WO2021250186A1 - 3d bioprinting method for forming a cell specific tissue construct - Google Patents

3d bioprinting method for forming a cell specific tissue construct Download PDF

Info

Publication number
WO2021250186A1
WO2021250186A1 PCT/EP2021/065669 EP2021065669W WO2021250186A1 WO 2021250186 A1 WO2021250186 A1 WO 2021250186A1 EP 2021065669 W EP2021065669 W EP 2021065669W WO 2021250186 A1 WO2021250186 A1 WO 2021250186A1
Authority
WO
WIPO (PCT)
Prior art keywords
bioink
peptide
cells
extracellular matrix
meth
Prior art date
Application number
PCT/EP2021/065669
Other languages
French (fr)
Inventor
Hugo Fernando CAMALHAO LOPES DE OLIVEIRA
Chantal MEDINA
Bruno PAIVA DOS SANTOS
Nathalie DUSSERRE
Jean-Christophe Fricain
Marie-Laure STACHOWICZ
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Université De Bordeaux
Chu De Bordeaux
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Université De Bordeaux, Chu De Bordeaux filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2021250186A1 publication Critical patent/WO2021250186A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0062General methods for three-dimensional culture
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B33ADDITIVE MANUFACTURING TECHNOLOGY
    • B33YADDITIVE MANUFACTURING, i.e. MANUFACTURING OF THREE-DIMENSIONAL [3-D] OBJECTS BY ADDITIVE DEPOSITION, ADDITIVE AGGLOMERATION OR ADDITIVE LAYERING, e.g. BY 3-D PRINTING, STEREOLITHOGRAPHY OR SELECTIVE LASER SINTERING
    • B33Y80/00Products made by additive manufacturing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M25/00Means for supporting, enclosing or fixing the microorganisms, e.g. immunocoatings
    • C12M25/14Scaffolds; Matrices
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0619Neurons
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0656Adult fibroblasts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/069Vascular Endothelial cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0697Artificial constructs associating cells of different lineages, e.g. tissue equivalents
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B33ADDITIVE MANUFACTURING TECHNOLOGY
    • B33YADDITIVE MANUFACTURING, i.e. MANUFACTURING OF THREE-DIMENSIONAL [3-D] OBJECTS BY ADDITIVE DEPOSITION, ADDITIVE AGGLOMERATION OR ADDITIVE LAYERING, e.g. BY 3-D PRINTING, STEREOLITHOGRAPHY OR SELECTIVE LASER SINTERING
    • B33Y70/00Materials specially adapted for additive manufacturing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/13Coculture with; Conditioned medium produced by connective tissue cells; generic mesenchyme cells, e.g. so-called "embryonic fibroblasts"
    • C12N2502/1323Adult fibroblasts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/28Vascular endothelial cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2513/003D culture
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/30Synthetic polymers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/52Fibronectin; Laminin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/54Collagen; Gelatin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/70Polysaccharides
    • C12N2533/80Hyaluronan
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2537/00Supports and/or coatings for cell culture characterised by physical or chemical treatment
    • C12N2537/10Cross-linking

