matrices with varied HA concentrations were prepared.
  • Three-dimensional engineered ECMs were prepared similar to those described in Example 13 at a constant collagen type I concentration (2 mg/ml) and hyaluronic acid concentrations of between 0 and 1.0 mg/ml.
  • the polymerization buffer consisted of 1OX phosphate buffered saline (PBS) with an ionic strength of 0.14 M and a pH of 7.4. All 3D engineered ECMs and tissue constructs were polymerized in vitro within a humidified environment at 37 0 C. To determine the cellular signaling capacity of each 3D microenvironment, 3D tissue constructs were formed by first harvesting NHDFs in complete media and then adding the cells (5 x 10 4 cells/ml) as the last component to the collagen solutions prior to polymerization. Immediately following polymerization complete media was added and the constructs were maintained in a humidified atmosphere of 5% CO 2 in air at 37 0 C. Qualitative and Quantitative Analysis of 3D ECM Microstructure
  • Fibril diameter measurements were made by applying Imaris 4.0 (Bitplane hie, Saint Paul, MN) to both confocal reflection and SEM images of engineered ECM constructs.
  • Imaris 4.0 Bendin hie, Saint Paul, MN
  • engineered ECM constructs were fixed in 3% glutaraldehyde in 0.1M cacodylate at pH 7.4, dehydrated with ethanol, and critical point dried.
  • Samples were sputter-coated with gold/palladium prior to imaging.
  • Samples were imaged in at least duplicate with a JEOL (Peabody, MA) JSM-840 SEM. From each image obtained, twenty fibrils were chosen at random (5 fibrils per quadrant). Five lines were drawn perpendicular to the long axis of each fibril using the measurement tool in Imaris. The average number of pixels representing the fibril diameter was then converted into ⁇ m based upon the known pixel size.
  • a shear creep test was conducted with a shear stress of 1 Pa for 120 seconds. Creep data was interpreted with a standard four- element Voigt spring dashpot model. Next a frequency sweep of controlled-strain oscillatory shear was made at 0.1 % strain, from 0.01 to 20 Hz. Following the frequency sweep, a continuous shear stress ramp from 0.1 to 10.0 Pa over 2 minutes was applied. Finally, the specimen was subjected to unconfined compression at a rate of lO ⁇ m/sec.
  • Tissue constructs representing NHDFs seeded at 5 x 10 4 cells/ml within 3D engineered ECMs with defined microstructural and biochemical compositions were evaluated using time-lapse confocal microscopy. Beginning 1 hour after polymerization, 2 to 3 cells were repeatedly monitored using the confocal microscope in a reflection (back-scattered light) mode to obtain image stacks of the individual cell and its surrounding matrix as described previously (Voytik-Harbin, et al., Microscopy and Microanalysis, 9:74-85, 2003). Images were collected at 30- minute intervals and a z-step of 0.5 mm to minimize exposure of the tissue constructs to radiation from the argon laser. Determination of Volumetric Strain
  • Consecutive confocal reflection images representing temporal deformation induced by a resident cell on its surrounding ECM microstructure provided the basis for the quantification of local displacements and strains in 3D.
  • subvolumes of 32 x 32 x 20 pixels in the x, y, and z directions, respectively, were established.
  • Each subvolume represented a group of voxels centered around a given point at which displacement values were sought.
  • Each image subvolume provided a unique 3D voxel intensity pattern that allowed correlation pattern matching between consecutive images using an incremental digital volume correlation (IDVC) algorithm developed previously by our laboratory (Roeder et al., J. Biomech. Eng. vol. 124, pp. 214-222 (2002)).
  • IDVC incremental digital volume correlation
  • the IDVC algorithm provided strain-state data, including principal strains and their associated directions, for all grid point locations.
  • Grid points were established in 512 ' 512-pixel images that were 32 pixels apart in both x- and y-directions, with 24-pixel spacing in the z-direction.
  • tissue constructs Prior to imaging at either 6 or 12 hours after construct polymerization, tissue constructs were stained with the vital dye Cell Tracker Green (Molecular Probes, Eugene, OR) to facilitate discrimination of the cell from the surrounding collagen ECM. Confocal image stacks were then collected in a combined reflection- epifluorescence mode for determination of cell morphology and fibril microstructural organization. Results
  • Fibril diameter distribution was measured, as determined from scanning electron microscopy images, for engineered matrices prepared from type I collagen in the presence of varied amounts of hyaluronic acid. Over the range of hyaluronic acid concentrations tested, no significant difference was observed in mean fibril diameter. Mean fibril diameter measurements were 80.8 ⁇ 18.3 ⁇ m, 72.2 ⁇ 13.0 ⁇ m, and 72.1 ⁇ 11.8 ⁇ m ( ⁇ standard deviation) for engineered matrices prepared from 2 mg/ml type I collagen containing 0, 0.5 mg/ml, and 1.0 mg/ml hyaluronic acid, respectively. Interestingly, it did appear that the variation (standard deviation) of fibril diameter measurement decreased with increasing hyaluronic acid content.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Transplantation (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Dermatology (AREA)
  • Medicinal Chemistry (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Botany (AREA)
  • Biophysics (AREA)
  • Developmental Biology & Embryology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Materials For Medical Uses (AREA)

