US20130323708A1 - Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same - Google Patents

Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same Download PDF

Info

Publication number
US20130323708A1
US20130323708A1 US13/392,661 US201013392661A US2013323708A1 US 20130323708 A1 US20130323708 A1 US 20130323708A1 US 201013392661 A US201013392661 A US 201013392661A US 2013323708 A1 US2013323708 A1 US 2013323708A1
Authority
US
United States
Prior art keywords
organ
cells
perfusate
liver
recellularized
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US13/392,661
Inventor
Martin Yarmush
Basak E. Uygun
Mustafa Korkut Uygun
Maria Louisa Izamis
Alejandro Soto-Gutierrez
Herman Tolboom
Hiroshi Yagi
Carley Shulman
John Miles Milwid
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
General Hospital Corp
Massachusetts Institute of Technology
Original Assignee
General Hospital Corp
Massachusetts Institute of Technology
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 General Hospital Corp, Massachusetts Institute of Technology filed Critical General Hospital Corp
Priority to US13/392,661 priority Critical patent/US20130323708A1/en
Assigned to THE GENERAL HOSPITAL CORPORATION reassignment THE GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: UYGUN, BASAK E., UYGUN, MUSTAFA KORKUT, YARMUSH, MARTIN, TOLBOOM, HERMAN, YAGI, HIROSHI, SOTO-GUTIERREZ, ALEJANDRO
Assigned to MASSACHUSETTS INSTITUTE OF TECHNOLOGY reassignment MASSACHUSETTS INSTITUTE OF TECHNOLOGY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: IZAMIS, MARIA LOUISA, MILWID, JOHN MILES
Publication of US20130323708A1 publication Critical patent/US20130323708A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/067Hepatocytes
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0205Chemical aspects
    • A01N1/021Preservation or perfusion media, liquids, solids or gases used in the preservation of cells, tissue, organs or bodily fluids
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0236Mechanical aspects
    • A01N1/0242Apparatuses, i.e. devices used in the process of preservation of living parts, such as pumps, refrigeration devices or any other devices featuring moving parts and/or temperature controlling components
    • A01N1/0247Apparatuses, i.e. devices used in the process of preservation of living parts, such as pumps, refrigeration devices or any other devices featuring moving parts and/or temperature controlling components for perfusion, i.e. for circulating fluid through organs, blood vessels or other living parts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/37Digestive system
    • A61K35/407Liver; Hepatocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/14Dialysis systems; Artificial kidneys; Blood oxygenators ; Reciprocating systems for treatment of body fluids, e.g. single needle systems for hemofiltration or pheresis