Definitions

  • the present invention relates a 3D bioprinting method and to hydrogel compositions for use as bioink gel formulations.
  • the instant invention relates to a method for forming a 3D cell or tissue construct with a desired biological function useful for tissue engineering, organ on a chip or in vitro tissue/organ model applications.
  • Bioprinting technology offers the possibility for controlled deposition and patterning of cells, polymers, composites, and/or hydrogels to create structurally defined scaffolds.
  • This approach takes advantage of rapid prototyping, assisted by computer-assisted design (CAD) and/or computer-assisted manufacturing (CAM) procedures, to build a cellularized scaffold with geometric control of its internal structure and external shape.
  • CAD computer-assisted design
  • CAM computer-assisted manufacturing
  • bioprinting Different technologies of bioprinting have been developed, including inkjet or microjet, microextrusion and laser-assisted bioprinting [2, 3] Different technologies enable spatial control over bioink deposition with inherent different resolutions, cell survival and speed. From the 3 main bioprinting technologies, microextrusion is the most common, due to its ease of operation and affordability. It allows to create, layer-by-layer, the deposition of a wide range of bioinks, where the viscosity of the chosen bioinks often play a key role in their printability [4] Usually, high viscosity biomaterials (e.g . high concentrations or high molecular weight) provide better printability higher structural integrity to the printed geometry as they can easily support their own weight but upon chemical and/or physical gelation.
  • high viscosity biomaterials e.g . high concentrations or high molecular weight
  • bioinks for bioprinting [6, 7], including natural polymers like gelatin, type I collagen, hyaluronic acid, fibrin, agarose, alginate, or synthetic polymers such as polyethylene glycol, pluronics, among others [8]
  • natural polymers like gelatin, type I collagen, hyaluronic acid, fibrin, agarose, alginate, or synthetic polymers such as polyethylene glycol, pluronics, among others
  • Optimal bioink biomaterial properties include printability, mechanical properties, biodegradation, modifiable functional groups on the surface and post printing maturation.
  • extracellular matrix-derived polymers have inherent beneficial properties, like ease of degradation, better cell attachment, growth and proliferation [9] Such examples are collagen and hyaluronic acid.
  • Collagen a major component of the extracellular matrix (ECM), is commonly used as bioink alone or in combination with other biomaterials [10-12], due to its excellent biocompatible properties [13] Nonetheless, the slow physical gelation of collagen and low mechanical bearing properties poses several limitations for 3D bioprinting. In this sense the combination with other biomaterials increases its extrusion properties in view of bioprinting, as for instance fibrin [14], alginate [11] or hyaluronic acid [15]
  • Hyaluronic acid also a natural ECM major component, is abundantly present in cartilage, skin and in connective tissues [16] Also it has shown relevant key functions that make it a key material for tissue engineering, namely wound healing properties [17], reduce inflammation [18], induce cell proliferation and migration [19], key role in embryogenesis and morphogenesis [20] and angiogenesis [21] Thanks to these inherent properties, HA is one of the prominent biomaterials which are used in 3D bioprinting alone or in combination with other biomaterials).
  • HA shows a weak post print stability and in this sense its chemical modification by methacrylation [22], thiolation [15], modification with phenolic hydroxyl moieties [23], among others, have shown improvements on the long terms stability and on the capacity to sustain both osteogenesis, stromal cell elongation or for the culture of chondrocytes.
  • Poldervaart et al. [22] describe methacrylated hyaluronic acid hydrogels for 3D printing intended to promote osteogenicity.
  • the aim of the present invention is to provide a bioink that is easily printable, exhibits mechanical properties relevant for structural stability and pertinent for the final application, has good gelling capacity, is biodegradable, and comprises surface modifiable functional groups or cellular adhesion sequences, and has the capacity to sustain the maturation of the printed structure in time.
  • the instant invention provides a method for forming a cell specific 3D cell or tissue construct with a desired biological function comprising the steps of: a) Providing a (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel support medium; b) Reacting said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel with a thiol flanked functionalized adhesion peptide to obtain a (meth) acrylate crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide, via a thiol-ene reaction; cl) Either combining said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide with a plurality of specific living cells to provide a printable bioink, dl) Printing the said bioink into a first layer of printed bioink with a defined design; el) Submitting the said first layer of bioink to a polymerization reaction, under controlled conditions in order
  • the present invention provides a method for forming a cell specific 3D cell or tissue construct with a desired biological function comprising the steps of: a) Providing a (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel support medium; b) Reacting said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel with a thiol flanked functionalized adhesion peptide to obtain a (meth) acrylate crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide, via a thiol-ene reaction; cl) Either, combining said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide with a plurality of specific living cells to provide a printable bioink, dl) Printing the said bioink into a first layer of printed bioink with a defined design; el) Submitting the said first layer of bioink to a polymerization reaction, under controlled conditions in
  • the method of the invention is comprising the following steps: al) Providing a (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel support medium; bl) Reacting said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel with a thiol flanked functionalized adhesion peptide to obtain a (meth) acrylate crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide, via a thiol-ene reaction; cl) Combining said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide with a plurality of specific living cells to provide a printable bioink, dl) Printing the said bioink into a first layer of printed bioink with a defined design; el) Submitting the said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crosslinked bioink; and fl) Repeating
  • the method of the invention is comprising the following steps: a2) Providing a (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel support medium; b2) Reacting said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel with a thiol flanked functionalized adhesion peptide to obtain a (meth) acrylate crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide, via a thiol-ene reaction c2) Printing said bioink into a first layer of printed bioink with a defined design; d2) Submitting said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crosslinked bioink; and e2) Repeating the printing step and said polymerization step until a 3D construct with the desired shape is obtained, and f2), adding to said printed (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said
  • Bioprinting or “3D bioprinting” is the process of creating cell patterns in a confined space using 3D printing technologies, where cell function and viability are preserved within the printed construct.
  • 3D bioprinting utilizes the layer-by-layer method to deposit materials known as Bioinks to create tissue-like structures that are later used in medical and tissue engineering fields.
  • Bioinks consist on the combination of materials that mimic an extracellular matrix environment to support the adhesion, proliferation, and differentiation of living cells, and living cells. Layers of bioink are deposited, following a defined geometry, to create a cell laden scaffold.
  • the “storage modulus” (G’), also called “elastic modulus”, represents the elastic portion of the viscoelastic behavior of a hydrogel and corresponds to the amount of energy stored and released in each oscillation.
  • the “loss modulus” (G”) or viscous modulus characterizes the viscous portion of the viscoelastic behavior, which can be seen as the liquid-state behavior of the hydrogel sample. It characterizes the deformation energy lost (dissipated) through internal friction when the gel is flowing.
  • G’ values exceed G” values over the entire frequency range, showing the elastic character of these materials.
  • gels may have G’ values comprised between 50 Pa and 250 kPa and G” values comprised between 5 Pa and 80 Pa, following photopolymerization
  • the hydrogel may comprise an aqueous solution.
  • the aqueous solution may comprise a salt chosen in the group comprising lithium, sodium, potassium, rubidium, cesium, or ammonium chloride, preferably sodium chloride.
  • the concentration of lithium, sodium, potassium, rubidium, cesium or ammonium chloride in the aqueous solution may be comprised between 0.5 to 1.5 wt.%, preferably is 0.9 wt. %.
  • the aqueous solution may be a 0.9% aqueous sodium chloride solution.
  • biological function can refer to the modulation, growth, and/or proliferation of at least one cell, such as a progenitor cell and/or differentiated cell, the modulation of the state of differentiation of at least one cell, and/or the induction of a pathway in at least one cell, which directs the cell to grow, proliferate, and/or differentiate along a desired pathway, e.g., leading to a desired cell phenotype, cell migration, angiogenesis, apoptosis, etc.
  • a progenitor cell and/or differentiated cell the modulation of the state of differentiation of at least one cell, and/or the induction of a pathway in at least one cell, which directs the cell to grow, proliferate, and/or differentiate along a desired pathway, e.g., leading to a desired cell phenotype, cell migration, angiogenesis, apoptosis, etc.
  • trimer can refer to any natural polymer or oligomer.
  • peptide refers to an oligopeptide, peptide, polypeptide, or protein sequence, or to a fragment, portion, or subunit of any of these, and to naturally occurring or synthetic molecules.
  • cell can refer to any progenitor cell, such as totipotent stem cells, pluripotent stem cells, and multipotent stem cells, as well as any of their lineage descendant cells, including more differentiated cells.
  • the cells can derive from embryonic, fetal, or adult tissues.
  • Exemplary progenitor cells can be selected from, but not restricted to, totipotent stem cells, induced pluripotent or progenitor stem cells, pluripotent stem cells, multipotent stem cells, mesenchymal stem cells (MSCs), hematopoietic stem cells, neuronal stem cells, hematopoietic stem cells, pancreatic stem cells, cardiac stem cells, embryonic stem cells, embryonic germ cells, neural crest stem cells, kidney stem cells, hepatic stem cells, lung stem cells, hemangioblast cells, endothelial progenitor cells, and cancer stem cells.
  • MSCs mesenchymal stem cells
  • hematopoietic stem cells neuronal stem cells
  • hematopoietic stem cells pancreatic stem cells
  • cardiac stem cells embryonic stem cells, embryonic germ cells, neural crest stem cells, kidney stem cells, hepatic stem cells, lung stem cells, hemangioblast cells, endothelial progenitor cells, and cancer stem cells
  • Other cells are for example, fibroblasts, endothelial cells, nerve cells, schwan cells, mesenchymal stromal cells, primary or cell line cancer cells, hepatocytes, stellate cells, pericytes, macrophages, lymphocytes, chondrocytes, osteoblasts, osteoblasts, osteoclasts and epithelial cells.
  • Embodiments described herein relate to methods for three dimensional (3D) bioprinting of living cells to form scaffold-free 3D cell or tissue constructs.
  • the 3D tissue constructs can be used in regenerative medicine, cell-based technologies, tissue engineering, and bioprinting applications.
  • the systems and methods described herein use a sheer thinning, crosslinkable, biocompatible hydrogel support medium to provide a structural support role for the printed cell constructs, allowing media provision, and long term culture.
  • Precise maintenance of the structure for example, mirroring an original computer aided design (CAD) file, can also be achieved even after maturation of the tissue by cell proliferation, differentiation, and extracellular matrix (ECM) production.
  • CAD computer aided design
  • the printing step is performed through microextrusion
  • the hydrogel support medium can behave as a viscous fluid during printing and be resistant to flow before and after printing. For example, initially, the hydrogel support medium is in a flow-resistant or solid-like state before being printed with the bioink. The hydrogel support medium becomes fluidized under the increased shear stress caused by printing the bioink into the hydrogel support medium. Then, after the printing is finished the hydrogel support medium can be polymerized to form a solid-like stable support medium.
  • the extracellular matrix derived hydrogel is cytocompatible and, upon degradation produces substantially non-toxic products.
  • the living cells are added to the (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide prior to the printing of the bioink and the polymerization reaction is performed subsequently.
  • a culture medium in which the hydrogel support medium can be cultured can be provided after the bioink is printed and the hydrogel support medium is further crosslinked.
  • the culture medium can in addition include bioactive agents for promoting growth and/or differentiation of the cells of the printed bioink.
  • the culture medium can include a cell differentiation medium.
  • the hydrogel support medium can include a plurality of hydrogel particles that include a plurality of crosslinkable biodegradable extracellular matrix derived polymer macromers.
  • the hydrogel particles can have an average diameter of about 100 nm to about 100 pm.
  • the crosslinkable biocompatible extracellular matrix derived hydrogel support medium of the invention comprised a combination of natural polymer such as hyaluronic acid and collagen.
  • the natural polymer include a plurality of acrylated and/or methacrylated natural polymer macromers, preferably methacrylated polymers.
  • the acrylated and/or methacrylated, polymer macromers extracellular matrix derived polymers are advantageously comprised of a combination of collagen and hyaluronic acid.
  • the degree of (meth) acrylation of the natural polymer macromers such as collagen and hyaluronic acid is advantageously comprised between 5 to 75%, preferably between 15 and 70%.
  • the degree of (meth) acrylation of collagen is advantageously comprised between 40 to 70% more preferably between 40 to 60%. As described in the experimental section, and the degree of methacrylation of collagen is 57% (+/- 15%)
  • the degree of (meth) acrylation of hyaluronic acid is advantageously comprised between 10 to 30%. As described in the experimental section, the degree of methacrylation of hyaluronic acid is 20%
  • the ratio of collagen and hyaluronic acid is adjusted according to the desired properties of the bioink and hence the desired biological function of the 3D scaffold that is ultimately obtained.
  • hyaluronic acid concentration plays a key role on the overall bioink viscosity and therefore in the printability of the final formulation.
  • the concentration of collagen, a major component of the extracellular matrix is adjusted to enable optimal cell adhesion and survival.
  • the mass ratio, hyaluronic acid/ collagen is advantageously comprised between 0.125 and 100, preferably between 5 and 20.
  • the mass ratio, hyaluronic acid / collagen is 10 (as described in the experimental section)
  • the concentration of collagen in said hydrogel in the range from 0.5 to 5 mg/mL more preferably between 1 and 2 mg/mL and the concentration of hyaluronic acid in said hydrogel is in the range from 0.5 to 50 mg/ml, more preferably between 10 and 20 mg/mL.
  • Both collagen and hyaluronic acid are advantageously methacrylated, to form methacrylated collagen and methacrylated hyaluronic acid (CollMA and HAMA, respectively) and combined for instance with laminin-derived thiolated peptides.
  • the thiol-ene reaction, between the acrylate groups of CollMA and HAMA, and the thiol moieties of the peptides is achieved before bioprinting.
  • subsequent radical polymerization between the excess acrylate groups of the methacrylated polymers, of the composite hydrogel is accomplished following bioprinting.
  • This approach enables to separate the optimization regarding printability, namely by changing the CollMA and HAMA concentrations and ratio and the cell active peptide sequence spatial density, drastically increasing the tunability of the developed bioinks and allowing to easily adapt to other peptide sequences or to create complex mixtures and/or gradients.
  • the adhesion peptide is a laminin derived peptide.
  • laminin derived peptides are the peptides containing the following sequences: YIGSR and IKVAV, that can be flanked by a spacer sequence (i.e. beta alanine or glycine repeats) and that can be flanked by terminal cysteines, at the amino and carboxyl terminal chains.
  • a spacer sequence i.e. beta alanine or glycine repeats
  • the density of said peptide within the said extracellular matrix is adjusted with respect to the desired properties of the bioink and hence the desired biological function of the 3D scaffold that is ultimately obtained.
  • the density inside the final composite hydrogel is comprised between 0.5 mg/mL and 10 mg/mL.
  • the plurality of living cells can include progenitor cells, undifferentiated cells, differentiated cells, and/or cancer cells.
  • the cells may be selected from apical papilla stem cells, human bone marrow cells, human skin fibroblast, human gingival fibroblast, human umbilical vein cells, endothelial cells, neuronal cells, hematopoietic stem cells (FISC), human induced pluripotent stem cells (HiPSC) and mesenchymal stem cells (MFISC).
  • FISC hematopoietic stem cells
  • HiPSC human induced pluripotent stem cells
  • MFISC mesenchymal stem cells
  • the gel may comprise a concentration of cells between 0.01 million to 100 million of cells/mL of gel, preferably between 0.05 and to 20 million of cells/mL of gel.
  • the concentration of cell that can be used is 0.05 million of cells/ and for the endothelial the concentration of cell that can be used is 20 M/mL.
  • the living cells comprise fibroblasts, preferably human skin fibroblasts and/or human umbilical vein endothelial cells.
  • the sequence of the peptide is YIGSR and the concentration of the peptide in the hydrogel comprised between 0.5 mg/ml and 6 mg/ml.
  • the living cells are neurons, preferably primary sensory neurons.
  • the sequence of the peptide is IKVAV and the concentration of the peptide in the hydrogel comprised between 0.5 mg/ml and 6 mg/ml.
  • the polymerization step is performed through crosslinking of the acrylate or methacrylate groups of the acrylated and/or methacrylated natural polymer macromer of the hydrogel particles.
  • the crosslinking is advantageously performed using UV light or visible light in the presence of adapted photoinitiators (see the review Lim KS et al “Fundamentals and Applications of Photo-Cross-Linking in Bioprinting” Chem. Rev. April 2020 https://doi.org/10.1021/acs.chemrev.9b00812).
  • acrylated and/or methacrylated natural polymer macromers of the hydrogel particles can be photocrosslinked with a photoinitiator that is provided in the hydrogel support medium.
  • the hydrogel particles can be exposed to a light source at a wavelength and for a time to promote crosslinking of the acrylate groups of the polymers and form the photocrosslinked biodegradable hydrogel particles.
  • a photoinitiator can include any photo-initiator that can initiate or induce polymerization of the acrylate or methacrylate macromer.