Abstract

L'invention concerne une composition permettant la culture de cellules souches. Cette composition comprend une matrice de synthèse à base de collagène purifié, qui a été formée dans des conditions contrôlées, de manière à présenter la microstructure et les caractéristiques mécaniques recherchées. Ces compositions de matrice de synthèse à base de collagène purifié peuvent être utilisées seules ou combinées à des cellules, en tant que greffon tissulaire reconstitué, pour favoriser la réparation de tissus endommagés ou malades.
PCT/US2006/019130 2005-05-16 2006-05-16 Matrices extracellulaires de synthese regulant le comportement des cellules souches WO2006125025A2 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CA2608422A CA2608422C (fr) 2005-05-16 2006-05-16 Matrices extracellulaires de synthese regulant le comportement des cellules souches
AU2006247228A AU2006247228B2 (en) 2005-05-16 2006-05-16 Engineered extracellular matrices control stem cell behavior
GB0724345A GB2441268B (en) 2005-05-16 2006-05-16 Engineered extracellular matrices control stem cell behavior

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US68151105P 2005-05-16 2005-05-16
US68168905P 2005-05-16 2005-05-16
US68152205P 2005-05-16 2005-05-16
US68127805P 2005-05-16 2005-05-16
US60/681,522 2005-05-16
US60/681,278 2005-05-16
US60/681,511 2005-05-16
US60/681,689 2005-05-16

Publications (2)

Publication Number Publication Date
WO2006125025A2 true WO2006125025A2 (fr) 2006-11-23
WO2006125025A3 WO2006125025A3 (fr) 2007-06-07

Family

ID=37106236

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/019130 WO2006125025A2 (fr) 2005-05-16 2006-05-16 Matrices extracellulaires de synthese regulant le comportement des cellules souches

Country Status (4)

Country Link
AU (1) AU2006247228B2 (fr)
CA (1) CA2608422C (fr)
GB (1) GB2441268B (fr)
WO (1) WO2006125025A2 (fr)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2453452B (en) * 2006-05-16 2010-12-01 Purdue Research Foundation Three dimensional purified collagen matrices
US8084055B2 (en) 2006-09-21 2011-12-27 Purdue Research Foundation Collagen preparation and method of isolation
US9315778B2 (en) 2006-05-16 2016-04-19 Purdue Research Foundation Engineered extracellular matrices control stem cell behavior
US9867905B2 (en) 2007-12-10 2018-01-16 Purdue Research Foundation Collagen-based matrices with stem cells
US9878071B2 (en) 2013-10-16 2018-01-30 Purdue Research Foundation Collagen compositions and methods of use
CN109224129A (zh) * 2018-09-30 2019-01-18 四川大学华西医院 一种皮肤缺损修复材料
WO2021202619A1 (fr) * 2020-03-30 2021-10-07 Northeastern University Inhibiteurs de la nucléation du collagène
US11739291B2 (en) 2017-04-25 2023-08-29 Purdue Research Foundation 3-dimensional (3D) tissue-engineered muscle for tissue restoration
US11919941B2 (en) 2015-04-21 2024-03-05 Purdue Research Foundation Cell-collagen-silica composites and methods of making and using the same

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002102237A2 (fr) * 2001-06-15 2002-12-27 The Cleveland Clinic Foundation Cordes de valvule mitrale obtenues par genie tissulaire; methodes de realisation et d'utilisation
WO2003068287A1 (fr) * 2002-02-11 2003-08-21 Neocutis Sa Compositions comprenant des cellules embryonnaires indifferenciees destinees au traitement d'affections de la peau
WO2003087337A2 (fr) * 2002-04-12 2003-10-23 Yale University Equivalent de peau humaine vascularisee
WO2006003442A2 (fr) * 2004-07-05 2006-01-12 Ucl Business Plc Fabrication independante des cellules d'equivalents tissulaires
EP1674116A2 (fr) * 2004-12-22 2006-06-28 DePuy Products, Inc. Matrice de protéines auto-assemblante obtenue à partir de matrices extracellulaires naturelles

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7615373B2 (en) * 1999-02-25 2009-11-10 Virginia Commonwealth University Intellectual Property Foundation Electroprocessed collagen and tissue engineering