Definitions

  • HHs human hepatocytes
  • ADMET toxicology
  • Cell transplantation is a new and elegant approach that can address the need for organs and can also be used for patients that are severely ill or as a preventative therapy.
  • the direct delivery of cells is inefficient due to low engraftment.
  • a tissue-engineered vehicle designed to ensure long-term cell engraftment, viability, and functionality is a necessity to make cell transplantation work clinically.
  • tissue engineering has so far had limited success in many tissues, including the liver.
  • the key factor that prevents advancement of the field is the lack of an ideal transplantable scaffold that has all the necessary microstructure and extracellular cues for cell attachment, differentiation, functioning, as well as vascularization, which has so far proven to be difficult to manufacture in vitro.
  • hepatocytes the major cell type in the liver
  • the only reliable source of cells and tissues is human donor livers.
  • Stem-cell-derived hepatocytes are not fully equivalent to primary human hepatocytes (adult hepatocytes) and are time consuming and very expensive to produce in mass quantities.
  • organs in good condition are used for transplantation. Therefore, only the worst quality livers are available for hepatocyte isolation.
  • the available livers typically are either excessively fatty or are recovered from donors after cardiac death. In either case, the hepatocytes suffer extended ischemic damage, and the quantity and quality of the cells isolated is greatly reduced compared to ideal livers.
  • One aspect of the invention provides a method for harvesting adult cells from an organ.
  • the method includes perfusing the organ with a perfusate and isolating adult cells from the organ, thereby harvesting the adult cells from the organ.
  • the organ is a mammalian organ. In another embodiment, the organ is a human organ. In still another embodiment, the organ is a liver.
  • the adult cells can be selected from the group consisting of: hepatocytes, endothelial cells, cholangiocytes (bile duct cells), Kupffer cells, stellate cells, and smooth muscle cells.
  • the organ can be selected from the group consisting of: pancreas, spleen, heart, lung, and kidney.
  • the organ can be ischemic.
  • the perfusate includes one or more oxygen carriers.
  • the one or more oxygen carriers can include erythrocytes.
  • the perfusate can include one or more anti-inflammatory agents.
  • the one or anti-inflammatory agents can include hydrocortisone.
  • the perfusate can include collagenase.
  • the perfusate can be a normothermic perfusate, a room temperature perfusate, or a hypothermic perfusate.
  • the method includes culturing the harvested adult cells.
  • the method can include providing the organ.
  • the step of isolating the adult cells from the organ can include filtering the perfusate.
  • the filtering the perfusate step can include filtering the perfusate through one or more mesh filters.
  • the one or more mesh filters can have pore sizes ranging from about 1 ⁇ m to about 1000 ⁇ m.
  • the step of isolating the adult cells from the organ can include performing density centrifugation on the perfusate.
  • the method can include circulating the perfusate through a dialyzer.
  • the organ can be a damaged organ.
  • the perfusion step can improve the vitality of the adult cells.
  • Another aspect of the invention provides a method for rehabilitating an organ.
  • the method includes: dividing the organ into a first portion and a second portion, perfusing the first portion with a decellularization medium, isolating adult cells from the second portion, and recellularizing the first portion with a suspension of the adult cells, thereby rehabilitating the organ.
  • the method includes preserving the first portion.
  • the preserving step can include washing the first portion with one or more isotonic washes.
  • the preserving step can include storing the first portion in a storage medium.
  • the storage medium can include one or more antimicrobial agents.
  • the one or more antimicrobial agents can be selected from the group consisting of antibiotics and fungicides.
  • the method includes culturing the adult cells.
  • the method can include perfusing the recellularized first portion.
  • the method can include transplanting the first portion into a subject.
  • the organ can be previously harvested from the subject.
  • the organ can be a damaged organ.
  • the method can include utilizing the first portion as an in vitro model.
  • the in vitro model can be an in vitro model of organ function.
  • the method can include utilizing the first portion as an in vitro drug model.
  • the in vitro drug model can be an in vitro model of drug absorption, drug metabolism, drug excretion, and/or drug toxicity.
  • the first portion and the second portion can be substantially equal in size.
  • the decellularization medium includes one or more agents selected from the group consisting of detergents, vasodilators, buffers, inorganic salts, and enzymes.
  • the organ is mammalian and/or human.
  • the organ can be selected from the group consisting of liver, pancreas, kidney, spleen, heart, and lung.
  • Another aspect of the invention provides a rehabilitated organ prepared by a method including: dividing an organ into a first portion and a second portion, perfusing the first portion with a decellularization medium, isolating adult cells from the second portion, and recellularizing the first portion with a suspension of the adult cells, thereby preparing a rehabilitated organ.
  • the method includes preserving the first portion.
  • the method can include washing the first portion with one or more isotonic washes.
  • the method can include storing the first portion in a storage medium.
  • the storage medium can include one or more antimicrobial agents.
  • the one or more antimicrobial agents can be selected from the group consisting of antibiotics and fungicides.
  • the organ can be a damaged organ.
  • the first portion and the second portion can be substantially equal in size.
  • the decellularization medium can include one or more agents selected from the group consisting of detergents, vasodilators, buffers, inorganic salts, and enzymes.
  • the organ can be mammalian and/or human.
  • the organ can be selected from the group consisting of liver, pancreas, kidney, spleen, heart, and lung.
  • an artificial organ including: a bioscaffold comprising an extracellular matrix having a vascular network and a plurality of adult cells.
  • the adult cells are engrafted to the matrix and are vascularized such that the adult cells are capable of organ function.
  • the plurality of adult cells were previously harvested from one or more other organs.
  • the bioscaffold can be derived from a previously decellularized organ.
  • the bioscaffold can be bioartificial organ device.
  • the bioscaffold and the adult cells can be mammalian-derived.
  • the bioscaffold and the adult cells can be human-derived.
  • the bioscaffold and the adult cells can be derived from an organ selected from the group consisting of: liver, pancreas, kidney, spleen, heart, and lung.
  • the bioscaffold and the adult cells can be derived from a liver.
  • the adult cells are harvested from an organ by any method described herein.
  • the bioscaffold is prepared according to a method including: cannulating one or more vessels of an organ and decellularizing the organ, thereby preparing a bioscaffold comprising a decellularized extracellular matrix having a vascular network.
  • the decellularizing step can include perfusing the organ with a decellularization medium.
  • the decellularization medium can include one or more agents selected from the group consisting of detergents, vasodilators, buffers, inorganic salts, and enzymes.
  • the perfusion can be single or multidirectional perfusion.
  • the perfusion is can be machine- or gravity-driven perfusion.
  • the temperature of the decellularization medium can be maintained between about 4° C. and about 42° C.
  • the organ can be perfused from about 4 hours to about 14 days.
  • the organ can be perfused for between about 4 days and about 7 days.
  • the method includes preserving the bioscaffold.
  • the preserving step can include washing the bioscaffold with one or more isotonic washes.
  • the preserving step can include storing the bioscaffold in a storage medium.
  • the storage medium can include one or more antimicrobial agents.
  • the one or more antimicrobial agents can be selected from the group consisting of antibiotics and fungicides.
  • the bioscaffold can be stored at a temperature less than about 4° C.
  • the organ is mammalian and/or human.
  • the organ can be selected from the group consisting of liver, pancreas, kidney, spleen, heart, and lung.
  • the organ can be a liver.
  • Another aspect of the invention provides a method for fabricating an artificial organ.
  • the method includes: cannulating one or more vessels in a bioscaffold and cellularizing the bioscaffold by introducing a suspension of adult organ cells into the bioscaffold, thereby preparing an artificial organ.
  • the method includes perfusing the cellularized bioscaffold.
  • the perfusion can be machine- or gravity-driven perfusion.
  • the suspension can include a solution of osmolality ranging from about 100 mOsm to about 500 mOsm, one or more oxygen carriers, one or more antioxidants, one or more anti-inflammatory agents, one or more vasodilators, one or more amino acids, one or more buffers, one or more inorganic salts, one or more substrates for metabolism, and one or more agents to maintain oncotic pressure between about 15 to about 45 mm Hg.
  • the suspension of adult organ cells is introduced into the bioscaffold by injection or perfusion.
  • the method includes preserving the artificial organ.
  • the preserving step can include washing the artificial organ with one or more isotonic washes.
  • the preserving step can include storing the artificial organ in a storage medium.
  • the storage medium can include one or more antimicrobial agents.
  • the one or more antimicrobial agents can be selected from the group consisting of antibiotics and fungicides.
  • the artificial organ can be stored at a temperature less than about 4° C.
  • the bioscaffold and the adult cells are mammalian-derived and/or human-derived.
  • the bioscaffold and the adult cells can be derived from an organ selected from the group consisting of liver, pancreas, kidney, spleen, heart, and lung.
  • the bioscaffold and the adult cells can be derived from a liver.
  • the adult cells can be harvested from an organ by any method described herein.
  • the method includes preparing the bioscaffold according to a method including: cannulating one or more vessels of an organ and decellularizing the organ, thereby preparing a bioscaffold comprising a decellularized extracellular matrix having a vascular network.
  • Decellularizing the organ can include perfusing the organ with a decellularization medium.
  • the decellularization medium can include one or more agents selected from the group consisting of detergents, vasodilators, buffers, inorganic salts, and enzymes.
  • the perfusion can be single or multidirectional perfusion.
  • the perfusion can be machine- or gravity-driven perfusion.
  • the temperature of the decellurization medium can be maintained between about 4° C. and about 42° C.
  • the organ can be perfused from about 4 hours to about 14 days.
  • the organ can be perfused for between about 4 days and about 7 days.
  • the method includes preserving the bioscaffold.
  • the preserving step can include washing the bioscaffold with one or more isotonic washes.
  • the preserving step can include storing the bioscaffold in a storage medium.
  • the storage medium can include one or more antimicrobial agents.
  • the one or more antimicrobial agents can be selected from the group consisting of antibiotics and fungicides.
  • the bioscaffold can be stored at a temperature less than about 4° C.
  • the organ is mammalian and/or human.
  • the organ can be selected from the group consisting of liver, pancreas, kidney, spleen, heart, and lung.
  • the organ can be a liver.
  • an organ encasement including: a first membrane and a second membrane adapted for coupling with the first membrane to encase an organ.
  • the organ encasement also includes coupling means for coupling the first and the second membrane.
  • the coupling means can be selected from the group consisting of: magnets, screws, bolts, nuts, rivets, adhesives, and zipper storage assemblies.
  • first membrane and the second membrane are flexible.
  • the first membrane and the second membrane can be gas permeable.
  • the first membrane and the second membrane can be biocompatible.
  • the first membrane and the second membrane can include one or more biocompatible materials selected from the group consisting of: silicone, thermoset polymers, thermoplastic polymers, amniotic membranes, small intestinal submucosa (SIS), fascia, dura mater, peritoneum, and pericardium.
  • an organ perfusion system including: an organ encasement and a primary circuit in fluid communication with an organ encasement for circulating a perfusate.
  • the primary circuit includes a pressure sensor.
  • the primary circuit can include a sampling port, a heat exchanger, a bubble trap, an oxygenator, and/or one or more pumps.
  • the primary circuit can include a first cannula in communication with the organ and a second cannula in communication with the organ.
  • the organ perfusion system includes: a dialyzer in communication with the primary circuit and a secondary circuit in communication with the dialyzer.
  • the secondary circuit can include: a pump and a dialysate reservoir.
  • the primary circuit includes one or more decellularization modules and/or one or more recellularization modules.
  • the organ perfusion system can be adapted to preserve the organ.
  • Another aspect of the invention provides a method of perfusing an organ.
  • the method includes: providing an organ perfusion system including an organ encasement and a primary circuit in fluid communication with an organ encasement for circulating a perfusate; placing an organ within the organ encasement; and circulating the perfusate.
  • the perfusate is a normothermic perfusate.
  • the perfusate can be a room temperature perfusate.
  • the perfusate can be a hypothermic perfusate.
  • kits for perfusing an organ includes: an organ encasement, a perfusate, and instructions for use.
  • kits for preparing a recellularized bioscaffold includes: a plurality of adult cells, a perfusate, and instructions for use.
  • the kit includes a bioscaffold encasement.
  • an artificial organ production system including: an organ encasement and a primary circuit in fluid communication with an organ encasement for circulating a perfusate.
  • the primary circuit includes: one or more decellularization modules and one or more recellularization modules.
  • Another aspect of the invention provides a method for producing a research tool.
  • the method includes: slicing a decellularized organ to produce a decellularized organ slice, plating the decellularized organ slice, seeding the decellularized organ slice with one or more cells to produce a recellularized organ slice, and culturing the recellularized organ slice.
  • the method includes storing the recellularized organ slice.
  • the method can include using the recellularized organ slice in an experiment.
  • the one or more cells can be adult cells and/or stem cells.
  • Another aspect of the invention provides a research tool prepared by a method including: slicing a decellularized organ to produce a decellularized organ slice, plating the decellularized organ slice, seeding the decellularized organ slice with one or more cells to produce a recellularized organ slice, and culturing the recellularized organ slice.
  • Another aspect of the invention provides a method for producing a research tool.
  • The includes: slicing an organ to produce an organ slice; decellularizing the organ slice to produce a decellularized organ slice; seeding the decellularized organ slice with one or more cells to produce a recellularized organ slice; and culturing the recellularized organ slice.
  • the method includes storing the recellularized organ slice.
  • the method can include using the recellularized organ slice in an experiment.
  • the one or more cells can be adult cells.
  • the one or more cells can be stem cells.
  • the method can include plating the decellularized organ slice.
  • Another aspect of the invention provides a research tool prepared by a method including: slicing an organ to produce an organ slice; decellularizing the organ slice to produce a decellularized organ slice; seeding the decellularized organ slice with one or more cells to produce a recellularized organ slice; and culturing the recellularized organ slice.
  • Another aspect of the invention provides a method for producing a research tool.
  • the method includes: slicing a recellularized organ to produce a recellularized organ slice and plating the recellularized organ slice.
  • the method includes storing the recellularized organ slice.
  • the method can include using the recellularized organ slice in an experiment.
  • Another aspect of the invention provides a research tool prepared by a method including: slicing a recellularized organ to produce a recellularized organ slice and plating the recellularized organ slice.
  • the research tool includes a recellularized organ slice.
  • the research tool can also include a vessel.
  • the recellularized organ slice can be received within the vessel.
  • the vessel can contain an agar and wherein the recellularized organ slice is mounted on the agar.
  • the recellularized organ slice can be seeded with cells before slicing.
  • the recellularized organ slice can be seeded with cells after slicing.
  • Another aspect of the invention provides an assay method including: applying one or more test agents to a recellularized organ slice, incubating the recellularized organ slice, and analyzing one or more metabolites of the test agent.
  • FIG. 1 depicts a perfusion system according to an embodiment of the invention.
  • FIGS. 2A and 2B depict organ encasement system according to an embodiment of the invention.
  • FIG. 3 depicts a method of harvesting adult cells according to an embodiment of the invention.
  • FIG. 4 depicts a method of preparing a bioscaffold according to an embodiment of the invention.
  • FIG. 5 depicts a method of recellularizing a bioscaffold according to an embodiment of the invention.
  • FIG. 6 depicts a method of organ rehabilitation according to an embodiment of the invention.
  • FIGS. 7A and 7B depict methods of creating a research tool according to embodiments of the invention.
  • FIGS. 8A-8E depict functional parameters of perfused ischemic livers during normothermic perfusion as compared to perfused fresh livers.
  • FIG. 9 depicts survival rates of transplant recipients of perfused ischemic rat livers compared to survival rates of recipients of fresh (normal) and warm ischemic livers with different modalities of preservation.
  • FIG. 10 depicts a comparison of survival rates for transplant recipients of perfused ischemic livers compared with different versions of the perfusion systems described herein.
  • FIGS. 11A and 11B depict cell yields and viability for harvested livers.
  • FIGS. 12A-12C depict phase contrast microscopy of cells obtained from ischemic livers after perfusion ( FIG. 12A ), healthy control livers ( FIG. 12B ), and one-hour ischemic livers directly isolated ( FIG. 12C ) in a collagen double gel, respectively, at two weeks post-seeding.
  • FIG. 12D depicts urea production rates for hepatocytes from perfused ischemic livers compared to fresh livers during six hours of suspension culture.
  • FIG. 12E depicts albumin secretion for hepatocytes from perfused ischemic livers compared to fresh livers during six hourse of suspension culture as evaluated with an enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • FIG. 12F depicts urea production rates for hepatocytes from perfused ischemic livers compared to fresh livers when cultures in collagen double gel.
  • FIG. 12G depicts albumin secretion for hepatocytes from perfused ischemic livers compared to fresh livers when cultured in collagen double gel.
  • FIGS. 13A-13E are representative photographs of livers during the decellularization process.
  • FIGS. 14A-14D are representative photographs of liver during infusion with isolated rat hepatocytes.
  • FIGS. 15A-15C are representative photographs of H&E-stained recellularized livers.
  • FIGS. 16A-16C are representative photographs of TUNEL-stained recellularized livers after eight hours of perfusion.
  • FIG. 16A depicts a negative control of TUNEL staining without antibodies.
  • FIG. 16 b depicts the staining of apoptic cells in green.
  • FIG. 16C depicts TUNEL staining of a DNAse exposed liver sample as a positive control. DAPI staining of nuclei is presented in the bottom left inset of each figure.
  • FIG. 17A-17C depict albumin, urea, and lactate dehydrogenase levels for recellularized livers compared to hepatocytes in collagen culture.
  • FIGS. 18A-18F are representative photographs of H&E-stained liver matrices.
  • FIGS. 18A and 18D depict normal livers.
  • FIGS. 18B and 18E depict recellularized livers 24 hours after recellularization.
  • FIGS. 18C and 18F depict recellularized livers 48 hours after recellularization.
  • FIG. 19A is a photograph of a transplanted liver bioscaffold.
  • FIG. 19B is a schematic of a transplanted recellularized liver.
  • FIGS. 20A-20E are representative photographs of ischemic rat livers during a decellularization process at 0 hours, 18 hours, 48 hours, 52 hours, and 72 hours, respectively.
  • the scale bars represent 10 mm.
  • FIG. 21 is a comparison of a normal liver (top row) and a decellularized liver matrix (bottom row) produced through staining for (from left to right across each row) hematoxylin and eosin, collagen I (red), collagen IV (red), fibronectin (red), and lamin (red). Sections were counterstained with DAPI (blue). The scale bars represented 100 ⁇ m.
  • FIGS. 22A and 22B are representative photographs of decellularized left lateral and median lobes of a decellularized liver matrix with the vascular tree visible.
  • FIG. 22A depicts the decellularized liver matrix before perfusion with Allura Red AC dye and
  • FIG. 22B depicts the decellularized liver matrix after perfusion with Allura Red AC dye.
  • FIGS. 23A and 23B are representative photographs of corrosion cast models the vascular tree of a normal liver ( FIG. 23A ) and a decellularized liver matrix ( FIG. 23B ) with portal (red) and venous (blue) vasculature.
  • FIGS. 24A-C are scanning electron microscopy (SEM) micrographs of a decellularized liver matrix.
  • FIG. 24A is an SEM image of a blood vessel.
  • FIG. 24B is a section featuring bile-duct-like small vessels identified with white arrows.
  • FIG. 24C displays the extracellular matrix within the parenchyma with hepatocyte-size free spaces.
  • the scale bar in each of FIGS. 24A-C represents 20 ⁇ m.
  • FIG. 25 depicts an exemplary method of recellularizing a decellularized organ (e.g., a decellularized liver matrix).
  • a decellularized organ e.g., a decellularized liver matrix
  • FIG. 26 is bar graph depicting the percentage of TUNEL-positive cells in recellularized liver grafts as a function of perfusion-culture time.
  • FIG. 27 provides three images of TUNEL-stained recellularized liver grafts.
  • the left photograph depicts a recellularized graft after 48 hours in culture.
  • the center photograph depicts a freshly isolated liver, which was used as a negative control.
  • the right photograph depicts a DNAse-treated normal live, which was used as a positive control.
  • Arrows indicate TUNEL-positive cells.
  • Each photograph contains an inset photograph depicting a Hoeschst 33258 counterstain of the respective sections.
  • the scale bar in each of the large photographs represents 200 ⁇ m.
  • FIG. 28 is a line graph of lactate dehydrogenase release from recellularized liver grafts during perfusion culture as compared with a static sandwich culture.
  • FIGS. 29A and 29B are SEM micrographs of recellularized liver grafts after two days in culture.
  • FIG. 30 is a comparison of a normal liver (top row) and a recellularized liver graft (bottom row) produced through immunostaining after two days of culture for (from left to right across each row) hematoxylin and eosin, albumin (red), G6 pc (red), and Ugt1a (green).
  • the scale bars represent 100 ⁇ m.
  • FIGS. 31A and 31B are line graphs depicting albumin synthesis and urea secretion, respectively, for recellularized liver grafts in comparison with a static sandwich culture.
  • FIG. 32A depicts a gene expression analysis of hepatocytes in a recellularized liver graft after two days of culture compared to a normal liver, fresh hepatocytes, and sandwich culture hepatocytes after two days of culture for phase I and phase II drug metabolism enzymes.
  • FIG. 32B is a scatter plot comparing gene expression of phase I and phase II drug metabolism enzymes for a recellularized liver graft and sandwich culture hepatocytes after two days of culture.
  • FIG. 34 is a series of representative images of recellularized liver graft transplantation.
  • the top row depicts (from left to right): a transplant site, a recellularized graft at the transplant site, a transplanted graft before blood reperfusion, and a graft after declamping the renal artery.