  • the photoinitiator can include camphor Quinone, benzoin methyl ether, 2-hydroxy-2-methyl- 1 -phenyl- 1-propanone, diphenyl(2,4,6-trimethylbenzoyl)phosphine oxide, lithium phenyl-2, 4,6,- trimethylbenzoylphosphinate, benzoin ethyl ether, benzophenone, 9,10-anthraquinone, ethyl-4- N,N-dimethylaminobenzoate, diphenyliodonium chloride, roboflavin and derivatives thereof.
  • the polymerization conditions are preferably selected in order to obtain a polymerization degree between 70 and 98%.
  • the invention provides a method for forming a 3D tissue construct for promoting angiogenesis, wherein the living cells comprise fibroblasts, preferably human skin fibroblasts and/or human umbilical vein endothelial cells and the sequence of the peptide is YIGSR at a concentration of 2 mg/mL.
  • the invention provides a method for forming a 3D tissue construct for promoting neurogenesis, wherein the living cells are neurons, preferably primary sensory neurons and the sequence of the peptide is IKVAV at a concentration of 2 mg/mL.
  • the printing step can be performed through several techniques as: inkjet, laser and extrusion-based bioprinting.
  • Extrusion-based bioprinting (EBB) is part of the most widespread tools used in additive manufacturing, mainly due to its capabilities for building large volume constructs such as tissue or organ equivalents.
  • This technique offers a large opportunity in terms of bioink as it can be adaptable to a wide range of materials.
  • the 3D printer is a bioprinter that can dispense the bioink from a cartridge or container in a specific pattern and at specific positions in the hydrogel support medium as directed by a computer aided design (CAD) software in order to form a specific cellular construct, tissue, or organ.
  • CAD computer aided design
  • a cartridge containing the bioink comprises one dispensing orifice. In various other embodiments, a cartridge comprises from 1 to 100 or more dispensing orifices.
  • the hydrogel can further include a culture medium for modulating a function or characteristic of a cell and/or for promoting growth and/or differentiation of the cells of the printed bioink.
  • the culture medium can include a cell differentiation medium, such as an osteogenic differentiation media or chondrogenic differentiation media.
  • the hydrogel support medium containing the printed bioink can be provided in the culture medium after crosslinking or before further crosslinking the hydrogel support medium to enhance the stability of the hydrogel support medium.
  • growth factors can be added to the medium to enhance or stimulate cell growth.
  • growth factors include vascular endothelial growth factor, platelet-derived growth factor, brain derived growth factor, nerve growth factor, transforming growth factor, platelet-derived growth factor, insulin-like growth factor, acid fibroblast growth factor, basic fibroblast growth factor, epidermal growth factor, hepatocytic growth factor, keratinocyte growth factor, and bone morphogenic protein.
  • agents such as cytokines, hormones (e.g., parathyroid hormone, parathyroid hormone-related protein, hydrocortisone, thyroxine, insulin), fatty acids (e.g., Omega-3 fatty acids such as al8:3 linolenate), and/or vitamins (e.g., vitamin D), may also be added or removed from the culture medium to promote cell growth.
  • hormones e.g., parathyroid hormone, parathyroid hormone-related protein, hydrocortisone, thyroxine, insulin
  • fatty acids e.g., Omega-3 fatty acids such as al8:3 linolenate
  • vitamins e.g., vitamin D
  • the gel may comprise between 0.1 and 10 % w/w concentration of hyaluronic acid and collagen, and between 90 and 99.9 % of an aqueous solution comprising lithium, sodium, potassium, rubidium, cesium or ammonium chloride
  • the invention also provides a bioink comprising a biocompatible (meth) acrylated extracellular matrix optionally partially crosslinked with an adhesion peptide and a plurality of cells (as above describe), wherein the extracellular matrix is comprised of a combination of hyaluronic acid and collagen, and hyaluronic acid at a mass ratio of hyaluronic acid /collagen comprised between 0.125 and 100, preferably between 5 and 20 BRIEF DESCRIPTION OF THE DRAWINGS
  • Figure 1 - (A) Viscosity evaluation, in function of the shear rate (0.01-500 s 1 ), of HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite, or HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of YIGSR peptide, or HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of IKVAV peptide. All bioinks were evaluated prior photopolymerization and at 25°C (Average ⁇ SD, n 3).
  • (B) Storage modulus (G’) of polymerized HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite, HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of YIGSR peptide, and HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of IKVAV. All composition contained 0.1 % (w/v) of LAP (Lithium phenyl-2, 4, 6-trimethylbenzoylphosphinate) as photoinitiator. Evaluated following 2 min photopolymerization, measured at 37 °C (Aver ⁇ SD, n 3). Statistically significant differences between indicated groups was analyzed by the non-parametric Kruskal- Wallis test, followed by a Dunns post-test (NS denotes Non Significant).
  • LAP Lithium phenyl-2, 4, 6-trimethylbenzoylphosphinate
  • FIG. 2 Fidelity of bioprinting struts composed of HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite, or HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of YIGSR peptide, using a 210 pm microextrusion tip diameter.
  • HAMA methacrylated hyaluronic acid
  • CollMA methacrylated type I collagen
  • DDG dorsal root ganglia
  • Methacrylation of hyaluronic acid The procedure was adapted from Poldervaart et al. [22] Briefly, one gram ofhyaluronic acid sodium salt from Streptococcus Equi (1.5-1.8 x 10E6 Da, Sigma-Aldrich, France) was dissolved in 75 mL of mQ grade water, overnight at room temperature. Then, 50 mL of dimethylformamide (Sigma-Aldrich, France) was added and the pH was adjusted to 8 using NaOH 1M. Then, 1.12 mL of methacrylic anhydride (Sigma- Aldrich) was added to the solution the pH was again adjusted to 8 and the solution was left under constant stirring overnight at room temperature.
  • Methacrylation of type I collagen The procedure was adapted from Ravichandran et al. [27] Briefly, type I collagen from bovine skin (Sigma-Aldrich) was adjusted to pH 10 using 2N NaOH and kept on ice, under mild agitation. Then, methacrylic anhydride at a molar ratio of 5:1 (with respect to number of lysine amine groups in collagen) was added subsequently drop-wise and left to react for 4 hours. Then the obtained solution was dialyzed (12-14 kDa, Sigma- Aldrich) for five days against 5L mQ water and with 2 daily water changes. The obtained solution was then freeze-dried for 5 days, and the polymer recovered and stored at -20 °C until further use. The methacrylated type I collagen (CollMA) stock solution was prepared at 6 mg/mL in 0.02 M Acetic acid and stored at 4 °C until further use.
  • Methacrylation degree was determined using the Tri-nitro benzene sulfonic acid (TNBS) assay [28] Briefly, neutralized native collagen or CollMA were dissolved in 1 mL of carbonate buffer (0.1M, pH 8.5) at 0.1 mg/mL NaHC03 (pH 8.5). Then to 500 pL of collagen or CollMA solution, we added 250 pL of a 1:500 dilution, in carbonate buffer, of TNBSA solution (5% w/v, Sigma-Aldrich) and let to react for 2 hours at 37°C. The reaction was then stopped by the addition of 250pL of 10% (w/v) of SDS (Sigma-Aldrich) and 125pL of HC1 1M.
  • TNBS Tri-nitro benzene sulfonic acid
  • the OD335 of both the blanc, collagen and CollMA was determined using a UV-Vis spectrophotometer (Nanodrop, ThermoFisher). The percentage of remaining free primary amines of CollMA was calculated in relation to the total amount of the pristine collagen. The degree of methacrylation was then determined using the loss of primary amines percentage loss (% DM).
  • HAMA at 1% (w/v) was prepared by diluting HAMA 2% (w/v) using DMEM HG (Gibco) and mixed with 0.1 % (w/v) of the photonitiator tithium phenyl-2, 4, 6-trimethylbenzoylphosphinate (LAP, TCI Chemicals).
  • CollMA 0.1% (w/v) was prepared by diluting the stock 0.6% (w/v) CollMA solution (dissolved in 0.02 M Acetic acid) with DMEM HG (Gibco), neutralizing with NaOH 0.1M and mixed with 0.1 % (w/v) of LAP photonitiator.
  • the adhesion peptides Cys- (Beta-Ala)-Ile-Lys-Val-Ala-Val-(Beta-Ala)-Cys (IKVAV) and Cys-(Beta-Ala)-Tyr-Ile-Gly- Ser-Arg-(Beta-Ala)-Cys (YIGSR) (Biomatik, Canada) were dissolved at 25 mg/mL in acetic acid 0.02 M aliquoted and stored at -20 °C untill further use.
  • composite bioink was obtained by diluting and neutralizing the stock 0.6% (w/v) CollMA, then HAMA 2% (w/v) was added, the solution was mixed with 0.1 % (w/v) of LAP and the volume adjusted with DMEM HG.
  • the peptide solution containing the laminin derived peptides was homogeneously added, at defined final concentrations, to the composite polymer solution prior use. All formulation were prepared fresh and kept protected from light.
  • HUVEECs Human umbilical vein endothelial cells
  • IMDM Gibco
  • SVF + ECGS/H ECGS/H
  • fluorescent microscopy cells were stably transduced with lenti virus bearing the RFP gene, under the EFla promoter sequence (Vectalys, France), following the manufacturer’s instructions. Infection efficiency was evaluated by flow cytometry and was determined to be superior to 98% (data not shown).
  • Human skin fibroblasts were isolated as previously described [30], and cultured, under standard conditions, in DMEM:F12 (Gibco), 20% (v/v) FBS, until passage 10.
  • DRG dorsal root ganglion
  • digestion product was washed twice with culture medium supplemented with 2 % (v/v) B-27 Serum-Free Supplement® (B-27, Gibco®) and 1 % (v/v) Penicilin Streptomycin, and mechanically dissociated using fire-polished glass Pasteur pipettes (full diameter and 1 ⁇ 2 diameter).
  • B-27 B-27 Serum-Free Supplement®
  • Penicilin Streptomycin was mechanically dissociated using fire-polished glass Pasteur pipettes (full diameter and 1 ⁇ 2 diameter).
  • the cell suspension was finally washed three more times with medium and resuspended in complete culture medium.
  • Nerve growth factor (NGF, 0.1 pg/mL, Sigma Aldrich) was added to the culture medium at 48 hrs of culture.
  • HUVEC cells The capacity of HUVEC cells to form capillary like structures in 3D was assessed by confocal imaging of RFP+ HUVEC cells at days 7 and 14, inside the different bioinks in 3D culture or after 3D bioprinting.
  • a 150 pm Z axis height was acquired using confocal microscopy.
  • a 500 pm Z axis height was acquired.
  • DRG cells The capacity of DRG cells to extend neurites was assessed by confocal imaging following Calcein-AM (1 pL/mL, Invitrogen) staining for 45 minutes at day 14, inside the different bioinks in 3D culture or after 3D bioprinting.
  • Calcein-AM 1 pL/mL, Invitrogen
  • For the 3D culture a 150 pm Z axis height was acquired using confocal microscopy.
  • bioprinted structures a 500 pm Z axis height was acquired.
  • the 3D Discovery bioprinting platform (RegenHU) was used in this work. It is composed of two independent pressure activated microextrusion heads and two microvalve heads, automated tip length and substrate height calibration and integrated ultraviolet (UV, 365 nm, 2W) photopolymerization diode. Also, the integrated computer assisted design software (BioCAD, RegenHU) enable to create the desired geometry of printing. The chosen geometry consisted on 5 parallel lines, spaced of 2.5 mm, and intercrossed with same geometry with a 90-degree angle rotation. The same geometry was repeated to a total of 4 layers. The geometry was adapted to print inside 6 well plate wells and repeated in series.
  • HUVEC+HSF bioprinting a conic microextrusion tip of 0.21mm diameter was used. Both HUVECs and HSF were both loaded together inside the designated bioink (containing or not the YIGSR peptide), both at 10 million/mL. The ink was homogenized and loaded inside the bioprinting syringe.
  • the bioprinting process consisted on the deposition of the first layer with a pressure of 0.008 MPa, speed of 140 mm/s and a distance to the substrate of 0.075 mm. Then the first layer was polymerized during 15 seconds using the UV source. The second perpendicular layer was then deposited using the same pressure and speed conditions at a distance of 0.1 mm of the first layer. The process was then repeated for a total of 4 layers, before adding 3 mL of cell culture media.
  • DRG bioprinting a conic microextrusion tip of 0.41mm diameter was used. DRG were loaded inside the designated bioink (containing or not the IKVAV peptide), at 50 000 cells/mL. The ink was homogenized and loaded inside the bioprinting syringe.
  • the bioprinting process consisted on the deposition of the first layer with a pressure of 0.005 MPa, speed of 160 mm/s and a distance to the substrate of 0.075 mm. Then the first layer was polymerized during 15 seconds using the UV source. The second perpendicular layer was then deposited using the same pressure and speed conditions at a distance of 0.1 mm of the first layer. The process was then repeated for a total of 4 layers, before adding 3 mL of cell culture media.
  • HUVECs The capacity of HUVECs to form capillary -like structures in 3D culture was quantified by image analysis of confocal stacking images using the Angiogenesis Analyzer [33] plugin for Image!
  • the software allowed to assess relevant parameters as number of junctions, number of segments and total segment length.
  • HUVEC evaluation upon 3D printing was achieved by 3D image analysis of the volumetric confocal acquisition, at day 14, and using the Imaris software (9.0, Bitplane, UK).
  • the 3D segmentation of independent structures, inside the bioprinted matrix were evaluated in terms of their length in the Z axis (Bounding Box length Z axis), as to further demonstrate the capacity of HUVECs to migrate inside the structure, and the volume of each one of these independent 3D structures (volume bounding box), as means to evaluate the capacity to interconnect and form a complex network.
  • the capacity of DRGs to extend neurites in 3D culture was quantified by image analysis of confocal stacking images using the NeuriteQuant [34] plugin for Image!
  • the software allowed to assess relevant parameters as number of neurites per neuron, maximum neurite length and neurite total length/per neuron.
  • DRG evaluation upon 3D printing was achieved by 3D image analysis of the volumetric confocal acquisition, at day 14, and using the Imaris software (9.0, Bitplane, UK).
  • the 3D segmentation of independent structures, inside the bioprinted matrix, were evaluated by segmental analysis and allow to determine the dendrite length distribution for different compositions of bioink.
  • the chemical approach for the creation of a printable composite hydrogel was based on an initial methacrylation of the base polymers, namely hyaluronic acid and type I collagen, both natural components of the extracellular matrix.
  • the inclusion of laminin-derived peptides was achieved by the inclusion of cysteine groups at both the extremities of the peptides, what permits to graft, via a thiol-ene UV initiated radical addition, the peptides to the methacrylated matrix.
  • the excess methacrylate moieties enables the photopolymerization of the matrix by radical polymerization via UV irradiation.
  • Shear-viscosity measurements were performed, prior photopolymerization, for HAMA (1% (w/v)) CollMA (0.1% (w/v)) alone or containing the highest peptide formulations (2 mg/mL of YIGSR or IKVAV). In the shear range tested, all formulations had similar viscosity values and showed a shear-thinning character. This fact is particularly relevant as shows that with the selected chemical peptide grafting approach one can achieve independent formulations, while maintaining viscosity unaltered.
  • Rheological characterization for polymerized methacrylated hyaluronic acid (HAMA) at 1% (w/v) show a G’ of 202 ⁇ 25 Pa (at 37°C), for methacrylated type 1 collagen (CollMA) at 0.1% (w/v) show a G’ of 114 ⁇ 12 Pa (at 37°C), for HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite show a G’ of 120 ⁇ 12 Pa (at 37°C), for HAMA(1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of YIGSR peptide a G’ of 197 ⁇ 85 Pa (at 37°C), and for HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of IKVAV a G’ of 169 ⁇ 24 Pa (at 37°C).
  • HAMA polymerized methacrylated hyaluronic acid
  • bioprinting strut dimensions show that the gel could respect the expected dimensions with an observable dispersion of 1.3x in relation to the expected dimensions and where no significant differences could be observed for the formulation with or without peptide. Additionally, the established conditions enabled to reach filament stability on air and to achieve the fabrication of complex structures, like a hollow tube (data not shown) or a parallelepiped (data not shown).
  • the quantification of the capillary-like structure was achieved by image analysis (Angiogenesis Analyser, ImageJ) and as seen in Figure 4 A,B,C, a significant increase in terms of the number of junctions, number of segments and total segment length was observed at 14 days of culture and for the HAMA /CollMA + 2 mg/mL YIGSR peptide.
  • the obtained results allowed the selection of the HAMA/CollMA + 2 mg/mL YIGSR peptide as optimal formulation for the maturation of HUVEC cells and was compared with the basal formulation, HAMA/CollMA.
  • the chosen assisted computer design (CAD) consisted on 5 parallel lines, spaced of 2.5 mm, and intercrossed with same geometry with a 90 degree angle rotation. The same geometry was repeated to a total of 4 layers. Both the speed of printing (140 mm/s), the diameter of the tip (0.21 mm) and the distance between the tip and the substrate (0.075 mm) were controlled in order to attain a reproducible geometry.
  • the capacity of the composite hydrogel containing YIGSR was tested to further sustain the maturation of HUVECs in coculture with HSFs, following bioprinting and using the same CAD. Both composition permitted the microextrusion bioprinting of the scaffold containing both HUVECs and HSFs. Additionally, one could observe that the composition containing the YIGSR peptide could further support HUVECs maturation, particularly relevant at day 14.
  • the HUVEC network was then evaluated by confocal microscopy on a thickness of 0.5 mm, and reconstructed using imaging analysis software (Imaris) in order to quantify its interconnectivity and dimensions.
  • Software analysis enabled to identify and create a tridimensional box that includes each interconnected structure (bounding box). Then, the tridimensional dimensions of this bounding box give a quantification of the interconnection and distribution inside the bioprinted matrix. Both the thickness (Z axis length) and interconnection structure volume was shown to be significantly increased for the bioprinted structure containing the laminin-derived YIGSR peptide.
  • the chosen CAD consisted on 5 parallel lines, spaced of 2.5 mm, and intercrossed with same geometry with a 90-degree angle rotation, as before. The same geometry was repeated to a total of 4 layers. Both the speed of printing (160 mm/s), the diameter of the tip (0.41 mm) and the distance between the tip and the substrate (0.075 mm) were controlled in order to attain a reproducible geometry. Live-dead assay staining assessed that the bioprinting process did not significantly affect the viability of the printed DRG neurons, as observe 14 days post printing, and the inclusion of the IKVAV peptide could improve neurite formation.
  • bioink formulations based on modular approaches considering at the same time the printability and rigidity (enabling to easily adapt components concentrations), while enabling to adjust for optimal cell adhesion sequence density or even on the combination or gradient of sequences, represents an added value in future developments in bioprinting.
  • the approach follows this line of thought, enabling inherent modularity and tunability while respecting the aimed application simplicity.
  • Phage display technology and the rampant field of biotechnology have already identified a wide range of small peptide sequences with inherent biological activity [56]
  • the simple approach of the invention enables to easily adapt to other bioactive peptide sequences, opening the field of application of such bioinks to a wide range of cell types and applications.
  • the adaptation of these novel formulations to other major bioprinting technologies, namely inkjet or laser-assisted bioprinting, are currently being explored opening the range of applications of this approach.