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002102237A2 (fr) * 2001-06-15 2002-12-27 The Cleveland Clinic Foundation Cordes de valvule mitrale obtenues par genie tissulaire; methodes de realisation et d'utilisation
WO2003068287A1 (fr) * 2002-02-11 2003-08-21 Neocutis Sa Compositions comprenant des cellules embryonnaires indifferenciees destinees au traitement d'affections de la peau
WO2003087337A2 (fr) * 2002-04-12 2003-10-23 Yale University Equivalent de peau humaine vascularisee
WO2006003442A2 (fr) * 2004-07-05 2006-01-12 Ucl Business Plc Fabrication independante des cellules d'equivalents tissulaires
EP1674116A2 (fr) * 2004-12-22 2006-06-28 DePuy Products, Inc. Matrice de protéines auto-assemblante obtenue à partir de matrices extracellulaires naturelles

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BRIGHTMAN ET AL.: "Time-lapse confocal reflection microscopy of collagen fibrillogenesis and extracellular matrix assembly in vitro" BIOPOLYMERS, vol. 54, 2000, pages 222-234, XP002428748 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2453452B (en) * 2006-05-16 2010-12-01 Purdue Research Foundation Three dimensional purified collagen matrices
US9315778B2 (en) 2006-05-16 2016-04-19 Purdue Research Foundation Engineered extracellular matrices control stem cell behavior
US8084055B2 (en) 2006-09-21 2011-12-27 Purdue Research Foundation Collagen preparation and method of isolation
US8512756B2 (en) 2006-09-21 2013-08-20 Purdue Research Foundation Collagen preparation and method of isolation
US9867905B2 (en) 2007-12-10 2018-01-16 Purdue Research Foundation Collagen-based matrices with stem cells
US9878071B2 (en) 2013-10-16 2018-01-30 Purdue Research Foundation Collagen compositions and methods of use
US11478574B2 (en) 2013-10-16 2022-10-25 Purdue Research Foundation Collagen compositions and methods of use
US11919941B2 (en) 2015-04-21 2024-03-05 Purdue Research Foundation Cell-collagen-silica composites and methods of making and using the same
US11739291B2 (en) 2017-04-25 2023-08-29 Purdue Research Foundation 3-dimensional (3D) tissue-engineered muscle for tissue restoration
CN109224129A (zh) * 2018-09-30 2019-01-18 四川大学华西医院 一种皮肤缺损修复材料
WO2021202619A1 (fr) * 2020-03-30 2021-10-07 Northeastern University Inhibiteurs de la nucléation du collagène

Also Published As

Publication number Publication date
GB2441268B (en) 2009-10-21
CA2608422C (fr) 2014-10-28
CA2608422A1 (fr) 2006-11-23
GB2441268A (en) 2008-02-27
GB0724345D0 (en) 2008-01-30
AU2006247228A1 (en) 2006-11-23
AU2006247228B2 (en) 2012-04-26
WO2006125025A3 (fr) 2007-06-07

Similar Documents

Publication Publication Date Title
US9315778B2 (en) Engineered extracellular matrices control stem cell behavior
CA2652138C (fr) Matrices tridimensionnelles de collagene purifie
CA2608422C (fr) Matrices extracellulaires de synthese regulant le comportement des cellules souches
US8512756B2 (en) Collagen preparation and method of isolation
CA2608708C (fr) Matrice extracellulaire fabriquee ayant une zone de fibrile specifique
RU2645473C2 (ru) Тканевые конструкции, полученные с помощью биоинженерии, и способы их получения и применения
JP6196217B2 (ja) 筋線維断片を利用した筋組織再生
US20200179567A1 (en) Regionally specific tissue-derived extracellular matrix
US20140271454A1 (en) Cell-synthesized particles
EP2780048B1 (fr) Petit tissu à base de dextrane contenant un lysat à plasma riche en plaquettes pour la réparation des cartilages
Rameshbabu et al. Bioinspired 3D porous human placental derived extracellular matrix/silk fibroin sponges for accelerated bone regeneration
Nie et al. Engineering a multiphasic, integrated graft with a biologically developed cartilage–bone interface for osteochondral defect repair
US20220378844A1 (en) Rotator cuff therapy using muscle fiber fragments
TW201545779A (zh) 製備去細胞化生物材料之方法及由其製備之去細胞化生物材料
US11998658B2 (en) Injectable porous hydrogels
US20200254142A1 (en) Injectable porous hydrogels
Nguyen et al. Fabrication of Injectable Hydrogel from Decellularized Adipose Tissue
Harvanová et al. Collagen/hyaluronan membrane as a scaffold for chondrocytes cultivation
GB2463811A (en) Collagen matrices

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2608422

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006247228

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 0724345

Country of ref document: GB

Kind code of ref document: A

Free format text: PCT FILING DATE = 20060516

WWE Wipo information: entry into national phase

Ref document number: 0724345.4

Country of ref document: GB

NENP Non-entry into the national phase

Ref country code: RU

ENP Entry into the national phase

Ref document number: 2006247228

Country of ref document: AU

Date of ref document: 20060516

Kind code of ref document: A

122 Ep: pct application non-entry in european phase

Ref document number: 06760040

Country of ref document: EP

Kind code of ref document: A2