  • the bottom row depicts (from left to right): a transplanted graft at one minute after clamping, a transplanted graft at two minutes after clamping, a transplanted graft at four minutes after clamping, and an auxiliary recellularized liver graft in contrast with the native liver.
  • FIG. 35 is a comparison of a normal liver (top row) and a recellularized liver graft (bottom row) produced through immunostaining eight hours after transplantation for (from left to right across each row) hematoxylin and eosin, albumin (red), G6pc (red), and Ugt1a (green). Sections were counterstained with Hoescht 33258 (blue). The scale bars represent 100 ⁇ m.
  • An “adult cell” shall be understood to mean any cell capable of performing essential organ functions.
  • Biocompatible shall be understood to refer to any material that is nontoxic and, optionally, does not provoke an immunological rejection.
  • Biocompatible materials can be naturally occurring or synthetic and permit diffusion of liquids, gases, and/or ions.
  • Biocompatible materials in accordance with the invention include, for example, membranous materials selected from the group consisting of silicone, thermoset polymers, thermoplastic polymers, amniotic membranes, small intestinal submucosa (SIS), fascia, dura mater, peritoneum, and pericardium.
  • a “bioscaffold” shall be understood to include any structure that provides an extracellular matrix sufficient to support one or more cells.
  • a “damaged organ” shall be understood to indicate an organ that is in less than ideal condition for transplantation, such that the expected probability of transplant success is reduced.
  • damaged organs include, but are not limited to, organs that suffer warm ischemia for more than 30 minutes, organs that suffer cold ischemia for more than 12 hours, moderate or highly steatotic livers (e.g., livers with greater than 30% fat), fibrotic livers, cirrhotic livers, livers from patients afflicted with hepatitis C or HIV, and the like.
  • decellularize shall be understood to mean any process for the removing one or more cells from an organ.
  • a decellularization process maintains the extracellular matrix and vascular structure of the organ.
  • Essential hepatic function shall be understood to include one or more of the following: protein synthesis; protein storage; transformation of carbohydrates; synthesis of cholesterol, bile salts, and/or phospholipids; and detoxification, modification, and/or excretion of exogenous and endogenous substances.
  • extracellular matrix shall be understood to be the portion of an organ and/or a bioscaffold that provides structural and life support for one or more cells.
  • a “health care provider” shall be understood to mean any person providing medical care to a patient. Such persons include, but are not limited to, medical doctors, physician's assistants, nurse practitioners (e.g., an Advanced Registered Nurse Practitioner (ARNP)), nurses, residents, interns, medical students, or the like. Although various licensure requirements may apply to one or more of the occupations listed above in various jurisdictions, the term health care provider is unencumbered for the purposes of this patent application.
  • ALTP Advanced Registered Nurse Practitioner
  • “Hypothermic” shall be understood to mean temperatures below room temperature.
  • “hypothermic” temperatures include, but are not limited to, temperatures between about 0° C. to about 15° C., temperatures between about 1° C. to about 8° C., temperatures between about 3° C. to about 5° C., and the like.
  • an “in vitro model” shall be understood to be a model created outside of an organism of a subset of the organism's constituent parts, for example, for studying the function of the constituent parts.
  • An “in vitro drug model” shall be understood to be an in vitro model created to study the interaction of a drug, pharmaceutical, test agent, and the like with a subset of an organism's constituent parts. Interactions can include absorption, distribution, metabolism, toxicity, and the like.
  • “Normothermic” shall be understood to mean temperatures above room temperature.
  • “normothermic” temperatures include, but are not limited to, temperatures between about 25° C. and about 42° C., temperatures between about 30° C. and about 38° C., temperatures between about 37° C. and about 37.5° C., and the like.
  • organ is a tissue that performs a specific function or group of functions within an organism.
  • the term organ includes the heart, lungs, stomach, liver, gallbladder, pancreas, spleen, kidneys, and the like.
  • a “perfusate” shall be understood to be any fluid capable of improving or maintaining the vitality of a cell, tissue, organ (including decellularized and recellularized organs), bioscaffold, and the like. Improving or maintaining vitality can include one or more of the following: maintenance of appropriate osmotic pressure, maintenance of appropriate oncotic pressure, maintenance of appropriate temperature, inhibition of decay, inhibition of microbial growth, and the like.
  • recellularize shall be understood to be any process for engrafting one or more cells within a decellularized organ or bioscaffold.
  • Room temperature shall be understood to mean a temperature between about 15° C. and about 25° C.
  • room temperature includes, but is not limited to, temperatures between about 18° C. and about 23° C., temperature between about 19° C. and about 21° C., temperatures between about 24° C. and about 25° C., temperatures between about 20° C. and about 21° C., and the like.
  • a “storage medium” shall be understood to be any substance for preserving vitality of a cell, tissue, organ (including decellularized and recellularized organs), bioscaffold, and the like.
  • Preservation of vitality can include one or more of the following: maintenance of appropriate osmotic pressure, maintenance of appropriate oncotic pressure, maintenance of appropriate temperature, inhibition of decay, inhibition of microbial growth, and the like.
  • a “subject” shall be understood to include any mammal including, but not limited to, humans, primates, swine, cows, sheep, and rats.
  • test agent shall be understood to include any substance that is evaluated for its ability to diagnose, cure, mitigate, treat, or prevent disease in a subject, or is intended to alter the structure or function of the body of a subject.
  • a test agent in an embodiment can be a “drug” as that term is defined under the Federal Food, Drug, and Cosmetic Act at 21 U.S.C. ⁇ 321(g)(1).
  • Test agents include, but are not limited to, chemical compounds, biologic agents, proteins, peptides, nucleic acids, lipids, polysaccharides, supplements, diagnostic agents and immune modulators.
  • a “vascular network” shall be understood to be the portion of an organ that provides fluid (e.g., blood) access to one or more cells.
  • the vascular network can form part of the extracellular matrix and is capable of handling the pressure and viscosity of physiological fluids.
  • aspects of the invention provide new methods and devices for harvesting cells from and rehabilitating damaged organs.
  • a perfusion system 100 is provided for perfusing an organ prior to cell isolation.
  • An organ 102 e.g., a liver
  • the perfusion chamber 102 can be any vessel capable of holding an organ and a perfusate.
  • the perfusion chamber 102 is a vessel such as a glass or plastic bowl.
  • the perfusion chamber 102 is an organ encasement 200 as described herein.
  • a primary circuit 106 circulates a perfusate through the organ 102 and perfusion chamber 104 .
  • the primary circuit 106 can include one or more pumps 108 a , 108 b to promote perfusate circulation. Alternatively, the perfusion can be gravity-driven.
  • the primary circuit can also include an oxygenator 110 , a bubble trap 112 , a heat exchanger 114 , a pressure sensor 116 , and one or more sampling ports 118 a , 118 b.
  • the perfusion system 100 include a secondary circuit 120 that interfaces with the primary circuit 106 via a dialyzer 122 .
  • the dialyzer is a hollow fiber dialyzer with a 30 kD nominal molecular weight cut-off membrane.
  • the secondary circuit 120 can include a pump 124 for circulating a dialysate 126 through the dialyzer 122 .
  • a control unit monitors operating parameters (e.g., pressure, flow rate, temperature, oxygen levels, electrolyte levels, and the like) and/or organ function and viability indicators (e.g., production of albumin, urea, nitrogen oxide, and the like) via sample ports 118 a , 118 b .
  • the control unit can control the operation of pumps 108 a , 108 b , 124 and heat exchanger 114 .
  • the control unit can include a user interface for displaying one or more parameters and indicators and/or allowing a user to alter one or more parameters.
  • control unit can include storage means for storing one or more parameters and indicators and/or communication means for communicating one or more one or more parameters and indicators with another system (e.g., a personal computer, a cellular phone, a personal digital assistant, and the like).
  • control unit includes an audible and/or visual alarm to alert users when one or more parameters or indicators exceeds a threshold.
  • organ perfusion system 100 is modified to create an artificial organ production system.
  • organ perfusion system 100 includes one or more decellularization or recellularization modules (not depicted) to implement the methods described herein.
  • decellularization or recellularization modules can include appropriate media as described herein to achieve the decellularization of an organ and/or recellularization of a bioscaffold.
  • organ perfusion system can be configured to perfuse an organ to maintain vitality before actuating the decellularization module to perfuse with a detergent as described herein to decellularize the organ.
  • the recellularization module can be actuated to recellularize the organ with cells harvested from another source.
  • an organ encasement 200 is provided in FIG. 2A .
  • the organ encasement 200 a includes a first membrane 202 and a second membrane 204 .
  • the first membrane 202 and the second membrane 204 are pressed together to encase an organ 206 a .
  • the first membrane 202 and the second membrane 204 are flexible and therefor conform to the geometry of the organ 206 a .
  • the organ 206 a is uniformly supported by one or more of the membranes 202 , 204 in a manner similar to a human lying in a hammock.
  • membranes 202 and 204 are formed by a single sheet of a biocompatible material that is folded upon itself.
  • FIG. 2B depicts an embodiment of an organ encasement 200 b encasing a rat liver 206 b.
  • membranes 202 and 204 are biocompatible materials.
  • the membranes 202 , 204 are gas permeable.
  • the membranes 202 , 204 can also be elastic and/or deformable to better compliment the geometry of the organ 206 .
  • Suitable membrane materials 202 , 204 include medical-grade silicone membranes (also known as polydimethylsiloxane or PDMS).
  • the membranes 202 , 204 can be fabricated from USP Class VI silicon. Silicone membranes are available from manufacturers such as McMaster-Carr Supply Company of Santa Fe Springs, Calif. and MedArray, Inc. of Ann Arbor, Mich.
  • Other suitable membranes 202 , 204 include thermoplastic or thermoset polymers, amniotic membranes, small intestinal submucosa (SIS), fascia, dura mater, peritoneum, pericardium, and the like.
  • SIS small intestinal submucosa
  • the membranes 202 , 204 are transparent, translucent, or opaque.
  • the membranes 202 , 204 can be sterilized through various techniques (e.g., heat, steam, gases such as ethylene oxide or propylene oxide, and the like).
  • Membranes 202 , 204 can be water-repellant, capable of forming watertight seals, non-stick, have low chemical reactivity, and/or have low toxicity.
  • the membranes 202 , 204 do not support microbiological growth.
  • Membranes 202 and 204 can be the same material or can be different materials. For example, if organ encasement 200 is designed for orientation wherein the first membrane 202 is the upper membrane and the second membrane 204 is the lower membrane, first membrane 202 can be a membrane selected for maximal gas permeability and second membrane 204 can be selected for maximal compliance to better support organ 206 .
  • the organ encasement 200 is suspended so that the organ encasement 200 does not rest on a rigid surface, thereby causing pressure points on the organ 206 via one of membranes 202 , 204 .
  • Suspension can be accomplished via a variety of devices.
  • one or more clips can attach to one or more of membranes 202 , 204 to suspend the organ encasement device 200 .
  • a plurality of grommets can be formed one or more of membranes 202 , 204 to suspend the organ encasement device 200 .
  • a rigid or substantially rigid frame is interposed between membranes 202 , 204 . The rigid frame can then be supported or suspended on one or more supports.
  • Membranes 202 , 204 can be coupled via a variety of means.
  • the membranes 202 , 204 (and, in some embodiments, the rigid or substantially rigid frame) can be coupled by magnets, screws, bolts, nuts, rivets, adhesives, and/or zipper storage assemblies.
  • Membranes 202 , 204 can be coupled to allow for one or more cannulae and/or lumens to pass between membranes 202 , 204 for perfusion of organ 206 .
  • membranes 202 , 204 are fabricated with one or more access ports to receive one or more cannulae and/or lumens.
  • a method 300 of harvesting adult cells is provided.
  • an organ is perfused with a perfusate to improve the vitality of the adult cells.
  • Perfusion enhances the recovery of cells from damaged organs by improving cell health before the cells are isolated from the organ. Additionally, perfusion resuscitates damaged organs.
  • the organ culture perfusion step S 302 can be accomplished in the organ perfusion system 100 described herein or in similar devices.
  • the organ culture perfusate in step S 302 can vary to reflect varying species, organs, and organ health. For rat livers, about 50 mL of perfusate is sufficient. Several litters of perfusate may be required for human livers.
  • the perfusate include a solution having an osmolality of about 100 mOsm to about 500 mOsm, one or more oxygen carriers, one or more antioxidants, one or more anti-inflammatory agents, one or more vasodilators, one or more amino acids, one or more buffers, one or more inorganic salts, one or more substrates for metabolism, one or more hormones, one or more antibiotics, plasma, one or more anticoagulants, and/or one or more agents to maintain oncotic pressure for the perfusate between about 15 mm Hg and about 45 mm Hg.
  • the base solution can be Williams' Medium E solution (available from Sigma-Aldrich Corp. of St. Louis, Mo.).
  • the one or more oxygen carriers can include erythrocytes (e.g., about 20% hematocrit).
  • the one or more antioxidants can include bucillamine. See Farin Amersi, et al, “Bucillamine, a thiol antioxidant, prevents transplantation-associate reperfusion injury,” 99(13) Proc. Nat'l Acad. Sci. 8915-20 (2002).
  • the one or more anti-inflammatory agents can include hydrocortisone.
  • the one or more vasodilators can include alpha-adrenoceptor antagonists (“alpha-blockers”), endothelin receptor antagonists (“ERAs”), angiotensin converting enzyme inhibitors (“ACE inhibitors”), and the like.
  • the one or more amino acids can include L-argenine, L-glutamine, and the like.
  • the one or more buffers can include phosphate buffered saline (“PBS”), Krebs-Ringer buffer (“KRB”) (available from Sigma Aldrich, Inc. of St. Louis, Mo.), and the like.
  • the one or more inorganic salts can include sodium, calcium, potassium, and the like.
  • the one or more substrates for metabolism can include glucose and other carbohydrates, lactate, fatty acids, other energy sources, vitamins, and the like.
  • the one or more hormones can include insulin (e.g., about 2 U/L).
  • the one or more antibiotics can include penicillin (e.g., about 40,000 U/L) and/or streptomycin (e.g., about 40 mg/L).
  • the plasma can have a volume-volume percentage of about 10%.
  • the one or more anticoagulants can include heparin (e.g., about 1000 U/L).
  • the one or more agents for maintaining oncotic pressure can include albumin, polyethylene glycol, and the like.
  • the oxygen tension of the perfusate is maintained between about 50 mm Hg and about 150 mm Hg. In some embodiments, the temperature of the perfusate is maintained between about 4° C. and about 42° C.
  • the organ culture perfusion step S 302 includes circulating a dialysate through a secondary circuit 120 (S 302 a ).
  • the operating parameters for the organ perfusions system 100 are as follows:
  • step S 304 adult cells are isolated from the organ.
  • the adult cells are isolated by perfusing the organ with a collagenase perfusate (step 304 a ).
  • the collagenase perfusate can include a solution having an osmolality of about 100 mOsm to about 500 mOsm, one or more enzymes (e.g., collagenase I, collagenase II, collagenase III, collagenase IV, collagenase V, collagenase VI, trypsin, hyaluronidase and the like), one or more oxygen carriers, one or more antioxidants, one or more anti-inflammatory agents, one or more vasodilators, one or more amino acids, one or more buffers, one or more inorganic salts, one or more substrates for metabolism, one or more hormones, one or more antibiotics, plasma, and one or more anticoagulants.
  • Collagenase IV can be obtained from clostridium histolyticum bacteria. The base
  • the collagenase perfusate breaks the peptide bonds in collagen molecules in the organ to release the adult cells.
  • the adult cells can be recovered from the collagenase perfusate through a variety of tissue culture methods.
  • the collagenase perfusate is filtered to isolate the adult cells (step 304 b ).
  • the collagenase perfusate can be filtered through one or more filters having pore sizes ranging from about 1 ⁇ m to about 1,000 ⁇ m.
  • density centrifugation can be performed on the perfusate to isolate the adult cells from the collagenase perfusate.
  • step S 306 the adult cells are cultured in accordance with existing tissue culture methods.
  • a variety of hepatocyte isolation and preservation protocols are described in U.S. Pat. Nos. 5,602,026 and 5,942,436, U.S. Patent Application Publication No. 2006/0019326, and James C. Y. Dunn et al., “Long-Term in Vitro Function of Adult Hepatocytes in a Collagen Sandwich Configuration,” 7(3) Biotechnology Progress 237-45 (1991). Hepatocytes can be cryopreserved for later use as described in Claire Terry et al., “Cryopreservation of isolated human hepatocytes for transplantation: State of the art,” 53 Cryobiology 149-59 (2006).
  • the cell isolation method 300 can recover a single type of cell or can recover multiple types of cells.
  • hepatocytes, endothelial cells, cholangiocytes (bile duct cells), Kupffer cells, stellate cells, and smooth muscle cells can be isolated from the liver.
  • Other organs will yield different types of cells as will be appreciated by one of skill in the art.
  • islet cells e.g., insulin-producing beta cells
  • acinar cells can be isolated from the pancreas.
  • a method 400 of preparing a bioscaffold is provided.
  • step S 402 one or more vessels of the organ are cannulated.
  • the organ is perfused with a decellularization medium.
  • the decellularization medium can include one or more detergents, one or more vasodilators, one or more amino acids, one or more buffers, one or more inorganic salts, one or more substrates for metabolism, and/or one or more enzymes.
  • the one or more detergents can include sodium dodecyl sulfate (SDS) and Triton X-100 (C 14 H 22 O(C 2 H 4 O) n ) (available from Sigma Aldrich, Inc. of St. Louis, Mo.).
  • the one or more vasodilators can include alpha-adrenoceptor antagonists (“alpha-blockers”), endothelin receptor antagonists (“ERAs”), angiotensin converting enzyme inhibitors (“ACE inhibitors”), and the like.
  • the one or more amino acids can include L-glutamine, L-arginine, and the like.
  • the one or more buffers can include phosphate buffered saline (“PBS”).
  • the one or more inorganic salts can include sodium, calcium, potassium, and the like.
  • the one or more substrates for metabolism can include glucose and other carbohydrates, lactate, fatty acids, other energy sources, vitamins, and the like.
  • the one or more substrates for enzymes can include collagenase I, II, III, IV, V, or VI.
  • the decellularization medium can also include a serum as discussed in Liqiong Gui et al., “Novel Utilization of Serum in Tissue Decellularization,” 16(2) Tissue Engineering 173-84 (2010).
  • Suitable serums include human serum and non-human serums such as fetal bovine serum (FBS), porcine serum, and the like.
  • Serums can be selected from the same or different species as the organ to be decellularized and can even be provided from the donor of the organ and/or recipient of a recellularized organ to be fabricated from the organ.
  • One or more serums can be mixed with other materials as discussed herein and in Gui et al. and can be administered as part of a single decellularization medium or as part of a follow-on decellularization medium to further remove DNA after an initial decellularization.
  • the temperature of the decellularization medium can be maintained between about 4° C. and about 42° C.
  • the decellularization process may last between about 4 hours and about one month.
  • the decellularization process may last for about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, about 14 days, about 21 days, about 28 days, and the like.
  • step S 406 the bioscaffold is preserved.
  • the bioscaffold can be washed with one or more isotonic wash mediums (e.g., saline, phosphate buffered saline, and the like).
  • the bioscaffold can be placed in storage medium that includes one or more antimicrobial agents.
  • the storage medium can include one or more antimicrobial agents.
  • the one or more antimicrobial agents can include one or more antibiotics and/or fungicides.
  • the one or more antibiotics and/or fungicides can include penicillin (e.g., about 40,000 U/L), streptomycin (e.g., about 40 mg/L), (amphotericin B (e.g., about 2.5 ⁇ g/mL), and the like.
  • penicillin e.g., about 40,000 U/L
  • streptomycin e.g., about 40 mg/L
  • amphotericin B e.g., about 2.5 ⁇ g/mL
  • the bioscaffold can be stored.
  • a variety of storage techniques can be employed.
  • the bioscaffold is stored at room temperature, at less than about 4° C., at about ⁇ 20° C. or about ⁇ 40° C. (e.g., in a freezer), or at about ⁇ 180° C. (e.g., in liquid nitrogen).
  • the bioscaffold can be perfused indefinitely.
  • the bioscaffold is dried.
  • the bioscaffold is stored using subzero nonfreezing techniques as described in publications such as Joaquin V.
  • a method 500 of preparing an artificial organ from a bioscaffold is provided.
  • the bioscaffold can be a decellularized organ, for example, a decellularized organ prepared according to the methods provided herein.
  • the bioscaffold is a bioartificial organ device such as those described in U.S. Pat. No. 7,371,400 and U.S. Patent Application Publication Nos. 2006/0019326 and 2007/0148139.
  • step S 502 one or more vessels of the bioscaffold are cannulated.
  • the bioscaffold is seeded with a cell suspension.
  • the cell suspension includes a solution having an osmolality ranging from about 100 mOsm to about 500 mOsm, a plurality of cells of one or more types, one or more oxygen carriers, one or more antioxidants, one or more anti-inflammatory agents, one or more vasodilators, one or more amino acids, one or more buffers, one or more inorganic salts, one or more substrates for metabolism, one or more hormones, one or more antibiotics, plasma, one or more anti-coagulants, and/or one or more agents to maintain oncotic pressure of the suspension between about 15 and about 45 mm Hg.
  • the cell suspension can include a single type of cells or multiple types of cells.
  • a liver bioscaffold can be seeded with hepatocytes, endothelial cells, cholangiocytes (bile duct cells), Kupffer cells, stellate cells, and/or smooth muscle cells
  • a pancreas bioscaffold can be seeded with islet cells (e.g., insulin-producing beta cells) or acinar cells.
  • islet cells e.g., insulin-producing beta cells
  • acinar cells e.g., insulin-producing beta cells
  • Particular cell types can be seeded in specific locations of the bioscaffold.
  • each cell type can be generally perfused throughout the bioscaffold.
  • the cell suspension can, but need not, include cells harvested from the same species as the bioscaffold and/or the recipient of the recellularized bioscaffold.
  • the base solution can be Williams' Medium E solution (available from Sigma-Aldrich Corp. of St. Louis, Mo.).
  • the one or more oxygen carriers can include erythrocytes (e.g., about 20% hematocrit).
  • the one or more antioxidants can include bucillamine.
  • the one or more anti-inflammatory agents can include hydrocortisone.
  • the one or more vasodilators can include alpha-adrenoceptor antagonists (“alpha-blockers”), endothelin receptor antagonists (“ERAs”), angiotensin converting enzyme inhibitors (“ACE inhibitors”), and the like.
  • the one or more amino acids can include L-glutamine, L-arginine, and the like.
  • the one or more buffers can include phosphate buffered saline (“PBS”).
  • PBS phosphate buffered saline
  • the one or more inorganic salts can include sodium, calcium, potassium, and the like.
  • the one or more substrates for metabolism can include glucose and other carbohydrates, lactate, fatty acids, other energy sources, vitamins, and the like.
  • the one or more hormones can include insulin (e.g., about 2 U/L).