Abstract

The aim of the present invention is to provide a bioink that is easily printable, exhibits mechanical properties relevant for structural stability and pertinent for the final application, has good gelling capacity, is biodegradable, and comprises surface modifiable functional groups or cellular adhesion sequences, and has the capacity to sustain the maturation of the printed structure in time. The present invention accordingly relates to a method for forming a cell specific 3D cell or tissue construct with a desired biological function wherein the hydrogel support medium, the sequence of the said peptide, the ratio of the said peptide to the said extracellular matrix derived hydrogel and the polymerization conditions are selected with respect to cell specificity so as to obtain a 3D tissue construct with the desired biological function (such as promoting angiogenesis or neurogenesis) useful for tissue engineering, organ on a chip or in vitro tissue/organ model applications.

Description

3D BIOPRINTING METHOD FOR FORMING A CELL SPECIFIC TISSUE
CONSTRUCT
FIELD OF THE INVENTION:
The present invention relates a 3D bioprinting method and to hydrogel compositions for use as bioink gel formulations.
Specifically, the instant invention relates to a method for forming a 3D cell or tissue construct with a desired biological function useful for tissue engineering, organ on a chip or in vitro tissue/organ model applications.
BACKGROUND OF THE INVENTION
3D bioprinting has emerged as a technological approach to address unsolved question in tissue engineering and regenerative medicine [1, 2] Bioprinting technology offers the possibility for controlled deposition and patterning of cells, polymers, composites, and/or hydrogels to create structurally defined scaffolds. This approach takes advantage of rapid prototyping, assisted by computer-assisted design (CAD) and/or computer-assisted manufacturing (CAM) procedures, to build a cellularized scaffold with geometric control of its internal structure and external shape. The bioprinting of cell-laden biomaterials, termed bioinks, enables the deposition of cells encapsulated in a 3D construct and provides a procedure to develop complex synthetic biological systems and tissue-engineered constructs. However, the number of biomaterials that are bioprintable, enable high cell viability, are biocompatible and biodegradable, allow cells to migrate and maturate and permit defined customization remains limited.
Different technologies of bioprinting have been developed, including inkjet or microjet, microextrusion and laser-assisted bioprinting [2, 3] Different technologies enable spatial control over bioink deposition with inherent different resolutions, cell survival and speed. From the 3 main bioprinting technologies, microextrusion is the most common, due to its ease of operation and affordability. It allows to create, layer-by-layer, the deposition of a wide range of bioinks, where the viscosity of the chosen bioinks often play a key role in their printability [4] Usually, high viscosity biomaterials ( e.g . high concentrations or high molecular weight) provide better printability higher structural integrity to the printed geometry as they can easily support their own weight but upon chemical and/or physical gelation. Nonetheless, higher viscosity usually induces cell toxicity during bioprinting, due to the induced shear stress when extruded, and the tight matrix can also limit the mobility of encapsulated cells and their capacity to restructure their surrounding matrix. In this sense a huge effort has been dedicated to optimize at the same time the printability of a bioink while providing an optimal environment to live cells [5]
Various hydrogel compositions have been developed as bioinks for bioprinting [6, 7], including natural polymers like gelatin, type I collagen, hyaluronic acid, fibrin, agarose, alginate, or synthetic polymers such as polyethylene glycol, pluronics, among others [8] Different chemistries can be envisaged to achieve cross-linking of the bioprinted scaffold, one popular choice considers photoinitiated radical polymerization of (meth)acrylate groups, due to spatial and temporal control of reaction [8] Optimal bioink biomaterial properties include printability, mechanical properties, biodegradation, modifiable functional groups on the surface and post printing maturation. Also, extracellular matrix-derived polymers have inherent beneficial properties, like ease of degradation, better cell attachment, growth and proliferation [9] Such examples are collagen and hyaluronic acid.
Collagen, a major component of the extracellular matrix (ECM), is commonly used as bioink alone or in combination with other biomaterials [10-12], due to its excellent biocompatible properties [13] Nonetheless, the slow physical gelation of collagen and low mechanical bearing properties poses several limitations for 3D bioprinting. In this sense the combination with other biomaterials increases its extrusion properties in view of bioprinting, as for instance fibrin [14], alginate [11] or hyaluronic acid [15]
Hyaluronic acid (HA), also a natural ECM major component, is abundantly present in cartilage, skin and in connective tissues [16] Also it has shown relevant key functions that make it a key material for tissue engineering, namely wound healing properties [17], reduce inflammation [18], induce cell proliferation and migration [19], key role in embryogenesis and morphogenesis [20] and angiogenesis [21] Thanks to these inherent properties, HA is one of the prominent biomaterials which are used in 3D bioprinting alone or in combination with other biomaterials). Nonetheless, HA shows a weak post print stability and in this sense its chemical modification by methacrylation [22], thiolation [15], modification with phenolic hydroxyl moieties [23], among others, have shown improvements on the long terms stability and on the capacity to sustain both osteogenesis, stromal cell elongation or for the culture of chondrocytes. Poldervaart et al. [22] describe methacrylated hyaluronic acid hydrogels for 3D printing intended to promote osteogenicity.
Nonetheless, cell-specific tailored bioinks that can target specific functions have not yet been developed. Focusing on the field of tissue engineering and on tissue repair, the support of the angiogenesis process is widely accepted as key in order to attain tissue repair in most tissues. Additionally, more recent studies have shed light on the importance and implication of the innervation in tissue remodelling and repair [24] In this sense, the development of bioinks that can support both of these processes are strongly awaited in the field of biofabrication, bioprinting and tissue engineering.
The aim of the present invention is to provide a bioink that is easily printable, exhibits mechanical properties relevant for structural stability and pertinent for the final application, has good gelling capacity, is biodegradable, and comprises surface modifiable functional groups or cellular adhesion sequences, and has the capacity to sustain the maturation of the printed structure in time.
SUMMARY OF THE INVENTION:
The instant invention provides a method for forming a cell specific 3D cell or tissue construct with a desired biological function comprising the steps of: a) Providing a (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel support medium; b) Reacting said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel with a thiol flanked functionalized adhesion peptide to obtain a (meth) acrylate crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide, via a thiol-ene reaction; cl) Either combining said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide with a plurality of specific living cells to provide a printable bioink, dl) Printing the said bioink into a first layer of printed bioink with a defined design; el) Submitting the said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crosslinked bioink; and fl) Repeating the printing step and said polymerization step until a 3D construct with the desired shape is obtained, c2) Or printing said bioink into a first layer of printed bioink with a defined design; d2) Submitting said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crosslinked bioink; and e2) Repeating the printing step and said polymerization step until a 3D construct with the desired shape is obtained, and f2), adding to said printed (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide a plurality of specific living cells, g) Allowing the growth of the living cells in 3D-construct obtained thereby under suitable conditions until the desired biological function is achieved; wherein the hydrogel support medium, the sequence of the said peptide, the ratio of the said peptide to the said extracellular matrix derived hydrogel and the polymerization conditions are selected with respect to cell specificity so as to obtain a 3D tissue construct with the desired biological function.
DETAILED DESCRIPTION OF THE INVENTION:
The present invention provides a method for forming a cell specific 3D cell or tissue construct with a desired biological function comprising the steps of: a) Providing a (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel support medium; b) Reacting said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel with a thiol flanked functionalized adhesion peptide to obtain a (meth) acrylate crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide, via a thiol-ene reaction; cl) Either, combining said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide with a plurality of specific living cells to provide a printable bioink, dl) Printing the said bioink into a first layer of printed bioink with a defined design; el) Submitting the said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crosslinked bioink; and fl) Repeating the printing step and said polymerization step until a 3D construct with the desired shape is obtained c2) Or printing said bioink into a first layer of printed bioink with a defined design; d2) Submitting said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crosslinked bioink; and e2) Repeating the printing step and said polymerization step until a 3D construct with the desired shape is obtained, and f2), adding to said printed (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide a plurality of specific living cells, g) Allowing the growth of the living cells in 3D-construct obtained thereby under suitable conditions until the desired biological function is achieved; wherein the hydrogel support medium, the sequence of the said peptide, the ratio of the said peptide to the said extracellular matrix derived hydrogel and the polymerization conditions are selected with respect to cell specificity so as to obtain a 3D tissue construct with the desired biological function.
Accordingly in first alternative embodiment, the method of the invention is comprising the following steps: al) Providing a (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel support medium; bl) Reacting said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel with a thiol flanked functionalized adhesion peptide to obtain a (meth) acrylate crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide, via a thiol-ene reaction; cl) Combining said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide with a plurality of specific living cells to provide a printable bioink, dl) Printing the said bioink into a first layer of printed bioink with a defined design; el) Submitting the said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crosslinked bioink; and fl) Repeating the printing step and said polymerization step until a 3D construct with the desired shape is obtained, and gl) Allowing the growth of the living cells in 3D-construct obtained thereby under suitable conditions until the desired biological function is achieved;
Accordingly in a second alternative embodiment, the method of the invention is comprising the following steps: a2) Providing a (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel support medium; b2) Reacting said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel with a thiol flanked functionalized adhesion peptide to obtain a (meth) acrylate crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide, via a thiol-ene reaction c2) Printing said bioink into a first layer of printed bioink with a defined design; d2) Submitting said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crosslinked bioink; and e2) Repeating the printing step and said polymerization step until a 3D construct with the desired shape is obtained, and f2), adding to said printed (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide a plurality of specific living cells, and g2) Allowing the growth of the living cells in 3D-construct obtained thereby under suitable conditions until the desired biological function is achieved.
Throughout this specification, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated step or element or integer or group of steps or elements or integers, but not the exclusion of any other step or element or integer or group of elements or integers.
“Bioprinting or “3D bioprinting” is the process of creating cell patterns in a confined space using 3D printing technologies, where cell function and viability are preserved within the printed construct. Generally, 3D bioprinting utilizes the layer-by-layer method to deposit materials known as Bioinks to create tissue-like structures that are later used in medical and tissue engineering fields.
“Bioinks” consist on the combination of materials that mimic an extracellular matrix environment to support the adhesion, proliferation, and differentiation of living cells, and living cells. Layers of bioink are deposited, following a defined geometry, to create a cell laden scaffold.
The “storage modulus” (G’), also called “elastic modulus”, represents the elastic portion of the viscoelastic behavior of a hydrogel and corresponds to the amount of energy stored and released in each oscillation.
The “loss modulus” (G”) or viscous modulus characterizes the viscous portion of the viscoelastic behavior, which can be seen as the liquid-state behavior of the hydrogel sample. It characterizes the deformation energy lost (dissipated) through internal friction when the gel is flowing. Advantageously, G’ values exceed G” values over the entire frequency range, showing the elastic character of these materials. In the case of the present invention, gels may have G’ values comprised between 50 Pa and 250 kPa and G” values comprised between 5 Pa and 80 Pa, following photopolymerization
Advantageously, the hydrogel may comprise an aqueous solution. The aqueous solution may comprise a salt chosen in the group comprising lithium, sodium, potassium, rubidium, cesium, or ammonium chloride, preferably sodium chloride. The concentration of lithium, sodium, potassium, rubidium, cesium or ammonium chloride in the aqueous solution may be comprised between 0.5 to 1.5 wt.%, preferably is 0.9 wt. %.
Advantageously, the aqueous solution may be a 0.9% aqueous sodium chloride solution.
The term “biological function” can refer to the modulation, growth, and/or proliferation of at least one cell, such as a progenitor cell and/or differentiated cell, the modulation of the state of differentiation of at least one cell, and/or the induction of a pathway in at least one cell, which directs the cell to grow, proliferate, and/or differentiate along a desired pathway, e.g., leading to a desired cell phenotype, cell migration, angiogenesis, apoptosis, etc.
The term “macromer” can refer to any natural polymer or oligomer.
The term “peptide” refers to an oligopeptide, peptide, polypeptide, or protein sequence, or to a fragment, portion, or subunit of any of these, and to naturally occurring or synthetic molecules.
The term “cell” can refer to any progenitor cell, such as totipotent stem cells, pluripotent stem cells, and multipotent stem cells, as well as any of their lineage descendant cells, including more differentiated cells. The cells can derive from embryonic, fetal, or adult tissues. Exemplary progenitor cells can be selected from, but not restricted to, totipotent stem cells, induced pluripotent or progenitor stem cells, pluripotent stem cells, multipotent stem cells, mesenchymal stem cells (MSCs), hematopoietic stem cells, neuronal stem cells, hematopoietic stem cells, pancreatic stem cells, cardiac stem cells, embryonic stem cells, embryonic germ cells, neural crest stem cells, kidney stem cells, hepatic stem cells, lung stem cells, hemangioblast cells, endothelial progenitor cells, and cancer stem cells.
Other cells are for example, fibroblasts, endothelial cells, nerve cells, schwan cells, mesenchymal stromal cells, primary or cell line cancer cells, hepatocytes, stellate cells, pericytes, macrophages, lymphocytes, chondrocytes, osteoblasts, osteoblasts, osteoclasts and epithelial cells.
Embodiments described herein relate to methods for three dimensional (3D) bioprinting of living cells to form scaffold-free 3D cell or tissue constructs. The 3D tissue constructs can be used in regenerative medicine, cell-based technologies, tissue engineering, and bioprinting applications. Unlike previous 3D bioprinting techniques which depend on external solid materials for structural maintenance or additional process for prefabrication of cell aggregates, the systems and methods described herein use a sheer thinning, crosslinkable, biocompatible hydrogel support medium to provide a structural support role for the printed cell constructs, allowing media provision, and long term culture. Precise maintenance of the structure, for example, mirroring an original computer aided design (CAD) file, can also be achieved even after maturation of the tissue by cell proliferation, differentiation, and extracellular matrix (ECM) production.
Different technologies of bioprinting have been developed, including inkjet or microjet, microextrusion and laser-assisted bioprinting [2, 3]
In a specific embodiment, in the method of the present invention, the printing step is performed through microextrusion
The hydrogel support medium can behave as a viscous fluid during printing and be resistant to flow before and after printing. For example, initially, the hydrogel support medium is in a flow-resistant or solid-like state before being printed with the bioink. The hydrogel support medium becomes fluidized under the increased shear stress caused by printing the bioink into the hydrogel support medium. Then, after the printing is finished the hydrogel support medium can be polymerized to form a solid-like stable support medium.
The extracellular matrix derived hydrogel is cytocompatible and, upon degradation produces substantially non-toxic products.
In a preferred embodiment, the living cells are added to the (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide prior to the printing of the bioink and the polymerization reaction is performed subsequently. In some embodiments, a culture medium in which the hydrogel support medium can be cultured can be provided after the bioink is printed and the hydrogel support medium is further crosslinked.
The culture medium can in addition include bioactive agents for promoting growth and/or differentiation of the cells of the printed bioink. For example, the culture medium can include a cell differentiation medium.
In some embodiments, the hydrogel support medium can include a plurality of hydrogel particles that include a plurality of crosslinkable biodegradable extracellular matrix derived polymer macromers. The hydrogel particles can have an average diameter of about 100 nm to about 100 pm. In particular embodiment, the crosslinkable biocompatible extracellular matrix derived hydrogel support medium of the invention comprised a combination of natural polymer such as hyaluronic acid and collagen.
The natural polymer (macromers) include a plurality of acrylated and/or methacrylated natural polymer macromers, preferably methacrylated polymers. The acrylated and/or methacrylated, polymer macromers extracellular matrix derived polymers are advantageously comprised of a combination of collagen and hyaluronic acid.
The degree of (meth) acrylation of the natural polymer macromers such as collagen and hyaluronic acid is advantageously comprised between 5 to 75%, preferably between 15 and 70%.
In particular embodiment, the degree of (meth) acrylation of collagen is advantageously comprised between 40 to 70% more preferably between 40 to 60%. As described in the experimental section, and the degree of methacrylation of collagen is 57% (+/- 15%)
In particular embodiment the degree of (meth) acrylation of hyaluronic acid is advantageously comprised between 10 to 30%. As described in the experimental section, the degree of methacrylation of hyaluronic acid is 20%
The ratio of collagen and hyaluronic acid is adjusted according to the desired properties of the bioink and hence the desired biological function of the 3D scaffold that is ultimately obtained. In sum, hyaluronic acid concentration plays a key role on the overall bioink viscosity and therefore in the printability of the final formulation. According to the concentration of hyaluronic acid used, the concentration of collagen, a major component of the extracellular matrix, is adjusted to enable optimal cell adhesion and survival.
The mass ratio, hyaluronic acid/ collagen, is advantageously comprised between 0.125 and 100, preferably between 5 and 20.
In a specific embodiment, the mass ratio, hyaluronic acid / collagen is 10 (as described in the experimental section)
Preferably, the concentration of collagen in said hydrogel in the range from 0.5 to 5 mg/mL more preferably between 1 and 2 mg/mL and the concentration of hyaluronic acid in said hydrogel is in the range from 0.5 to 50 mg/ml, more preferably between 10 and 20 mg/mL.
Both collagen and hyaluronic acid (CollMA and HAMA, respectively) are advantageously methacrylated, to form methacrylated collagen and methacrylated hyaluronic acid (CollMA and HAMA, respectively) and combined for instance with laminin-derived thiolated peptides. The thiol-ene reaction, between the acrylate groups of CollMA and HAMA, and the thiol moieties of the peptides is achieved before bioprinting. Then, subsequent radical polymerization, between the excess acrylate groups of the methacrylated polymers, of the composite hydrogel is accomplished following bioprinting. This approach enables to separate the optimization regarding printability, namely by changing the CollMA and HAMA concentrations and ratio and the cell active peptide sequence spatial density, drastically increasing the tunability of the developed bioinks and allowing to easily adapt to other peptide sequences or to create complex mixtures and/or gradients.