  • the one or more antibiotics can include penicillin (e.g., about 40,000 U/L) and/or streptomycin (e.g., about 40 mg/L).
  • the plasma can have a volume-volume percentage of about 10%.
  • the one or more agents for maintaining oncotic pressure can include albumin, polyethylene glycol, and the like.
  • the bioscaffold can be seeded by perfusing the bioscaffold with the cell suspension. In other embodiments, the bioscaffold is seeded by injecting the bioscaffold with the cell suspension in one or more locations. In still other embodiments, the bioscaffold is seeded by injecting cells in multiple phases as discussed herein in the context of FIG. 25 .
  • the recellularized bioscaffold is perfused with an organ culture perfusate to promote engrafting of the cells to the bioscaffold and organ function.
  • the organ culture perfusate can include the same or similar components of the perfusate for enhancing cell vitality as described herein.
  • the recellularized bioscaffold can be perfused for hours, days, weeks, months, or indefinitely.
  • the recellularized bioscaffold can be perfused for about 4 hours, about 8 hours, about 12 hours, about 16 hours, about 20 hours, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, about two weeks, about three weeks, and about one month.
  • the recellularized bioscaffold is preserved.
  • the recellularized bioscaffold can be preserved in a manner similar to the preservation of a decellularized organ as discussed herein, namely washing with one or more isotonic wash mediums, continuous perfusion, drying, storage in a medium including one or more antimicrobial agents, storage at less than about 4° C., about ⁇ 20° C., about ⁇ 40° C., and about ⁇ 180° C., and/or subzero nonfreezing storage.
  • recellularized bioscaffold can be transplanted into a subject by a health care provider in accordance with known surgical techniques.
  • the recellularized bioscaffold can be used an in vitro model (e.g., an in vitro pharmacologic model) in accordance with known laboratory techniques such as those described in U.S. Patent Application Publication No. 2006/0019326.
  • Organ rehabilitation can be particularly advantageous because, in some embodiments, a rehabilitated organ can be fabricated solely from a subject's own organ, thereby minimizing the risk of transplant rejection. Accordingly, in some embodiments the organ can be harvested from the subject, rehabilitated according to the method below, and transplanted to the same subject. During the interim period when the rehabilitation method is performed, the subject can be sustained with one or more external support systems (e.g. dialysis machines) and/or internal bioartificial organ devices (e.g. bioartificial liver devices) such as those described in U.S. Patent Application Publication No. 2007/0148139.
  • external support systems e.g. dialysis machines
  • bioartificial organ devices e.g. bioartificial liver devices
  • step S 602 the organ is perfused to improve the vitality of adult cells as described herein.
  • the organ is divided into first portion and a second portion.
  • Division of the organ into the first and section portion can be accomplished using existing surgical techniques, including, but not limited to living donor transplantation techniques.
  • living donor liver transplantation LDLT
  • either the right lobe or the left lobe of the liver is resected from the donor for transplantation to the recipient.
  • LDLT techniques are described in publications such as S. A. Shah et al., “Adult-to-adult living donor liver transplantation,” 20(5) Can. J. Gastroenterol.
  • the first portion and the second portion are processed independently.
  • the first portion is perfused with a decellularization medium and preserved, as described herein.
  • the adult cells are cultured as described herein.
  • the first portion is recellularized with a suspension of adult cells as described herein and perfused with an organ culture perfusate as described herein.
  • the first portion is preserved.
  • the first portion can be preserved in a manner similar to the preservation of a decellularized organ as discussed herein, namely washing with one or more isotonic wash mediums, continuous perfusion, drying, storage in a medium including one or more antimicrobial agents, storage at less than about 4° C., about ⁇ 20° C., about ⁇ 40° C., and about ⁇ 180° C., and/or subzero nonfreezing storage.
  • step S 620 the first portion can be transplanted into a subject by a health care provider in accordance with known surgical techniques.
  • the first portion and the second portion are substantially equal in size (e.g. mass and/or volume).
  • the first portion is larger than the first portion to provide for a larger vascular structure to support an increased number of cells.
  • the first and the second portion are selected based on the relative health of the vascular structure and the target cells in the various regions of the organ. For example, if “region A” of an organ had relatively intact vasculature and “region B” of the organ had relatively healthy target cells, region A could be selected decellularization and region B could be selected for cell isolation.
  • Recellularized organs can be used in a variety of research applications.
  • recellularized organs e.g., recellularized livers
  • drug discovery e.g., ADMET testing
  • Entire recellularized organs or portions thereof can be used, depending on the needs of a particular application.
  • a recellularized bioscaffold or recellularized first portion of an organ can be used as an in vitro model.
  • the entire recellularized bioscaffold can be used as an in vitro model.
  • the recellularized bioscaffold can be sliced accordingly to existing laboratory techniques and plated for use as an in vitro model.
  • FIG. 7A another method 700 A of creating a research tool is provided.
  • a decellularized organ is sliced according to existing laboratory techniques.
  • the decellularized organ can be produced according to the methods described herein or other methods. Additionally or alternatively, one or more mechanical methods such as agitation and/or sonication can be used to decellularize the organ.
  • step S 704 the decellularized organ slice is plated in a vessel such as a Petri dish.
  • the Petri dish can, in some embodiments, contain an agar to support the organ slice and cells added in step S 706 .
  • the decellularized organ slice is seeded with cells.
  • the cells can be seeded by placing the cells on top of the decellularized organ slice (e.g., by pouring or transferring a recellularization medium containing the cells with a pipette or other device).
  • the recellularized organ slices are cultured, for example, with a culture medium described herein.
  • the recellularized organ slices are stored according to existing laboratory protocols.
  • the recellularized organ slices are used in an experiment (e.g., a pharmacologic experiment).
  • one or more test agents can be applied to the recellularized organ slice.
  • the one or more test agents and the recellularized organ slice can then be incubated to form an enzyme-substrate complex between the one or more test agents and one or more enzymes contained in the recellularized organ slice.
  • One or more metabolites of the test agent can be then be detected with various laboratory techniques (e.g., mass spectrometry, fluorescence screening, and the like).
  • FIG. 7B another method 700 B of creating a research tool is provided.
  • step S 714 an organ is sliced according to existing laboratory techniques.
  • the organ slices are placed in a vessel such as a Petri dish.
  • the Petri dish can, in some embodiments, contain an agar to support the organ slice and cells added in step S 720 .
  • the organ slice is decellularized, for example, by exposing the organ slice to a decellularization medium as discussed herein. Additionally or alternatively, one or more mechanical methods such as agitation and/or sonication can be used to decellularize the organ slice.
  • steps S 716 and S 718 can be performed in any order (i.e., the organ can be plated before decellularization or can be decellularized after plating).
  • the decellularized organ slice is seeded with cells.
  • the cells can be seeded by placing the cells on top of the decellularized organ slice (e.g., by pouring or transferring a recellularization medium containing the cells with a pipette or other device).
  • the recellularized organ slices are cultured, for example, with a culture medium described herein.
  • the recellularized organ slices are stored according to existing laboratory protocols.
  • the recellularized organ slices are used in an experiment (e.g., a pharmacologic experiment).
  • one or more test agents can be applied to the recellularized organ slice.
  • the one or more test agents and the recellularized organ slice can then be incubated to form an enzyme-substrate complex between the one or more test agents and one or more enzymes contained in the recellularized organ slice.
  • One or more metabolites of the test agent can be then be detected with various laboratory techniques (e.g., mass spectrometry, fluorescence screening, and the like).
  • Test agents can include, but are not limited to, opioid analgesics, anti-inflammatory drugs such as antihistamines and non-steroidal anti-inflammatory drugs (NSAIDs), diuretics such as carbonic anhydrase inhibitors, loop diuretics, high-ceiling diuretics, thiazide and thiazide-like agents, and potassium-sparing diuretics, agents that impinge on the renal and cardiovascular systems such as angiotensin-converting enzyme inhibitors, cardiac drugs such as organic nitrates, calcium channel blockers, sympatholytic agents, vasodilators, ⁇ -adrenergic receptor agonists and antagonists, ⁇ -adrenergic receptor agonists and antagonists, cardiac glycosides, anti-arrhythmic drugs, agents that affect hyperlipoproteinemias such as 3-hydroxymethylglutaryl-coenzyme A (HMG-CoA) inhibitors, anti-neoplastic agents such as alkylating agents, antimetabolites, natural products, antibiotics, and other
  • Enzymes can include, but are not limited to, cytochrome P450, alkaline phosphatase, ⁇ -galactosidase, ⁇ -galactosidase, ⁇ -glucosidase, ⁇ -glucosidase, ⁇ -glucuronidase, ⁇ -glucuronidase, ⁇ -amylase, NADPH-cytochrome P450 reductase, cytochrome b 5 , N-demethylase, O-demethylase, acetylcholinesterase, pseudocholinesterase, epoxide hydrolase, amidases, uridine diphosphate (UDP)-glucuronosyltransferases, phenol sulfotransferase, alcohol sulfotransferase, sterid sulfotransferase, and arylamine sulfotransferase, UDP-glycosyltransferases, purine
  • FIGS. 8A-8E depict functional parameters of harvested livers during normothermic perfusion.
  • Alanine transaminase (ALT) and aspartate transaminase (AST) activities are depicted in FIGS. 8A and 8B as indicators of hepatocellular damage.
  • Both AST and ALT accumulated during the first 180 minutes of perfusion and then decreased. These values were severalfold higher than those for the control group of freshly isolated livers not subjected to any warm ischemia. Neither ALT nor AST were detected in the dialysate (data not shown).
  • Bile secretion and oxygen consumption describe the metabolic state of the liver. Bile was produced at a constant rate throughout the perfusion as depicted in FIG. 8C . This rate was 40% lower compared to freshly isolated livers.
  • the Oxygen Uptake Rate (OUR) of warm ischemic livers declined rapidly during the first 60 minutes of the perfusion and then remained stable as depicted in FIG. 8D . This behavior was very similar to the control group.
  • the OURs of the perfused warm ischemic and freshly isolated livers were very similar in the plateau region beyond 60 minutes.
  • SCS Static Cold Storage
  • livers were harvested from heparinized Lewis rats and held at 34° C. for 60 minutes. After one hour of ischemia, one group of livers was perfused in the organ perfusion system depicted in FIG. 1 , and cell isolation was performed with livers in the other group. Hepatocytes were isolated using a two-step collagenase perfusion procedure as described previously James C. Y. Dunn et al., “Long-Term in Vitro Function of Adult Hepatocytes in a Collagen Sandwich Configuration,” 7(3) Biotechnology Progress 237-45 (1991).
  • recovered hepatocytes were cultured in suspensions (1 ⁇ 10 6 cells/mL) for six hours with continuous gentle mixing in an incubator in 90% air/10% CO 2 at 37° C.
  • tissue culture dishes were coated with a mixed solution of nine parts type 1 rat tail collagen (1.1 mg/mL) and one part 10 ⁇ Dulbecco's Modified Eagle's Medium (DMEM) and incubated for one hour at 37° C. to form a collagen gel. After gelation, 125,000 cells per well were seeded in nine-well culture dishes, and incubated in 90% air/10% CO 2 at 37° C. The following day, after aspirating the culture medium from the dishes, the second collagen gel layer was overlaid on the hepatocytes and incubated for one hour at 37° C. After gelation, 1 mL of hepatocyte culture medium was applied, and then changed daily.
  • DMEM Dulbecco's Modified Eagle's Medium
  • Standard hepatocyte culture medium used both in suspension and collagen gel cultures includes DMEM with 10% fetal bovine serum, 7 ng/mL glucagon, 7.5 ⁇ g/mL hydrocortisone, 0.5 U/mL insulin, 20 ng/mL epidermal growth factor, 200 U/mL penicillin, and 200 ⁇ g/mL streptomycin. Group comparisons were performed via a t-test.
  • the cell yield is increased from 15 million to 100 million.
  • the viability of these recovered cells are well in the acceptable range (>85%), although slightly lower than obtained with non-ischemic livers.
  • a single asterisk (*) denotes a statistically significant difference as compared to normal livers.
  • a double asterisk (**) denotes a statistically significant difference as compared to ischemic, non-perfused livers (p ⁇ 0.05).
  • FIG. 12A depicts phase contrast microscopy of cells obtained from one-hour ischemic livers after perfusion in a collagen double gel at two weeks post-seeding. Compared to cells obtained from normal (fresh) livers depicted in FIG. 12B , there were no remarkable differences. By comparison, cells obtained from untreated ischemic livers depicted in FIG. 12C did not reach confluency, appeared steatotic and unhealthy, and dedifferentiation was observed. As depicted in FIGS.
  • urea synthesis by the hepatocytes from perfusion-treated ischemic cells when cultured in suspension for six hours was similar to those obtained from fresh livers, whereas albumin synthesis was approximately 50% of normal hepatocytes for the duration of the experiment (statistically different at p ⁇ 0.05).
  • urea and albumin synthesis by the hepatocytes from perfusion-treated ischemic cells when culture in collagen sandwich culture for 14 days was similar to that obtained from fresh livers. There was a slight drop in albumin, but the difference did not reach statistical significance for any day.
  • FIGS. 13A-13E are representative photographs of livers during the decellularization process.
  • FIGS. 14A-14D depict representative livers at 0, 10, 20, 30, and 40 minutes post-seeding, respectively.
  • the recellularized liver native matrices were then cultured for 8 hours under continuous perfusion with oxygenated culture medium.
  • a time period of eight hours was chosen as the initial period of testing because direct cell death due to perfusion damage or poor oxygenation typically kills most cells within two to four hours post-cell-isolation. Thus, eight hours would provide a good indication of the damage induced by the repopulation process.
  • the infused hepatocytes were counted to evaluate cell engraftment immediately after cell seeding and it was found that around 70% of the seeded cells remained in the livers and engrafted around the vessels with most hepatocytes in the parenchyma.
  • FIG. 15A depicts a hematoxylin and eosin (H&E) stained decellularized liver matrix.
  • FIG. 15B depicts an H&E-stained recellularized liver matrix.
  • FIG. 15C depicts a PAS-stained recellularized liver matrix. It was also notable that the vascular structure seemed to be preserved.
  • FIG. 16A depicts a negative control of TUNEL staining without antibody.
  • FIG. 16B apoptotic cells stained in green.
  • FIG. 16C depicts positive control of TUNEL staining of a DNAse exposed liver sample.
  • DAPI (4′,6-diamidino-2-phenylindole) staining is depicted in the lower left corner of FIGS. 16A-16C .
  • Less than 20% cells were positive for apoptosis. Stable oxygen uptake rates were also observed during this period.
  • FIGS. 17A and 17B To assess viability after repopulation, recellularized livers were perfusion-cultured for seven days. As depicted in FIGS. 17A and 17B , there was steady production of albumin (measured by ELISA) and urea (measured with a BUN assay kit) proving hepatic functionality in the recellularized liver. As depicted in FIG. 17B , extracellular lactate dehydrogenase levels (measured with an LDH kit) during first 80 hours of perfusion are identical to cell culture levels, indicating no significant damage to the cells in the system. In FIGS. 17A-17C , data for liver grafts repopulated with primary rat hepatocytes are represented with closed circles, while data for collagen sandwiched cultured primary rat hepatocytes are represented with open circles.
  • FIGS. 18A-18F are photographs of H&E-stained liver matrices.
  • FIGS. 18A and 18D depict an H&E-stained normal liver matrix at 20 ⁇ and 40 ⁇ power, respectively.
  • the cells were well distributed and healthy after 48 hours, with polygonal shaped cells containing multiple nuclei present. These results demonstrate that hepatocytes can be seeded in the recellularized grafts and the hepatocyte function can be maintained up to a week.
  • the decellularized grafts were transplanted to test whether the liver extracellular matrix (ECM) was capable of handling the pressure and viscosity of blood flow.
  • ECM extracellular matrix
  • the decellularized liver was subjected to resection of the right superior lobe, right inferior lobe, left lateral lobe, and the inferior and superior caudate lobes with piercing sutures to avoid damage to the stump and paracaval circulation circuit.
  • the hepatic artery, superior vena cava (SVC), and bile duct were closed.
  • the decellularized liver graft which was reduced to about 30% of the original size, was implanted into the left upper quadrant of the recipient's abdomen.
  • the inferior vena cava (IVC) was anastomosed by a stent technique.
  • the donor's portal vein was completely arterialized to the recipient's left renal artery in a stent technique.
  • a stent with an internal diameter of 0.3 mm the flow in the arterialized portal vein was regulated to achieve physiologic parameters.
  • FIGS. 23A and B The intricate vascular tree was further characterized by generating a corrosion cast of the DLM as depicted in FIGS. 23A and B.
  • Portal (red) and venous (blue) vasculature casting of both normal ( FIG. 23A ) and decellularized ( FIG. 23B ) livers showed that the larger portal and venous circulation system of vessels and the vast majority of the smaller microcirculatory branches were preserved, indicating that physiologic flow could be achieved by traversing the portal venous system and emptying into the systemic venous circulation via the hepatic vein and inferior vena cava (IVC).
  • IVC inferior vena cava
  • FIG. 24A scanning electron microscopy (SEM) images confirmed the presence of large vessels within the DLM as depicted in FIG. 24A . Remnants of small vessels closely resembling a portal triad were also observed as depicted in FIG. 24B .
  • the honeycomb of about 30- ⁇ m diameter gaps in the ECM depicted in FIG. 24C is thought to be the footprint of hepatocytes removed during decellularization.
  • Presence of a functional vascular bed in the DLM offers the ability to control hepatocyte engraftment and characterize liver-specific metabolic function in vitro prior to transplantation of the recellularized graft.
  • Primary rat hepatocytes were introduced via portal vein perfusion recirculation.
  • the recellularized liver grafts were transferred into a specially-designed perfusion chamber as depicted in FIG. 2B for in vitro culture.
  • the perfusion chamber features two hermetically-sealed silicon sheets, forming a pouch filled with culture medium. This design avoids rigid surfaces, thereby preventing development of pressure spots, while enabling sterile culture of the recellularized grafts up to two weeks in vitro. Histological staining of recellularized sections at 4 hours and at 1 day, 2 days and 5 days revealed that at 4 hours, a majority of the cells remain in and around the vessels, whereas at 1 day and 2 days, the cells leave the vessels and are distributed throughout the matrix. No changes in the distribution of the cells were observed within the matrix at 5 days, as compared to previous days
  • hepatocyte albumin production and urea synthesis was quantified as depicted in FIGS. 31A and 31B .
  • Albumin production rate by the recellularized liver graft was 27.6 ⁇ 7.0 ⁇ g million per cells per day as compared to 140 ⁇ g million per cells per day by the normal adult rat liver as discussed in R. Hoffenberg, “Measurement of the synthesis of liver-produced plasma proteins with particular reference to dietary protein and amino acid supply,” 123 Biochem. J. 3P (1972).
  • RT-PCR Real Time Polymerase Chain Reaction
  • Cyp2c11 (encoding cytochrome P450, subfamily 2, polypeptide 11) Gstm2 (glutathione S-transferase mu 2), Ugt1a1 (encoding UDP glucuronosyltransferase-1 family, polypeptide A1) and Cyplal (encoding cytochrome P450, family-1, subfamily a, polypeptide 1) were expressed in the recellularized liver at similar levels to those in normal liver as depicted in FIGS. 33A-33D .
  • Adh1 encoding alcohol dehydrogenase-1
  • Cyp3a18 encoding cytochrome P450, family 3, subfamily a, polypeptide 18 expression levels were higher in recellularized liver than in the sandwich plate culture control, although they were much lower than in normal liver as depicted in FIGS. 33E and 33F .
  • Primary hepatocytes are known to require between 7 to 10 days in culture before their metabolic activity and gene transcription levels stabilize as discussed in S. R. Khetani & S, N. Bhatia, “Microscale culture of human liver cells for drug development,” 26 Nature Biochemistry 120-26 (2008).
  • the median lobe of DLM was repopulated with 200 million hepatocytes and perfused with a culture medium for 10 days. This cellular amount corresponds to approximately 20% of rat's liver mass and is more than double the minimum hepatic mass necessary for therapeutic interventions. In these studies, 50 million hepatocytes were injected at each of the four steps mentioned above in the context of FIG. 25 .
  • the functional vascular structure demonstrated herein enables transplantation of the recellularized liver graft by connecting the graft to the blood supply, thereby allowing for the transplantation of a critical hepatocyte mass while avoiding ischemic damage due to poor graft perfusion.
  • recipient animals underwent unilateral nephrectomy to prepare a viable site for auxiliary liver graft transplantation.
  • the renal vein and artery were used to as ports to create blood flow within the graft.
  • the graft was perfused, quickly filling with blood, and appropriate efflux was established within 5 minutes as depicted in FIG. 34 .
  • the recellularized graft was kept in vivo for 8 hours prior to harvesting for further analysis. TUNEL staining revealed that there was minimal damage to the hepatocytes due to the arterial blood flow and consequent shear stress during 8 h of transplantation.
  • Ex vivo whole blood perfusion technique that has been shown to be representative of early graft performance and predict primary graft failure in an orthotopic rat liver transplant model in H. Tolbloom, et al., “Recovery of Warm Ischemic Rat Liver Grafts by Normothermic Extracorporeal Perfusion,” 87 Transplantation 170-77 (2009) was adopted as a surrogate model of transplantation.
  • Ex vivo perfusion system was similar to the in vitro perfusion described herein except that the perfusate consisted of whole rat blood diluted with perfusion medium (hematocrit 20%).
  • the recellularized liver graft was perfused for 24 hours during which the blood was replenished three times at eight-hour intervals to maintain the hematocrit level at 20%.
  • TUNEL staining and histological analysis showed that hepatocytes remained viable (22.6 ⁇ 13.8% TUNEL positive cells) and preserved their morphology and parenchymal position at the end of ex vivo blood perfusion.
  • hepatocytes remained metabolically active during the 24 hours of ex vivo blood perfusion as evidenced by urea and albumin secretion.
  • any functional element may perform fewer, or different, operations than those described with respect to the illustrated embodiment.
  • functional elements e.g., modules, computers, and the like
  • shown as distinct for purposes of illustration may be incorporated within other functional elements, separated in different hardware or distributed in a particular implementation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Environmental Sciences (AREA)
  • Dentistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Thermal Sciences (AREA)
  • Developmental Biology & Embryology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Virology (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Physiology (AREA)
  • Nutrition Science (AREA)
  • Mechanical Engineering (AREA)
  • Physics & Mathematics (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Agricultural Chemicals And Associated Chemicals (AREA)
  • Materials For Medical Uses (AREA)

Abstract

One aspect of the invention provides a method for harvesting adult cells from an organ. The method includes perfusing the organ with a perfusate and isolating adult cells from the organ, thereby harvesting the adult cells from the organ. Another aspect of the invention provides a method for rehabilitating an organ. The method includes: dividing the organ into a first portion and a second portion, perfusing the first portion with a decellularization medium, isolating adult cells from the second portion, and recellularizing the first portion with a suspension of the adult cells, thereby rehabilitating the organ.

Description

    RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Patent Application Ser. No. 61/222, 266, filed Jul. 1, 2009; U.S. Provisional Patent Application Ser. No. 61/304,860, filed Feb. 16, 2010; and U.S. Provisional Patent Application Ser. No. 61/330,959, filed May 4, 2010. The entire contents of each application are incorporated herein by reference.
  • STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERAL SPONSORED RESEARCH AND DEVELOPMENT
  • This invention was made with government support under Grant Nos. DK059766 and DK080942 awarded by the National Institutes of Health and Grant No. NSF CBET-0853569 awarded by the National Science Foundation. The U.S. Government has certain rights in this invention.
  • BACKGROUND
  • About thirty million people in the United States suffer from liver disorders of varying causes and about 27,000 deaths are registered annually in the United States due to liver disease. At this time, the only definitive treatment of hepatic failure is orthotopic liver transplantation. However, there is a critical shortage of organs with the total organ waiting list currently including about 100,000 requests (for all organs) and increasing at about 5% annually. Given that only organs in pristine condition are transplantable and the hidden demand for organs as an anti-aging solution will be many times the current figures, there will always be a limited pool of organs for orthotopic transplantation.
  • In addition, human hepatocytes (HHs) are in increasing demand as an ideal model for absorption, distribution, metabolism, excretion, and toxicology (ADMET) studies in drug development. HHs are ideal for drug development and testing as they enable a realistic estimation of the response of the human liver, whereas animal models are greatly limited because detoxification metabolism varies between species. Further, as liver replacement therapies such as cell transplantation, engineered hepatic tissues, and bioartificial livers approach clinical realization, the demand for human cells is expected to increase drastically.
  • Cell transplantation is a new and elegant approach that can address the need for organs and can also be used for patients that are severely ill or as a preventative therapy. However, the direct delivery of cells is inefficient due to low engraftment. A tissue-engineered vehicle designed to ensure long-term cell engraftment, viability, and functionality is a necessity to make cell transplantation work clinically. Unfortunately, tissue engineering has so far had limited success in many tissues, including the liver. The key factor that prevents advancement of the field is the lack of an ideal transplantable scaffold that has all the necessary microstructure and extracellular cues for cell attachment, differentiation, functioning, as well as vascularization, which has so far proven to be difficult to manufacture in vitro.
  • Currently, the availability and quality of human cells and tissues, including hepatocytes (the major cell type in the liver) is very limited. The only reliable source of cells and tissues is human donor livers. Stem-cell-derived hepatocytes are not fully equivalent to primary human hepatocytes (adult hepatocytes) and are time consuming and very expensive to produce in mass quantities. However, organs in good condition are used for transplantation. Therefore, only the worst quality livers are available for hepatocyte isolation. The available livers typically are either excessively fatty or are recovered from donors after cardiac death. In either case, the hepatocytes suffer extended ischemic damage, and the quantity and quality of the cells isolated is greatly reduced compared to ideal livers.
  • Accordingly, there is a need for new methods and devices for harvesting cells from damaged organs, rehabilitating damaged organs with harvested cells, and providing artificial organs containing harvested cells.
  • SUMMARY OF THE INVENTION
  • One aspect of the invention provides a method for harvesting adult cells from an organ. The method includes perfusing the organ with a perfusate and isolating adult cells from the organ, thereby harvesting the adult cells from the organ.
  • In one embodiment, the organ is a mammalian organ. In another embodiment, the organ is a human organ. In still another embodiment, the organ is a liver. The adult cells can be selected from the group consisting of: hepatocytes, endothelial cells, cholangiocytes (bile duct cells), Kupffer cells, stellate cells, and smooth muscle cells. The organ can be selected from the group consisting of: pancreas, spleen, heart, lung, and kidney. The organ can be ischemic.
  • In another embodiment, the perfusate includes one or more oxygen carriers. The one or more oxygen carriers can include erythrocytes. The perfusate can include one or more anti-inflammatory agents. The one or anti-inflammatory agents can include hydrocortisone. The perfusate can include collagenase. The perfusate can be a normothermic perfusate, a room temperature perfusate, or a hypothermic perfusate.
  • In another embodiment, the method includes culturing the harvested adult cells. The method can include providing the organ. The step of isolating the adult cells from the organ can include filtering the perfusate. The filtering the perfusate step can include filtering the perfusate through one or more mesh filters. The one or more mesh filters can have pore sizes ranging from about 1 μm to about 1000 μm. The step of isolating the adult cells from the organ can include performing density centrifugation on the perfusate. The method can include circulating the perfusate through a dialyzer. The organ can be a damaged organ. The perfusion step can improve the vitality of the adult cells.
  • Another aspect of the invention provides a method for rehabilitating an organ. The method includes: dividing the organ into a first portion and a second portion, perfusing the first portion with a decellularization medium, isolating adult cells from the second portion, and recellularizing the first portion with a suspension of the adult cells, thereby rehabilitating the organ.
  • In one embodiment, the method includes preserving the first portion. The preserving step can include washing the first portion with one or more isotonic washes. The preserving step can include storing the first portion in a storage medium. The storage medium can include one or more antimicrobial agents. The one or more antimicrobial agents can be selected from the group consisting of antibiotics and fungicides.
  • In another embodiment, the method includes culturing the adult cells. The method can include perfusing the recellularized first portion. The method can include transplanting the first portion into a subject. The organ can be previously harvested from the subject. The organ can be a damaged organ. The method can include utilizing the first portion as an in vitro model. The in vitro model can be an in vitro model of organ function. The method can include utilizing the first portion as an in vitro drug model. The in vitro drug model can be an in vitro model of drug absorption, drug metabolism, drug excretion, and/or drug toxicity. The first portion and the second portion can be substantially equal in size.
  • In another embodiment, the decellularization medium includes one or more agents selected from the group consisting of detergents, vasodilators, buffers, inorganic salts, and enzymes.
  • In another embodiment, the organ is mammalian and/or human. The organ can be selected from the group consisting of liver, pancreas, kidney, spleen, heart, and lung.
  • Another aspect of the invention provides a rehabilitated organ prepared by a method including: dividing an organ into a first portion and a second portion, perfusing the first portion with a decellularization medium, isolating adult cells from the second portion, and recellularizing the first portion with a suspension of the adult cells, thereby preparing a rehabilitated organ.
  • In one embodiment, the method includes preserving the first portion. The method can include washing the first portion with one or more isotonic washes. The method can include storing the first portion in a storage medium. The storage medium can include one or more antimicrobial agents. The one or more antimicrobial agents can be selected from the group consisting of antibiotics and fungicides.
  • The organ can be a damaged organ. The first portion and the second portion can be substantially equal in size. The decellularization medium can include one or more agents selected from the group consisting of detergents, vasodilators, buffers, inorganic salts, and enzymes. The organ can be mammalian and/or human. The organ can be selected from the group consisting of liver, pancreas, kidney, spleen, heart, and lung.
  • Another aspect of the invention provides an artificial organ including: a bioscaffold comprising an extracellular matrix having a vascular network and a plurality of adult cells. The adult cells are engrafted to the matrix and are vascularized such that the adult cells are capable of organ function.
  • In one embodiment, the plurality of adult cells were previously harvested from one or more other organs. The bioscaffold can be derived from a previously decellularized organ. The bioscaffold can be bioartificial organ device. The bioscaffold and the adult cells can be mammalian-derived. The bioscaffold and the adult cells can be human-derived. The bioscaffold and the adult cells can be derived from an organ selected from the group consisting of: liver, pancreas, kidney, spleen, heart, and lung. The bioscaffold and the adult cells can be derived from a liver. The adult cells are harvested from an organ by any method described herein.
  • In another embodiment, the bioscaffold is prepared according to a method including: cannulating one or more vessels of an organ and decellularizing the organ, thereby preparing a bioscaffold comprising a decellularized extracellular matrix having a vascular network. The decellularizing step can include perfusing the organ with a decellularization medium. The decellularization medium can include one or more agents selected from the group consisting of detergents, vasodilators, buffers, inorganic salts, and enzymes. The perfusion can be single or multidirectional perfusion. The perfusion is can be machine- or gravity-driven perfusion. The temperature of the decellularization medium can be maintained between about 4° C. and about 42° C. The organ can be perfused from about 4 hours to about 14 days. The organ can be perfused for between about 4 days and about 7 days.
  • In another embodiment, the method includes preserving the bioscaffold. The preserving step can include washing the bioscaffold with one or more isotonic washes. The preserving step can include storing the bioscaffold in a storage medium. The storage medium can include one or more antimicrobial agents. The one or more antimicrobial agents can be selected from the group consisting of antibiotics and fungicides. The bioscaffold can be stored at a temperature less than about 4° C.
  • In another embodiment, the organ is mammalian and/or human. The organ can be selected from the group consisting of liver, pancreas, kidney, spleen, heart, and lung. The organ can be a liver.
  • Another aspect of the invention provides a method for fabricating an artificial organ. The method includes: cannulating one or more vessels in a bioscaffold and cellularizing the bioscaffold by introducing a suspension of adult organ cells into the bioscaffold, thereby preparing an artificial organ.
  • In one embodiment, the method includes perfusing the cellularized bioscaffold. The perfusion can be machine- or gravity-driven perfusion. The suspension can include a solution of osmolality ranging from about 100 mOsm to about 500 mOsm, one or more oxygen carriers, one or more antioxidants, one or more anti-inflammatory agents, one or more vasodilators, one or more amino acids, one or more buffers, one or more inorganic salts, one or more substrates for metabolism, and one or more agents to maintain oncotic pressure between about 15 to about 45 mm Hg.
  • In one embodiment, the suspension of adult organ cells is introduced into the bioscaffold by injection or perfusion.
  • In another embodiment, the method includes preserving the artificial organ. The preserving step can include washing the artificial organ with one or more isotonic washes. The preserving step can include storing the artificial organ in a storage medium. The storage medium can include one or more antimicrobial agents. The one or more antimicrobial agents can be selected from the group consisting of antibiotics and fungicides. The artificial organ can be stored at a temperature less than about 4° C.
  • In another embodiment, the bioscaffold and the adult cells are mammalian-derived and/or human-derived. The bioscaffold and the adult cells can be derived from an organ selected from the group consisting of liver, pancreas, kidney, spleen, heart, and lung. The bioscaffold and the adult cells can be derived from a liver. The adult cells can be harvested from an organ by any method described herein.
  • In another embodiment, the method includes preparing the bioscaffold according to a method including: cannulating one or more vessels of an organ and decellularizing the organ, thereby preparing a bioscaffold comprising a decellularized extracellular matrix having a vascular network. Decellularizing the organ can include perfusing the organ with a decellularization medium. The decellularization medium can include one or more agents selected from the group consisting of detergents, vasodilators, buffers, inorganic salts, and enzymes. The perfusion can be single or multidirectional perfusion. The perfusion can be machine- or gravity-driven perfusion. The temperature of the decellurization medium can be maintained between about 4° C. and about 42° C. The organ can be perfused from about 4 hours to about 14 days. The organ can be perfused for between about 4 days and about 7 days.
  • In another embodiment, the method includes preserving the bioscaffold. The preserving step can include washing the bioscaffold with one or more isotonic washes. The preserving step can include storing the bioscaffold in a storage medium. The storage medium can include one or more antimicrobial agents. The one or more antimicrobial agents can be selected from the group consisting of antibiotics and fungicides. The bioscaffold can be stored at a temperature less than about 4° C.
  • In another embodiment, the organ is mammalian and/or human. The organ can be selected from the group consisting of liver, pancreas, kidney, spleen, heart, and lung. The organ can be a liver.
  • Another aspect of the invention provides an organ encasement including: a first membrane and a second membrane adapted for coupling with the first membrane to encase an organ.
  • In one embodiment, the organ encasement also includes coupling means for coupling the first and the second membrane. The coupling means can be selected from the group consisting of: magnets, screws, bolts, nuts, rivets, adhesives, and zipper storage assemblies.
  • In another embodiment, the first membrane and the second membrane are flexible. The first membrane and the second membrane can be gas permeable. The first membrane and the second membrane can be biocompatible. The first membrane and the second membrane can include one or more biocompatible materials selected from the group consisting of: silicone, thermoset polymers, thermoplastic polymers, amniotic membranes, small intestinal submucosa (SIS), fascia, dura mater, peritoneum, and pericardium.
  • Another aspect of the invention provides an organ perfusion system including: an organ encasement and a primary circuit in fluid communication with an organ encasement for circulating a perfusate.
  • In one embodiment, the primary circuit includes a pressure sensor. In other embodiments, the primary circuit can include a sampling port, a heat exchanger, a bubble trap, an oxygenator, and/or one or more pumps. The primary circuit can include a first cannula in communication with the organ and a second cannula in communication with the organ.
  • In another embodiment, the organ perfusion system includes: a dialyzer in communication with the primary circuit and a secondary circuit in communication with the dialyzer. The secondary circuit can include: a pump and a dialysate reservoir.
  • In another embodiment, the primary circuit includes one or more decellularization modules and/or one or more recellularization modules. The organ perfusion system can be adapted to preserve the organ.
  • Another aspect of the invention provides a method of perfusing an organ. The method includes: providing an organ perfusion system including an organ encasement and a primary circuit in fluid communication with an organ encasement for circulating a perfusate; placing an organ within the organ encasement; and circulating the perfusate.
  • In one embodiment, the perfusate is a normothermic perfusate. The perfusate can be a room temperature perfusate. The perfusate can be a hypothermic perfusate.
  • Another aspect of the invention provides a kit for perfusing an organ. The kit includes: an organ encasement, a perfusate, and instructions for use.
  • Another aspect of the invention provides a kit for preparing a recellularized bioscaffold. The kit includes: a plurality of adult cells, a perfusate, and instructions for use.
  • In one embodiment, the kit includes a bioscaffold encasement.
  • Another aspect of the invention provides an artificial organ production system including: an organ encasement and a primary circuit in fluid communication with an organ encasement for circulating a perfusate. The primary circuit includes: one or more decellularization modules and one or more recellularization modules.
  • Another aspect of the invention provides a method for producing a research tool. The method includes: slicing a decellularized organ to produce a decellularized organ slice, plating the decellularized organ slice, seeding the decellularized organ slice with one or more cells to produce a recellularized organ slice, and culturing the recellularized organ slice.
  • In one embodiment, the method includes storing the recellularized organ slice. The method can include using the recellularized organ slice in an experiment. The one or more cells can be adult cells and/or stem cells.
  • Another aspect of the invention provides a research tool prepared by a method including: slicing a decellularized organ to produce a decellularized organ slice, plating the decellularized organ slice, seeding the decellularized organ slice with one or more cells to produce a recellularized organ slice, and culturing the recellularized organ slice.
  • Another aspect of the invention provides a method for producing a research tool. The includes: slicing an organ to produce an organ slice; decellularizing the organ slice to produce a decellularized organ slice; seeding the decellularized organ slice with one or more cells to produce a recellularized organ slice; and culturing the recellularized organ slice.
  • In one embodiment, the method includes storing the recellularized organ slice. The method can include using the recellularized organ slice in an experiment. The one or more cells can be adult cells. The one or more cells can be stem cells. The method can include plating the decellularized organ slice.
  • Another aspect of the invention provides a research tool prepared by a method including: slicing an organ to produce an organ slice; decellularizing the organ slice to produce a decellularized organ slice; seeding the decellularized organ slice with one or more cells to produce a recellularized organ slice; and culturing the recellularized organ slice.
  • Another aspect of the invention provides a method for producing a research tool. The method includes: slicing a recellularized organ to produce a recellularized organ slice and plating the recellularized organ slice.
  • In one embodiment, the method includes storing the recellularized organ slice. The method can include using the recellularized organ slice in an experiment.
  • Another aspect of the invention provides a research tool prepared by a method including: slicing a recellularized organ to produce a recellularized organ slice and plating the recellularized organ slice.
  • In one embodiment, the research tool includes a recellularized organ slice. The research tool can also include a vessel. The recellularized organ slice can be received within the vessel. The vessel can contain an agar and wherein the recellularized organ slice is mounted on the agar. The recellularized organ slice can be seeded with cells before slicing. The recellularized organ slice can be seeded with cells after slicing.
  • Another aspect of the invention provides an assay method including: applying one or more test agents to a recellularized organ slice, incubating the recellularized organ slice, and analyzing one or more metabolites of the test agent.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • For a fuller understanding of the nature and desired objects of the present invention, reference is made to the following detailed description taken in conjunction with the accompanying drawing figures wherein like reference characters denote corresponding parts throughout the several views and wherein:
  • FIG. 1 depicts a perfusion system according to an embodiment of the invention.
  • FIGS. 2A and 2B depict organ encasement system according to an embodiment of the invention.
  • FIG. 3 depicts a method of harvesting adult cells according to an embodiment of the invention.
  • FIG. 4 depicts a method of preparing a bioscaffold according to an embodiment of the invention.
  • FIG. 5 depicts a method of recellularizing a bioscaffold according to an embodiment of the invention.
  • FIG. 6 depicts a method of organ rehabilitation according to an embodiment of the invention.
  • FIGS. 7A and 7B depict methods of creating a research tool according to embodiments of the invention.
  • FIGS. 8A-8E depict functional parameters of perfused ischemic livers during normothermic perfusion as compared to perfused fresh livers.
  • FIG. 9 depicts survival rates of transplant recipients of perfused ischemic rat livers compared to survival rates of recipients of fresh (normal) and warm ischemic livers with different modalities of preservation.
  • FIG. 10 depicts a comparison of survival rates for transplant recipients of perfused ischemic livers compared with different versions of the perfusion systems described herein.
  • FIGS. 11A and 11B depict cell yields and viability for harvested livers.
  • FIGS. 12A-12C depict phase contrast microscopy of cells obtained from ischemic livers after perfusion (FIG. 12A), healthy control livers (FIG. 12B), and one-hour ischemic livers directly isolated (FIG. 12C) in a collagen double gel, respectively, at two weeks post-seeding. FIG. 12D depicts urea production rates for hepatocytes from perfused ischemic livers compared to fresh livers during six hours of suspension culture. FIG. 12E depicts albumin secretion for hepatocytes from perfused ischemic livers compared to fresh livers during six hourse of suspension culture as evaluated with an enzyme-linked immunosorbent assay (ELISA). FIG. 12F depicts urea production rates for hepatocytes from perfused ischemic livers compared to fresh livers when cultures in collagen double gel. FIG. 12G depicts albumin secretion for hepatocytes from perfused ischemic livers compared to fresh livers when cultured in collagen double gel.
  • FIGS. 13A-13E are representative photographs of livers during the decellularization process.
  • FIGS. 14A-14D are representative photographs of liver during infusion with isolated rat hepatocytes.
  • FIGS. 15A-15C are representative photographs of H&E-stained recellularized livers.
  • FIGS. 16A-16C are representative photographs of TUNEL-stained recellularized livers after eight hours of perfusion. FIG. 16A depicts a negative control of TUNEL staining without antibodies. FIG. 16 b depicts the staining of apoptic cells in green. FIG. 16C depicts TUNEL staining of a DNAse exposed liver sample as a positive control. DAPI staining of nuclei is presented in the bottom left inset of each figure.
  • FIG. 17A-17C depict albumin, urea, and lactate dehydrogenase levels for recellularized livers compared to hepatocytes in collagen culture.
  • FIGS. 18A-18F are representative photographs of H&E-stained liver matrices. FIGS. 18A and 18D depict normal livers. FIGS. 18B and 18E depict recellularized livers 24 hours after recellularization. FIGS. 18C and 18F depict recellularized livers 48 hours after recellularization.
  • FIG. 19A is a photograph of a transplanted liver bioscaffold. FIG. 19B is a schematic of a transplanted recellularized liver.
  • FIGS. 20A-20E are representative photographs of ischemic rat livers during a decellularization process at 0 hours, 18 hours, 48 hours, 52 hours, and 72 hours, respectively. The scale bars represent 10 mm.
  • FIG. 21 is a comparison of a normal liver (top row) and a decellularized liver matrix (bottom row) produced through staining for (from left to right across each row) hematoxylin and eosin, collagen I (red), collagen IV (red), fibronectin (red), and lamin (red). Sections were counterstained with DAPI (blue). The scale bars represented 100 μm.
  • FIGS. 22A and 22B are representative photographs of decellularized left lateral and median lobes of a decellularized liver matrix with the vascular tree visible. FIG. 22A depicts the decellularized liver matrix before perfusion with Allura Red AC dye and FIG. 22B depicts the decellularized liver matrix after perfusion with Allura Red AC dye.
  • FIGS. 23A and 23B are representative photographs of corrosion cast models the vascular tree of a normal liver (FIG. 23A) and a decellularized liver matrix (FIG. 23B) with portal (red) and venous (blue) vasculature.
  • FIGS. 24A-C are scanning electron microscopy (SEM) micrographs of a decellularized liver matrix. FIG. 24A is an SEM image of a blood vessel. FIG. 24B is a section featuring bile-duct-like small vessels identified with white arrows. FIG. 24C displays the extracellular matrix within the parenchyma with hepatocyte-size free spaces. The scale bar in each of FIGS. 24A-C represents 20 μm.
  • FIG. 25 depicts an exemplary method of recellularizing a decellularized organ (e.g., a decellularized liver matrix).
  • FIG. 26 is bar graph depicting the percentage of TUNEL-positive cells in recellularized liver grafts as a function of perfusion-culture time.
  • FIG. 27 provides three images of TUNEL-stained recellularized liver grafts. The left photograph depicts a recellularized graft after 48 hours in culture. The center photograph depicts a freshly isolated liver, which was used as a negative control. The right photograph depicts a DNAse-treated normal live, which was used as a positive control. Arrows indicate TUNEL-positive cells. Each photograph contains an inset photograph depicting a Hoeschst 33258 counterstain of the respective sections. The scale bar in each of the large photographs represents 200 μm.
  • FIG. 28 is a line graph of lactate dehydrogenase release from recellularized liver grafts during perfusion culture as compared with a static sandwich culture.
  • FIGS. 29A and 29B are SEM micrographs of recellularized liver grafts after two days in culture.
  • FIG. 30 is a comparison of a normal liver (top row) and a recellularized liver graft (bottom row) produced through immunostaining after two days of culture for (from left to right across each row) hematoxylin and eosin, albumin (red), G6 pc (red), and Ugt1a (green). The scale bars represent 100 μm.
  • FIGS. 31A and 31B are line graphs depicting albumin synthesis and urea secretion, respectively, for recellularized liver grafts in comparison with a static sandwich culture.
  • FIG. 32A depicts a gene expression analysis of hepatocytes in a recellularized liver graft after two days of culture compared to a normal liver, fresh hepatocytes, and sandwich culture hepatocytes after two days of culture for phase I and phase II drug metabolism enzymes. FIG. 32B is a scatter plot comparing gene expression of phase I and phase II drug metabolism enzymes for a recellularized liver graft and sandwich culture hepatocytes after two days of culture.
  • FIGS. 33A-33F are bar graphs of gene expression of Cyp2c11, Gstm2, Ugt1a1, Cyp1a1, Adh1, and Cyp3a18, respectively. All error bars represent the standard error of the mean (n=3).
  • FIG. 34 is a series of representative images of recellularized liver graft transplantation. The top row depicts (from left to right): a transplant site, a recellularized graft at the transplant site, a transplanted graft before blood reperfusion, and a graft after declamping the renal artery. The bottom row depicts (from left to right): a transplanted graft at one minute after clamping, a transplanted graft at two minutes after clamping, a transplanted graft at four minutes after clamping, and an auxiliary recellularized liver graft in contrast with the native liver.
  • FIG. 35 is a comparison of a normal liver (top row) and a recellularized liver graft (bottom row) produced through immunostaining eight hours after transplantation for (from left to right across each row) hematoxylin and eosin, albumin (red), G6pc (red), and Ugt1a (green). Sections were counterstained with Hoescht 33258 (blue). The scale bars represent 100 μm.
  • DETAILED DESCRIPTION
  • The instant invention is most clearly understood with reference to the following definitions:
  • As used in the specification and claims, the singular form “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise.
  • An “adult cell” shall be understood to mean any cell capable of performing essential organ functions. The term “adult cell,” as used in the specification and claims, shall be distinguished from the term “stem cell.”
  • The term “biocompatible” shall be understood to refer to any material that is nontoxic and, optionally, does not provoke an immunological rejection. Biocompatible materials can be naturally occurring or synthetic and permit diffusion of liquids, gases, and/or ions. Biocompatible materials in accordance with the invention include, for example, membranous materials selected from the group consisting of silicone, thermoset polymers, thermoplastic polymers, amniotic membranes, small intestinal submucosa (SIS), fascia, dura mater, peritoneum, and pericardium.
  • A “bioscaffold” shall be understood to include any structure that provides an extracellular matrix sufficient to support one or more cells.
  • A “damaged organ” shall be understood to indicate an organ that is in less than ideal condition for transplantation, such that the expected probability of transplant success is reduced. Examples of damaged organs include, but are not limited to, organs that suffer warm ischemia for more than 30 minutes, organs that suffer cold ischemia for more than 12 hours, moderate or highly steatotic livers (e.g., livers with greater than 30% fat), fibrotic livers, cirrhotic livers, livers from patients afflicted with hepatitis C or HIV, and the like.
  • The term “decellularize” shall be understood to mean any process for the removing one or more cells from an organ. Preferably, a decellularization process maintains the extracellular matrix and vascular structure of the organ.
  • “Essential hepatic function” shall be understood to include one or more of the following: protein synthesis; protein storage; transformation of carbohydrates; synthesis of cholesterol, bile salts, and/or phospholipids; and detoxification, modification, and/or excretion of exogenous and endogenous substances.