In a specific embodiment, the adhesion peptide is a laminin derived peptide.
Examples of said laminin derived peptides are the peptides containing the following sequences: YIGSR and IKVAV, that can be flanked by a spacer sequence (i.e. beta alanine or glycine repeats) and that can be flanked by terminal cysteines, at the amino and carboxyl terminal chains.
The density of said peptide within the said extracellular matrix is adjusted with respect to the desired properties of the bioink and hence the desired biological function of the 3D scaffold that is ultimately obtained.
Advantageously, the density inside the final composite hydrogel is comprised between 0.5 mg/mL and 10 mg/mL.
In some embodiments, the plurality of living cells can include progenitor cells, undifferentiated cells, differentiated cells, and/or cancer cells.
The cells may be selected from apical papilla stem cells, human bone marrow cells, human skin fibroblast, human gingival fibroblast, human umbilical vein cells, endothelial cells, neuronal cells, hematopoietic stem cells (FISC), human induced pluripotent stem cells (HiPSC) and mesenchymal stem cells (MFISC).
Advantageously, the gel may comprise a concentration of cells between 0.01 million to 100 million of cells/mL of gel, preferably between 0.05 and to 20 million of cells/mL of gel.
As described in the Experimental data, in the case of neurons the concentration of cell that can be used is 0.05 million of cells/ and for the endothelial the concentration of cell that can be used is 20 M/mL.
According to a first embodiment, the living cells comprise fibroblasts, preferably human skin fibroblasts and/or human umbilical vein endothelial cells.
Advantageously, the sequence of the peptide is YIGSR and the concentration of the peptide in the hydrogel comprised between 0.5 mg/ml and 6 mg/ml.
According to a second embodiment, the living cells are neurons, preferably primary sensory neurons. Advantageously, the sequence of the peptide is IKVAV and the concentration of the peptide in the hydrogel comprised between 0.5 mg/ml and 6 mg/ml.
The polymerization step is performed through crosslinking of the acrylate or methacrylate groups of the acrylated and/or methacrylated natural polymer macromer of the hydrogel particles.
The crosslinking is advantageously performed using UV light or visible light in the presence of adapted photoinitiators (see the review Lim KS et al “Fundamentals and Applications of Photo-Cross-Linking in Bioprinting” Chem. Rev. April 2020 https://doi.org/10.1021/acs.chemrev.9b00812). For example, acrylated and/or methacrylated natural polymer macromers of the hydrogel particles can be photocrosslinked with a photoinitiator that is provided in the hydrogel support medium. The hydrogel particles can be exposed to a light source at a wavelength and for a time to promote crosslinking of the acrylate groups of the polymers and form the photocrosslinked biodegradable hydrogel particles.
A photoinitiator can include any photo-initiator that can initiate or induce polymerization of the acrylate or methacrylate macromer. Examples of the photoinitiator can include camphor Quinone, benzoin methyl ether, 2-hydroxy-2-methyl- 1 -phenyl- 1-propanone, diphenyl(2,4,6-trimethylbenzoyl)phosphine oxide, lithium phenyl-2, 4,6,- trimethylbenzoylphosphinate, benzoin ethyl ether, benzophenone, 9,10-anthraquinone, ethyl-4- N,N-dimethylaminobenzoate, diphenyliodonium chloride, roboflavin and derivatives thereof.
The polymerization conditions are preferably selected in order to obtain a polymerization degree between 70 and 98%.
According to one aspect , the invention provides a method for forming a 3D tissue construct for promoting angiogenesis, wherein the living cells comprise fibroblasts, preferably human skin fibroblasts and/or human umbilical vein endothelial cells and the sequence of the peptide is YIGSR at a concentration of 2 mg/mL.
According to a second aspect, the invention provides a method for forming a 3D tissue construct for promoting neurogenesis, wherein the living cells are neurons, preferably primary sensory neurons and the sequence of the peptide is IKVAV at a concentration of 2 mg/mL.
The printing step can be performed through several techniques as: inkjet, laser and extrusion-based bioprinting. Extrusion-based bioprinting (EBB) is part of the most widespread tools used in additive manufacturing, mainly due to its capabilities for building large volume constructs such as tissue or organ equivalents. This technique offers a large opportunity in terms of bioink as it can be adaptable to a wide range of materials. In some embodiments, the 3D printer is a bioprinter that can dispense the bioink from a cartridge or container in a specific pattern and at specific positions in the hydrogel support medium as directed by a computer aided design (CAD) software in order to form a specific cellular construct, tissue, or organ. In order to fabricate complex tissue constructs, the bioprinter deposits the bioink at precise speeds and in uniform amounts. In some embodiments, a cartridge containing the bioink comprises one dispensing orifice. In various other embodiments, a cartridge comprises from 1 to 100 or more dispensing orifices.
The hydrogel can further include a culture medium for modulating a function or characteristic of a cell and/or for promoting growth and/or differentiation of the cells of the printed bioink. For example, the culture medium can include a cell differentiation medium, such as an osteogenic differentiation media or chondrogenic differentiation media. The hydrogel support medium containing the printed bioink can be provided in the culture medium after crosslinking or before further crosslinking the hydrogel support medium to enhance the stability of the hydrogel support medium.
It will be appreciated that growth factors can be added to the medium to enhance or stimulate cell growth. Examples of growth factors include vascular endothelial growth factor, platelet-derived growth factor, brain derived growth factor, nerve growth factor, transforming growth factor, platelet-derived growth factor, insulin-like growth factor, acid fibroblast growth factor, basic fibroblast growth factor, epidermal growth factor, hepatocytic growth factor, keratinocyte growth factor, and bone morphogenic protein. It will also be appreciated that other agents, such as cytokines, hormones (e.g., parathyroid hormone, parathyroid hormone-related protein, hydrocortisone, thyroxine, insulin), fatty acids (e.g., Omega-3 fatty acids such as al8:3 linolenate), and/or vitamins (e.g., vitamin D), may also be added or removed from the culture medium to promote cell growth.
Advantageously, the gel may comprise between 0.1 and 10 % w/w concentration of hyaluronic acid and collagen, and between 90 and 99.9 % of an aqueous solution comprising lithium, sodium, potassium, rubidium, cesium or ammonium chloride
According to another aspect, the invention also provides a bioink comprising a biocompatible (meth) acrylated extracellular matrix optionally partially crosslinked with an adhesion peptide and a plurality of cells (as above describe), wherein the extracellular matrix is comprised of a combination of hyaluronic acid and collagen, and hyaluronic acid at a mass ratio of hyaluronic acid /collagen comprised between 0.125 and 100, preferably between 5 and 20 BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 - (A) Viscosity evaluation, in function of the shear rate (0.01-500 s 1), of HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite, or HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of YIGSR peptide, or HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of IKVAV peptide. All bioinks were evaluated prior photopolymerization and at 25°C (Average ± SD, n=3). (B) Storage modulus (G’) of polymerized HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite, HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of YIGSR peptide, and HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of IKVAV. All composition contained 0.1 % (w/v) of LAP (Lithium phenyl-2, 4, 6-trimethylbenzoylphosphinate) as photoinitiator. Evaluated following 2 min photopolymerization, measured at 37 °C (Aver±SD, n=3). Statistically significant differences between indicated groups was analyzed by the non-parametric Kruskal- Wallis test, followed by a Dunns post-test (NS denotes Non Significant).
Figure 2 - Fidelity of bioprinting struts composed of HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite, or HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of YIGSR peptide, using a 210 pm microextrusion tip diameter. The thickness of the bioprinted struts was evaluated by image analysis using ImageJ (Aver±SD, n=3). Statistically significant differences between indicated groups were analyzed by the non-parametric Mann- Whitney test (NS denotes Non Significant). The initial tip diameter is indicated in the correspondent graph using dotted line.
Figure 3 - (A) Quantification of percent viability of the live/dead assay with HUVECs cultured inside polymerized HAMA or HAMA /CollMA composite (a), at 24 hrs of culture (Aver±SD, n=4, ND denotes non-significant). (B) Metabolic activity of HUVECs when cultured inside HAMA /CollMA composite gels containing increased concentrations of the YIGSR peptide (0.25 - 2 mg/mL) at days 0, 3, 7 and 14 (b; Aver±SD, n=4, ND denotes non significant.
Figure 4 - Capillary-like structure maturation evaluation of human umbilical vein endothelial cells, in coculture with human skin fibroblasts, inside methacrylated hyaluronic acid (HAMA) and methacrylated type I collagen (CollMA) hydrogels containing increasing concentration of laminin derived peptides (i.e. YIGSR) for number of junctions (A), number of segments (B) and total segment length (C), at 7 and 14 days (Aver±SD, n=4, * and ** denotes p<0.05 and p<0.01, respectively). Figure 5 - (A) Metabolic activity of dorsal root ganglia (DRG) neurons cultured inside polymerized methacrylated hyaluronic acid (HAMA) and methacrylated collagen (CollMA) (at 1 and 0.1% (w/v), respectively) (HAMA/CollMA) and the composite with increased concentration of the IKVAV peptide (0, 1 and 2 mg/mL), at days 0, 3, 7, 14 and 21 (Aver±SD, n=4, ND denotes non-significant). Image analysis quantification (Neurite tracer, ImageJ) of the number of neurites (B), maximum neurite length (C) and neurite total length per neuron (D) (Aver±SD, n=6, ** and *** denotes p<0.01 and pO.001, respectively).
EXAMPLES
Materials and Methods
Hydrogels synthesis
Methacrylation of hyaluronic acid: The procedure was adapted from Poldervaart et al. [22] Briefly, one gram ofhyaluronic acid sodium salt from Streptococcus Equi (1.5-1.8 x 10E6 Da, Sigma-Aldrich, France) was dissolved in 75 mL of mQ grade water, overnight at room temperature. Then, 50 mL of dimethylformamide (Sigma-Aldrich, France) was added and the pH was adjusted to 8 using NaOH 1M. Then, 1.12 mL of methacrylic anhydride (Sigma- Aldrich) was added to the solution the pH was again adjusted to 8 and the solution was left under constant stirring overnight at room temperature. Then, we added 50 mL of NaCl 5M and the volume of the solution was adjusted to 500 mL with mQ grade water. The solution was filtered using a 0.2 pm filter and then dialyzed (12-14 kDa, Sigma-Aldrich) for five days against 5L mQ water and with 2 daily water changes. The obtained solution was then freeze-dried for 5 days, the polymer recovered and stored at -20 °C until further use. Hyaluronic acid methacrylate (HAMA) stock solution was prepared by dissolving the polymer at 2% (w/v) in DMEM HG (Gibco) and then stored at 4 °C until further use. Methacrylation degree was determined using nuclear magnetic resonance (NMR). Briefly, for 1H-NMR a 6% (w/v) solution of HAMA was prepared in D20 (Sigma-Aldrich) and analyzed at 25 °C on a Bruker Avance I NMR spectrometer operating at 400 MHz. 1H NMR spectra were recorded with an acquisition time of 4 sec, a relaxation delay of 2 and 64 scans. Methacrylate modification was determined by integration of the vinyl singlets relative to the ring ofhyaluronic acid.
Methacrylation of type I collagen: The procedure was adapted from Ravichandran et al. [27] Briefly, type I collagen from bovine skin (Sigma-Aldrich) was adjusted to pH 10 using 2N NaOH and kept on ice, under mild agitation. Then, methacrylic anhydride at a molar ratio of 5:1 (with respect to number of lysine amine groups in collagen) was added subsequently drop-wise and left to react for 4 hours. Then the obtained solution was dialyzed (12-14 kDa, Sigma- Aldrich) for five days against 5L mQ water and with 2 daily water changes. The obtained solution was then freeze-dried for 5 days, and the polymer recovered and stored at -20 °C until further use. The methacrylated type I collagen (CollMA) stock solution was prepared at 6 mg/mL in 0.02 M Acetic acid and stored at 4 °C until further use.
Methacrylation degree was determined using the Tri-nitro benzene sulfonic acid (TNBS) assay [28] Briefly, neutralized native collagen or CollMA were dissolved in 1 mL of carbonate buffer (0.1M, pH 8.5) at 0.1 mg/mL NaHC03 (pH 8.5). Then to 500 pL of collagen or CollMA solution, we added 250 pL of a 1:500 dilution, in carbonate buffer, of TNBSA solution (5% w/v, Sigma-Aldrich) and let to react for 2 hours at 37°C. The reaction was then stopped by the addition of 250pL of 10% (w/v) of SDS (Sigma-Aldrich) and 125pL of HC1 1M. The OD335 of both the blanc, collagen and CollMA was determined using a UV-Vis spectrophotometer (Nanodrop, ThermoFisher). The percentage of remaining free primary amines of CollMA was calculated in relation to the total amount of the pristine collagen. The degree of methacrylation was then determined using the loss of primary amines percentage loss (% DM).
Bioinks preparation
HAMA at 1% (w/v) was prepared by diluting HAMA 2% (w/v) using DMEM HG (Gibco) and mixed with 0.1 % (w/v) of the photonitiator tithium phenyl-2, 4, 6-trimethylbenzoylphosphinate (LAP, TCI Chemicals). CollMA 0.1% (w/v) was prepared by diluting the stock 0.6% (w/v) CollMA solution (dissolved in 0.02 M Acetic acid) with DMEM HG (Gibco), neutralizing with NaOH 0.1M and mixed with 0.1 % (w/v) of LAP photonitiator. The adhesion peptides Cys- (Beta-Ala)-Ile-Lys-Val-Ala-Val-(Beta-Ala)-Cys (IKVAV) and Cys-(Beta-Ala)-Tyr-Ile-Gly- Ser-Arg-(Beta-Ala)-Cys (YIGSR) (Biomatik, Canada) were dissolved at 25 mg/mL in acetic acid 0.02 M aliquoted and stored at -20 °C untill further use.
HAMA/CollMA, 1 and 0.1% (w/v) respectively, composite bioink was obtained by diluting and neutralizing the stock 0.6% (w/v) CollMA, then HAMA 2% (w/v) was added, the solution was mixed with 0.1 % (w/v) of LAP and the volume adjusted with DMEM HG. The peptide solution containing the laminin derived peptides was homogeneously added, at defined final concentrations, to the composite polymer solution prior use. All formulation were prepared fresh and kept protected from light. Rheological characterization
For viscosity determination, different formulations of the hydrogels were prepared, loaded in a Kinexus pro+ rheometer and rotational shear-viscosity measurements were performed in flow mode with shear rate ranging from 0.01 to 500 s 1. Measurements were performed in triplicate and at 25 °C.
For storage and loss modulus determination, different formulations of the hydrogels were prepared, loaded and directly polymerized (a 2w, 365 nm UV source and an exposure time of 2 minutes) in a Kinexus pro+ rheometer (Malvern Instruments, UK), and stabilized at 37°C prior to the determination of the storage moduli (G’) and loss moduli (G”). Frequency sweeps were performed at a constant strain (0.1%) in the angular frequency range 0.1-100 rad/s. Measurements were performed in triplicate and at 37 °C.
Primary cell culture
Human umbilical vein endothelial cells (HUVECs) were isolated as previously described [29] HUVECS were cultured, under standard conditions, in IMDM (Gibco), 20% SVF + ECGS/H, until passage 9. As means to enable their visualization by fluorescent microscopy cells were stably transduced with lenti virus bearing the RFP gene, under the EFla promoter sequence (Vectalys, France), following the manufacturer’s instructions. Infection efficiency was evaluated by flow cytometry and was determined to be superior to 98% (data not shown). Human skin fibroblasts were isolated as previously described [30], and cultured, under standard conditions, in DMEM:F12 (Gibco), 20% (v/v) FBS, until passage 10.
Primary sensory neurons were obtained from dorsal root ganglion (DRG) from healthy 5-9 weeks old Wistar rats as previously described [31] Briefly, following animal sacrifice with C02, spinal columns were removed and opened from the caudal to the rostral in order to expose the DRGs. They were individually harvested and digested with 1 mg/mL Collagenase Type IV (Gibco®) for 2 h at 37 °C. Subsequently, digestion product was washed twice with culture medium supplemented with 2 % (v/v) B-27 Serum-Free Supplement® (B-27, Gibco®) and 1 % (v/v) Penicilin Streptomycin, and mechanically dissociated using fire-polished glass Pasteur pipettes (full diameter and ½ diameter). The cell suspension was finally washed three more times with medium and resuspended in complete culture medium. Nerve growth factor (NGF, 0.1 pg/mL, Sigma Aldrich) was added to the culture medium at 48 hrs of culture.
Viability evaluation Cell viability percentage was determined using the live-dead assay. Briefly, at defined time points samples were stained with calcein-AM (“live”, 1 pL/mL, Invitrogen) and ethidium homodimer (“dead”, 4 pL/mL, Invitrogen) for 45 min, following manufacturer’s instructions. Samples were then analysed by confocal laser-scanning microscopy (SPE7, Leica Microsystems). Image analysis of the percentage of viable cells was performed using ImageJ (n=4 replicates) by determining the total number of cells per field (cells positive for calcein + cells positive for ethidium homodimer) and determining the percentage of “live” cells (cells positive for calcein).
Cell imaging
The capacity of HUVEC cells to form capillary like structures in 3D was assessed by confocal imaging of RFP+ HUVEC cells at days 7 and 14, inside the different bioinks in 3D culture or after 3D bioprinting. For the 3D culture a 150 pm Z axis height was acquired using confocal microscopy. For the bioprinted structures a 500 pm Z axis height was acquired.
The capacity of DRG cells to extend neurites was assessed by confocal imaging following Calcein-AM (1 pL/mL, Invitrogen) staining for 45 minutes at day 14, inside the different bioinks in 3D culture or after 3D bioprinting. For the 3D culture a 150 pm Z axis height was acquired using confocal microscopy. For the bioprinted structures a 500 pm Z axis height was acquired.
Cell metabolic activity evaluation
Metabolic activity of ECS and DRGs, when cultured inside the hydrogels, was evaluated using a resazurin-based assay [32] Briefly, cells were seeded inside the different formulation hydrogels at 2 million cells/mL for ECS and 1 million/mL for DRGs, respectively, in a 96 well plate. 150 pL of culture medium containing resazurin (0.01 mg/mL) was added to each well and the microplate was incubated at 37 °C for 3 h. Subsequently, 100 pL of supernatant was transferred to another 96 wells microplate and fluorescence was measured (exc = 530 nm, em = 590 nm, Victor X3, Perkin Elmer). Metabolic activity was measured at days 0, 3, 7 and 14 for ECS and days 0, 3, 7, 14 and 21 for DRGs, respectively, (n=6 replicates) % metabolic activity was calculated in relation to day 0.
Bioprinting
The 3D Discovery bioprinting platform (RegenHU) was used in this work. It is composed of two independent pressure activated microextrusion heads and two microvalve heads, automated tip length and substrate height calibration and integrated ultraviolet (UV, 365 nm, 2W) photopolymerization diode. Also, the integrated computer assisted design software (BioCAD, RegenHU) enable to create the desired geometry of printing. The chosen geometry consisted on 5 parallel lines, spaced of 2.5 mm, and intercrossed with same geometry with a 90-degree angle rotation. The same geometry was repeated to a total of 4 layers. The geometry was adapted to print inside 6 well plate wells and repeated in series.
In the case of HUVEC+HSF bioprinting, a conic microextrusion tip of 0.21mm diameter was used. Both HUVECs and HSF were both loaded together inside the designated bioink (containing or not the YIGSR peptide), both at 10 million/mL. The ink was homogenized and loaded inside the bioprinting syringe. The bioprinting process consisted on the deposition of the first layer with a pressure of 0.008 MPa, speed of 140 mm/s and a distance to the substrate of 0.075 mm. Then the first layer was polymerized during 15 seconds using the UV source. The second perpendicular layer was then deposited using the same pressure and speed conditions at a distance of 0.1 mm of the first layer. The process was then repeated for a total of 4 layers, before adding 3 mL of cell culture media.
In the case of DRG bioprinting, a conic microextrusion tip of 0.41mm diameter was used. DRG were loaded inside the designated bioink (containing or not the IKVAV peptide), at 50 000 cells/mL. The ink was homogenized and loaded inside the bioprinting syringe. The bioprinting process consisted on the deposition of the first layer with a pressure of 0.005 MPa, speed of 160 mm/s and a distance to the substrate of 0.075 mm. Then the first layer was polymerized during 15 seconds using the UV source. The second perpendicular layer was then deposited using the same pressure and speed conditions at a distance of 0.1 mm of the first layer. The process was then repeated for a total of 4 layers, before adding 3 mL of cell culture media.
Image analysis
The capacity of HUVECs to form capillary -like structures in 3D culture was quantified by image analysis of confocal stacking images using the Angiogenesis Analyzer [33] plugin for Image! The software allowed to assess relevant parameters as number of junctions, number of segments and total segment length.