  • An “extracellular matrix” shall be understood to be the portion of an organ and/or a bioscaffold that provides structural and life support for one or more cells.
  • A “health care provider” shall be understood to mean any person providing medical care to a patient. Such persons include, but are not limited to, medical doctors, physician's assistants, nurse practitioners (e.g., an Advanced Registered Nurse Practitioner (ARNP)), nurses, residents, interns, medical students, or the like. Although various licensure requirements may apply to one or more of the occupations listed above in various jurisdictions, the term health care provider is unencumbered for the purposes of this patent application.
  • “Hypothermic” shall be understood to mean temperatures below room temperature. For example, “hypothermic” temperatures include, but are not limited to, temperatures between about 0° C. to about 15° C., temperatures between about 1° C. to about 8° C., temperatures between about 3° C. to about 5° C., and the like.
  • An “in vitro model” shall be understood to be a model created outside of an organism of a subset of the organism's constituent parts, for example, for studying the function of the constituent parts. An “in vitro drug model” shall be understood to be an in vitro model created to study the interaction of a drug, pharmaceutical, test agent, and the like with a subset of an organism's constituent parts. Interactions can include absorption, distribution, metabolism, toxicity, and the like.
  • “Normothermic” shall be understood to mean temperatures above room temperature. For example, “normothermic” temperatures include, but are not limited to, temperatures between about 25° C. and about 42° C., temperatures between about 30° C. and about 38° C., temperatures between about 37° C. and about 37.5° C., and the like.
  • An “organ” is a tissue that performs a specific function or group of functions within an organism. The term organ includes the heart, lungs, stomach, liver, gallbladder, pancreas, spleen, kidneys, and the like.
  • A “perfusate” shall be understood to be any fluid capable of improving or maintaining the vitality of a cell, tissue, organ (including decellularized and recellularized organs), bioscaffold, and the like. Improving or maintaining vitality can include one or more of the following: maintenance of appropriate osmotic pressure, maintenance of appropriate oncotic pressure, maintenance of appropriate temperature, inhibition of decay, inhibition of microbial growth, and the like.
  • The term “recellularize” shall be understood to be any process for engrafting one or more cells within a decellularized organ or bioscaffold.
  • “Room temperature” shall be understood to mean a temperature between about 15° C. and about 25° C. For example, “room temperature” includes, but is not limited to, temperatures between about 18° C. and about 23° C., temperature between about 19° C. and about 21° C., temperatures between about 24° C. and about 25° C., temperatures between about 20° C. and about 21° C., and the like.
  • A “storage medium” shall be understood to be any substance for preserving vitality of a cell, tissue, organ (including decellularized and recellularized organs), bioscaffold, and the like. Preservation of vitality can include one or more of the following: maintenance of appropriate osmotic pressure, maintenance of appropriate oncotic pressure, maintenance of appropriate temperature, inhibition of decay, inhibition of microbial growth, and the like.
  • A “subject” shall be understood to include any mammal including, but not limited to, humans, primates, swine, cows, sheep, and rats.
  • A “test agent” shall be understood to include any substance that is evaluated for its ability to diagnose, cure, mitigate, treat, or prevent disease in a subject, or is intended to alter the structure or function of the body of a subject. A test agent in an embodiment can be a “drug” as that term is defined under the Federal Food, Drug, and Cosmetic Act at 21 U.S.C. §321(g)(1). Test agents include, but are not limited to, chemical compounds, biologic agents, proteins, peptides, nucleic acids, lipids, polysaccharides, supplements, diagnostic agents and immune modulators.
  • A “vascular network” shall be understood to be the portion of an organ that provides fluid (e.g., blood) access to one or more cells. The vascular network can form part of the extracellular matrix and is capable of handling the pressure and viscosity of physiological fluids.
  • Aspects of the invention provide new methods and devices for harvesting cells from and rehabilitating damaged organs.
  • Organ Perfusion Systems
  • Referring to FIG. 1, a perfusion system 100 is provided for perfusing an organ prior to cell isolation. An organ 102 (e.g., a liver) is received within a perfusion chamber 104. The perfusion chamber 102 can be any vessel capable of holding an organ and a perfusate. In some embodiments, the perfusion chamber 102 is a vessel such as a glass or plastic bowl. In other embodiments, the perfusion chamber 102 is an organ encasement 200 as described herein.
  • A primary circuit 106 circulates a perfusate through the organ 102 and perfusion chamber 104. The primary circuit 106 can include one or more pumps 108 a, 108 b to promote perfusate circulation. Alternatively, the perfusion can be gravity-driven. The primary circuit can also include an oxygenator 110, a bubble trap 112, a heat exchanger 114, a pressure sensor 116, and one or more sampling ports 118 a, 118 b.
  • Some embodiments of the perfusion system 100 include a secondary circuit 120 that interfaces with the primary circuit 106 via a dialyzer 122. In some embodiments, the dialyzer is a hollow fiber dialyzer with a 30 kD nominal molecular weight cut-off membrane. The secondary circuit 120 can include a pump 124 for circulating a dialysate 126 through the dialyzer 122.
  • In some embodiments, a control unit (not depicted) monitors operating parameters (e.g., pressure, flow rate, temperature, oxygen levels, electrolyte levels, and the like) and/or organ function and viability indicators (e.g., production of albumin, urea, nitrogen oxide, and the like) via sample ports 118 a, 118 b. The control unit can control the operation of pumps 108 a, 108 b, 124 and heat exchanger 114. The control unit can include a user interface for displaying one or more parameters and indicators and/or allowing a user to alter one or more parameters.
  • Additionally or alternatively, the control unit can include storage means for storing one or more parameters and indicators and/or communication means for communicating one or more one or more parameters and indicators with another system (e.g., a personal computer, a cellular phone, a personal digital assistant, and the like). In some embodiments, the control unit includes an audible and/or visual alarm to alert users when one or more parameters or indicators exceeds a threshold.
  • In some embodiments, organ perfusion system 100 is modified to create an artificial organ production system. In such an embodiment, organ perfusion system 100 includes one or more decellularization or recellularization modules (not depicted) to implement the methods described herein. Such modules can include appropriate media as described herein to achieve the decellularization of an organ and/or recellularization of a bioscaffold. For example, organ perfusion system can be configured to perfuse an organ to maintain vitality before actuating the decellularization module to perfuse with a detergent as described herein to decellularize the organ. Finally, the recellularization module can be actuated to recellularize the organ with cells harvested from another source.
  • Organ Encasements
  • Conventional organ perfusion systems utilize a rigid vessel such as a glass bowl as a perfusion chamber. Such rigid vessels can be problematic for several reasons. First, the geometry of rigid vessels typically does not compliment the geometry of the organ, which can cause pressure points where the organ contacts the vessel. Second, gravitational forces can deform the organ to fit into corners of the vessel.
  • In order to improve the function of organ perfusion systems, an organ encasement 200 is provided in FIG. 2A. The organ encasement 200 a includes a first membrane 202 and a second membrane 204. The first membrane 202 and the second membrane 204 are pressed together to encase an organ 206 a. The first membrane 202 and the second membrane 204 are flexible and therefor conform to the geometry of the organ 206 a. Instead of resting on a few pressure points in a rigid vessel, the organ 206 a is uniformly supported by one or more of the membranes 202, 204 in a manner similar to a human lying in a hammock.
  • In some embodiments, membranes 202 and 204 are formed by a single sheet of a biocompatible material that is folded upon itself.
  • FIG. 2B depicts an embodiment of an organ encasement 200 b encasing a rat liver 206 b.
  • Preferably, membranes 202 and 204 are biocompatible materials. In some embodiments, the membranes 202, 204 are gas permeable. The membranes 202, 204 can also be elastic and/or deformable to better compliment the geometry of the organ 206.
  • Suitable membrane materials 202, 204 include medical-grade silicone membranes (also known as polydimethylsiloxane or PDMS). For example, the membranes 202, 204 can be fabricated from USP Class VI silicon. Silicone membranes are available from manufacturers such as McMaster-Carr Supply Company of Santa Fe Springs, Calif. and MedArray, Inc. of Ann Arbor, Mich. Other suitable membranes 202, 204 include thermoplastic or thermoset polymers, amniotic membranes, small intestinal submucosa (SIS), fascia, dura mater, peritoneum, pericardium, and the like.
  • In some embodiments, the membranes 202, 204 are transparent, translucent, or opaque. The membranes 202, 204 can be sterilized through various techniques (e.g., heat, steam, gases such as ethylene oxide or propylene oxide, and the like). Membranes 202, 204 can be water-repellant, capable of forming watertight seals, non-stick, have low chemical reactivity, and/or have low toxicity. In some embodiments, the membranes 202, 204 do not support microbiological growth.
  • Membranes 202 and 204 can be the same material or can be different materials. For example, if organ encasement 200 is designed for orientation wherein the first membrane 202 is the upper membrane and the second membrane 204 is the lower membrane, first membrane 202 can be a membrane selected for maximal gas permeability and second membrane 204 can be selected for maximal compliance to better support organ 206.
  • In some embodiments, the organ encasement 200 is suspended so that the organ encasement 200 does not rest on a rigid surface, thereby causing pressure points on the organ 206 via one of membranes 202, 204. Suspension can be accomplished via a variety of devices. For example, one or more clips can attach to one or more of membranes 202, 204 to suspend the organ encasement device 200. In another example, a plurality of grommets can be formed one or more of membranes 202, 204 to suspend the organ encasement device 200. In still another example, a rigid or substantially rigid frame is interposed between membranes 202, 204. The rigid frame can then be supported or suspended on one or more supports.
  • Membranes 202, 204 can be coupled via a variety of means. For example, the membranes 202, 204 (and, in some embodiments, the rigid or substantially rigid frame) can be coupled by magnets, screws, bolts, nuts, rivets, adhesives, and/or zipper storage assemblies.
  • Membranes 202, 204 can be coupled to allow for one or more cannulae and/or lumens to pass between membranes 202, 204 for perfusion of organ 206. In some embodiments, membranes 202, 204 are fabricated with one or more access ports to receive one or more cannulae and/or lumens.
  • Methods of Harvesting Adult Cells
  • Referring to FIG. 3, a method 300 of harvesting adult cells is provided. In step S302, an organ is perfused with a perfusate to improve the vitality of the adult cells. Perfusion enhances the recovery of cells from damaged organs by improving cell health before the cells are isolated from the organ. Additionally, perfusion resuscitates damaged organs.
  • The organ culture perfusion step S302 can be accomplished in the organ perfusion system 100 described herein or in similar devices.
  • The organ culture perfusate in step S302 can vary to reflect varying species, organs, and organ health. For rat livers, about 50 mL of perfusate is sufficient. Several litters of perfusate may be required for human livers. In some embodiments, the perfusate include a solution having an osmolality of about 100 mOsm to about 500 mOsm, one or more oxygen carriers, one or more antioxidants, one or more anti-inflammatory agents, one or more vasodilators, one or more amino acids, one or more buffers, one or more inorganic salts, one or more substrates for metabolism, one or more hormones, one or more antibiotics, plasma, one or more anticoagulants, and/or one or more agents to maintain oncotic pressure for the perfusate between about 15 mm Hg and about 45 mm Hg.
  • The base solution can be Williams' Medium E solution (available from Sigma-Aldrich Corp. of St. Louis, Mo.). The one or more oxygen carriers can include erythrocytes (e.g., about 20% hematocrit). The one or more antioxidants can include bucillamine. See Farin Amersi, et al, “Bucillamine, a thiol antioxidant, prevents transplantation-associate reperfusion injury,” 99(13) Proc. Nat'l Acad. Sci. 8915-20 (2002). The one or more anti-inflammatory agents can include hydrocortisone. The one or more vasodilators can include alpha-adrenoceptor antagonists (“alpha-blockers”), endothelin receptor antagonists (“ERAs”), angiotensin converting enzyme inhibitors (“ACE inhibitors”), and the like. The one or more amino acids can include L-argenine, L-glutamine, and the like. The one or more buffers can include phosphate buffered saline (“PBS”), Krebs-Ringer buffer (“KRB”) (available from Sigma Aldrich, Inc. of St. Louis, Mo.), and the like. The one or more inorganic salts can include sodium, calcium, potassium, and the like. The one or more substrates for metabolism can include glucose and other carbohydrates, lactate, fatty acids, other energy sources, vitamins, and the like. The one or more hormones can include insulin (e.g., about 2 U/L). The one or more antibiotics can include penicillin (e.g., about 40,000 U/L) and/or streptomycin (e.g., about 40 mg/L). The plasma can have a volume-volume percentage of about 10%. The one or more anticoagulants can include heparin (e.g., about 1000 U/L). The one or more agents for maintaining oncotic pressure can include albumin, polyethylene glycol, and the like.
  • In some embodiments, the oxygen tension of the perfusate is maintained between about 50 mm Hg and about 150 mm Hg. In some embodiments, the temperature of the perfusate is maintained between about 4° C. and about 42° C.
  • In some embodiments, the organ culture perfusion step S302 includes circulating a dialysate through a secondary circuit 120 (S302 a).
  • In one embodiment, the operating parameters for the organ perfusions system 100 are as follows:
  • Flow rate=1.84±0.05 ml/min/g;
  • Portal hydrostatic pressure=12-16 cm H2O (8-12 mm Hg);
  • Hematocrit=17.8%±0.8%;
  • Inlet oxygen pressure=128.4±8.1 mm Hg;
  • Outlet oxygen pressure=47.9±1.7 mm Hg;
  • Inlet carbon dioxide pressure=30.1±1.1 mm Hg; and
  • Outlet carbon dioxide pressure=34.6±1.6 mm Hg.
  • In step S304, adult cells are isolated from the organ. In one embodiment, the adult cells are isolated by perfusing the organ with a collagenase perfusate (step 304 a). The collagenase perfusate can include a solution having an osmolality of about 100 mOsm to about 500 mOsm, one or more enzymes (e.g., collagenase I, collagenase II, collagenase III, collagenase IV, collagenase V, collagenase VI, trypsin, hyaluronidase and the like), one or more oxygen carriers, one or more antioxidants, one or more anti-inflammatory agents, one or more vasodilators, one or more amino acids, one or more buffers, one or more inorganic salts, one or more substrates for metabolism, one or more hormones, one or more antibiotics, plasma, and one or more anticoagulants. Collagenase IV can be obtained from clostridium histolyticum bacteria. The base solution and the components of the collagenase perfusate can be the same or similar to the organ culture perfusate described above.
  • The collagenase perfusate breaks the peptide bonds in collagen molecules in the organ to release the adult cells. The adult cells can be recovered from the collagenase perfusate through a variety of tissue culture methods. In one embodiment, the collagenase perfusate is filtered to isolate the adult cells (step 304 b). For example, the collagenase perfusate can be filtered through one or more filters having pore sizes ranging from about 1 μm to about 1,000 μm. Additionally or alternatively, density centrifugation can be performed on the perfusate to isolate the adult cells from the collagenase perfusate.
  • In step S306, the adult cells are cultured in accordance with existing tissue culture methods. A variety of hepatocyte isolation and preservation protocols are described in U.S. Pat. Nos. 5,602,026 and 5,942,436, U.S. Patent Application Publication No. 2006/0019326, and James C. Y. Dunn et al., “Long-Term in Vitro Function of Adult Hepatocytes in a Collagen Sandwich Configuration,” 7(3) Biotechnology Progress 237-45 (1991). Hepatocytes can be cryopreserved for later use as described in Claire Terry et al., “Cryopreservation of isolated human hepatocytes for transplantation: State of the art,” 53 Cryobiology 149-59 (2006).
  • The cell isolation method 300 can recover a single type of cell or can recover multiple types of cells. For example, hepatocytes, endothelial cells, cholangiocytes (bile duct cells), Kupffer cells, stellate cells, and smooth muscle cells can be isolated from the liver. Other organs will yield different types of cells as will be appreciated by one of skill in the art. For example, islet cells (e.g., insulin-producing beta cells) or acinar cells can be isolated from the pancreas.
  • Preparation of Bioscaffolds
  • Referring to FIG. 4, a method 400 of preparing a bioscaffold is provided. In step S402, one or more vessels of the organ are cannulated.
  • In step S404, the organ is perfused with a decellularization medium. The decellularization medium can include one or more detergents, one or more vasodilators, one or more amino acids, one or more buffers, one or more inorganic salts, one or more substrates for metabolism, and/or one or more enzymes. The one or more detergents can include sodium dodecyl sulfate (SDS) and Triton X-100 (C14H22O(C2H4O)n) (available from Sigma Aldrich, Inc. of St. Louis, Mo.). The one or more vasodilators can include alpha-adrenoceptor antagonists (“alpha-blockers”), endothelin receptor antagonists (“ERAs”), angiotensin converting enzyme inhibitors (“ACE inhibitors”), and the like. The one or more amino acids can include L-glutamine, L-arginine, and the like. The one or more buffers can include phosphate buffered saline (“PBS”). The one or more inorganic salts can include sodium, calcium, potassium, and the like. The one or more substrates for metabolism can include glucose and other carbohydrates, lactate, fatty acids, other energy sources, vitamins, and the like. The one or more substrates for enzymes can include collagenase I, II, III, IV, V, or VI.
  • The decellularization medium can also include a serum as discussed in Liqiong Gui et al., “Novel Utilization of Serum in Tissue Decellularization,” 16(2) Tissue Engineering 173-84 (2010). Suitable serums include human serum and non-human serums such as fetal bovine serum (FBS), porcine serum, and the like. Serums can be selected from the same or different species as the organ to be decellularized and can even be provided from the donor of the organ and/or recipient of a recellularized organ to be fabricated from the organ.
  • One or more serums can be mixed with other materials as discussed herein and in Gui et al. and can be administered as part of a single decellularization medium or as part of a follow-on decellularization medium to further remove DNA after an initial decellularization.
  • The temperature of the decellularization medium can be maintained between about 4° C. and about 42° C. The decellularization process may last between about 4 hours and about one month. For example, the decellularization process may last for about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, about 14 days, about 21 days, about 28 days, and the like.
  • In step S406, the bioscaffold is preserved. In step S406 a, the bioscaffold can be washed with one or more isotonic wash mediums (e.g., saline, phosphate buffered saline, and the like). In step S406 b, the bioscaffold can be placed in storage medium that includes one or more antimicrobial agents. The storage medium can include one or more antimicrobial agents. The one or more antimicrobial agents can include one or more antibiotics and/or fungicides. The one or more antibiotics and/or fungicides can include penicillin (e.g., about 40,000 U/L), streptomycin (e.g., about 40 mg/L), (amphotericin B (e.g., about 2.5 μg/mL), and the like.
  • In step S406 c, the bioscaffold can be stored. A variety of storage techniques can be employed. In one embodiment, the bioscaffold is stored at room temperature, at less than about 4° C., at about −20° C. or about −40° C. (e.g., in a freezer), or at about −180° C. (e.g., in liquid nitrogen). In another embodiment, the bioscaffold can be perfused indefinitely. In other embodiment, the bioscaffold is dried. In still another embodiment, the bioscaffold is stored using subzero nonfreezing techniques as described in publications such as Joaquin V. Rodríguez et al., “Subzero nonfreezing storage of rat hepatocytes using modified University of Wisconsin solution (mUW) and 1,4-butanediol. I—effects on cellular metabolites during cold storage,” 8(1) Annals of Hepatology 57-62 (2009).
  • Methods of Preparing Artificial Organs
  • Referring to FIG. 5, a method 500 of preparing an artificial organ from a bioscaffold is provided.
  • In some embodiments, the bioscaffold can be a decellularized organ, for example, a decellularized organ prepared according to the methods provided herein. In other embodiments, the bioscaffold is a bioartificial organ device such as those described in U.S. Pat. No. 7,371,400 and U.S. Patent Application Publication Nos. 2006/0019326 and 2007/0148139.
  • In step S502, one or more vessels of the bioscaffold are cannulated.
  • In step S504, the bioscaffold is seeded with a cell suspension. In some embodiments, the cell suspension includes a solution having an osmolality ranging from about 100 mOsm to about 500 mOsm, a plurality of cells of one or more types, one or more oxygen carriers, one or more antioxidants, one or more anti-inflammatory agents, one or more vasodilators, one or more amino acids, one or more buffers, one or more inorganic salts, one or more substrates for metabolism, one or more hormones, one or more antibiotics, plasma, one or more anti-coagulants, and/or one or more agents to maintain oncotic pressure of the suspension between about 15 and about 45 mm Hg.
  • The cell suspension can include a single type of cells or multiple types of cells. For example, a liver bioscaffold can be seeded with hepatocytes, endothelial cells, cholangiocytes (bile duct cells), Kupffer cells, stellate cells, and/or smooth muscle cells In another example, a pancreas bioscaffold can be seeded with islet cells (e.g., insulin-producing beta cells) or acinar cells. Particular cell types can be seeded in specific locations of the bioscaffold. Alternatively, each cell type can be generally perfused throughout the bioscaffold.
  • The cell suspension can, but need not, include cells harvested from the same species as the bioscaffold and/or the recipient of the recellularized bioscaffold.
  • The base solution can be Williams' Medium E solution (available from Sigma-Aldrich Corp. of St. Louis, Mo.). The one or more oxygen carriers can include erythrocytes (e.g., about 20% hematocrit). The one or more antioxidants can include bucillamine. The one or more anti-inflammatory agents can include hydrocortisone. The one or more vasodilators can include alpha-adrenoceptor antagonists (“alpha-blockers”), endothelin receptor antagonists (“ERAs”), angiotensin converting enzyme inhibitors (“ACE inhibitors”), and the like. The one or more amino acids can include L-glutamine, L-arginine, and the like. The one or more buffers can include phosphate buffered saline (“PBS”). The one or more inorganic salts can include sodium, calcium, potassium, and the like. The one or more substrates for metabolism can include glucose and other carbohydrates, lactate, fatty acids, other energy sources, vitamins, and the like. The one or more hormones can include insulin (e.g., about 2 U/L). The one or more antibiotics can include penicillin (e.g., about 40,000 U/L) and/or streptomycin (e.g., about 40 mg/L). The plasma can have a volume-volume percentage of about 10%. The one or more agents for maintaining oncotic pressure can include albumin, polyethylene glycol, and the like.
  • The bioscaffold can be seeded by perfusing the bioscaffold with the cell suspension. In other embodiments, the bioscaffold is seeded by injecting the bioscaffold with the cell suspension in one or more locations. In still other embodiments, the bioscaffold is seeded by injecting cells in multiple phases as discussed herein in the context of FIG. 25.
  • In step S506, the recellularized bioscaffold is perfused with an organ culture perfusate to promote engrafting of the cells to the bioscaffold and organ function. The organ culture perfusate can include the same or similar components of the perfusate for enhancing cell vitality as described herein. The recellularized bioscaffold can be perfused for hours, days, weeks, months, or indefinitely. For example, the recellularized bioscaffold can be perfused for about 4 hours, about 8 hours, about 12 hours, about 16 hours, about 20 hours, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, about two weeks, about three weeks, and about one month.
  • In step S508, the recellularized bioscaffold is preserved. The recellularized bioscaffold can be preserved in a manner similar to the preservation of a decellularized organ as discussed herein, namely washing with one or more isotonic wash mediums, continuous perfusion, drying, storage in a medium including one or more antimicrobial agents, storage at less than about 4° C., about −20° C., about −40° C., and about −180° C., and/or subzero nonfreezing storage.
  • In step S510, recellularized bioscaffold can be transplanted into a subject by a health care provider in accordance with known surgical techniques. Alternatively, in step S512, the recellularized bioscaffold can be used an in vitro model (e.g., an in vitro pharmacologic model) in accordance with known laboratory techniques such as those described in U.S. Patent Application Publication No. 2006/0019326.
  • Methods of Organ Rehabilitation
  • Referring to FIG. 6, a method 600 of organ rehabilitation is provided. Organ rehabilitation can be particularly advantageous because, in some embodiments, a rehabilitated organ can be fabricated solely from a subject's own organ, thereby minimizing the risk of transplant rejection. Accordingly, in some embodiments the organ can be harvested from the subject, rehabilitated according to the method below, and transplanted to the same subject. During the interim period when the rehabilitation method is performed, the subject can be sustained with one or more external support systems (e.g. dialysis machines) and/or internal bioartificial organ devices (e.g. bioartificial liver devices) such as those described in U.S. Patent Application Publication No. 2007/0148139.
  • In step S602, the organ is perfused to improve the vitality of adult cells as described herein.
  • In step S604, the organ is divided into first portion and a second portion. Division of the organ into the first and section portion can be accomplished using existing surgical techniques, including, but not limited to living donor transplantation techniques. For example, in living donor liver transplantation (LDLT) techniques, either the right lobe or the left lobe of the liver is resected from the donor for transplantation to the recipient. LDLT techniques are described in publications such as S. A. Shah et al., “Adult-to-adult living donor liver transplantation,” 20(5) Can. J. Gastroenterol. 339-43 (2006); and Chi Leung Liu et al., “Operative Outcomes of Adult-to-Adult Right Lobe Live Donor Liver Transplantation: A Comparative Study With Cadaveric Whole-Graft Liver Transplantation in a Single Center,” 243(3) Annals of Surgery 404-10 (2006).
  • The first portion and the second portion are processed independently. In steps S606 and S608, the first portion is perfused with a decellularization medium and preserved, as described herein. In step S610 and S612, the adult cells are cultured as described herein.
  • In steps S614 and S616, the first portion is recellularized with a suspension of adult cells as described herein and perfused with an organ culture perfusate as described herein.
  • In step S618, the first portion is preserved. The first portion can be preserved in a manner similar to the preservation of a decellularized organ as discussed herein, namely washing with one or more isotonic wash mediums, continuous perfusion, drying, storage in a medium including one or more antimicrobial agents, storage at less than about 4° C., about −20° C., about −40° C., and about −180° C., and/or subzero nonfreezing storage.
  • In step S620, the first portion can be transplanted into a subject by a health care provider in accordance with known surgical techniques.
  • In some embodiments, the first portion and the second portion are substantially equal in size (e.g. mass and/or volume). In another embodiment, the first portion is larger than the first portion to provide for a larger vascular structure to support an increased number of cells. In still another embodiment, the first and the second portion are selected based on the relative health of the vascular structure and the target cells in the various regions of the organ. For example, if “region A” of an organ had relatively intact vasculature and “region B” of the organ had relatively healthy target cells, region A could be selected decellularization and region B could be selected for cell isolation.
  • Use of Recellularized Organs for Research
  • Recellularized organs can be used in a variety of research applications. For example, recellularized organs (e.g., recellularized livers) can be used for drug discovery (e.g., ADMET testing). Entire recellularized organs or portions thereof can be used, depending on the needs of a particular application.
  • Referring again to step S512 of FIG. 5 and S622 of FIG. 6, a recellularized bioscaffold or recellularized first portion of an organ (collectively referred to as a “recellularized bioscaffold”) can be used as an in vitro model. The entire recellularized bioscaffold can be used as an in vitro model. Alternatively, the recellularized bioscaffold can be sliced accordingly to existing laboratory techniques and plated for use as an in vitro model.
  • Referring now to FIG. 7A, another method 700A of creating a research tool is provided.
  • In step S702, a decellularized organ is sliced according to existing laboratory techniques. The decellularized organ can be produced according to the methods described herein or other methods. Additionally or alternatively, one or more mechanical methods such as agitation and/or sonication can be used to decellularize the organ.
  • In step S704, the decellularized organ slice is plated in a vessel such as a Petri dish. The Petri dish can, in some embodiments, contain an agar to support the organ slice and cells added in step S706.
  • In step S706, the decellularized organ slice is seeded with cells. The cells can be seeded by placing the cells on top of the decellularized organ slice (e.g., by pouring or transferring a recellularization medium containing the cells with a pipette or other device). In step S708, the recellularized organ slices are cultured, for example, with a culture medium described herein.
  • In step S710, the recellularized organ slices are stored according to existing laboratory protocols. In step S712, the recellularized organ slices are used in an experiment (e.g., a pharmacologic experiment). For example, one or more test agents can be applied to the recellularized organ slice. The one or more test agents and the recellularized organ slice can then be incubated to form an enzyme-substrate complex between the one or more test agents and one or more enzymes contained in the recellularized organ slice. One or more metabolites of the test agent can be then be detected with various laboratory techniques (e.g., mass spectrometry, fluorescence screening, and the like).
  • Referring now to FIG. 7B, another method 700B of creating a research tool is provided.
  • In step S714, an organ is sliced according to existing laboratory techniques.
  • In step S716, the organ slices are placed in a vessel such as a Petri dish. The Petri dish can, in some embodiments, contain an agar to support the organ slice and cells added in step S720.
  • In step S718, the organ slice is decellularized, for example, by exposing the organ slice to a decellularization medium as discussed herein. Additionally or alternatively, one or more mechanical methods such as agitation and/or sonication can be used to decellularize the organ slice.
  • As depicted in FIG. 7B, steps S716 and S718 can be performed in any order (i.e., the organ can be plated before decellularization or can be decellularized after plating).
  • In step S720, the decellularized organ slice is seeded with cells. The cells can be seeded by placing the cells on top of the decellularized organ slice (e.g., by pouring or transferring a recellularization medium containing the cells with a pipette or other device). In step S722, the recellularized organ slices are cultured, for example, with a culture medium described herein.
  • In step S724, the recellularized organ slices are stored according to existing laboratory protocols. In step S726, the recellularized organ slices are used in an experiment (e.g., a pharmacologic experiment). For example, one or more test agents can be applied to the recellularized organ slice. The one or more test agents and the recellularized organ slice can then be incubated to form an enzyme-substrate complex between the one or more test agents and one or more enzymes contained in the recellularized organ slice. One or more metabolites of the test agent can be then be detected with various laboratory techniques (e.g., mass spectrometry, fluorescence screening, and the like).
  • Test agents can include, but are not limited to, opioid analgesics, anti-inflammatory drugs such as antihistamines and non-steroidal anti-inflammatory drugs (NSAIDs), diuretics such as carbonic anhydrase inhibitors, loop diuretics, high-ceiling diuretics, thiazide and thiazide-like agents, and potassium-sparing diuretics, agents that impinge on the renal and cardiovascular systems such as angiotensin-converting enzyme inhibitors, cardiac drugs such as organic nitrates, calcium channel blockers, sympatholytic agents, vasodilators, β-adrenergic receptor agonists and antagonists, α-adrenergic receptor agonists and antagonists, cardiac glycosides, anti-arrhythmic drugs, agents that affect hyperlipoproteinemias such as 3-hydroxymethylglutaryl-coenzyme A (HMG-CoA) inhibitors, anti-neoplastic agents such as alkylating agents, antimetabolites, natural products, antibiotics, and other drugs, immunomodulators, anti-diabetic agents, and anti-microbial agents such as antibacterial agents, antiviral agents, antifungal agents, antiprotozoal agents, and antihelminthic agents.
  • Enzymes can include, but are not limited to, cytochrome P450, alkaline phosphatase, α-galactosidase, β-galactosidase, α-glucosidase, β-glucosidase, α-glucuronidase, β-glucuronidase, α-amylase, NADPH-cytochrome P450 reductase, cytochrome b5, N-demethylase, O-demethylase, acetylcholinesterase, pseudocholinesterase, epoxide hydrolase, amidases, uridine diphosphate (UDP)-glucuronosyltransferases, phenol sulfotransferase, alcohol sulfotransferase, sterid sulfotransferase, and arylamine sulfotransferase, UDP-glycosyltransferases, purine phosphoribosyltransferase, N-acetyltransferases, glutathione S-transferase, phenylethanolamine N-methyltransferase, non-specific N-methyltransferase, imidazole N-methyltransferase, catechol-O-methyltransferase, hydroxyindole-O-methyltransferase, S-methyltransferase, alcohol dehydrogenase, aldehyde dehydrogenase, xanthine oxidase, monoamine oxidases, diamine oxidases, flavoprotein N-oxidases, hydroxylases, aromatases, cysteine conjugate β-lyase, and alkylhydrazine oxidase. The enzyme can be endogenously expressed in the recellularized organ slice, and can have either normal enzymatic activity or altered enzymatic activity, for example, such as where the enzyme contains a polymorphism or mutation.
  • Working Example #1 Use of Normothermic Perfusion to Restore Organ Vitality
  • As a model of Donors after Cardiac Death (DCD), lean livers were harvested from rats (inbred Lewis strain) and held at 34° C. for 60 min. This temperature is slightly lower than physiological, which is justified by the fact that core body temperature tends to decrease after death. After one hour of ischemia, the livers were perfused for five hours in the organ perfusion system depicted in FIG. 1 using the methods described above. Perfusate samples (1 ml) were collected from the inlet of the liver and analyzed using a PICCOLO® miniature blood chemistry analyzer available from Abaxis, Inc. of Union City, Calif.
  • FIGS. 8A-8E depict functional parameters of harvested livers during normothermic perfusion. Alanine transaminase (ALT) and aspartate transaminase (AST) activities are depicted in FIGS. 8A and 8B as indicators of hepatocellular damage. Both AST and ALT accumulated during the first 180 minutes of perfusion and then decreased. These values were severalfold higher than those for the control group of freshly isolated livers not subjected to any warm ischemia. Neither ALT nor AST were detected in the dialysate (data not shown).
  • Bile secretion and oxygen consumption describe the metabolic state of the liver. Bile was produced at a constant rate throughout the perfusion as depicted in FIG. 8C. This rate was 40% lower compared to freshly isolated livers. The Oxygen Uptake Rate (OUR) of warm ischemic livers declined rapidly during the first 60 minutes of the perfusion and then remained stable as depicted in FIG. 8D. This behavior was very similar to the control group. The OURs of the perfused warm ischemic and freshly isolated livers were very similar in the plateau region beyond 60 minutes. The urea level in the perfusate showed a steady increase from 4.20 mg/dL at t=0 to 8.60 mg/dL at t=300 minutes, indicating a constant rate of urea production. This rate was consistently higher than that observed in perfused healthy livers as depicted in FIG. 8E. The bile and oxygen uptake values were significantly lower than the control group and the urea levels were significantly higher than the control group (p<0.01 by ANOVA). Asterisks (*) in FIGS. 8A-4E indicate a statistical difference compared to healthy perfused livers asp <0.1
  • Warm ischemic livers were transplanted into recipient rats after five hours of normothermic perfusion (n=13) or five hours of Static Cold Storage (SCS) in University of Wisconsin solution at 0° C. (n=6). In addition, freshly isolated livers not subjected to any warm ischemia were transplanted after six hours of SCS (n=6) or normothermic perfusion (n=11) and ischemic livers were transplanted directly without having undergone preservation (n=9).
  • Transplantation of perfusion-treated ischemic livers was uneventful in all but one case, where bleeding at the anastomosis occurred. All animals recovered from anesthesia rapidly. The animal that bled during surgery died on day four postoperatively. The other recipient animals survived beyond one month and did not exhibit external signs of liver failure, such as jaundice.
  • No surgical complications occurred during transplantation of ischemic livers preserved by SCS and recipients recovered rapidly from anesthesia, but within six hours, all developed symptoms of primary nonfunction and died within 12 hours. Autopsy revealed patchy livers and serous fluid in the abdomen. All recipients of directly transplanted ischemic livers died in a similar way within 24 hours post-operatively. All controls that received freshly isolated livers preserved for six hours by SCS recovered rapidly from surgery and survived beyond one month. Survival rates are depicted in FIG. 9.
  • Because primary nonfunction indicates disruption of liver microcirculation and death of parenchymal and nonparenchymal cells, in this context recipient death due to primary nonfunction is an indicator of poor conditions for hepatocyte isolation compared to livers that demonstrate no such symptoms. Accordingly, these results provide strong evidence that normothermic perfusion enhances viability of damaged livers for hepatocyte isolation.
  • Additionally, as depicted in FIG. 10, the use of an organ perfusion system 100 with a secondary dialysis circuit 120 yields significantly better results than perfusion without a secondary dialysis circuit 120. (Survival rates are shown at four weeks post-operation; n>6 for all groups).
  • Working Example #2 Isolation of Hepatocytes from Livers after Normothermic Perfusion
  • Lean livers were harvested from heparinized Lewis rats and held at 34° C. for 60 minutes. After one hour of ischemia, one group of livers was perfused in the organ perfusion system depicted in FIG. 1, and cell isolation was performed with livers in the other group. Hepatocytes were isolated using a two-step collagenase perfusion procedure as described previously James C. Y. Dunn et al., “Long-Term in Vitro Function of Adult Hepatocytes in a Collagen Sandwich Configuration,” 7(3) Biotechnology Progress 237-45 (1991).
  • For short-term function evaluation, recovered hepatocytes were cultured in suspensions (1×106 cells/mL) for six hours with continuous gentle mixing in an incubator in 90% air/10% CO2 at 37° C.
  • For long-term function evaluation, recovered hepatocytes were cultured in the double gel sandwich configuration to maintain stable liver specific function. Briefly, tissue culture dishes were coated with a mixed solution of nine parts type 1 rat tail collagen (1.1 mg/mL) and one part 10× Dulbecco's Modified Eagle's Medium (DMEM) and incubated for one hour at 37° C. to form a collagen gel. After gelation, 125,000 cells per well were seeded in nine-well culture dishes, and incubated in 90% air/10% CO2 at 37° C. The following day, after aspirating the culture medium from the dishes, the second collagen gel layer was overlaid on the hepatocytes and incubated for one hour at 37° C. After gelation, 1 mL of hepatocyte culture medium was applied, and then changed daily.
  • Standard hepatocyte culture medium used both in suspension and collagen gel cultures includes DMEM with 10% fetal bovine serum, 7 ng/mL glucagon, 7.5 μg/mL hydrocortisone, 0.5 U/mL insulin, 20 ng/mL epidermal growth factor, 200 U/mL penicillin, and 200 μg/mL streptomycin. Group comparisons were performed via a t-test.
  • As depicted in FIG. 11A, the cell yield is increased from 15 million to 100 million. As depicted in FIG. 11B, the viability of these recovered cells are well in the acceptable range (>85%), although slightly lower than obtained with non-ischemic livers. A single asterisk (*) denotes a statistically significant difference as compared to normal livers. A double asterisk (**) denotes a statistically significant difference as compared to ischemic, non-perfused livers (p<0.05). These results demonstrate that normothermic perfusion increases the number of cells isolated from ischemic livers by approximately sixfold.
  • Hepatocytes obtained from perfusion-treated livers were compared to cells obtained from fresh rat livers. FIG. 12A depicts phase contrast microscopy of cells obtained from one-hour ischemic livers after perfusion in a collagen double gel at two weeks post-seeding. Compared to cells obtained from normal (fresh) livers depicted in FIG. 12B, there were no remarkable differences. By comparison, cells obtained from untreated ischemic livers depicted in FIG. 12C did not reach confluency, appeared steatotic and unhealthy, and dedifferentiation was observed. As depicted in FIGS. 12D and 12E, urea synthesis by the hepatocytes from perfusion-treated ischemic cells when cultured in suspension for six hours was similar to those obtained from fresh livers, whereas albumin synthesis was approximately 50% of normal hepatocytes for the duration of the experiment (statistically different at p<0.05).
  • As depicted in FIGS. 12F and 12G, urea and albumin synthesis by the hepatocytes from perfusion-treated ischemic cells when culture in collagen sandwich culture for 14 days was similar to that obtained from fresh livers. There was a slight drop in albumin, but the difference did not reach statistical significance for any day.
  • These results demonstrate that with the organ perfusion system 100 described herein, the recovery of hepatocytes can be increased significantly and that the recovered hepatocytes are capable of performing essential hepatic functions at a level lower, but still comparable to cells obtained from healthy livers.
  • Working Example #3 Decellularization of Ischemic Livers
  • As depicted in FIG. 13A-13E, ischemic rat livers (one hour of warm ischemia at 34° C. in a saline solution) can be decellularized in intact form with the vasculature generally well-preserved. Livers were perfused through the portal vein with 0.1% sodium dodecyl sulfate (SDS) for four hours. FIGS. 13A-13E are representative photographs of livers during the decellularization process. FIG. 13A depicts a liver at t=0 minutes. FIG. 13A depicts a liver at t=25 minutes. FIG. 13C depicts a liver at t=1 hour. FIG. 13D depicts a liver at t=2 hours. FIG. 13E depicts a liver at t=4 hours. This procedure yielded a fully decellularized construct suitable for use as a bioscaffold for recellularization.
  • Working Example #4 Recellularization of Decellularized Livers with Primary Rat Hepatocytes
  • After storage in an antibiotic solution at 4° C., the decellularized matrices were washed thoroughly and brought to room temperature by perfusion with phosphate buffered saline (“PBS”). After four hours of PBS perfusion, the portal vein of each decellularized liver was cannulated with a sterile 18-gauge cannulae and sutured. The livers were then mounted in the perfusion system and infused with 100×106 isolated rat hepatocytes for over one hour. FIGS. 14A-14D depict representative livers at 0, 10, 20, 30, and 40 minutes post-seeding, respectively.
  • The recellularized liver native matrices were then cultured for 8 hours under continuous perfusion with oxygenated culture medium. A time period of eight hours was chosen as the initial period of testing because direct cell death due to perfusion damage or poor oxygenation typically kills most cells within two to four hours post-cell-isolation. Thus, eight hours would provide a good indication of the damage induced by the repopulation process. The infused hepatocytes were counted to evaluate cell engraftment immediately after cell seeding and it was found that around 70% of the seeded cells remained in the livers and engrafted around the vessels with most hepatocytes in the parenchyma.
  • To assess viability, recellularized livers were stained for glycogen and the majority of the cells were positive for PAS (Periodic Acid Schiff) staining demonstrating that infused hepatocytes were viable and active in the short term following recellularization. FIG. 15A depicts a hematoxylin and eosin (H&E) stained decellularized liver matrix. FIG. 15B depicts an H&E-stained recellularized liver matrix. FIG. 15C depicts a PAS-stained recellularized liver matrix. It was also notable that the vascular structure seemed to be preserved.
  • To further assess the success of reseeding, apoptosis in the seeded cells was evaluated via TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) staining after eight hours of perfusion. FIG. 16A depicts a negative control of TUNEL staining without antibody. In FIG. 16B, apoptotic cells stained in green. FIG. 16C depicts positive control of TUNEL staining of a DNAse exposed liver sample. DAPI (4′,6-diamidino-2-phenylindole) staining is depicted in the lower left corner of FIGS. 16A-16C. Less than 20% cells were positive for apoptosis. Stable oxygen uptake rates were also observed during this period.
  • To assess viability after repopulation, recellularized livers were perfusion-cultured for seven days. As depicted in FIGS. 17A and 17B, there was steady production of albumin (measured by ELISA) and urea (measured with a BUN assay kit) proving hepatic functionality in the recellularized liver. As depicted in FIG. 17B, extracellular lactate dehydrogenase levels (measured with an LDH kit) during first 80 hours of perfusion are identical to cell culture levels, indicating no significant damage to the cells in the system. In FIGS. 17A-17C, data for liver grafts repopulated with primary rat hepatocytes are represented with closed circles, while data for collagen sandwiched cultured primary rat hepatocytes are represented with open circles.
  • In addition, hepatocytes demonstrated the ability to home to and engraft in the liver matrix and around the vessels as depicted by FIGS. 18A-18F, which are photographs of H&E-stained liver matrices. FIGS. 18A and 18D depict an H&E-stained normal liver matrix at 20× and 40× power, respectively. FIGS. 18B and 18E depict an H&E-stained repopulated liver matrix at 20× and 40× power, respectively, after 24 hours of perfusion culturing. FIGS. 18C and 18F depict an H&E-stained repopulated liver matrix at 20× and 40× power, respectively, after 48 hours of perfusion culturing. The cells were well distributed and healthy after 48 hours, with polygonal shaped cells containing multiple nuclei present. These results demonstrate that hepatocytes can be seeded in the recellularized grafts and the hepatocyte function can be maintained up to a week.
  • Working Example #5 In Vivo Transplantation of Decellularized Liver Graft
  • As depicted in FIGS. 19A and 19B, the decellularized grafts were transplanted to test whether the liver extracellular matrix (ECM) was capable of handling the pressure and viscosity of blood flow. The decellularized liver was subjected to resection of the right superior lobe, right inferior lobe, left lateral lobe, and the inferior and superior caudate lobes with piercing sutures to avoid damage to the stump and paracaval circulation circuit. The hepatic artery, superior vena cava (SVC), and bile duct were closed. After a left nephrectomy, the decellularized liver graft, which was reduced to about 30% of the original size, was implanted into the left upper quadrant of the recipient's abdomen. The inferior vena cava (IVC) was anastomosed by a stent technique. The donor's portal vein was completely arterialized to the recipient's left renal artery in a stent technique. Using a stent with an internal diameter of 0.3 mm, the flow in the arterialized portal vein was regulated to achieve physiologic parameters.
  • Overall, the decellularized graft was well perfused, able to sustain the flow, and no occlusion of blood circulation was observed in the bioscaffold.
  • Working Example #6 Characterization of Decellularized Liver Matrix (DLM)
  • Whole organ decellularization was achieved by portal perfusion with sodium dodecyl sulfate (SDS), an anionic detergent that lyses cells and solubilizes cytoplasmic components. Following 72 hours of decellularization, a translucent acellular scaffold, which retained the gross shape of liver, was generated as depicted in FIGS. 20A-20E. Histological evaluation revealed no nuclei or cytoplasmic staining in the DLM compared to normal rat liver as depicted in the leftmost segments of FIG. 21. Immunostaining for extracellular matrix (ECM) proteins, collagen type I, collagen type IV, fibronectin and laminin-β1, indicated that both structural and basement membrane components of the ECM were retained similarly to native liver as depicted in the remaining panes of FIG. 21. The lack of DAPI staining in DLM confirms the absence of cells. Both collagen type IV and fibronectin were observed in decellularized sinusoidal spaces, while laminin-β1 is seen in the basement membrane of the larger vessels. 100% of the fibrillar collagen and approximately 50% of the glycosaminoglycans of native liver were retained following decellularization. Residual DNA content in the DLM was less than 3%.
  • To demonstrate that decellularization protocol was successful in retaining a functional vascular bed, its perfusion was characterized using Allura Red dye. Structural components of the vascular tree were clearly apparent in the translucent matrix as depicted in FIGS. 22A and 22B. The dye injected through the portal vein flowed as would be expected inside the vascular network, gradually moving from larger vessels to smaller capillaries, suggesting the microvasculature remained intact.
  • The intricate vascular tree was further characterized by generating a corrosion cast of the DLM as depicted in FIGS. 23A and B. Portal (red) and venous (blue) vasculature casting of both normal (FIG. 23A) and decellularized (FIG. 23B) livers showed that the larger portal and venous circulation system of vessels and the vast majority of the smaller microcirculatory branches were preserved, indicating that physiologic flow could be achieved by traversing the portal venous system and emptying into the systemic venous circulation via the hepatic vein and inferior vena cava (IVC).
  • Moreover, scanning electron microscopy (SEM) images confirmed the presence of large vessels within the DLM as depicted in FIG. 24A. Remnants of small vessels closely resembling a portal triad were also observed as depicted in FIG. 24B. The honeycomb of about 30-μm diameter gaps in the ECM depicted in FIG. 24C is thought to be the footprint of hepatocytes removed during decellularization.
  • Working Example #7 Recellularization of a Decellularized Liver Matrix
  • Presence of a functional vascular bed in the DLM offers the ability to control hepatocyte engraftment and characterize liver-specific metabolic function in vitro prior to transplantation of the recellularized graft. Primary rat hepatocytes were introduced via portal vein perfusion recirculation.
  • As depicted in FIG. 25, roughly 12.5 million cells were introduced at each step, for a total of four steps, with 10 minute intervals between each step. Cell viability and distribution in the parenchyma using this four-step seeding protocol was superior to single step infusion of 50 million cells for 40 min. Engraftment efficiency using the four-step protocol was 95.6±3.4%.
  • Following seeding, the recellularized liver grafts were transferred into a specially-designed perfusion chamber as depicted in FIG. 2B for in vitro culture. The perfusion chamber features two hermetically-sealed silicon sheets, forming a pouch filled with culture medium. This design avoids rigid surfaces, thereby preventing development of pressure spots, while enabling sterile culture of the recellularized grafts up to two weeks in vitro. Histological staining of recellularized sections at 4 hours and at 1 day, 2 days and 5 days revealed that at 4 hours, a majority of the cells remain in and around the vessels, whereas at 1 day and 2 days, the cells leave the vessels and are distributed throughout the matrix. No changes in the distribution of the cells were observed within the matrix at 5 days, as compared to previous days
  • The recellularized liver graft was continuously perfused for 5 days. Hepatocyte viability was maintained during culture. As depicted in FIG. 26, quantification of TUNEL-positive cells (depicted in FIG. 27) revealed that less than 20% of the cells were apoptotic on the first 2 days of culture. LDH release during perfusion was comparable and statistically not different between groups indicating minimal cell death (p=0.0455) as depicted in FIG. 28. SEM and histological analysis depicted in FIG. 29 demonstrated that hepatocytes engrafted around the larger vessels, populating the surrounding parenchyma and suggesting that the cells migrated beyond the matrix barrier to reach decellularized sinusoidal spaces. Functional characteristics of engrafted hepatocytes in the decellularized matrix were assessed via immunostaining at 2 days of culture using Ugt1a (a sensitive enzyme with a short half-life (about 50 min) whose presence demonstrates hepatocyte viability and function), G6 pc, and albumin as depicted in FIG. 30. The level of immunostaining for these markers in engrafted hepatocytes was similar to normal livers.
  • To assess the metabolic activity of engrafted hepatocytes, hepatocyte albumin production and urea synthesis was quantified as depicted in FIGS. 31A and 31B. The cumulative urea levels in the recellularized liver system were statistically higher than in hepatocyte sandwich culture during the 5 day culture period (p=0.0017). The difference in cumulative albumin levels was statistically not significant (p=0.0176). Albumin production rate by the recellularized liver graft was 27.6±7.0 μg million per cells per day as compared to 140 μg million per cells per day by the normal adult rat liver as discussed in R. Hoffenberg, “Measurement of the synthesis of liver-produced plasma proteins with particular reference to dietary protein and amino acid supply,” 123 Biochem. J. 3P (1972).
  • As depicted in FIGS. 32A and 32B, analysis of the expression of drug metabolism enzymes via quantitative Real Time Polymerase Chain Reaction (RT-PCR) at 2 days revealed that expression levels of phase I and phase II drug metabolism enzymes in the recellularized liver were similar to those measured in sandwich hepatocyte cultures (p=0.0499). As expected at this stage in culture and depicted in FIG. 32A, gene transcription levels were overall much lower than those of freshly isolated hepatocytes. However, Cyp2c11 (encoding cytochrome P450, subfamily 2, polypeptide 11) Gstm2 (glutathione S-transferase mu 2), Ugt1a1 (encoding UDP glucuronosyltransferase-1 family, polypeptide A1) and Cyplal (encoding cytochrome P450, family-1, subfamily a, polypeptide 1) were expressed in the recellularized liver at similar levels to those in normal liver as depicted in FIGS. 33A-33D. Adh1 (encoding alcohol dehydrogenase-1) and Cyp3a18 (encoding cytochrome P450, family 3, subfamily a, polypeptide 18) expression levels were higher in recellularized liver than in the sandwich plate culture control, although they were much lower than in normal liver as depicted in FIGS. 33E and 33F. Primary hepatocytes are known to require between 7 to 10 days in culture before their metabolic activity and gene transcription levels stabilize as discussed in S. R. Khetani & S, N. Bhatia, “Microscale culture of human liver cells for drug development,” 26 Nature Biochemistry 120-26 (2008).
  • Working Example #8 Seeding of Non-Parenchymal Cells
  • Seeding of a non-parenchymal component to the recellularized liver graft was demonstrated by incorporating microvascular endothelial cells (ECs) to the hepatocyte-repopulated graft, and testing by perfusion-culture for up to 5 days to allow for EC engraftment. Histological analysis showed that endothelial cells were capable of lining the vasculature encircled by hepatocytes at 3 days of culture. Hepatocytes remained viable as indicated by TUNEL staining and immunohistochemistry.
  • In order to test the scalability of the seeding approach as well as the limits of the perfusion-culture system, the median lobe of DLM was repopulated with 200 million hepatocytes and perfused with a culture medium for 10 days. This cellular amount corresponds to approximately 20% of rat's liver mass and is more than double the minimum hepatic mass necessary for therapeutic interventions. In these studies, 50 million hepatocytes were injected at each of the four steps mentioned above in the context of FIG. 25. The metabolic activity of the recellularized liver graft was assessed through albumin production, urea secretion and total bile acid synthesis and the cumulative levels of production of these metabolites were found to be at similar levels to static sandwich culture controls (p=0.5249, 0.5271, and 0.0114, respectively) indicating the scalability of methods described herein as well as the usability of the perfusion-culture system described herein as an in vitro model.
  • Working Example #9 Transplantation of Recellularized Liver Grafts
  • The functional vascular structure demonstrated herein enables transplantation of the recellularized liver graft by connecting the graft to the blood supply, thereby allowing for the transplantation of a critical hepatocyte mass while avoiding ischemic damage due to poor graft perfusion.
  • Briefly, recipient animals underwent unilateral nephrectomy to prepare a viable site for auxiliary liver graft transplantation. The renal vein and artery were used to as ports to create blood flow within the graft. Upon unclamping of the artery, the graft was perfused, quickly filling with blood, and appropriate efflux was established within 5 minutes as depicted in FIG. 34. The recellularized graft was kept in vivo for 8 hours prior to harvesting for further analysis. TUNEL staining revealed that there was minimal damage to the hepatocytes due to the arterial blood flow and consequent shear stress during 8 h of transplantation. 19.7±13.7% of cells were TUNEL-positive after transplantation as compared 21.4±5.8% of cells before transplantation (p=0.55). Histological staining indicated that the hepatocytes retained both normal morphology and their parenchymal positions as depicted in the leftmost panes of FIG. 35. Immunohistochemical staining for albumin, G6 pc, and Ugt1a confirmed that hepatic function was also retained in the transplanted grafts with minimal indications of ischemic damage as depicted in FIG. 35.
  • To investigate early graft function beyond 8 hours, an ex vivo whole blood perfusion technique that has been shown to be representative of early graft performance and predict primary graft failure in an orthotopic rat liver transplant model in H. Tolbloom, et al., “Recovery of Warm Ischemic Rat Liver Grafts by Normothermic Extracorporeal Perfusion,” 87 Transplantation 170-77 (2009) was adopted as a surrogate model of transplantation. Ex vivo perfusion system was similar to the in vitro perfusion described herein except that the perfusate consisted of whole rat blood diluted with perfusion medium (hematocrit 20%). The recellularized liver graft was perfused for 24 hours during which the blood was replenished three times at eight-hour intervals to maintain the hematocrit level at 20%. TUNEL staining and histological analysis showed that hepatocytes remained viable (22.6±13.8% TUNEL positive cells) and preserved their morphology and parenchymal position at the end of ex vivo blood perfusion. In addition, hepatocytes remained metabolically active during the 24 hours of ex vivo blood perfusion as evidenced by urea and albumin secretion.
  • EQUIVALENTS
  • The functions of several elements may, in alternative embodiments, be carried out by fewer elements, or a single element. Similarly, in some embodiments, any functional element may perform fewer, or different, operations than those described with respect to the illustrated embodiment. Also, functional elements (e.g., modules, computers, and the like) shown as distinct for purposes of illustration may be incorporated within other functional elements, separated in different hardware or distributed in a particular implementation.
  • While certain embodiments according to the invention have been described, the invention is not limited to just the described embodiments. Various changes and/or modifications can be made to any of the described embodiments without departing from the spirit or scope of the invention. Also, various combinations of elements, steps, features, and/or aspects of the described embodiments are possible and contemplated even if such combinations are not expressly identified herein.
  • INCORPORATION BY REFERENCE
  • The entire contents of all patents, published patent applications, and other references cited herein are hereby expressly incorporated herein in their entireties by reference.