HUVEC evaluation upon 3D printing was achieved by 3D image analysis of the volumetric confocal acquisition, at day 14, and using the Imaris software (9.0, Bitplane, UK). The 3D segmentation of independent structures, inside the bioprinted matrix, were evaluated in terms of their length in the Z axis (Bounding Box length Z axis), as to further demonstrate the capacity of HUVECs to migrate inside the structure, and the volume of each one of these independent 3D structures (volume bounding box), as means to evaluate the capacity to interconnect and form a complex network.
The capacity of DRGs to extend neurites in 3D culture was quantified by image analysis of confocal stacking images using the NeuriteQuant [34] plugin for Image! The software allowed to assess relevant parameters as number of neurites per neuron, maximum neurite length and neurite total length/per neuron. DRG evaluation upon 3D printing was achieved by 3D image analysis of the volumetric confocal acquisition, at day 14, and using the Imaris software (9.0, Bitplane, UK). The 3D segmentation of independent structures, inside the bioprinted matrix, were evaluated by segmental analysis and allow to determine the dendrite length distribution for different compositions of bioink.
Statistical analysis
Using the Graphpad Prism 5.0 software, a D’Agostino and Pearson omnibus normality test was used in order to test if data obeyed to a Gaussian distribution. Statistically significant differences between several groups were analysed by the non-parametric Kruskal -Wallis test, followed by a Dunns post-test. The non-parametric Mann-Whitney test was used to compare two groups. A p value lower than 0.05 was considered to be statistically significant.
Results
Hydrogel Characterization
The chemical approach for the creation of a printable composite hydrogel was based on an initial methacrylation of the base polymers, namely hyaluronic acid and type I collagen, both natural components of the extracellular matrix. The inclusion of laminin-derived peptides was achieved by the inclusion of cysteine groups at both the extremities of the peptides, what permits to graft, via a thiol-ene UV initiated radical addition, the peptides to the methacrylated matrix. Also, the excess methacrylate moieties enables the photopolymerization of the matrix by radical polymerization via UV irradiation.
Chemical characterization of both CollMA and HAMA consisted on the quantification of acrylate groups introduced in the correspondent polymer backbone. CollMA showed a methacrylation degree was 57 ± 15 % (n=3), as determined by TNBS assay. For HAMA, NMR analysis by the integration of the vinyl singlets (1H each) relative to the ring of hyaluronic acid (10H) showed a degree of methacrylation of 20±1% (n=3). The following HAMA (1% (w/v)) and CollMA (0.1% (w/v)) formulations were selected based on previous unpublished results as a base for this study. The obtained results were based on the survival and proliferation of HUVECs and DRGs and on their printability character.
Shear-viscosity measurements (Figure 1A) were performed, prior photopolymerization, for HAMA (1% (w/v)) CollMA (0.1% (w/v)) alone or containing the highest peptide formulations (2 mg/mL of YIGSR or IKVAV). In the shear range tested, all formulations had similar viscosity values and showed a shear-thinning character. This fact is particularly relevant as shows that with the selected chemical peptide grafting approach one can achieve independent formulations, while maintaining viscosity unaltered.
Rheological characterization for polymerized methacrylated hyaluronic acid (HAMA) at 1% (w/v) show a G’ of 202±25 Pa (at 37°C), for methacrylated type 1 collagen (CollMA) at 0.1% (w/v) show a G’ of 114±12 Pa (at 37°C), for HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite show a G’ of 120±12 Pa (at 37°C), for HAMA(1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of YIGSR peptide a G’ of 197±85 Pa (at 37°C), and for HAMA (1% (w/v)) and CollMA (0.1% (w/v)) composite with 2 mg/mL of IKVAV a G’ of 169±24 Pa (at 37°C). Based on the unique chemistry, we could establish that by using the optimized HAMA and CollMA ratios, and based on the peptide grafting via two thiols per molecule, we could establish bioinks viscosity and rheology independently of its peptide inclusion/concentration. This has great implications in terms of bioink optimization, as rheological properties can have great impact on cell maturation.
Both HAMA (1% (w/v)) and CollMA (0.1% (w/v)) based hydrogels showed modest G’ values, with no significant differences when the composite (HAMA/CollMA) was tested. Additionally, and as observed in Figure IB, the G’ values were at the same range for HAMA (1% (w/v)) and CollMA (0.1% (w/v)) alone or for the highest peptide formulations (2 mg/mL of YIGSR or IKVAV). In all formulations tested the storage modulus (G’) was in the range of 100 to 200 Pa.
Additionally, we have also evaluated bioink fidelity using the described bioprinting conditions. As observed in Figure 2 bioprinting strut dimensions show that the gel could respect the expected dimensions with an observable dispersion of 1.3x in relation to the expected dimensions and where no significant differences could be observed for the formulation with or without peptide. Additionally, the established conditions enabled to reach filament stability on air and to achieve the fabrication of complex structures, like a hollow tube (data not shown) or a parallelepiped (data not shown).
HUVEC culture in 3D gels
Based on previous results of the optimization of the concentration of HAMA in the behavior of encapsulated HUVEC cells (data not shown) the concentration of 1% (w/v) was established as the basis for this study. Also, it was previously show that the inclusion of CollMA could further support HUVECs migration. Here, and as observed in Figure 3A, it could be shown that in the present formulations and polymerization conditions that cells showed a percent viability of 67 and 77%, for HAMA and HAMA/CollMA, respectively, at 24 hours of culture. Based on this profile, the HAMA/CollMA formulation were selected in the subsequent studies. As means to evaluate the impact of the inclusion of the laminin-derived YIGSR peptide on HUVECs their metabolic activity progression up to 14 days of culture was then evaluated. As seen in Figure 3B, no significant differences could be observed for the different HAMA/CollMA formulations containing increased concentrations of the YIGSR peptide. Also, it could be seen a significant progression of the metabolic activity of all formulations towards time, up to 14 days.
The maturation of endothelial cells (HUVECs), when in coculture with HSF, inside the composite hydrogels containing different concentrations of the laminin-derived peptide YIGSR was then evaluated. For all formulations tested no significant maturation was observed at day 7. At day 14 of culture a significant increase on capillary-like structures was observed for the formulation containing the highest YIGSR concentration (2 mg/mL, Figure 4). The quantification of the capillary-like structure was achieved by image analysis (Angiogenesis Analyser, ImageJ) and as seen in Figure 4 A,B,C, a significant increase in terms of the number of junctions, number of segments and total segment length was observed at 14 days of culture and for the HAMA /CollMA + 2 mg/mL YIGSR peptide.
Bioprinting of HUVEC cells
The obtained results allowed the selection of the HAMA/CollMA + 2 mg/mL YIGSR peptide as optimal formulation for the maturation of HUVEC cells and was compared with the basal formulation, HAMA/CollMA. The chosen assisted computer design (CAD) consisted on 5 parallel lines, spaced of 2.5 mm, and intercrossed with same geometry with a 90 degree angle rotation. The same geometry was repeated to a total of 4 layers. Both the speed of printing (140 mm/s), the diameter of the tip (0.21 mm) and the distance between the tip and the substrate (0.075 mm) were controlled in order to attain a reproducible geometry. Additionally, the capacity of the composite hydrogel containing YIGSR was tested to further sustain the maturation of HUVECs in coculture with HSFs, following bioprinting and using the same CAD. Both composition permitted the microextrusion bioprinting of the scaffold containing both HUVECs and HSFs. Additionally, one could observe that the composition containing the YIGSR peptide could further support HUVECs maturation, particularly relevant at day 14.
At 14 days post printing, the HUVEC network was then evaluated by confocal microscopy on a thickness of 0.5 mm, and reconstructed using imaging analysis software (Imaris) in order to quantify its interconnectivity and dimensions. Software analysis enabled to identify and create a tridimensional box that includes each interconnected structure (bounding box). Then, the tridimensional dimensions of this bounding box give a quantification of the interconnection and distribution inside the bioprinted matrix. Both the thickness (Z axis length) and interconnection structure volume was shown to be significantly increased for the bioprinted structure containing the laminin-derived YIGSR peptide.
Dorsal root ganglia sensory neuron culture in 3D gels
Based on the previous results of the optimal bioink formulation for HUVECs the same base of HAMA/CollMA was established to which a new laminin-derived peptide, IKVAV was combined. Important to refer that the same YIGSR formulations were equally tested for DRG culture and were found no optimal (data not shown).
Again, and means to evaluate the impact of the inclusion of the laminin-derived IKVAV peptide on DRGs their viability at 7 days of culture and metabolic activity progression up to 21 days of culture were evaluated. No signs of cell mortality could be observed, for all formulations tested. Additionally, the metabolic activity follow-up, up to day 21, show no significant differences between formulations or a decrease in time (Figure 5A), indicating that in the use conditions the different composition could support the 3D culture of DRG neurons. Finally, based on image analysis we quantified the number of neurites, maximum neurite length and total neurite length per neuron (Figure 5B, C and D, respectively). We could observe a significant increase on all parameters for the highest peptide concentration, in relation with the formulation without peptide, and consistent with a dose response effect.
Bioprinting of dorsal root ganglia sensory neurons
The chosen CAD consisted on 5 parallel lines, spaced of 2.5 mm, and intercrossed with same geometry with a 90-degree angle rotation, as before. The same geometry was repeated to a total of 4 layers. Both the speed of printing (160 mm/s), the diameter of the tip (0.41 mm) and the distance between the tip and the substrate (0.075 mm) were controlled in order to attain a reproducible geometry. Live-dead assay staining assessed that the bioprinting process did not significantly affect the viability of the printed DRG neurons, as observe 14 days post printing, and the inclusion of the IKVAV peptide could improve neurite formation. As such, image analysis of calcein stained DRG neurons at 14 days post printing, was performed and the formation of dendrite processes was quantified using segment analysis with Imaris software. Dendrite length quantification could assert that for the formulation containing 2 mg/mL of IKVAV peptide a significant increase could be observed.
It is currently accepted that different matrix compositions have to be determined to individual cell types and for specific biological applications. In this sense, bioink formulations based on modular approaches considering at the same time the printability and rigidity (enabling to easily adapt components concentrations), while enabling to adjust for optimal cell adhesion sequence density or even on the combination or gradient of sequences, represents an added value in future developments in bioprinting. The approach follows this line of thought, enabling inherent modularity and tunability while respecting the aimed application simplicity.
Phage display technology and the rampant field of biotechnology have already identified a wide range of small peptide sequences with inherent biological activity [56] The simple approach of the invention enables to easily adapt to other bioactive peptide sequences, opening the field of application of such bioinks to a wide range of cell types and applications. The adaptation of these novel formulations to other major bioprinting technologies, namely inkjet or laser-assisted bioprinting, are currently being explored opening the range of applications of this approach.
References
[1] Mandrycky C, Wang Z, Kim K, Kim DH. 3D bioprinting for engineering complex tissues. Biotechnol Adv 2016;34:422-34.
[2] Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nat Biotechnol 2014;32:773-85.
[3] Holzl K, Lin S, Tytgat L, Van Vlierberghe S, Gu L, Ovsianikov A. Bioink properties before, during and after 3D bioprinting. Biofabrication 2016;8: 032002. [4] Malda J, Visser J, Melchels FP, Jiingst T, Hennink WE, Dhert WJA, Groll J, Hutmacher DW. 25th Anniversary Article: Engineering Hydrogels for Biofabrication. Advanced Materials 2013;25:5011-28.
[5] Donderwinkel I, van Hest JCM, Cameron NR. Bio-inks for 3D bioprinting: recent advances and future prospects. Polymer Chemistry 2017;8:4451-71.
[6] Hospodiuk M, Dey M, Sosnoski D, Ozbolat IT. The bioink: A comprehensive review on bioprintable materials. Biotechnology Advances 2017;35:217-39.
[7] Gungor-Ozkerim PS, Inci I, Zhang YS, Khademhosseini A, Dokmeci MR. Bioinks for 3D bioprinting: an overview. Biomaterials Science 2018;6:915-46.
[8] Gopinathan J, Noh I. Recent trends in bioinks for 3D printing. Biomater Res 2018;22:11.
[9] Park KD, Wang X, Lee JY, Park KM, Zhang S, Noh I. Research trends in biomimetic medical materials for tissue engineering: commentary. Biomaterials Research 2016;20:8.
[10] Keriquel V, Oliveira H, Remy M, Ziane S, Delmond S, Rousseau B, Rey S, Catros S, Amedee J, Guillemot F, Fricain JC. In situ printing of mesenchymal stromal cells, by laser- assisted bioprinting, for in vivo bone regeneration applications. Sci Rep 2017;7:1778.
[ 11 ] Y ang X, Lu Z, Wu H, Li W, Zheng L, Zhao J. Collagen-alginate as bioink for three- dimensional (3D) cell printing based cartilage tissue engineering. Materials Science and Engineering: C 2018;83:195-201.
[12] Jang J, Park JY, Gao G, Cho D-W. Biomaterials-based 3D cell printing for next- generation therapeutics and diagnostics. Biomaterials 2018;156:88-106.
[13] Kleinman HK, Klebe RJ, Martin GR. Role of collagenous matrices in the adhesion and growth of cells. J Cell Biol 1981;88:473-85.
[14] Lee YB, Polio S, Lee W, Dai G, Menon L, Carroll RS, Yoo SS. Bio-printing of collagen and VEGF-releasing fibrin gel scaffolds for neural stem cell culture. Exp Neurol 2010;223:645-52.
[15] Mazzocchi A, Devarasetty M, Huntwork R, Soker S, Skardal A. Optimization of collagen type I-hyaluronan hybrid bioink for 3D bioprinted liver microenvironments. Biofabrication 2018;11:015003.
[16] Fraser JR, Laurent TC, Laurent UB. Hyaluronan: its nature, distribution, functions and turnover. J Intern Med 1997;242:27-33.
[17] Li H, Xue Y, Jia B, Bai Y, Zuo Y, Wang S, Zhao Y, Yang W, Tang H. The preparation of hyaluronic acid grafted pullulan polymers and their use in the formation of novel biocompatible wound healing film. Carbohydrate Polymers 2018;188:92-100. [18] Chan DD, Xiao WF, Li J, de la Motte CA, Sandy JD, Plaas A. Deficiency of hyaluronan synthase 1 (Hasl) results in chronic joint inflammation and widespread intra- articular fibrosis in a murine model of knee joint cartilage damage. Osteoarthritis Cartilage 2015;23:1879-89.
[19] Murakami T, Otsuki S, Okamoto Y, Nakagawa K, Wakama H, Okuno N, Neo M. Hyaluronic acid promotes proliferation and migration of human meniscus cells via a CD44- dependent mechanism. Connect Tissue Res 2019;60: 117-27.
[20] Toole BP. Hyaluronan: from extracellular glue to pericellular cue. Nat Rev Cancer 2004;4:528-39.
[21] Pardue EL, Ibrahim S, Ramamurthi A. Role of hyaluronan in angiogenesis and its utility to angiogenic tissue engineering. Organogenesis 2008;4:203-14.
[22] Poldervaart MT, Goversen B, de Ruijter M, Abbadessa A, Melchels FPW, Oner FC, Dhert WJA, Vermonden T, Alblas J. 3D bioprinting of methacrylated hyaluronic acid (MeHA) hydrogel with intrinsic osteogenicity. PLoS One 2017;12:e0177628.
[23] Sakai S, Ohi H, Hotta T, Kamei H, Taya M. Differentiation potential of human adipose stem cells bioprinted with hyaluronic acid/gelatin-based bioink through microextrusion and visible light-initiated crosslinking. Biopolymers 2018;109:e23080.
[24] Farkas JE, Monaghan JR. A brief history of the study of nerve dependent regeneration. Neurogenesis (Austin) 2017;4:el302216.
[25] Grant DS, Tashiro K-I, Segui-Real B, Yamada Y, Martin GR, Kleinman HK. Two different laminin domains mediate the differentiation of human endothelial cells into capillary- like structures in vitro. Cell 1989;58:933-43.
[26] Patel R, Santhosh M, Dash JK, Karpoormath R, Jha A, Kwak J, Patel M, Kim JH. Ile-Lys-Val-ala-Val (IKVAV) peptide for neuronal tissue engineering. Polymers for Advanced Technologies 2019;30:4-12.
[27] Ravichandran R, Islam MM, Alarcon El, Samanta A, Wang S, Lundstrom P, Hilbom J, Griffith M, Phopase J. Functionalised type-I collagen as a hydrogel building block for bio-orthogonal tissue engineering applications. Journal of Materials Chemistry B 2016;4:318-26.
[28] Tronci G, Doyle A, Russell SJ, Wood DJ. Triple-helical collagen hydrogels via covalent aromatic functionalisation with 1,3-phenylenediacetic acid. Journal of Materials Chemistry B 2013;1:5478-88. [29] Bordenave L, Baquey C, Bareille R, Lefebvre F, Lauroua C, Guerin V, Rouais F, More N, Vergnes C, Anderson JM. Endothelial cell compatibility testing of three different Pellethanes. J Biomed Mater Res 1993;27:1367-81.
[30] Germain L, Guignard R, Rouabhia M, Auger FA. Early basement membrane formation following the grafting of cultured epidermal sheets detached with thermolysin or Dispase. Bums 1995;21:175-80.
[31] Malin SA, Davis BM, Molliver DC. Production of dissociated sensory neuron cultures and considerations for their use in studying neuronal function and plasticity. Nat Protoc 2007;2:152-60.
[32] O'Brien J, Wilson I, Orton T, Pognan F. Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity. Eur J Biochem 2000;267:5421-6.
[33] Carpentier G. ImageJ contribution: angiogenesis analyzer2012.
[34] Dehmelt L, Poplawski G, Hwang E, Halpain S. NeuriteQuant: an open source toolkit for high content screens of neuronal morphogenesis. BMC Neurosci 2011;12:100.
[35] Groll J, Boland T, Blunk T, Burdick JA, Cho DW, Dalton PD, Derby B, Forgacs G, Li Q, Mironov VA, Moroni L, Nakamura M, Shu W, Takeuchi S, Vozzi G, Woodfield TB, Xu T, Yoo JJ, Mai da J. Biofabrication: reappraising the definition of an evolving field. Biofabrication 2016;8:013001.
[36] Groll J, Burdick JA, Cho DW, Derby B, Gelinsky M, Heilshom SC, Jiingst T, Malda J, Mironov VA, Nakayama K, Ovsianikov A, Sun W, Takeuchi S, Yoo JJ, Woodfield TBF. A definition of bioinks and their distinction from biomaterial inks. Biofabrication 2018;11:013001.
[37] Jungst T, Smolan W, Schacht K, Scheibel T, Groll J. Strategies and Molecular Design Criteria for 3D Printable Hydrogels. Chem Rev 2016;116: 1496-539.
[38] Ashammakhi N, Ahadian S, Xu C, Montazerian H, Ko H, Nasiri R, Barros N, Khademhosseini A. Bioinks and bioprinting technologies to make heterogeneous and biomimetic tissue constructs. Materials Today Bio 2019;1:100008.
[39] Gopinathan J, Noh I. Recent trends in bioinks for 3D printing. Biomaterials Research 2018;22: 11.
[40] Burdick JA, Prestwich GD. Hyaluronic acid hydrogels for biomedical applications. Adv Mater 2011;23:H41-56. [41] Jiang Y, Chen J, Deng C, Suuronen EJ, Zhong Z. Click hydrogels, microgels and nanogels: emerging platforms for drug delivery and tissue engineering. Biomaterials 2014;35:4969-85.
[42] Hoyle CE, Bowman CN. Thiol-ene click chemistry. Angew Chem Int Ed Engl 2010;49:1540-73.
[43] Sasmal P, Datta P, Wu Y, Ozbolat IT. 3D bioprinting for modelling vasculature. Microphysiological Systems; November 20182018.
[44] Jia W, Gungor-Ozkerim PS, Zhang YS, Yue K, Zhu K, Liu W, Pi Q, Byambaa B, Dokmeci MR, Shin SR, Khademhosseini A. Direct 3D bioprinting of perfusable vascular constructs using a blend bioink. Biomaterials 2016;106:58-68.
[45] Kolesky DB, Homan KA, Skylar-Scott MA, Lewis JA. Three-dimensional bioprinting of thick vascularized tissues. Proceedings of the National Academy of Sciences 2016;113:3179.
[46] Kolesky DB, Truby RL, Gladman AS, Busbee TA, Homan KA, Lewis JA. 3D Bioprinting of Vascularized, Heterogeneous Cell-Laden Tissue Constructs. Advanced Materials 2014;26:3124-30.
[47] Miller JS, Stevens KR, Yang MT, Baker BM, Nguyen D-HT, Cohen DM, Toro E, Chen AA, Galie PA, Yu X, Chaturvedi R, Bhatia SN, Chen CS. Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues. Nature Materials 2012;11:768.
[48] Lee VK, Lanzi AM, Haygan N, Yoo SS, Vincent PA, Dai G. Generation of Multi- Scale Vascular Network System within 3D Hydrogel using 3D Bio-Printing Technology. Cell Mol Bioeng 2014;7:460-72.
[49] Caniggia I, Liu J, Han R, Wang J, Tans well AK, Laurie G, Post M. Identification of receptors binding fibronectin and laminin on fetal rat lung cells. American Journal of Physiology-Lung Cellular and Molecular Physiology 1996;270:L459-L68.
[50] Stupack DG, Cheresh DA. Integrins and Angiogenesis. Current Topics in Developmental Biology: Academic Press; 2004. p. 207-38.
[51] Beachley VZ, Wolf MT, Sadtler K, Manda SS, Jacobs H, Blatchley MR, Bader JS, Pandey A, Pardoll D, Elisseeff JH. Tissue matrix arrays for high-throughput screening and systems analysis of cell function. Nature Methods 2015;12:1197.
[52] Schmidt CE, Leach JB. Neural Tissue Engineering: Strategies for Repair and Regeneration. Annual Review of Biomedical Engineering 2003;5:293-347. [53] Lozano R, Stevens L, Thompson BC, Gilmore KJ, Gorkin R, Stewart EM, in het Panhuis M, Romero-Ortega M, Wallace GG. 3D printing of layered brain-like structures using peptide modified gellan gum substrates. Biomaterials 2015;67:264-73.
[54] Ning L, Sun H, Lelong T, Guilloteau R, Zhu N, Schreyer DJ, Chen X. 3D bioprinting of scaffolds with living Schwann cells for potential nerve tissue engineering applications. Biofabrication 2018;10:035014.
[55] Quaegebeur A, Lange C, Carmeliet P. The Neurovascular Link in Health and Disease: Molecular Mechanisms and Therapeutic Implications. Neuron 2011;71:406-24.
[56] Hamley IW. Small Bioactive Peptides for Biomaterials Design and Therapeutics. Chemical Reviews 2017;117:14015-41.

Claims

CLAIMS:
1. A method for forming a cell specific 3D cell or tissue construct with a desired biological function comprising the steps of: a) Providing a (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel support medium; b) Reacting said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel with a thiol flanked functionalized adhesion peptide to obtain a (meth) acrylate crosslinkable biocompatible extracellular matrix derived hydrogel partially crossbnked with said peptide, via a thiol-ene reaction; cl) Either, combining said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crossbnked with said peptide with a plurality of specific living cells to provide a printable bioink, dl) Printing the said bioink into a first layer of printed bioink with a defined design; el) Submitting the said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crossbnked bioink; and fl) Repeating the printing step and said polymerization step until a 3D construct with the desired shape is obtained c2) Or printing said bioink into a first layer of printed bioink with a defined design; d2) Submitting said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crossbnked bioink; and e2) Repeating the printing step and said polymerization step until a 3D construct with the desired shape is obtained, and f2), adding to said printed (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crossbnked with said peptide a plurality of specific living cells, g) Allowing the growth of the living cells in 3D-construct obtained thereby under suitable conditions until the desired biological function is achieved; wherein the hydrogel support medium, the sequence of the said peptide, the ratio of the said peptide to the said extracellular matrix derived hydrogel and the polymerization conditions are selected with respect to cell specificity so as to obtain a 3D tissue construct with the desired biological function.
2. The method according to claim 1 comprising the following steps: al) Providing a (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel support medium; bl) Reacting said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel with a thiol flanked functionalized adhesion peptide to obtain a (meth) acrylate crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide, via a thiol-ene reaction; cl) Combining said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide with a plurality of specific living cells to provide a printable bioink, dl) Printing the said bioink into a first layer of printed bioink with a defined design; el) Submitting the said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crosslinked bioink; and fl) Repeating the printing step and said polymerization step until a 3D construct with the desired shape is obtained, and gl) Allowing the growth of the living cells in 3D-construct obtained thereby under suitable conditions until the desired biological function is achieved.
3. The method according to claim 1 comprising the following steps: a2) Providing a (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel support medium; b2) Reacting said (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel with a thiol flanked functionalized adhesion peptide to obtain a (meth) acrylate crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide, via a thiol-ene reaction c2) Printing said bioink into a first layer of printed bioink with a defined design; d2) Submitting said first layer of bioink to a polymerization reaction, under controlled conditions in order to provide a crosslinked bioink; and e2) Repeating the printing step and said polymerization step until a 3D construct with the desired shape is obtained, and f2), adding to said printed (meth) acrylated crosslinkable biocompatible extracellular matrix derived hydrogel partially crosslinked with said peptide a plurality of specific living cells, and g2) Allowing the growth of the living cells in 3D-construct obtained thereby under suitable conditions until the desired biological function is achieved.
4. A method according to anyone of the preceding claims, wherein the polymerization conditions are selected in order to obtain a polymerization degree between 70 and 98 %.
5. The method according to claim 1 to 5, wherein the crosslinkable biocompatible extracellular matrix derived hydrogel support medium is comprised of a combination of the natural polymer macromers such as hyaluronic acid and collagen at a mass ratio comprised between 0.125 and 100, preferably between 5 and 20.
6. The method according to claim 5, wherein the concentration of collagen in said hydrogel is in the range from 0.5 to 5 mg/mL and the concentration of hyaluronic acid in said hydrogel is in the range from 0.5 to 50 mg/mL.
7. A method according to claims 5 or 6, wherein the degree of (meth) acrylation of the natural polymer macromers is comprised between 5 to 75 %.
8. A method according to claim 7, wherein the degree of (meth) acrylation of collagen is comprised between 40 to 70%.
9. A method according to claim 7 or 8, wherein the degree of (meth) acrylation of hyaluronic acid is comprised between 10 to 30%.
10. The method according to anyone of the preceding claims, wherein the peptide is a laminin derived peptide.
11. The method according to anyone of the preceding claims, wherein the density of said peptide within the said (meth) acrylated extracellular matrix derived hydrogel is between 0.5 and 10 mg/mL.
12. The method according to anyone of the preceding claims, wherein the cells are progenitor cells selected from the list consisting of totipotent stem cells, induced pluripotent or progenitor stem cells, pluripotent stem cells, multipotent stem cells, mesenchymal stem cells (MSCs), hematopoietic stem cells, neuronal stem cells, hematopoietic stem cells, pancreatic stem cells, cardiac stem cells, embryonic stem cells, embryonic germ cells, neural crest stem cells, kidney stem cells, hepatic stem cells, lung stem cells, hemangioblast cells, endothelial progenitor cells, and cancer stem cells.
13. A method according to anyone of the preceding claims, for forming a 3D tissue construct for promoting angiogenesis, wherein the living cells comprise fibroblasts, preferably human skin fibroblasts and/or human umbilical vein endothelial cells and the sequence of the peptide is YIGSR at a concentration comprised between 0.5 mg/mL and 6 mg/mL.
14. A method according to anyone of the claims 1 to 11 for forming a 3D tissue construct for promoting neurogenesis, wherein the living cells are neurons, preferably primary sensory neurons and the sequence of the peptide is IKVAV at a concentration comprised between 0.5 mg/mL and 6 mg/mL.
15. A method according to anyone of the preceding claims wherein the printing step is performed through microextrusion.
16. A bioink comprising a biocompatible (meth) acrylated extracellular matrix optionally partially crosslinked with an adhesion peptide and a plurality of cells, wherein the extracellular matrix is comprised of a combination of hyaluronic acid and collagen, at a mass ratio of hyaluronic acid /collagen comprised between 0.125 and 100, preferably between 5 and 20
17. A scaffold obtained by printing a bioink according to anyone of method of claims 1 to 14.
PCT/EP2021/065669 2020-06-11 2021-06-10 3d bioprinting method for forming a cell specific tissue construct WO2021250186A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP20305640 2020-06-11
EP20305640.3 2020-06-11

Publications (1)

Publication Number Publication Date
WO2021250186A1 true WO2021250186A1 (en) 2021-12-16

Family

ID=71575286

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2021/065669 WO2021250186A1 (en) 2020-06-11 2021-06-10 3d bioprinting method for forming a cell specific tissue construct

Country Status (1)

Country Link
WO (1) WO2021250186A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114438067A (en) * 2022-02-11 2022-05-06 中山大学·深圳 Method for fixing microorganism to produce hyaluronic acid with high yield by using 3D printing technology

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018225076A1 (en) * 2017-06-09 2018-12-13 Collplant Ltd. Additive manufacturing using recombinant collagen-containing formulation
US20190106673A1 (en) * 2017-10-11 2019-04-11 Wake Forest University Health Sciences Bioink compositions and methods of preparing and using the same

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018225076A1 (en) * 2017-06-09 2018-12-13 Collplant Ltd. Additive manufacturing using recombinant collagen-containing formulation
US20190106673A1 (en) * 2017-10-11 2019-04-11 Wake Forest University Health Sciences Bioink compositions and methods of preparing and using the same

Non-Patent Citations (61)

* Cited by examiner, † Cited by third party
Title
ASHAMMAKHI NAHADIAN SXU CMONTAZERIAN HKO HNASIRI RBARROS NKHADEMHOSSEINI A: "Bioinks and bioprinting technologies to make heterogeneous and biomimetic tissue constructs", MATERIALS TODAY BIO, vol. 1, 2019, pages 100008
BEACHLEY VZWOLF MTSADTLER KMANDA SSJACOBS HBLATCHLEY MRBADER JSPANDEY APARDOLL DELISSEEFF JH: "Tissue matrix arrays for high-throughput screening and systems analysis of cell function", NATURE METHODS, vol. 12, 2015, pages 1197
BORDENAVE LBAQUEY CBAREILLE RLEFEBVRE FLAUROUA CGUERIN VROUAIS FMORE NVERGNES CANDERSON JM: "Endothelial cell compatibility testing of three different Pellethanes", J BIOMED MATER RES, vol. 27, 1993, pages 1367 - 81
BURDICK JAPRESTWICH GD: "Hyaluronic acid hydrogels for biomedical applications", ADV MATER, vol. 23, 2011, pages H41 - 56, XP055011775, DOI: 10.1002/adma.201003963
CANIGGIA ILIU JHAN RWANG JTANSWELL AKLAURIE GPOST M: "Identification of receptors binding fibronectin and laminin on fetal rat lung cells", AMERICAN JOURNAL OF PHYSIOLOGY-LUNG CELLULAR AND MOLECULAR PHYSIOLOGY, vol. 270, 1996, pages L459 - L68
CARPENTIER G, IMAGEJ CONTRIBUTION: ANGIOGENESIS ANALYZER, 2012
CHAN DDXIAO WFLI JDE LA MOTTE CASANDY JDPLAAS A: "Deficiency of hyaluronan synthase 1 (Hasl) results in chronic joint inflammation and widespread intra-articular fibrosis in a murine model of knee joint cartilage damage", OSTEOARTHRITIS CARTILAGE, vol. 23, 2015, pages 1879 - 89, XP029321965, DOI: 10.1016/j.joca.2015.06.021
DEHMELT LPOPLAWSKI GHWANG EHALPAIN S: "NeuriteQuant: an open source toolkit for high content screens of neuronal morphogenesis", BMC NEUROSCI, vol. 12, 2011, pages 100, XP021112919, DOI: 10.1186/1471-2202-12-100
DONDERWINKEL IVAN HEST JCMCAMERON NR: "Bio-inks for 3D bioprinting: recent advances and future prospects", POLYMER CHEMISTRY, vol. 8, 2017, pages 4451 - 71
FARKAS JEMONAGHAN JR: "A brief history of the study of nerve dependent regeneration", NEUROGENESIS (AUSTIN, vol. 4, 2017, pages e1302216
FRASER JRLAURENT TCLAURENT UB: "Hyaluronan: its nature, distribution, functions and turnover", J INTERN MED, vol. 242, 1997, pages 27 - 33
GERMAIN LGUIGNARD RROUABHIA MAUGER FA: "Early basement membrane formation following the grafting of cultured epidermal sheets detached with thermolysin or Dispase", BURNS, vol. 21, 1995, pages 175 - 80, XP002240786, DOI: 10.1016/0305-4179(95)80004-8
GHILAN ALINA ET AL: "Trends in 3D Printing Processes for Biomedical Field: Opportunities and Challenges", JOURNAL OF POLYMERS AND THE ENVIRONMENT, SPRINGER NEW YORK LLC, US, vol. 28, no. 5, 31 March 2020 (2020-03-31), pages 1345 - 1367, XP037123053, ISSN: 1566-2543, [retrieved on 20200331], DOI: 10.1007/S10924-020-01722-X *
GOPINATHAN JNOH I: "Recent trends in bioinks for 3D printing", BIOMATER RES, vol. 22, 2018, pages 11
GOPINATHAN JNOH I: "Recent trends in bioinks for 3D printing", BIOMATERIALS RESEARCH, vol. 22, 2018, pages 11
GRANT D S ET AL: "Two different laminin domains mediate the differentiation of human endothelial cells into capillary-like structures in vitro", CELL, ELSEVIER, AMSTERDAM NL, vol. 58, no. 5, 8 September 1989 (1989-09-08), pages 933 - 943, XP024245400, ISSN: 0092-8674, [retrieved on 19890908], DOI: 10.1016/0092-8674(89)90945-8 *
GRANT DSTASHIRO K-ISEGUI-REAL BYAMADA YMARTIN GRKLEINMAN HK: "Two different laminin domains mediate the differentiation of human endothelial cells into capillary-like structures in vitro", CELL, vol. 58, 1989, pages 933 - 43, XP024245400, DOI: 10.1016/0092-8674(89)90945-8
GROLL JBOLAND TBLUNK TBURDICK JACHO DWDALTON PDDERBY BFORGACS GLI QMIRONOV VA: "Biofabrication: reappraising the definition of an evolving field", BIOFABRICATION, vol. 8, 2016, pages 013001
GROLL JBURDICK JACHO DWDERBY BGELINSKY MHEILSHORN SCJIINGST TMALDA JMIRONOV VANAKAYAMA K: "A definition of bioinks and their distinction from biomaterial inks", BIOFABRICATION, vol. 11, 2018, pages 013001
GUNGOR-OZKERIM PSINCI IZHANG YSKHADEMHOSSEINI ADOKMECI MR: "Bioinks for 3D bioprinting: an overview", BIOMATERIALS SCIENCE, vol. 6, 2018, pages 915 - 46, XP055708634, DOI: 10.1039/C7BM00765E
HAMLEY IW: "Small Bioactive Peptides for Biomaterials Design and Therapeutics", CHEMICAL REVIEWS, vol. 117, 2017, pages 14015 - 41
HOLZL KLIN STYTGAT LVAN VLIERBERGHE SGU LOVSIANIKOV A: "Bioink properties before, during and after 3D bioprinting", BIOFABRICATION, vol. 8, 2016, pages 032002
HOSPODIUK MDEY MSOSNOSKI DOZBOLAT IT: "The bioink: A comprehensive review on bioprintable materials", BIOTECHNOLOGY ADVANCES, vol. 35, 2017, pages 217 - 39
HOYLE CEBOWMAN CN: "Thiol-ene click chemistry", ANGEW CHEM INT ED ENGL, vol. 49, 2010, pages 1540 - 73
J P RANIERI ET AL: "Neuronal cell attachment to fluorinated ethylene propylene films with covalently immobilized laminin oligopeptides YIGSR and IKVAV. II", JOURNAL OF BIOMEDICAL MATERIALS RESEARCH, 1 June 1995 (1995-06-01), UNITED STATES, pages 779 - 785, XP055466504, Retrieved from the Internet <URL:https://onlinelibrary.wiley.com/doi/pdf/10.1002/jbm.820290614> [retrieved on 20201125], DOI: 10.1002/jbm.820290614 *
JANG JPARK JYGAO GCHO D-W: "Biomaterials-based 3D cell printing for next-generation therapeutics and diagnostics", BIOMATERIALS, vol. 156, 2018, pages 88 - 106, XP085314424, DOI: 10.1016/j.biomaterials.2017.11.030
JIA WGUNGOR-OZKERIM PSZHANG YSYUE KZHU KLIU WPI QBYAMBAA BDOKMECI MRSHIN SR: "Direct 3D bioprinting of perfusable vascular constructs using a blend bioink", BIOMATERIALS, vol. 106, 2016, pages 58 - 68, XP029718631, DOI: 10.1016/j.biomaterials.2016.07.038
JIANG YCHEN JDENG CSUURONEN EJZHONG Z: "Click hydrogels, microgels and nanogels: emerging platforms for drug delivery and tissue engineering", BIOMATERIALS, vol. 35, 2014, pages 4969 - 85, XP028841771, DOI: 10.1016/j.biomaterials.2014.03.001
JUNGST TSMOLAN WSCHACHT KSCHEIBEL TGROLL J: "Strategies and Molecular Design Criteria for 3D Printable Hydrogels", CHEM REV, vol. 116, 2016, pages 1496 - 539
KERIQUEL VOLIVEIRAHREMY MZIANE SDELMOND SROUSSEAU BREY SCATROS SAMEDEE JGUILLEMOT F: "In situ printing of mesenchymal stromal cells, by laser-assisted bioprinting, for in vivo bone regeneration applications", SCI REP, vol. 7, 2017, pages 1778
KLEINMAN HKKLEBE RJMARTIN GR: "Role of collagenous matrices in the adhesion and growth of cells", J CELL BIOL, vol. 88, 1981, pages 473 - 85
KOLESKY DBHOMAN KASKYLAR-SCOTT MALEWIS JA: "Three-dimensional bioprinting of thick vascularized tissues", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 113, 2016, pages 3179, XP055629148, DOI: 10.1073/pnas.1521342113
KOLESKY DBTRUBY RLGLADMAN ASBUSBEE TAHOMAN KALEWIS JA: "3D Bioprinting of Vascularized, Heterogeneous Cell-Laden Tissue Constructs", ADVANCED MATERIALS, vol. 26, 2014, pages 3124 - 30, XP055303134, DOI: 10.1002/adma.201305506
LEE VKLANZI AMHAYGAN NYOO SSVINCENT PADAI G: "Generation of Multi-Scale Vascular Network System within 3D Hydrogel using 3D Bio-Printing Technology", CELL MOL BIOENG, vol. 7, 2014, pages 460 - 72
LEE YBPOLIO SLEE WDAI GMENON LCARROLL RSYOO SS: "Bio-printing of collagen and VEGF-releasing fibrin gel scaffolds for neural stem cell culture", EXP NEUROL, vol. 223, 2010, pages 645 - 52, XP027038407
LI HXUE YJIA BBAI YZUO YWANG SZHAO YYANG WTANG H: "The preparation of hyaluronic acid grafted pullulan polymers and their use in the formation of novel biocompatible wound healing film", CARBOHYDRATE POLYMERS, vol. 188, 2018, pages 92 - 100
LIM KS ET AL.: "Fundamentals and Applications of Photo-Cross-Linking in Bioprinting", CHEM. REV., April 2020 (2020-04-01), Retrieved from the Internet <URL:https://doi.org/10.1021/acs.chemrev.9b00812>
LOZANO RSTEVENS LTHOMPSON BCGILMORE KJGORKIN RSTEWART EMPANHUIS MROMERO-ORTEGA MWALLACE GG: "3D printing of layered brain-like structures using peptide modified gellan gum substrates", BIOMATERIALS, vol. 67, 2015, pages 264 - 73
MALDA JVISSER JMELCHELS FPJIINGST THENNINK WEDHERT WJAGROLL JHUTMACHER DW, 25TH ANNIVERSARY ARTICLE: ENGINEERING HYDROGELS FOR BIOFABRICATION. ADVANCED MATERIALS, vol. 25, 2013, pages 5011 - 28
MALIN SADAVIS BMMOLLIVER DC: "Production of dissociated sensory neuron cultures and considerations for their use in studying neuronal function and plasticity", NAT PROTOC, vol. 2, 2007, pages 152 - 60
MANDRYCKY CWANG ZKIM KKIM DH: "3D bioprinting for engineering complex tissues", BIOTECHNOL ADV, vol. 34, 2016, pages 422 - 34, XP029549278, DOI: 10.1016/j.biotechadv.2015.12.011
MATTHEW L. BEDELL ET AL: "A high-throughput approach to compare the biocompatibility of candidate bioink formulations", BIOPRINTING, vol. 17, 1 March 2020 (2020-03-01), pages e00068, XP055752551, ISSN: 2405-8866, DOI: 10.1016/j.bprint.2019.e00068 *
MAZZOCCHI ADEVARASETTY MHUNTWORK RSOKER SSKARDAL A: "Optimization of collagen type I-hyaluronan hybrid bioink for 3D bioprinted liver microenvironments", BIOFABRICATION, vol. 11, 2018, pages 015003
MILLER JSSTEVENS KRYANG MTBAKER BMNGUYEN D-HTCOHEN DMTORO ECHEN AAGALIE PAYU X: "Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues", NATURE MATERIALS, vol. 11, 2012, pages 768
MURAKAMI TOTSUKI SOKAMOTO YNAKAGAWA KWAKAMA HOKUNO NNEO M: "Hyaluronic acid promotes proliferation and migration of human meniscus cells via a CD44-dependent mechanism", CONNECT TISSUE RES, vol. 60, 2019, pages 117 - 27
MURPHY SVATALA A: "3D bioprinting of tissues and organs", NAT BIOTECHNOL, vol. 32, 2014, pages 773 - 85, XP055244641, DOI: 10.1038/nbt.2958
NING LSUN HLELONG TGUILLOTEAU RZHU NSCHREYER DJCHEN X: "3D bioprinting of scaffolds with living Schwann cells for potential nerve tissue engineering applications", BIOFABRICATION, vol. 10, 2018, pages 035014
O'BRIEN JWILSON IORTON TPOGNAN F: "Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity", EUR J BIOCHEM, vol. 267, 2000, pages 5421 - 6, XP002968559, DOI: 10.1046/j.1432-1327.2000.01606.x
PARDUE ELIBRAHIM SRAMAMURTHI A: "Role of hyaluronan in angiogenesis and its utility to angiogenic tissue engineering", ORGANOGENESIS, vol. 4, 2008, pages 203 - 14, XP055113442, DOI: 10.4161/org.4.4.6926
PARK KDWANG XLEE JYPARK KMZHANG SNOH I: "Research trends in biomimetic medical materials for tissue engineering: commentary", BIOMATERIALS RESEARCH, vol. 20, 2016, pages 8
PATEL RSANTHOSH MDASH JKKARPOORMATH RJHA AKWAK JPATEL MKIM JH: "Ile-Lys-Val-ala-Val (IKVAV) peptide for neuronal tissue engineering", POLYMERS FOR ADVANCED TECHNOLOGIES, vol. 30, 2019, pages 4 - 12
POLDERVAART MTGOVERSEN BDE RUIJTER MABBADESSA AMELCHELS FPWONER FCDHERT WJAVERMONDEN TALBLAS J: "3D bioprinting of methacrylated hyaluronic acid (MeHA) hydrogel with intrinsic osteogenicity", PLOS ONE, vol. 12, 2017, pages e0177628
QUAEGEBEUR ALANGE CCARMELIET P: "The Neurovascular Link in Health and Disease: Molecular Mechanisms and Therapeutic Implications", NEURON, vol. 71, 2011, pages 406 - 24, XP028382852, DOI: 10.1016/j.neuron.2011.07.013
RAVICHANDRAN RISLAM MMALARCON EISAMANTA AWANG SLUNDSTROM PHILBORN JGRIFFITH MPHOPASE J: "Functionalised type-I collagen as a hydrogel building block for bio-orthogonal tissue engineering applications", JOURNAL OF MATERIALS CHEMISTRY B, vol. 4, 2016, pages 318 - 26, XP055556963, DOI: 10.1039/C5TB02035B
SAKAI SOHI HHOTTA TKAMEI HTAYA M: "Differentiation potential of human adipose stem cells bioprinted with hyaluronic acid/gelatin-based bioink through microextrusion and visible light-initiated crosslinking", BIOPOLYMERS, vol. 109, 2018, pages e23080
SASMAL PDATTA PWU YOZBOLAT IT: "3D bioprinting for modelling vasculature", MICROPHYSIOLOGICAL SYSTEMS, vol. 2018, November 2018 (2018-11-01)
SCHMIDT CELEACH JB: "Neural Tissue Engineering: Strategies for Repair and Regeneration", ANNUAL REVIEW OF BIOMEDICAL ENGINEERING, vol. 5, 2003, pages 293 - 347, XP002583326, DOI: 10.1146/ANNUREV.BIOENG.5.011303.120731
STUPACK DGCHERESH DA: "Current Topics in Developmental Biology", 2004, ACADEMIC PRESS, article "Integrins and Angiogenesis", pages: 207 - 38
TOOLE BP: "Hyaluronan: from extracellular glue to pericellular cue", NAT REV CANCER, vol. 4, 2004, pages 528 - 39, XP055089628, DOI: 10.1038/nrc1391
TRONCI GDOYLE ARUSSELL SJWOOD DJ: "Triple-helical collagen hydrogels via covalent aromatic functionalisation with 1,3-phenylenediacetic acid", JOURNAL OF MATERIALS CHEMISTRY B, vol. 1, 2013, pages 5478 - 88
YANG XLU ZWU HLI WZHENG LZHAO J: "Collagen-alginate as bioink for three-dimensional (3D) cell printing based cartilage tissue engineering", MATERIALS SCIENCE AND ENGINEERING: C, vol. 83, 2018, pages 195 - 201, XP085298393, DOI: 10.1016/j.msec.2017.09.002

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114438067A (en) * 2022-02-11 2022-05-06 中山大学·深圳 Method for fixing microorganism to produce hyaluronic acid with high yield by using 3D printing technology
CN114438067B (en) * 2022-02-11 2023-10-20 中山大学·深圳 Method for fixing microorganism to produce hyaluronic acid in high yield by using 3D printing technology

Similar Documents

Publication Publication Date Title
Ooi et al. Thiol–ene alginate hydrogels as versatile bioinks for bioprinting
Kim et al. Efficient myotube formation in 3D bioprinted tissue construct by biochemical and topographical cues
Petta et al. Hyaluronic acid as a bioink for extrusion-based 3D printing
Hoch et al. Chemical tailoring of gelatin to adjust its chemical and physical properties for functional bioprinting
Magno et al. Polymer hydrogels to guide organotypic and organoid cultures
Lim et al. New visible-light photoinitiating system for improved print fidelity in gelatin-based bioinks
Chung et al. Influence of three-dimensional hyaluronic acid microenvironments on mesenchymal stem cell chondrogenesis
Hanjaya-Putra et al. Spatial control of cell-mediated degradation to regulate vasculogenesis and angiogenesis in hyaluronan hydrogels
Uygun et al. Effects of immobilized glycosaminoglycans on the proliferation and differentiation of mesenchymal stem cells
Skardal et al. Photocrosslinkable hyaluronan-gelatin hydrogels for two-step bioprinting
Dash et al. Biofunctionalization of ulvan scaffolds for bone tissue engineering
Nedunchezian et al. Generating adipose stem cell-laden hyaluronic acid-based scaffolds using 3D bioprinting via the double crosslinked strategy for chondrogenesis
Prestwich et al. Injectable synthetic extracellular matrices for tissue engineering and repair
US20110274742A1 (en) Cartilage Repair Systems and Applications Utilizing A Glycosaminoglycan Mimic
US20210046220A1 (en) Hydrogel for tissue engineering and bioprinting
KR20220077156A (en) Biogum and botanical gum hydrogel bioinks for the physiological 3d bioprinting of tissue constructs for in vitro culture and transplantation
Oliveira et al. Extracellular matrix (ECM)-derived bioinks designed to foster vasculogenesis and neurite outgrowth: Characterization and bioprinting
Agarwal et al. Recent advances in chemically defined and tunable hydrogel platforms for organoid culture
Moghaddam et al. Review of bioprinting in regenerative medicine: naturally derived bioinks and stem cells
Binner et al. Cell-instructive starPEG-heparin-collagen composite matrices
WO2017187114A1 (en) Improvements in 3d printing
Ji et al. Novel bioinks from UV-responsive norbornene-functionalized carboxymethyl cellulose macromers
Ghorbani et al. Photo-cross-linkable hyaluronic acid bioinks for bone and cartilage tissue engineering applications
Jain et al. Impact of cell density on the bioprinting of gelatin methacrylate (GelMA) bioinks
Sekar et al. Hyaluronic acid as bioink and hydrogel scaffolds for tissue engineering applications

Legal Events

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

Ref document number: 21732014

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21732014

Country of ref document: EP

Kind code of ref document: A1