Claims (25)

1. A method for harvesting adult cells from an organ, the method comprising:
perfusing the organ with a perfusate; and
isolating adult cells from the organ;
thereby harvesting the adult cells from the organ.
2. The method of claim 1, wherein the organ is a mammalian organ.
3. The method claim 2, wherein the organ is a human organ.
4. The method of claim 1, wherein the organ is a liver.
5. The method of claim 4, wherein the adult cells are selected from the group consisting of: hepatocytes, endothelial cells, cholangiocytes (bile duct cells), Kupffer cells, stellate cells, and smooth muscle cells.
6. The method of claim 1, wherein the organ is one selected from the group consisting of: pancreas, spleen, heart, lung, and kidney.
7. The method of claim 1, wherein the organ is ischemic.
8. The method of claim 1, wherein the perfusate includes one or more oxygen carriers.
9. The method of claim 8, wherein the one or more oxygen carriers include erythrocytes.
10. The method of claim 1, wherein the perfusate includes one or more antiinflammatory agents.
11. The method claim 10, wherein the one or anti-inflammatory agents includes hydrocortisone.
12. The method of claim 1, wherein the perfusate includes collagenase.
13. The method of claim 1, wherein the perfusate is a normothermic perfusate.
14. The method of claim 1, wherein the perfusate is a room temperature perfusate.
15. The method of claim 1, wherein the perfusate is a hypothermic perfusate.
16. The method of claim 1, further comprising culturing the harvested adult cells.
17. The method of claim 1, further comprising providing the organ.
18. The method of claim 1, wherein the step of isolating the adult cells from the organ includes filtering the perfusate.
19. The method of claim 18, wherein the filtering the perfusate step including filtering the perfusate through one or more mesh filters.
20. The method of claim 19, wherein the one or more mesh filters have pore sizes ranging from about 1 μm to about 1000 μm.
21. The method of claim 1, wherein the step of isolating the adult cells from the organ includes performing density centrifugation on the perfusate.
22. The method of claim 1, further comprising circulating the perfusate through a dialyzer.
23. The method of claim 1, wherein the organ is a damaged organ.
24. The method of claim 1, wherein the perfusion step improves the vitality of the adult cells.
25-171. (canceled)
US13/392,661 2009-07-01 2010-06-30 Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same Abandoned US20130323708A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/392,661 US20130323708A1 (en) 2009-07-01 2010-06-30 Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US22226609P 2009-07-01 2009-07-01
US30486010P 2010-02-16 2010-02-16
US33095910P 2010-05-04 2010-05-04
US13/392,661 US20130323708A1 (en) 2009-07-01 2010-06-30 Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same
PCT/US2010/040663 WO2011002926A2 (en) 2009-07-01 2010-06-30 Isolated adult cells, artificial organs,rehabilitated organs, research rools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/040663 A-371-Of-International WO2011002926A2 (en) 2009-07-01 2010-06-30 Isolated adult cells, artificial organs,rehabilitated organs, research rools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/645,527 Continuation US11136553B2 (en) 2009-07-01 2015-04-07 Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same

Publications (1)

Publication Number Publication Date
US20130323708A1 true US20130323708A1 (en) 2013-12-05

Family

ID=43411735

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/392,661 Abandoned US20130323708A1 (en) 2009-07-01 2010-06-30 Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same
US14/645,527 Active US11136553B2 (en) 2009-07-01 2015-04-07 Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same
US17/449,762 Pending US20220098548A1 (en) 2009-07-01 2021-10-01 Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/645,527 Active US11136553B2 (en) 2009-07-01 2015-04-07 Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same
US17/449,762 Pending US20220098548A1 (en) 2009-07-01 2021-10-01 Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same

Country Status (2)

Country Link
US (3) US20130323708A1 (en)
WO (1) WO2011002926A2 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120064537A1 (en) * 2010-06-30 2012-03-15 Miromatrix Medical Inc. Use of perfusion decellularized organs for matched recellularization
WO2017070007A3 (en) * 2015-10-15 2017-06-15 Wake Forest University Health Sciences Methods of producing in vitro liver constructs and uses thereof
US9986734B2 (en) 2013-01-08 2018-06-05 Yale University Human and large-mammal lung bioreactor
US20200080050A1 (en) * 2016-07-11 2020-03-12 Yaakov Nahmias Systems and methods for growing cells in vitro
US10660329B2 (en) 2015-04-23 2020-05-26 Etablissement Francais Du Sang Method for preserving cells, tissues or organs in hypothermia
US20200352157A1 (en) * 2014-09-02 2020-11-12 United Therapeutics Corporation Automated bioreactor system, system for automatically implementing protocol for decellularizing organ, and waste decontamination system
CN112980683A (en) * 2021-03-12 2021-06-18 立沃生物科技(深圳)有限公司 In-vitro liver and kidney perfusion system and hepatocyte regeneration method
JP2023052212A (en) * 2018-08-03 2023-04-11 ベリグラフト アクティエボラーグ Methods of preparing personalized blood vessels
CN117264876A (en) * 2023-11-21 2023-12-22 妙顺(上海)生物科技有限公司 Method for preparing multi-donor liver cells
EP4069821A4 (en) * 2019-12-02 2024-04-03 Avant Meats Company Ltd System for producing cultivated meats, tissues and associated products from cells

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9078428B2 (en) 2005-06-28 2015-07-14 Transmedics, Inc. Systems, methods, compositions and solutions for perfusing an organ
US9457179B2 (en) 2007-03-20 2016-10-04 Transmedics, Inc. Systems for monitoring and applying electrical currents in an organ perfusion system
ES2620778T3 (en) 2009-02-04 2017-06-29 Yale University Lung tissue engineering
US10575515B2 (en) 2010-05-04 2020-03-03 The General Hospital Corporation Methods and compositions for preserving tissues and organs
CA3106469A1 (en) 2011-04-14 2012-10-18 Transmedics, Inc. Organ care solution for ex-vivo machine perfusion of donor lungs
WO2013114372A1 (en) 2012-02-01 2013-08-08 Nayacure Therapeutics Ltd. Method for inducing immune tolerance to organ transplants
EP2633755A1 (en) * 2012-03-02 2013-09-04 Rodos BioTarget GmbH Organ perfusion system
US9770472B1 (en) 2013-03-08 2017-09-26 Brahm Holdings, Llc Organ jacket and methods of use
KR102577616B1 (en) 2013-03-15 2023-09-11 미로매트릭스 메디칼 인크. Use of perfusion decellularized liver for islet cell recellularization
EP3033093A1 (en) 2013-08-16 2016-06-22 Alexion Pharmaceuticals, Inc. Treatment of graft rejection by administering a complement inhibitor to an organ prior to transplant
CN103719075B (en) * 2014-01-14 2015-01-28 吕凌 Isolated organ preserving device and preserving method thereof
WO2015138832A1 (en) 2014-03-13 2015-09-17 The General Hospital Corporation Devices and methods to improve and assess viability of human livers
IL303658B1 (en) 2014-06-02 2024-03-01 Transmedics Inc Ex vivo organ care system
CA2970117A1 (en) 2014-12-12 2016-06-16 Darren FREED Apparatus and method for organ perfusion
WO2016178980A1 (en) 2015-05-01 2016-11-10 Alexion Pharmaceuticals, Inc. Efficacy of an anti-c5 antibody in the prevention of antibody mediated rejection in sensitized recipients of kindney thansplant
CN104845878B (en) * 2015-05-28 2017-01-25 四川大学华西医院 In-vitro liver perfusion device and application thereof
WO2017075325A1 (en) 2015-10-30 2017-05-04 Alexion Pharmaceuticals, Inc. A method of inhibiting exacerbations of t cell-mediated allograft vasculopathy
US20170369849A1 (en) * 2016-05-11 2017-12-28 The University Of North Carolina At Chapel Hill Compositions and methods for bioengineered tissues
FR3067220B1 (en) 2017-06-13 2019-07-26 Centre National De La Recherche Scientifique DEVICE FOR INFUSION OF AN ORGAN
US11700848B2 (en) 2017-11-20 2023-07-18 Tevosol, Inc. Apparatus for perfusion of an excised organ
US20210315200A1 (en) * 2020-04-14 2021-10-14 Miromatrix Medical Inc. System for functional testing and transportation of biologically engineered organs
CN111643735B (en) * 2020-06-10 2022-05-13 中南大学湘雅二医院 Preparation method of acellular xenogeneic small blood vessel

Family Cites Families (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1682344A (en) 1928-08-28 lesieur
US1916658A (en) 1931-10-09 1933-07-04 Louis R Davidson Pneumothorax apparatus
US3183961A (en) 1960-09-08 1965-05-18 Brandt Herbert Method and apparatus for controlling the temperature and humidity of a regenerative air-heater
US3468136A (en) 1964-08-25 1969-09-23 Emil S Swenson Method for maintaining organs in a completely viable state
US3406531A (en) 1964-08-25 1968-10-22 Emil S. Swenson Apparatus for maintaining organs in a completely viable state
US3545221A (en) 1967-05-22 1970-12-08 Swenko Research & Dev Inc Apparatus for maintaining organs in vitro in a completely viable state
FR1577356A (en) 1968-04-04 1969-08-08
US3772153A (en) 1969-04-02 1973-11-13 Air Liquide Apparatus for the preservation of animal or human organs in living condition
BE755423A (en) 1969-10-06 1971-02-01 Baxter Laboratories Inc PROCESS AND APPARATUS FOR ORGAN PRESERVATION
US3660241A (en) 1970-01-12 1972-05-02 Baxter Laboratories Inc Container for organ perfusion or the like
US3810367A (en) 1970-07-16 1974-05-14 W Peterson Container for cooling, storage, and shipping of human organ for transplant
DE2241698C2 (en) 1971-09-02 1982-08-26 Roland Dr.med. Zürich Doerig Process for organ preservation and device for carrying out this process
US3881990A (en) 1971-11-24 1975-05-06 Waters Instr Inc Method of transporting and storing organs while retaining the organs in a viable condition
US3843455A (en) 1972-09-13 1974-10-22 M Bier Apparatus and technique for preservation of isolated organs through perfusion
US3877843A (en) 1973-05-21 1975-04-15 Baxter Laboratories Inc Pulsatile pumping system
US3995444A (en) 1974-11-08 1976-12-07 American Hospital Supply Corporation Organ perfusion system
US4186565A (en) 1978-05-19 1980-02-05 Henry Ford Hospital Perfusion system for organ preservation
US4242883A (en) 1979-04-02 1981-01-06 Henry Ford Hospital Liver preservation
US4798824A (en) 1985-10-03 1989-01-17 Wisconsin Alumni Research Foundation Perfusate for the preservation of organs
US4745759A (en) 1986-12-23 1988-05-24 Bauer Dan O Kidney preservation machine
US5602026A (en) 1988-10-14 1997-02-11 The General Hospital Corporation Culturing liver cells between two supports
US5356771A (en) 1993-03-11 1994-10-18 Board Of Regents, The University Of Texas System Combined perfusion and oxygenation organ preservation apparatus
US6642045B1 (en) * 1997-04-14 2003-11-04 Breonics, Inc. System for exsanguinous metabolic support of an organ or tissue
US8409846B2 (en) 1997-09-23 2013-04-02 The United States Of America As Represented By The Department Of Veteran Affairs Compositions, methods and devices for maintaining an organ
US6100082A (en) 1997-09-23 2000-08-08 Hassanein; Waleed H. Perfusion apparatus and method including chemical compositions for maintaining an organ
US6977140B1 (en) 1998-09-29 2005-12-20 Organ Recovery Systems, Inc. Method for maintaining and/or restoring viability of organs
US7749693B2 (en) 1998-09-29 2010-07-06 Lifeline Scientific, Inc. Method of determining that an organ is not suitable for transplantation and using it for testing substances
US6673594B1 (en) 1998-09-29 2004-01-06 Organ Recovery Systems Apparatus and method for maintaining and/or restoring viability of organs
FR2785501B1 (en) 1998-11-10 2001-01-05 Centre Nat Rech Scient PERFUSION AND / OR PRESERVATION AND / OR REPERFUSION SOLUTION DURING ORGAN TRANSPLANTATION
GB9908335D0 (en) 1999-04-12 1999-06-09 Univ Cambridge Tech Methods and means for extracorporeal organ perfusion
US20040038192A1 (en) 1999-04-14 2004-02-26 Breonics, Inc. System for exsanguinous metabolic support of an organ or tissue
CA2370781A1 (en) 1999-04-30 2000-11-09 Joseph P. Vacanti Fabrication of vascularized tissue using microfabricated two-dimensional molds
KR100304594B1 (en) 1999-06-12 2001-09-13 강흔수 Composition for the Preservation of Organs and Blood Cells
US6479064B1 (en) * 1999-12-29 2002-11-12 Children's Medical Center Corporation Culturing different cell populations on a decellularized natural biostructure for organ reconstruction
US20020039786A1 (en) * 2000-01-19 2002-04-04 Reid Lola M. Liver tissue source
WO2002053193A2 (en) 2001-01-02 2002-07-11 The Charles Stark Draper Laboratory, Inc. Tissue engineering of three-dimensional vascularized using microfabricated polymer assembly technology
US7572622B2 (en) 2002-08-14 2009-08-11 Transmedic, Inc. Heart preservation chamber
EP1589814B1 (en) 2003-01-16 2009-08-12 The General Hospital Corporation Use of three-dimensional microfabricated tissue engineered systems for pharmacologic applications
CA2521324C (en) 2003-04-04 2014-12-09 Organ Recovery Systems, Inc. Methods and apparatus for perfusion, diagnosis, storage and/or transport of an organ or tissue
NL1024022C2 (en) 2003-07-30 2005-02-01 Technologiestichting Stw Portable preservation device for a donor organ.
US20070042339A1 (en) 2003-08-08 2007-02-22 Massachusetts General Hospital Preservation of biomaterials with transported preservation agents
DE10340487B4 (en) 2003-09-03 2007-07-12 Technische Universität Dresden perfusion
US7504201B2 (en) * 2004-04-05 2009-03-17 Organ Recovery Systems Method for perfusing an organ and for isolating cells from the organ
NZ554543A (en) 2004-10-07 2011-03-31 Transmedics Inc Systems for ex-vivo organ care
US9078428B2 (en) 2005-06-28 2015-07-14 Transmedics, Inc. Systems, methods, compositions and solutions for perfusing an organ
KR20140040293A (en) 2005-08-26 2014-04-02 리전츠 오브 더 유니버스티 오브 미네소타 Decellularization and recellularization of organs and tissues
ES2537953T3 (en) 2006-03-28 2015-06-16 Organ Assist B.V. A method and system for prolonging the viability of an organ of a donor
BRPI0711599B8 (en) * 2006-05-11 2021-07-27 Hli Cellular Therapeutics Llc methods for collecting and using stem cells from placental umbilical cord blood
JP2010501486A (en) 2006-08-18 2010-01-21 リジェナークス・バイオファーマスーティカルズ・インコーポレイテッド Methods and compositions for storing and / or preparing organs or tissues for transplantation
CN101677526B (en) 2007-03-01 2014-07-09 生命线科学有限公司 Perfusion regulation
US9457179B2 (en) 2007-03-20 2016-10-04 Transmedics, Inc. Systems for monitoring and applying electrical currents in an organ perfusion system
US8765364B2 (en) 2007-05-18 2014-07-01 Lifeline Scientific, Inc. Ex vivo methods for validating substance testing with human organs and/or tissues
US8771930B2 (en) * 2007-05-18 2014-07-08 Lifeline Scientific, Inc. Ex vivo methods for testing toxicity of substances using donated human organs or tissues
US8927257B2 (en) 2007-09-28 2015-01-06 Organ Assist B.V. Organ transport container system
US9814230B2 (en) 2008-01-31 2017-11-14 Transmedics, Inc. Systems and methods for ex vivo lung care
BRPI1008009B1 (en) 2009-02-23 2018-05-29 Cell & Tissue Systems, Inc. Methods for cryopreservation of tissue without ice

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120064537A1 (en) * 2010-06-30 2012-03-15 Miromatrix Medical Inc. Use of perfusion decellularized organs for matched recellularization
US9986734B2 (en) 2013-01-08 2018-06-05 Yale University Human and large-mammal lung bioreactor
US20200352157A1 (en) * 2014-09-02 2020-11-12 United Therapeutics Corporation Automated bioreactor system, system for automatically implementing protocol for decellularizing organ, and waste decontamination system
US10660329B2 (en) 2015-04-23 2020-05-26 Etablissement Francais Du Sang Method for preserving cells, tissues or organs in hypothermia
WO2017070007A3 (en) * 2015-10-15 2017-06-15 Wake Forest University Health Sciences Methods of producing in vitro liver constructs and uses thereof
US11732241B2 (en) 2015-10-15 2023-08-22 Wake Forest University Health Sciences Methods of producing in vitro liver constructs and uses thereof
AU2016340819B2 (en) * 2015-10-15 2023-04-06 Wake Forest University Health Sciences Methods of producing in vitro liver constructs and uses thereof
US20200080050A1 (en) * 2016-07-11 2020-03-12 Yaakov Nahmias Systems and methods for growing cells in vitro
JP7303952B2 (en) 2018-08-03 2023-07-05 ベリグラフト アクティエボラーグ Methods of making individualized blood vessels
JP2023052212A (en) * 2018-08-03 2023-04-11 ベリグラフト アクティエボラーグ Methods of preparing personalized blood vessels
EP4069821A4 (en) * 2019-12-02 2024-04-03 Avant Meats Company Ltd System for producing cultivated meats, tissues and associated products from cells
CN112980683A (en) * 2021-03-12 2021-06-18 立沃生物科技(深圳)有限公司 In-vitro liver and kidney perfusion system and hepatocyte regeneration method
CN117264876A (en) * 2023-11-21 2023-12-22 妙顺(上海)生物科技有限公司 Method for preparing multi-donor liver cells

Also Published As

Publication number Publication date
WO2011002926A3 (en) 2011-05-19
US20220098548A1 (en) 2022-03-31
WO2011002926A2 (en) 2011-01-06
US20150322404A1 (en) 2015-11-12
US11136553B2 (en) 2021-10-05

Similar Documents

Publication Publication Date Title
US20220098548A1 (en) Isolated adult cells, artificial organs, rehabilitated organs, research tools, organ encasements, organ perfusion systems, and methods for preparing and utilizing the same
Barakat et al. Use of decellularized porcine liver for engineering humanized liver organ
Uygun et al. Organ reengineering through development of a transplantable recellularized liver graft using decellularized liver matrix
Park et al. Bioartificial liver systems: current status and future perspective
Mirmalek-Sani et al. Immunogenicity of decellularized porcine liver for bioengineered hepatic tissue
Bao et al. Construction of a portal implantable functional tissue-engineered liver using perfusion-decellularized matrix and hepatocytes in rats
Mirmalek-Sani et al. Porcine pancreas extracellular matrix as a platform for endocrine pancreas bioengineering
Strom et al. Hepatocyte transplantation as a bridge to orthotopic liver transplantation in terminal liver failure
JP6636956B2 (en) Human liver scaffold
Gerlach et al. Use of primary human liver cells originating from discarded grafts in a bioreactor for liver support therapy and the prospects of culturing adult liver stem cells in bioreactors: a morphologic study
US20100093066A1 (en) Decellularization and recellularization apparatuses and systems containing the same
Orlando et al. Will regenerative medicine replace transplantation?
JP2017195900A (en) Decellularization and recellularization of organs and tissues
US20110190679A1 (en) Extracorporeal cell-based therapeutic device and delivery system
US20090081296A1 (en) Extracorporeal cell-based therapeutic device and delivery system
Hsu et al. Liver-assist device with a microfluidics-based vascular bed in an animal model
Booth et al. Liver bioengineering: current status and future perspectives
de Vries et al. Bulk droplet vitrification: an approach to improve large-scale hepatocyte cryopreservation outcome
Ambrosino et al. Hepatocyte transplantation in the treatment of acute liver failure: microencapsulated hepatocytes versus hepatocytes attached to an autologous biomatrix
JP2015515277A (en) Bioartificial filtration organ
Zhang et al. Integration of single-layer skin hollow fibers and scaffolds develops a three-dimensional hybrid bioreactor for bioartificial livers
US20050265979A1 (en) Method for cryopreserving microencapsulated living animal cells enclosed in immunoisolation membranes, such microencapsulated living animal cells in immunoisolation membranes, and biohybrid artificial organ modules using such microencapsulated living animal cells in immunoisolation membranes
Stefanovich et al. Extracorporeal plasma perfusion of cultured hepatocytes: effect of intermittent perfusion on hepatocyte function and morphology
Bikhchandani et al. Extracorporeal liver perfusion system for artificial liver support across a membrane
Fathi et al. Whole-liver decellularization: Advances and insights into current understanding

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:YARMUSH, MARTIN;UYGUN, BASAK E.;UYGUN, MUSTAFA KORKUT;AND OTHERS;SIGNING DATES FROM 20120529 TO 20120817;REEL/FRAME:029318/0108

Owner name: MASSACHUSETTS INSTITUTE OF TECHNOLOGY, MASSACHUSET

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:IZAMIS, MARIA LOUISA;MILWID, JOHN MILES;SIGNING DATES FROM 20120517 TO 20120519;REEL/FRAME:029318/0120

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION