Revision cards Flashcards

1
Q

What is tissue engineering?

A

Regenerative medicine is an umbrella term, underneath lies a number of approaches including tissue engineering. Tissue engineering is a multidisciplinary field aimed at development of biological substitutes to restore, maintain or improve tissue functions.
Uses cells and biomaterials to treat degenerative diseases or injury.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What is the clinical need for tissue engineering?

A

End-stage organ failure and tissue loss are often the most devastating. Major causes of organ failure include injury, disease and aging.
Current treatments for organ failure:
-Surgical reconstruction
-Mechanical devices
-Transplantation
Limitations include: poor success rate, surgical complications, morbidity at donor sites, only mechanical support unifunction, don’t grow with the tissue (require surgery in children as they grow), need for immunosuppressants.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are the fatal consequences of the Transplantation Crisis?

A

The transplantation crisis means that 3 people die a day in the UK waiting for a suitable organ. Aging population requires more and more organ donations. This crisis means that patients receive unfit unsuitable organs (subobtimal).
CASE STUDY 2 patients were each given a kidney but a couple of days later died of Meningitis. Donor died of Meningitis caused by a rare nematode which had been transferred to them.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

How can tissue engineering aid the transplantation crisis?

A

Tissue engineering could be a new solution for treatment of organ failure.
Needed because:
-Donor tissues and organs are in short supply
-Want to minimise immune system response/need for immunosuppressants
Goals of tissue engineering involve saving and improving lives by assembling functional constructs that can be placed into the body to restore or replace a damaged tissue/organ.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the history of tissue engineering?

A

The research on what we now know to be TE emerged in 1970s and 1980s. The term was coined in 1987. In the 1990s research accelerated and industry began to emerge partly due to the parallel development in the field of biomaterials and stem cell biology.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are the main components when building a tissue?

A
  • Cells to make up the tissue
  • Biomaterials/scaffolds to replace ECM
  • Bioactive molecules to direct fate of cells and promote integration of construct into body
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the 2 main approaches when building a tissue?

A

IN VITRO: combining cells with a biomaterial/scaffold then transplantation
IN VIVO: transplantation of scaffold into the body, endogenous host cells are recruited

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are the different disciplines involved in tissue engineering?

A
  • Cell biology to cell culture
  • Bioengineering to create and engineer the biomaterial
  • Immunology for transplantation/immunorejection
  • Surgeons to transplant
  • Clinicians to perform and organise clinical trials
  • Pharmacists for immunosuppressants
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What did Cao et al do in 1997 and what were their limitations?

A

Aim to assess the feasibility of growing tissue-engineered cartilage in the shape of a human ear.
A plaster mold of a 3 yr olds ear was cast from an impression of an ear. This was used as a scaffold for cartilage cells from a calf to be seeded and grown on. Nude mouse used as a bioreactor for the scaffold and cells to grow on. After 12 weeks the constructs were explanted, sectioned and stained.
Limitations: skin misssing, bovine chrondrocytes were used, scaffolds had to be refined for mechanical stability, implications on the growth rate of the artificial ear.
Media described it as a human ear grown on a genetically engineered mouse, none of which is true!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What makes up an organ structure?

A

Epithelial tissue, connective tissue, muscle tissue, nerve tissue.
Requires oxygen, discharge waste via blood, EC fluid and lymph. Vasculature is required for tissue perfusion.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What are the phases of wound healing?

A

Ordered sequence of events that will lead to healing of an insult to the skin.
-Inflammatory phase: stop the bleeding, inflammatory cells clear up debris caused by dead cells, injured cells and microbes.
-Proliferative phase: wound trying to close up, new tissue formed but in a disorganised manner.
-Remodelling phase: remodelling new tissue to return to normal organisation.
Overlapping processes that occur over days, weeks, months, years.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What is an overview of what occurs in wound healing?

A

Injury cuts the blood vessels which bleed into the wound. Blood clot forms and leukocytes clean the wound. Blood vessels regrow and granulation tissue forms (fibroblasts). Epithelium regenerates and scar forms, reforming 70-80% of original tensile strength.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is the difference between regeneration and repair?

A

Mild superficial injury will only affect the uppermost layer - epithelia. Skin, intestine and blood have a high turnover and have resident stem cells allowing them to regenerate quickly. However this is providing that the injury only affects cells and not ECM and tissue affected contains stem cells. Stable tissues like heart and brain cannot regenerate so repair their injury instead. It all depends on the extent of the injury (transient or not), the type of tissue and components affected.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

How are acute and chronic injuries different?

A

Acute injury occurs when the stimulus is removed quickly and there is cell death but there is still an intact tissue framework. This allows for regeneration and restitution of the normal structure.
If there is an acute injury with damaged tissue framework, it can be repaired but will leave scar tissue.
If there is a chronic injury (persistent tissue damage) it leads to fibrosis like pulmonary fibrosis (tissue scar).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is fibrous encapsulation?

A

Tissue response to implanted biomaterials is similar to foreign material response. The implantation of a biomaterial/medical device results in injury to the tissues and organs. Abundant deposition of extracellular matrix. Isolation of biomaterial from local tissue environment. Type of wound healing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are the different sources of cells for transplantation?

A
  • Autologous (cells from the same patient)
  • Allogeneic (same species, different individual)
  • Xenogeic (different species)
  • Syngenic or isogenic (genetically identical/twin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What are the different types of cells used for tissue engineering?

A

-Differentiated mature cells
-Mixture of differentiated cells
-Stem cells
Adult stem cells: can’t be kept in culture for long, but can be autologous, need a large number, difficult to proliferate in vitro.
Embryonic stem cells: ethical issues but can give almost any cell.
Induced pluripotent stem cells: can be autologous, almost any cell, difficult to control differentiation, unknown risks.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

How do autologous and allogenic cells compare?

A

Autologous cells don’t require tissue matching, no need for graft vs host response, faster engraftment, no disease transmission.

Allogeneic cells require tissue matching and graft vs host response, slower engraftment, possible disease transmission (HIV or hepatitis).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What are the problems with using differentiated cells in tissue engineering?

A

Examples of differentiated cells that may be used include fibroblasts, keratinocytes, osteoblasts, endothelial cells, chrondrocytes, preadipocytes and adipocytes.
Main problem is sourcing the cells.
Biopsies are intrusive and don’t give many cells.
Differentiated cells can’t really be expanded in vitro because they senesce after a while. If they do they change too much. Batch to batch variability.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

How to culture cells?

A
  • Growth medium: high energy, source of glucose, amino acids, GFs. Replacing blood so needs to be exchanged every day to mimic circulation.
  • Laminar hood to ensure aseptic conditions and to direct the airflow.
  • Incubator to keep cells at 37c and use water to humidify the environment.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is good manufacturing practice?

A

Ensures that medicinal products are consistently produced and controlled to the quality standards appropriate to their intended use.
Cell manufacturing must be free from animal products, eliminate batch-to-batch variability as much as possible and meet their storage needs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What are the roles of the ECM?

A

Most cells require attachment to a solid surface otherwise they undergo apoptosis.
Roles of ECM:
-Provides structural support for cells
-Contributes to the mechanical properties e.g. collagen bundles in tendon, collagen and elastin fibrils in skin for elasticity and toughness, calcified ECM in bone for strength.
-Provides bioactive cues for cells to modulate them
-Act as reservoirs of growth factors and potentiates their actions
-Scaffolding for orderly tissue renewal, avoiding scarring.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What are the components of the ECM?

A

Tends to differ from tissue to tissue.

  • Fibrous structural proteins like collagens and elastin for tensile strength and recoil
  • Water hydrated gels like proteoglycans and hyaluronon for resilience and lubrication
  • Adhesive glycoproteins like fibronectin and laminin for connection to ECM components and cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is collagen?

A

Most abundant protein. 80-90% of collagen in the body is either type 1, 2 or 3. Basic collagen unit has 3 helical structures, each composed of 1050 AAs. Helical due to the increase abundance of sequence of proline, hydroproline and glycine. Assemble into fibrils end to end side to side to make fibres.
Vit C required for collagen production.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What are proteoglycans?

A

Composed of glycosaminoglycan chains linked to a specific protein core. Very hydrophilic, form highly hydrated compressible gels.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What are adhesive molecules?

A

Very diverse
e.g. fibronectin contains a RGD sequence (Arg-Gly-Asp). The sequence is recognised by cells as a cell binding domain. If cells will not adhere to biomaterial you can add this sequence onto the biomaterial (structurally integrate).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

How do cells adhere to ECM/biomaterials?

A

Cells mechanically interact with ECM before making contact via receptors/signalling parts.
Cell flattens out to reach appropriate shape.
Can move around and explore their environment.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What are integrins?

A

19a and 8B subunits.
Form heterodimers with extracellular domain, transmembrane domain and short intracellular domain. ECD and ICD can be used in inside-out and outside-in signalling. a and B can make up many different combinations, different affinit depending on environment. Both specificity and redundancy allowing a robust system.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

How is integrin conformationally activated?

A

-Extrinsic ligand ‘outside-in’ (collagen, laminin, fibronectin) causes a conformational change in integrin, changing their affinities for specific ECM components, activates signalling pathways.
-Intrinsic ligand ‘inside-out’ (talin, kindins) causes changes which intiates the assembly of the actin cytoskeleton.
Mostly mediated by focal adhesion kinase which recruits a number of other proteins to the sites of focal adhesion triggering a cellular response.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What is machanotransduction?

A

Process by which external mechanical stimuli are transmitted into the nucleus. Cells can sense the environment and alter it accordingly.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is a Sheffied case study of tissue engineering?

A

Macular degeneration clinical trial with 2 patients. Treatment looks promising from Phase 1 trials. Stem cells used were SHEF-1hESC. Using fully differentiated embryonic stem cells placed on a coated, synthetic basement membrane.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What are biomaterials?

A

Nonviable materials used in a medical device intended to interact with biological systems. Can be used to develop scaffolds for tissue engineering. Developed from the field of medical devices.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is the history behind biomaterials?

A

Using hair, silk etc for suturing wounds.
Issues with infection and immune response.
Sir Harold Ridley examined eyes of pilots and saw that they had plastic splinters from the cockpit without an immune response. Developed intraocular lens for the treatment of cataracts.
Important development: a material can be integrated into the body without an immune response, making it biocompatible.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What is the biocompatibility of materials?

A

Ability of a material to perform with an appropriate host response in a specific application.
e.g. haemolyalysis membrane for patients with poor kidney function, the membrane is only in contact with blood for a couple of hours. Urinary catheter is implanted for days or weeks. Hip replacement is needed for life.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What are some examples of appropriate host response?

A

Resistance of blood clotting
Resistance to bacterial colonization
Normal healing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What is the evolution of the biomaterials field?

A

First generation: Bioinertness
Determined to be biocompatible if do not induce toxic reaction or carcinogenesis.
Second generation: Bioactivity
Bioglass is a ceramic implant that you can place in rats. Bonded with bone and could not be separated. Cannot respond to inputs.
Third generation: Functional tissue
Materials that can induce a cellular response at a molecular level.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What are polymers?

A

‘Poly’ (many) ‘meros’ (parts)
Large molecules made up of chains or rings of linked monomeric units. Mw 200,000 (compared to h20 18Da).
Can be linear, branched or networked.
Common polymer biomaterials include Polyethylene, Polytetrafluroethylene, Polypropylene, Polyvinylchloride, Polydimethylsiloxane, Poly (methyl methacrylate). Typically have carbon atoms plus side chains.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

How can polymers be structured?

A
Can be arranged in different shapes
-Homopolymer
-Random copolymer
-Alternating copolymers
-Block copolymer
-Graft copolymer
It is possible to mix two polymers e.g. blend of homopolymer A and B.
We can change their functional properties by changing how they are arranged.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What are hydrogels?

A

Crosslinked polymer networks that are insoluble but swell in aqueous medium. Offer an environment that resembles the highly hydrated state of natural tissues.
Can model soft tissue environments.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What are natural polymers?

A

Already exist in animal or plant tissue.
1) Protein-based natural polymers
Collagen - well integrated, highly common, high immunogeneity
Gelatin - less immunoggenic, denatured form of collagen
Fibrin - major component of blood clots
Elastin and Soybean
2) Polysaccharides
Chitosan - present in fungi and crustaceans
Aliginates - produced by seaweed and some bacteria
Hyaluronan - present in umblical cord, skin, cartiliage
Chondroitin sulfate - present in cartilage

Obtain the plant/animal tissue by extraction. Purify them from the tissue. Concentrate them to get good enough sample.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What are synthetic polymers?

A

Synthesise from scrath.
Polylactic acid
Polyglycolic acid
Poly (lactic-co-glycolic) acid

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What are semi-synthetic polymers?

A

Hybrid molecules made by incorporation of biologically active macromolecules onto the balance of synthetic polymers. e.g. semi-synthetic PEG (polyethylene glycol)-fibrinogen. PEG fives density, stiffness and biodegradability and fibrinogen gives biofunctional domains.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What are the advantages and disadvantages of natural polymers?

A
Advantages:
-Already exist, easily obtainable
-Existing bioactivity
-Can make the most of naturally-occuring properties
Disadvantages:
-May be difficult to harvest
-Potential ethics
-Batch-to-batch variability
-Shelf-life
-Immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What are the advantages and disadvantages of synthetic polymers?

A
Advantages:
-Can be tailored to specific properties
-Can control process and structure
Disadvantages:
-Diffferent structures that may not interact with tissues
-Difficult to predict bioactivity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What are the essential properties of biomaterials?

A
  • Physical/mechanical - strength, elasticity, architecture
  • Chemical - degradability, resorbtion, water content
  • Biological - interactions with cells, release of biologically acitve signals
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What is the difference between degradable and resorbabale materials?

A

Degradable materials:
Hydrolysis of a polymer require H20 to break down covalent bonds to give degradation products. For it to be biocompatible degradation products should be non-toxic.
Resorbable materials:
Total elimination of the initial foreign material and its byproducts. Broken down by the body, metabolised leaving no trace.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What are the bulk properties of biomaterials?

A

Strength, toughness, fatigue resistance, stability.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What are the surface properties of biomaterials?

A
  • Chemically or physically altering the atoms/molecules in the existing surface
  • Overcoating the existing surface with a material having a different composition
  • Creating surface textures or patterns
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

How does matrix direct stem cell fate?

A

Engler et al (2006) Matrix elasticity directs stem cell lineage specification. Mesenchymal stem cells were placed on substrates of different elasticity (soft tissue, muscle, collagenous bone resemblance). The cells were directed to different lineages (neural, muscle, bone) just based on stiffness, can sense mechanical environment,

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What are the consideration for creating surface modifications?

A
  • Thin surface modifications (3-10nm) to avoid changing the marker properties of the material
  • Delamination resistance
  • Surface analysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

How do cells interact with biomaterials?

A

Cells do not interact directly with materials.

  • A later of protein (from growth or plasma) adheres to the surface of biomaterials
  • Cells recognise these proteins and adsorb to them depending on the surface material properties.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What are non-fouling surfaces?

A

Surfaces that resist adsorption of proteins and/or adhesion of cells e.g. PEG and Zwitterionic. In medical devices this is good because may inhibit bacterial colonization.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

How to functionalise a surface?

A

Attachment of biomolecules to polymer surfaces

Immobilisation of integrin-binding peptides or entire proteins (RGD domains added).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

How do cells respond to substrate chemistry?

A

Cell adhesion to materials occurs through receptors in the cell membrane
Arginine-glycine-aspartic acid (RGD) domain in fibronectin and vitronectin
Cellular responses can vary with the surface density of RGD peptides immobilised

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What are Micrometer-scale chemical patterns?

A

Can be created using microstamping or microcontact printing (µCP)

  • Prepolymer is poured onto a structured master
  • Prepolymer is cured and the stamp is peeled off the mamster
  • Stamp is cut into smaller pieces using an enzyme
  • Stamp is inked by soaking in ink solution
  • Ink printed on suitable surface
  • A pattern of substrate is obtained, cells adhere to ECM area stamp
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

How does cell shape control cell fate of endothelial cells?

A

Chen et al 1997
It was already known that angiogenesis relies on interplay of chemical and mechanical signals. Endothelial cell growth is also critical, early experiments suggest that increases in spread area are accompanied by an increase in cell proliferation.
Hypothesis: cell shape per se controls cell fate of endothelial cells.
Approach: Micropatterning of fibronectin islands on a non-fouling surface of lots of different sizes
Result: The extent of cell spreading determined whether a cell underwent proliferation or apoptosis. Same GF, same genes, same ECM but different geometry, different fate outcome. Cells can filter the same set of chemical inputs to produce different functional outputs.
Warmflash et al (2014) used this to recapitulate gastrulation,
Teixeira et al (2006) used microstamping or microcontact printing. Thin lines, cells were perpendicular, thick lines cells were vertical.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What is the need for an artificial scaffold?

A

Scaffolds provide the support for cells to proliferate and maintain their functions.
They deliver and retain cells and bioactive molecules. Therefore when you are tissue engineering you need to develop an artificial extracellular matrix which would usually perform these functions in vivo.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What are the design considerations for a scaffold?

A
  • Material science: is it biodegradable?
  • Scaffold architecture: does it have adequate mechanical properties?
  • Scaffold-cell interaction: does it allow cell attachment/function?
  • Up-scaling production: can it be mass produced to meet clinical need?
  • 3D
  • Nutrient supply: is there capability for it to be vascularised to allow for O2 perfusion and tissue integration?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

How can a scaffold be degradable?

A

3D scaffold can be added to two types of cells to support the cells. If the scaffold is degradable, it will gradually degrade and the cells put down their own ECM, allowing for stability at all times. The scaffold itself and the degradation process should be non-toxic.
Degradation kinetics should be matched to the tissue you are trying to regenerate.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

How must a scaffold interact with cells?

A

The surface properties of the scaffold must allow for cell attachment. If they don’t (e.g synthetic materials) they can be made functional by adding RDG domains which create cell binding domains.
The scaffold mustn’t hinder the ability of cells to attach to the matrix and start signalling (functionality).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What are the different materials that can be used for scaffold fabrication?

A

Naturally derived materials
Synthetic materials
Semi-synthetic materials
Acellular tissue matrices

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What is acellular tissue matrices?

A

Can be considered as natural. However, all the cellular material is removed from the extracted tissue, either by physical scrapping, although this is often not enough. Chemical methods (acids/bases) or biological methods (enzymes) can be used instead. Removing acellular components also removes antibodies to prevents an immunogenic reaction to seeded cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What are the advantages and limitations of decellularising a tissue to make a scaffold?

A

Decellularisation maintains intricate structures that would otherwise be difficult to manufacture.
Exploiting the intact 3D structure of the ECM
Some are commercially available (alloderm)

Up-scaling may be a problem.
Acessibility is a potential issue.
However it is important to fully decellularize a tissue to avoid initiating the host response, bacteria contamination or crosslinking which will affect the tissue’s normal morphology.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What are the mechanics and architecture of a scaffold?

A

Macroscopically the final shape of the tissue engineered is defined by the scaffold.
Microscopically human skin and bone are usually very porous for infiltration of O2 and nutrients. However, there may be limited interconnectivity and some pores may not be accessible. Further more too many pores will decrease the mechanical stability of the tissue, however this depends on the tissue being engineered.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What is porogen leaching?

A

Porogen leaching is a method used to create an artificial porous scaffold. It involves mixing a polymer solution with salt particles and placing it into a mold. Solvent evaporation causes polymer to solidify which when placed in water causes the salt to leach out. This is then freeze-dried to create a porous scaffold where the salt particles used to be. You can control the size of the salt particles to control the pore size.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What is phase separation?

A

Phase separation is a method used to create an artificial porous scaffold based on properties of a mixed system. It involves mixing the polymer solution with a solvent, placing into a mold. Using temperature, you can separate a polymer-rich phase and a solvent-rich phase. Evaporate the solvent to cause the polymer to solidify. Pores are formed where the solvent used to be.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What is electrospinning?

A

Electrospinning is a method used to create an artificial porous scaffold. Polymer solution is placed into a syringe. It is linked to a high voltage power supply. An electromagnetic field is created between the tip of the syringe and the grounded collection plate. As the polymer is released from the syringe the electromagnetic field causes the polymer fibres to splay. Very fine micronanofibres are collected onto the grounded plate.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What is additive manufacturing?

A
Additive manufacturing (aka rapid prototyping or free form fabrication) is a method used to create an artificial porous scaffold. It is a collection of methods. Process of joining materials to make objects from 3D model data usually layer upon layer.
3d scaffold design is converted to particular file that makes virtual slices in 2D. This is then transferred to a rapid prototyping machine. Roller spreads polymer across the table, inkjet head secretes a polymer binder to specific areas according to the shape determined by the computer.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What are the advantages of additive manufacturing?

A

High control of architecture
Production of scaffolds with precise morphologies
Combines medical imagine like MRI to fabricate anatomically shaped implants.
Main limitation is that there is a limited number of biomaterials that can be used.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What is cell encapsulation?

A

Usually when you combine the artificial scaffold and seeded cells, it may be the case that the cells are not integrated or that they clog the pores.
Cell encapsulation provides an alternative way to provide a better integration in 3D printing. In this method, scaffolds and cells are already combined and gaps are free to act as pores.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

How was ITOP developed?

A

Kang et al (2016) sought to address the challenge of producing 3D, vascularised cellular constructs of clinically relevant size, shape and structural integrity. Designed the ITOP Integrated Tisuue-Organ Printer to overcome the limitation of currently used bioprinting technologies (structural integrity, mechanical stability, printability). Contains different bioinks: cells combined with hydrogels which are non-toxic and bio-compatible. New idea to print cell-laden hydrogels with sacrificial scaffold to provide initial mechanical integrity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What are the different biomolecules that can be used for inducing tissue regeneration?

A

Small molecules (corticosteroids and hormones)
Proteins and peptides
Oligonucleotides

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What are bone morphogenetic proteins?

A

Initially extracted from bone matrix.
Members of the TGFb family.
Made by osteoblasts (bone-forming cells)
Osteoinductive - able to recruit osteoblasts and initiate differentiation of mesenchymal stem cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What are the problems with using BMPs in tissue engineering?

A

The efficient clinical use of BMPs depends on the delivery strategy.
BMPs act locally so they easily dissipate.
The use of biomaterials that can retain and sequester BMPs will enhance efficacy in bone regeneration.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What method was developed to effectively use BMPs in tissue engineering?

A
Lutolf et al (2003) 
Polyethylene glycol (PEG) is non-fouling so it is functionalised using RDG domain. Crosslinked to create a network mesh using MMP substrate sites. Cells in the vicinity recognise sites and bind to the scaffold. Cells secrete MMPs that degrade MMP cleavable bonds serving as crosslinks for the matrix. rhBMP2 is physically entrapped into the pores of the PEG gel by mixing it with PEG precursor before gelation. BMP is liberated from the matrix and can diffuse from the site signalling osteoblast precursor cells to secrete bone marrow and regenerate the bone.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

How was ITOP used for skeletal muscle reconstruction?

A

3D muscle construct 15mm x 5mm x 1mm dimension containing mouse myoblasts.
Designed a fibre bundle structure for muscle organisation. PCL pillars were used to maintain the structure and to induce cell alignment.
The printed construct was cross-linked with thrombin solution to induce gelation of fibrinogen and uncross-linked sacrificial material was removed by dissolving in cold medium.
TE construct matured into a functional muscle in vivo in rats. High cell viability after the printing process

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What are the steps involved with ITOP?

A
Medical imaging
3D CAD model
Visualised motion program
3D printing process
3D bioprinted tissue product
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What were the results of Lutolf et al (2003) research to effectively use BMPs in tissue engineering?

A

First they tested the sensitivity of the gels to proteolysis by cell-secreted MMPs using an in vitro model system for cell invasion. Fibroblasts recognised the cleavage site and could invade.
Control: no MMP cleavage site or MMP inhibitor
Monitored by BMP release

Functional proof cam from testing it on critical injuries (large enough so that the bone cannot regenerate).
MMP-sensitive, no BMP
MMP-insensitive, BMP present
No regeneration however MMP-sensitive with BMP showed good regeneration of the bone.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

How does using bioreactors compare in industry and tissue engineering?

A

Classical bioreactors are fermentors used in industry to grow high numbers of eukaryotic and prokaryotic cells for the production of antibiotics, recombinant genes and metabolic products under controlled conditions.
In tissue engineering bioreactors enable us to direct, maintain and intiate 3D structures which would be very difficult otherwise.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What are the challenges associates with tissue engineering that are tackled with the use of a bioreactor?

A
  • The ability to grow 3D tissue structures of relevant clinical sizes, same as in vivo
  • The growth and 3D assembly of multiple cell types that are requires for more complex functional tissue.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What is static culturing and what are the associated issues?

A

Static culture is usually performed in a culture flask, no special equipment is required, it is relatively cheap and easy. Cells are grown statically without any mixture, causing concentration gradients to occur. Fed with medium every day/few days - no control over pH and environment. Diffusion of O2 and nutrients is inefficient so cells on the inside will become necrotic.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What is a dynamic culture system?

A

A dynamic culture is the mixing of the cells in culture. Gives rise to homogenous concentrations of nutrients, toxins and other components. Can control environment, pH, temperature etc. Cells are placed in the bioreactors, medium is pumped in and out from medium tank by peristatic pump providing perfusion.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What are the key roles of bioreactors?

A
  • To establish spatially uniform cell distributions on 3D scaffolds
  • To overcome mass transport limitations in 3D constructs
  • To expose the developing tissue to physical stimuli (physical conditioning)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

What are the requirements for cell distribution on 3D scaffolds (bioreactors)?

A

Usually you just place cells of choice on scaffold using a pipette, relying on gravity to pull the cells through the porous structure and distribute through 3D. This is an inefficient process so cells will ususally just stay on the top and many will be lost. This is a problem especially when many biopsies are painful.
Therefore you need a high seeding efficiency, short inoculation period and uniform distribution of cells within the scaffold since this is what will determine the biochemical activity and mechanical strength of final TE construct.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

Give an example of a study that measured cell distribution on 3D scaffolds.

A

Bone marrow stromal cells were seeded onto scaffolds and after 18 hours MTT assay was used. MTT is converted by the mitochondria from a soluble yellow salt into an insoluble purple formazan salt. Used to see if cells are alive and metabolically active and where they are distributed on the scaffold. Compared between static (white area present) and perfused (even distribution).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What is mass transfer in 3D structures (bioreactors)?

A

Once cells are delivered to a scaffold you have to make sure that they are viable and functional.
External mass transfer: delivery of nutrients and O2 from the inner bulk to exterior
Internal mass transfer: delivery of nutrients and O2 from the exterior to the inner bulk
Also involves the removal of metabolites and CO2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

Give an example of a study where mass transfer was measured in 3D structures.

A

Wendt et al 2008
Chrondrocytes seeded using perfusion cell seeding. Cultured for 2 weeks with OR without perfusion. In statically cultured scaffold, there is a large amount of empty space where cells have undergone necrosis. Cells are only at the edges. Whereas the perfused culture has lots of cells all spread across.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What is physical conditioning (bioreactors)?

A

Tissues and organs in the body are subject to complex biomechanical environment. These physical forces include hydrodynamic/hydrostatic, mechanical and electrical. The tissue engineered construct needs to be able to maintain these too.
Eg. Pulsatile flow in blood vessels causes erthrocytes mature to their appropriate phenotype.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

Why are there different types of bioreactors?

A

Diversity in bioreactor design reflects the range of signals needed for formation of various tissues e.g. heart, bone, valves.
Biomaterials making up bioreactors must not induce toxocity or unwanted effects, they must be biocompatible and non-fouling (don’t stick).
They must also be sterile so are either single use or can be sterilised in an autoclave.
Withstand 37 degrees.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What are Spinner Flask Bioreactors?

A

Magnetic stirrer creates dynamic culture, the turbulence can cause damage to cells. Scaffold is dispensed from needles into the bulk media. Internal transfer is still quite limited for centre.
Relatively cheap and simple.
Mass transfer in the flasks is not good enough to deliver homogeneous cell distribution throughout scaffolds and cells predominantly reside on the construct periphery.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What are Rotator Wall Bioreactors?

A

Centrifugal forces generated due to the rotation of the cylinder counterbalance the gravitational pull on the scaffolds. As tissue grows in the bioreactor, the rotational speed must be increased in order to balance the gravitational force and ensure the scaffold remains in suspension.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

What are Perfusion Bioreactors?

A

The culture medium continually circulates through the TE construct which stays static. Most mass transfer limitations are mitigated. The effects of direct perfusion can be highly dependent on the medium flow rate.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

What are Compression Bioreactors?

A

Used to apply mechanical stimulus to cell-seeded constructs. This can be acting constantly, intermittently or cyclically. Apply mechanical pressure and examine the effects of this.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

What are the applications of bioreactors in decellarisation?

A

Complex architecture of tissues makes decelluarising them difficult. Perfusion bioreactor has been used for the heart, lung, liver and pancreas, mostly used for whole organs.
Porcine heart
The barbed end of the tubing is inserted into the aorta of the native heart. The tubing must be secured with hose clamps or zip ties above the aortic valve to ensure perfusion through the coronary arteries. The hear is then submerged in a water in a 4L beaker and air bubbles must be removed from the tubing using a pump. As solutions are perfused through the coronary arteries, the heart will lose its native colour and become white. It can then be re-seeded with cells again.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What are the applications of bioreactors in cartilage tissue engineering?

A

Articular cartilage is a load-bearing tissue, it is exposed to cyclical stress. Bioreactors with mechanical loading have been used for tissue engineering of cartilage.
Mauck et al (2000) Loading applied in cycles improved biomechanical properties of engineered articular cartilage.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

What are the applications of bioreactors in microgravity?

A

Space flights represent the best environment to investigate near-zero gravity effects but there are major limitations for setting up experimental analysis. Rotating wall bioreactors have been developed by NASA.
Tamma et al (2009) studied osteoclasts and the effects of near-zero gravity, found there was a decrease in bone density.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

Where is the clinical need in tissue engineering of heart valves?

A

Heart valves are required for directing the flow of blood. May be causes that valves cannot open or close, causing reverse flow. Current treatment options are surgical repair or valve replacement. Prosthetic valve replacement isn’t ideal because you need anti-thrombotic drugs and they don’t grow with children.
Tissue engineering offers the potential of creating a valve that will grow and adapt with growth.
Sourcing cells for this is difficult as you need smooth muscle cells and endothelial cells - stem cells may be the main choice. Must sustain the biophysical forces that cells experience in vivo: mechanical strech and hydrodynamic shear.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What is a Flex-Stretch-Flow bioreactor?

A

Developed by Engelmayr et al (2008)
Hypothesised that mimicking the mechanical stimulation and perfusion in vitro could lead to further improvement of the engineered tissues.

Flex-stretch-slow bioreactor
Place a scaffold seeded with cells which is fixed onto 2 posts, one which is fixedd and another which is movable which is linked to a linear motor to drive its movement. Enables flexing and stretching of scaffolds and allows media flow over the scaffold. Culture media can be recirculated within the bioreactor chamber via a magnetically coupled paddle-wheal to provide a laminar flow and associated fluid shear stresses to scaffold specimens.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

How have mesenchymal stem cells been used in a flex-stretch-flow bioreactor?

A

Mesenchymal stem cells derived from sheep bone marrow were cultured on polyglycolic acid/polylactic acid scaffolds.
Test group under mechanical stimuli and hydrodynamic shear had increased collagen content and effective stiffness of engineered valves after 3 weeks in culture compared to controls.
By the end of the 3 weeks engineered valves had a modulus comparable to those generated from smooth muscle cells in previous studies. Cyclic flexure and laminar flow can synergistically accelerate MSC-mediated tissue formation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

Give examples of how tissue engineering can be used in blood vessels and vocal folds.

A

Nikiason et al (1999) Pulsatile bioreactors/pump mimic pulses found in blood vessels.

Vocal fold are subject to mechanical stimuli so bioreactor incorporate speakers to mimic vocal fold maturation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

What are the current challenges to bioreactor design?

A
  • Mimicking native cell behaviour - we need further understanding of tissue development and regeneration
  • Scaling up. Most bioreactors are specialised devices with a low volume output. Time consuming and labour intensive.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What is the history of skin substitutes?

A

Skin substitutes were the first tissue engineered created ex vivo. There is a huge clinical need for skin substitutes and keratinocytes have been well cultured since the mid 1970s.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What are the different types of tissues according to their structure?

A

-Flat e.g. epidermis, cornea
One dominant cell type, relatively simple
-Tube e.g. blood vessels and urethras
Several cell types, serve as conduits
-Hollow non-tubular organs e.g. bladder, stomach
-Solid organs e.g. kidneys, lungs, heart and liver
Complexity in structure, histology, function
More complex - more difficult to recreate. Need a good knowledge of their structure and function first.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

What are the properties and function of skin?

A

Largest organ in the body by surface area
About 10% of body mass
Functions of the skin:
-Protection against chemicals, UV light and microbes
-Regulation of water, temperature, sweating etc
-Sensation, contains sensory nerves
3 main layers: epidermis (neuroectoderm), dermis (mesoderm) and hypodermis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

What is the epidermis?

A

A thin layer that varies in thickness according to location.
Protects the body from environmental factors.
Consists only of cells:
-Keratinocytes (95%) produce keratin for toughness
-Melanocytes - produce melanin, located at the bottom
-Langerhans’ cells - dendritic cells, antigen properties
-Merkel cells - touch sensation, near neurons
Mature epidermis is a multilatered epithelium, upper layer is shed. Cells in basal layer are mitotic so will divide and move up and differentiate. Basal cells will secrete ECM matrix to create basement membrane.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What is the dermis?

A

Bulk of the skin
Composed of the skin with some elastin and glycosaminoglycans (mainly molecular).
Main cell types are fibroblasts - important for wound healing.
Also contains blood vessels, hair follicles, sebaceous and sweat glands.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

What is the hypodermis?

A

A network of adipose cells and collagen

Functions as a thermal insulator and shock absorber and stores fat as an energy reserve.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

When are skin substitutes needed?

A

There is a huge clinical need for skin substitues, namely in 4 instances:

  • Acute trauma (most common)
  • Chronic wounds
  • Surgery
  • Genetic diseases (bullous conditions)

Thermal trauma is one of the most common reasons for skin loss. Burns and scalds cause rapid and extensive wounds. Damaging large areas of skin can lead to death.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

What is the limitation of wound healing repair?

A

The wound healing process involves 3 stages: inflammatory response phase, fibroblastic repair phase and maturation-remodelling phase.
The remodelling of new tissue is never as good as the original, so has a detrimental to appearance, function and mechanical properties.

110
Q

What is the contracture of skin?

A

Consequence of the scarring.
During the healing process the myofibroblasts the cells which lay down collagen (phenotype similar to fibroblasts and smooth muscle cells) try to contract and close the wound. Overstimulation may lead to contracture whereby the limb cannot be moved.

111
Q

What are the 3 different types of skin wound classification?

A

Skin wounds are classified according to the depth.

  • Epidermal
  • Partial thickness (superficial or deep)
  • Full thickness
112
Q

What are epidermal injuries?

A
e.g. sunburn
Affects the epidermis
Characterised by erythema and minor pain
Do not require surgical treatment
No scarring
113
Q

What are superficial partial-thickness wounds?

A

Affect the epidermis and superficial part of the dermis.
Wound appearance: wet and weeping, red to pink blisters.
Very painful due to exposure of sensory nerves.
Heals spontaneously.

114
Q

What are deep partial-thickness wounds?

A

Involve greater dermal damage.
Wound appearance is moist white/red/pink.
Results in fewer skin appendages remaining.
Scarring is more pronounced.

115
Q

What are full thickness wounds?

A

Complete destruction of epithelial-regenerative elements.
Wound appearance is dry, leathery and rigid.
No spontaneous healing.

116
Q

How are major skin injuries usually treated?

A

1) An early excision of a dry scab (ESCHAR): dry scab contains denatured proteins that can trigger immune response. Good breeding ground for microbes.
2) Wound closure: reduces mortality from sepsis and morbidity from the scarring.
3) Skin grafts: gold standard treatment

117
Q

What are skin grafts?

A

Skin grafts are grafts of tissues of epidermis and varying amounts of dermis that is detached from its own blood supply and placed in a new area.
Types of skin graft:
-Split thickness - epidermis, some dermis
-Full thickness - epidermis, all dermis

118
Q

What are autologous skin grafts?

A

Autologous skin grafts or autografts are the gold standards for full-thickness skin wounds. Skin grafts are obtained from a non-injureed site of the patient. Obtained used a dermatome, graft may be meshed to cover a larger area.

119
Q

What is ‘graft take’?

A

Graft take refers to how the skin grafts intergrates to the new site.
Remove the graft from its old vascularisation, placed on the wound site. Initially (2-5 days) the graft will survive by diffusion of nutrients from the new site (plasmatic imbibition). Over time, the capillaries on wound bed start to grow and cause revascularisation.

120
Q

Why is preparation of the wound bed important?

A

Preparation of the wound bed is the most critical factor for a successful ‘graft take’. The graft has to adhere to the wound bed (no bleeding, infection or movement). The graft needs a thin layer of connective tissue that is vascularised.

121
Q

What are skin allografts?

A

The use of cadaveric skin for a temporary prevention of fluid loss or wound contamination.
Can be obtained from non-profit skin banks.
Possibility of pathogen transmission and immunogeneic rejection.

122
Q

Why are skin substitutes needed and what would be the ideal?

A

There is a great need for skin substitutes because there is a limited availability of skin grafts, there is often pain and scarring in the donor site area and there may be further pain inflicted on the patient.

An ideal skin substitute would be readily available, cause no immune response, cover and protect the wound, enhance the healing, lessen the pain and leave no scars.

123
Q

What are epidermal substitutes?

A

A key step is isolation of keratinocytes from a skin biopsy. Small biopsy taken from non-injured site and deattach the epidermis from the dermis. The keratinocytes are then isolated and expanded in culture. 3cm sqaured can be increased by 1000 fold after 3-4 weeks. Cultured keratinocytes are delivered onto the wound and form a new epidermal layer.
This formed cultured epithelium sheets e.g. Epicel, EPIBASE, MySkin.

124
Q

What is ‘MySkin’?

A
Celtran Ltd (Uni of Sheff spin-out company).
Subconfluent autologous keratinocytes.
Synthetic silicone delivery membrane to enable handling and manipulation.
Applications include using it to treat chronic wounds through repeated treatments - very successful.
125
Q

What are the pros and cons of epidermal substitutes?

A
  • Epidermal substitutes contain only keratinocytes grown in vitro and can be applied or sprayed onto the wound site
  • They are effective in treating chronic ulcers
  • Combination with a dermal substitute is needed to achieve full-thickness wound healing.
126
Q

What are dermal substitutes?

A
  • Provide a dermal alternative to facilitate the process of wound healing.
  • Acellular - require colonization from the wound bed.
  • After application to a prepared dermis, these substitutes are colonized and vascularized by the underlying cells.
  • 2 step process: first applying a dermal substitute followed by an epidermal cover.
127
Q

What is a Integra Dermal Regeneration Template?

A

Consists of two layers
-the dermal layer from bovine type I collagen and shark chondroitin-6-sulfate (xenogenic)
-epidermal later made of silicone (pseudoepidermis)
The collagen-chrondroitin matrix allows in-growth of cells from the wound bed and the artificial silicone epidermis regulates heat and fluid loss.
Semi-permanent as it is delivered to the wound bed, and the cells infiltrate the scaffold over 20 days. After vascularization and formation of the neodermis the membrane can be removed.

128
Q

What is ‘Alloderm’?

A

Acellular human allogenic dermal matrix preserved by freeze drying.
Used for full-thickness skin burns, alloplastic breast reconstruction, abdominal wall reconstruction, rhinoplasty.

129
Q

What are Composite substitutes?

A
  • Aim to mimic the histological structure of normal skin
  • Most of these products are based on allogeneic skin incorporated into a dermal scaffold.
  • Enable production of large batches of the products ‘off the shelf’
  • Temporary bioactive dressings: allogeneic will not last long and will be rejected.
  • Relieves pain and promotes wound healing
130
Q

What is ‘OrCel’?

A

Includes cultured allogeneic fibroblasts and heratinocytes obtained from the same neonatal foreskin.
Fibroblasts are seeded into a bovine type I collagen sponge and keratinocytes on top.
Cytokines and growth factors from the product promote host cell migration and wound healing.

131
Q

What are the limitations of current skin replacement strategies?

A
  • The average wait time ranges from 3-12 weeks after the biopsy is taken
  • Currently available composite skin substitutes use only 2 cell types: keratinocytes and fibroblasts but in normal skin there are other cells involved too (Merkel, Langerhans, melanocytes).
  • Clinical success but economic failure!
132
Q

What is Apligraf?

A

Composite skin consisiting of bovine type I collagen cultured with allogeneic male neonatal fibroblasts and keratinocytes - resembles normal skin structures.

133
Q

Give a case study that applied skin substitutes as a clinical success?

A

Hirch et al 2017
7 year old boy admitted to hospital in June 2015 with severe skin injuries (60% loss of epidermis). Suffering from epidermolysis bullosa, a genetic disorder affecting 1 in 50,000 live births. Had spontaneous (or result of minor injury) skin blistering which was painful and life-threatening due to the risk of sepsis.
High risk of skin cancer.
Had a splice site mutation in intron 14 of LAMB3 (laminin 332) causing poor attachment of epidermis to dermis. Suffered from infection from Staphylococcus and Pseudomonas.
Attempts with antibiotic treatment, topical dressings and ointments, skin transplant from father and split thick graft, but nothing worked.

134
Q

Why is there skin blistering in Epidermolysis Bullosa?

A

Epidermis and dermis are usually divided by basement membrane, but blisters form when the epidermis comes away from the dermis.
Basement membrane is a well-organised structure that contains a number of extracellular proteins. In the epidermis, keratinocytes are inserted with plectins and BPAGs. These interact with a6 B4 integrins and some types of collagens. Across the basement membrane they interact with laminin 332 and other collagens in the dermis. This usually keep them adherent, however, mutations in these components means the protein is absent or non-functional. Different groups of epidermolysis bullosa depending on non-functional protein.
German authorities allowed for a one-off compassionate treatments involving genetically modified stem cells. Proved successful, normal skin appearance and function. Procedure took 7-8 months.

135
Q

What previous research using tissue engineering in epidermolysis bullosa had been performed?

A

Mavilio et al (2006)
Italian group specialised in correcting junctional epidermolysis bullosa using genetically modified epidermal stem cells. Deemed successful in small areas. New EU regulations in GMP limited further research.

136
Q

Why would a autologous keratinocyte transplantation fail to work in epidermolysis bullosa patients?

A

Alone this wouldn’t work as there would still be no wildtype laminin produced, the basement membrane would not be adherent enough and the skin blistering would continue.
Needs a change in the genetics.

137
Q

What is gene therapy?

A

Gene therapy aims to repair or replace a mutated gene by integrating the correct gene into the patients cells.
This can be done in two ways:
EX VIVO: Cells taken from the patients, manipulated in culture and then placed back into the patient.
IN VIVO: Cells are manipulated within the body.
Vectors are used to carry the corrected gene to be integrated into the patient’s cells. This can be done using viral or non-viral (e.g. liposomes) methods.

138
Q

How was gene therapy skin transplantation performed in the boy with junctional epidermolysis bullosa?

A

They used ex vivo harvesting of cells whereby a skin biopsy from a safe area was taken and cultured on a feeder layer of fibroblasts. These were passaged and expanded over time. They are then transduced with a retroviral vector carrying the full-length of LAMB3 cDNA. The cells were passaged further so that the gene would incorporate into the patient’s DNA.
They made epidermal sheets from the cultured cells. For the first skin transplant they compared keratinocytes grown on plastic or fibrin. Fibrin performed better so this was used for second and third transplants.

139
Q

How were the new cells WT for LAMB3 tested before they were placed into the patient with EB?

A

The cells were characterised and next generation sequencing was performed to identify where the gene has integrated (exons, introns, intergenic, promotors). You can check to see if any of the other genes (tumour suppressors/oncogenes) have been disrupted which may be dangerous.
They found that very few landed in exons and none seemed to disrupt major regulatory genes so it was deemed safe to be used for the patient.

140
Q

How was the patient with gene therapy treatment for EB followed up?

A

Since this was an experimental treatment the patient was followed up by taking various biopsies at different time points to assess them for dangerous markers. Next generation sequencing, clonal analysis, immunoflurescence, histology, in situ hydridisation were all performed.

141
Q

What were the results from the follow-up tests in the patient with EB?

A

In situ hybridisation: Looking for the presence of the corrected gene. Used Cdh1 (control) and t-LAMB3 probes to see if the cells have integrated the correct gene. After 4 months post-transplant, the t-LAMB3 was present showing effective transduction.

Histology: at admission you can see the epidermis moving away from the dermis. But after the transplant, there is visible better attachment.

Immunofluorescene: Using laminin probes. At admission, the boy had no laminin332-B3 expression but 4 months post-transplantation there is good laminin-332 expression resembling the control.

Behaviour: laminin332-B3 null keratinocytes came away from the flask edge but laminin332-B3 corrected ketatinocytes stuck and required enzymatic treatment for deattachment.

142
Q

What did the gene therapy study in the EB patient reveal about the basic biology of the epidermis?

A

The epidermis has a high turnover, skin will rectify itself every month.
When placed in culture keratinocytes make 3 different clones: holoclones (clones of proliferative, undifferentiated cells), paraclones (clones of differentiated cells) and meroclones (intermediates).
The question is…
Is the epidermis renewal driven by a restricted number of long-lived stem cells (holoclones) OR equipotent progenitors?

143
Q

Is the epidermis renewal driven by a restricted number of long-lived stem cells (holoclones) OR equipotent progenitors?

A

An integration profiles of cells was performed to identify which cells were driving epidermal renewal.
The viral integration sites were mapped in holoclones and compared to the integration sites in biopsied cells (4m, 8m). Fewer integration sites were seen at 4 months than in the initial culture but many integration sites were preserved between 4 and 8 month samples. This is because most of the cells differentiated and died and the rest gave rise to differentiated cells (paraclones and meroclones). The remaining contribute to epidermis renewal (holoclones) over the long-term.

144
Q

What are the considerations for further work in gene therapy and skin transplantation?

A
  • Need to ensure a longer term follow up for the patient to avoid missing any dangerous signs. Particularly looking for skin cancer.
  • Further and wider clinical studies needed for safety and efficacy.
  • Alternative gene editing strategies could be used e.g. CRISPR
  • Does the patient’s age have an effect on efficacy? Younger patients have proliferative keratinocytes, would it work in adults?
145
Q

What are the in vitro applications for tissue engineered skin?

A
  • Disease modelling - replacements for animal models, ethical and representative. Diseases include epidermolysis bullosa, vitiligo, psoriasis, skin cancer and allergic contact dermatitis.
  • Drug discovery
  • Chemical testing - again avoids using animals

Better we can mimic in vivo the more accurate these studies/tests will be.

146
Q

How has epidermolysis bullosa been modelled?

A

Itoh et al (2011)
Disease modelling of EB where collagen 7 is lost. Somatic cells were taken from the patient and were induced to become pluripotent. They can differentiate into any cell (keratinocytes, fibroblasts etc.) WT and EB patient-specific iPSCs were generated and characterised. They were differentiated in keratinocytes and characterised.
This has the potential to be used to look for drugs that will alleviate phenotype.

147
Q

What is Psoriasis?

A

Psoriasis is a chronic inflammatory skin disease. Gives red plaques on the skin with inflamed appearance. Due to keratinocyte hyperproliferation, immune cell infiltration into epidermis and increased angiogenesis.

148
Q

How has psoriasis been modelled?

A

In vitro
2D: Use patient keratinocytes with addition of cytokines (TNFa, IL-1a, IL-17, IL-22) to mimic secretions of immune cells or via a co-culture model whereby immune cells themselves are integrated.
3D: Keratinocytes and fibroblasts are combined with the addition of cytokines to model the inflammatory response.

149
Q

What is allergic contact dermatitis?

A

15-20% of general population have ACD resulting from environmental chemical exposure. A consequence of a repeated exposure of the skin to an allergen.

150
Q

What is the future of next generation skin substitutes?

A

Need for vascularisation, immunity, pigmentation, appendages (sweat glands, hair follicles, dermal papillae), hypodermis, innervation.
To source these cells you could use tissue-specific stem cells but skin is derived from multiple embryonic germ layers so you need a common population that will give rise to all.
Pluripotent stem cells are promising.

151
Q

How have induced pluripotent stem cells been used to bioengineer an organ system?

A

Takagi et al (2016)
Fabrication of skin and its appendages could contribute to regenerative therapies. Difficult to create in vitro.
-Hypothesis: pluripotent stem cells can be used to mimic the developmental patterning.
-Experimental approach: Make embryoid bodies. Using iPS cells, cells will begin to organise/differentiate into different clusters. EBs placed in collagen gels and then transplanted further into SCID mice (in vivo organogenesis).
-Results: When treated with Wnt10, there was creation of hair which was dark, showing it was iPS cell-derived. iPS cell derived hair follicles had a correct structure and connected with local tissues.

152
Q

What was the significance of the study that used iPS to bioengineer an organ system from iPS cells?

A

The study produced fully-functional iPSC-derived explants including hair follicles and sabaceous glands with proper connections to the epithelium, dermis, fat, aneator pili muscles and nerve fibres.
A big step towards complete reconstruction of the skin.

153
Q

Where does the cornea fit in the eye’s structure?

A

Together with the other tissues of the eye, the cornea facilitates sight. The sclera and the cornea make up the spherical outer shell. The sclera covers 5/6th of the eye whereas the cornea covers the iris (1/6th).

154
Q

What is the function of the cornea?

A
  • Aiding sight: it is transparent to 99% of incident light. Has refractive power (2/3) along with the lens to focus light onto the retina.
  • Eye protection: barrier to external environment protecting against dust and microbes.
155
Q

What is the structure of the cornea?

A

Made up of three layers with 2 membranes.

  • Corneal epithelium
  • Bowman’s membrane
  • Corneal stroma
  • Descemet’s membrane
  • Corneal endothelium
156
Q

What is the structure of the corneal epithelium?

A

Outer most layer with a thickness of 50μm (10% of cornea). Highly innervated with nociceptors.
Functions include prevention of fluid loss, a barrier to pathogens and production of a rapid response to wound healing.
5-7 cell layers, entirely cellular.
Stratified and non-keratinized squamous epithelial cells.
Innermost layer is basal cells which are attached to bowman’s membrane via hemidesmosomes and to each other via desmosomes. Basal cells are mitotic and will give rise to wing cells which will give rise to superficial cells. Superficial cells have tight junctions to make the cornea impermeable to dust and glycocalyx present on microvilli to retain tea fluid on the eye.

157
Q

What is the corneal stroma?

A

Similar to the dermis of the skin. 90% of corneal thickness.
Contains collagens, proteoglycans and glycoproteins.
Relatively acellular but does contain keratocytes which are long thin flatterned cells. They synthesise and maintain ECM proteins in the stroma. To keep the shape correct and the appropriate distance between collagen fibres.
Functions:
-Provide strength, holding the shape of the eye, important for refractory power.
-Allows transparency due to the ordered collagen structure (bundled together in lamella). Distance between them and their location is important.

158
Q

What is the corneal endothelium?

A

A single layer of cells.
Acts to maintain the stromal hydration. Can’t be too hydrated or the collagen fibre structure will be disrupted by the water molecules, causing swelling.
Acts as a leaky pump, allows solutes and nutrients in to stroma from aqueous humor but also pumps out water from stroma using various channels and pumps.
Highly metabolically demanding process.

159
Q

How is the cornea innervated?

A

The cornea is the most densely innervated body structure. Filled with nociceptor endings from the ophthalmic branch of the trigeminal nerve. Important for blink reflex, wound healing and tear production.

160
Q

How is the cornea vascularised?

A

It is AVASCULAR
If it were to have blood vessels, it would no longer be transparent. For the diffusion of oxygen and delivery of nutrients the tear layer and transport from the humor is important.

161
Q

What are the four factors contributing to transparency?

A
  • The cornea is avascular
  • Collagen is organised into a specific way in the stroma
  • The stroma is kept in a dehydrated state
  • Very few cells present in the stroma and certainly no pigmented cells.
162
Q

What is corneal injury/disease and how is it treated?

A

Corneal disease/injury is the second leading cause of vision loss (first in bacterial infection in 3rd world countries).
10 million people suffer from vision loss due to corneal disease (e.g. trachoma) or injury (e.g. acid/base burns).
Traditionally treated by corneal transplantation whereby the cornea is exised and a new, usually cadaveric is sutured in to replace faulty version.
But there may be immune rejection, viral/disease transmission and the sourcing/logistics of storing and transporting corneas is difficult.
Need for regenerative therapy that will completely restore function of the eye.

163
Q

What is keratoprosthesis?

A

A synthetic medical device known as keratoprosthesis.
Used for patients with repeated failed organs.
Require life-long regime of antibiotics.
Medications needed to control inflammation and glaucoma (due to uncontrolled pressure in the eye).
Need for regenerative therapy that will completely restore function of the eye.

164
Q

What are limbal epithelial stem cells?

A

If the cornea epithelium is injured it can regenerate quite well without scarring. This is due to the limbal epithelial stem cells. They are located on the corneal rim at the border between the sclera and the cornea. Found in undulated niches.
Have the ability to undergo asymmetric division, some will stay in the limbus and others will give rise to daughter cells. These will migrate outwards and give rise to trans-amplifying cells (divide rapidly in the basal layer), undergo differentiation and become post-mitotic (wing cell layer) or terminally differentiated cells (squamous cells).

165
Q

What is limbal stem cell deficiency?

A

When numbers of limbal stem cells fall below a certain threshold due to injury/disease. Unable to regenerate.
May be due to congenital conditions such as aniridia or internal/external injury.
In the absence of LSCs cells from conjunctiva migrate to the cornea and create opaque and vascularised tissue (vision loss).

166
Q

How is limbal stem cell deficiency treated?

A

If the corneal disease is unilateral the easiest approach is to take part of the limbus from the other eye and transplant it directly or expand it to create a corneal epithelial sheet on amniotic layer/synthetic scaffold.

167
Q

How has research revealed limbal stem cell therapy?

A

Pellegrini et al 1997) performed studies using expanded limbal stem cells which demonstrated that the ocular surface in patients with ocular burns could be restored.

2015: Approval for stem cell therapy using limbal stem cells was given in Europe. Holoclar was developed.

168
Q

What is Holoclar?

A

Stem cell therapy using limbal stem cells.
Patient’s healthy eye has the limbal area explanted and shipped to a production facility whereby cells are cultures and expanded. Can be frozen at this stage or used directly. Cell culture is placed in a fibrin matrix to help deliver the cells to the patient.

169
Q

Is Holoclar successful?

A

75 out of 104 patients studied showed:

  • Stable corneal surface with no surface defects
  • Little or no ingrown blood vessels
  • Reductions in pain and inflammation
  • Improvements in vision

No immune rejection issues because it is an autologous approach.
But if both eyes are diseased, it cannot help. And you may damage the eye from which you are removing the limbal cells.

170
Q

What is an alternative source of cells for corneal epithelium transplantation?

A

In cases when autologous sourcing of limbal stem cells is not possible you can take cells from the muscosa tissue.
It doesn’t cause scarring in the eye or the mucosa but there is a risk of neovascularization of the cornea.

171
Q

What happens when the stroma is injured?

A

When collagen fibrils are disrupted they are very hard to recreate. When the stroma is injured the wound healing process begins. The keratocytes will differentiate into fibroblasts which will become myofibroblasts to close the wound. Then they should undergo apoptosis be in some conditions they become overactive leading to scarring. Cornea will no longer be transparent.

172
Q

How can biomaterials be used in stromal replacement?

A

They must be acellular at the time of implantation.
They are expected to promote repopulation by the host’s cells and reinnervation.
A range of materials have been tested for this purpose including decellularised cornea, collagen and recombinant collagen and self-assembling peptides.

173
Q

How did researcher use recombinant collagen in stromal replacement?

A

Fagerholm et al (2014)
Produced recombinant human collagen cell-free implants. Showed successful endogenous cell recruitment, regenerated neo-corneas that were stably integrated and nerve and stromal cell repopulation. There is no need for immunosuppression.
Visual acuity could be improved by using better materials and the scaffold didn’t have appropriate mechanical properties.

Islam et al (2018)
Used recombinant human collagen reinforced with a synthetic lipid. Tested in pigs and in 7 patients.
Implants were shown to improve vision and relieve pain.

174
Q

How can a cell-based approach be used in stromal replacement?

A

Take cells that usually make stroma to secrete a deposit ECM proteins. ECM sheet are produced populated with stromal cells. Cells secrete their own ECM that can be remodelled. Can be used with autologous cells. Allows for cell amplification and relatively large amounts of the fabricated tissue.
Obvious source of cells would be keratocytes. They can be isolated from the stroma, amplified in vitro, in some cases they will change their phenotype to become more fibroblastic but under serum and ascorbic acid they will lay down ECm proteins.
Others will change from keratocytes to fibroblasts to keratocyte-like cells.
Or you can use stem cells.

175
Q

What are the challenges in endothelial regeneration?

A

-Human corneal endothelium does not regenerate!
-Endothelial cells have a finite life span leading to a decrease in density with age. At birth 3500-4000 cells/mm2 but by 85 only have 2300 cells/mm2. Necessary to have 500 cells/mm2 to be functional.
-If endothelial cells are put in culture they don’t regenerate and if they are immortalised they lose their phenotype which has implications for research and clinical use.
Recent development suggests that endothelial cells may be able to be derived from pluripotent stem cells.

176
Q

What should a corneal substitute be?

A

It should allow for adequate mass transport, prevent angiogenesis, have tensile strength/mechanical properties, have low immunogenicity, support nerve regrowth.
For epithelium: continuous replacement of cells, maintenance of barrier integrity and optical transparency.
For stroma: high tensile strength, optical transparency.

177
Q

How can the cornea be re-innervated?

A

Patients with corneal transplants have limited re-innervation yet innervation is essential for normal function of cornea.
Can we use growth factors to encourage re-innervation?

178
Q

What is the incidence and impact of peripheral nerve injuries?

A

9000 cases in the UK per year
Mainly in young people (working population)
Main cause in peace time is car accidents.
Brings financial, healthcare and social burden.

179
Q

What is the nervous system?

A

The nervous system is divided into central nervous system (brain and spinal cord) and peripheral nervous system (paired, cranial, spinal nerves) which stem from the CNS and relay electrical signals to and from.
Cellular composition includes neurons and glial supporting cells but it varies across the CNS and PNS.

180
Q

What is the structure of the peripheral nerve?

A

Axons are surrounded by mylinating Schwann cells and are enclosed by endoneurium.
Individual axons are bound together by perineurium to form fascicles.

181
Q

What are the causes of peripheral nerve injury?

A
  • Elongation: the connective tissue of nerves allows 10-20% elongation before structural damage occurs. Severe lesions can disrupt the axon.
  • Laceration: 30% of nerve injuries
  • Compression: external mechanical pressure on the conductive membrane. If it’s persistent it leads to nerve injury.
182
Q

What are the grades of peripheral nerve injury?

A

Seddon, 1942
NEUROPRAXIA
No/little structural damage, no loss of nerve continuity. Symptoms are transient and reversible. Includes entrapment neuropathies like carpal tunnel syndrome and ‘Saturday Night Palsy’ (radial nerve).
AXONOTMESIS
Complete interruption of the axon and its myelin sheath.
Perineurium and epineurium remain intact.
NEUROTMESIS
Nerve and the surrounding stroma are completely disconnected. No spontaneous recovery. Weakness, atrophy and poor prognosis.

183
Q

What is Wallerian degeneration?

A

When axons are severed, axon distal to site of injury are now disconnected to the cell body and will begin to degenerate due to proteases and metabolic activity disconnect. This leads to cell debris which is cleared up by macrophages. Schwann cells take up regenerative phenotype. They proliferate and align into tracts called bands of Bungrer. They serve as a conduit for growing axons. Takes about 1-3mm a day.

184
Q

How is regeneration different in CNS and PNS?

A

Macrophages are crucial in nerve regeneration but in CNS macrophages infiltrate much more slowly because there is a blood spine barrier that macrophages cannot infiltrate. This delays the removal of inhibitory myelin. Astrocytes become ‘reactive astrocytes’ which produce glial scars that inhibit regeneration.
-In PNS: repair of damage is actively promoted
-In CNS: repair of damage is inhibited
CNS and PNS require different regenerative medicine strategies. More success in PNS.

185
Q

What are the approaches used to tackle neural tissue repair in PNS?

A

Surgical reconstruction
Grafts
Nerve conduits

186
Q

How is surgical reconstruction used in PNS tissue repair?

A

Surgical reconstruction has been used historically. Suturing the stumps of the nerves back together. Can only be used if the stumps are proximal, if the gap is larger than 1mm, it cannot be used because it will cause elongation injury.
Blood flow reduces by 50% when the nerve is stretched 8%, complete ichaemia is caused at 15%.

187
Q

How are grafts used in PNS tissue repair?

A

When the gap between the nerve stumps is too big, the gold standard are grafts.
Can be autologous: a nerve is taken from somewhere else in the patient’s body (usually sensory nerve). Gives low risk of immune rejection but causes loss of function at the donor site, two surgeries are required and it is limited by the size and type of nerve they can use.
Or can be allogeneic which is from the same species but different individual. Good because there is no secondary surgery and no loss of function at donor site. However, there is a higher risk of rejection and limited availability in cadavers.

188
Q

How are nerve conduits in PNS tissue repair?

A

Nerve conduits are devices where 2 stumps of a severed nerve are placed inside a conduit. It guides regenerating axons, prevents infiltration of scar tissue and increases concentration of intraluminal proteins.
Conduit fills with plasma from the severed nerves (hours). The plasma is filled with fibrin precursoes which will form a fibrin cable. There is promoted proliferation and migration. The resulting tissue is notably thinner. Can take months/years.

189
Q

What are the ideal properties for a nerve conduits?

A
  • Good mechanical strength to prevent collapse
  • Not too tough, shouldn’t affect function or surrounding soft tissue
  • Porous to nutrients and oxygen, prevents fibroblasts entering
  • Rate of degradation should match tissue
  • No immunogeneiticty
  • Bio-compatible
  • Suturable
  • Able to be sterilised to avoid bacterial infection
190
Q

How are decellularised nerve conduits used?

A

“Top down approach”
Take a nerve which already exists in nature and stripping it down to its bare bones.
Complete scaffold for the physical properties as seen in vivo. Aim is to retain the ECM architecture and remove antigens. Less immunogenic and hollow compared to an allograft.
-Provides a 3D scaffold to support nerve regeneration
-Clean pathways allow cell migration and axonal regeneration
-Axon regeneration is well distributed through nerve thickness
-Functional incorporation of the nerve conduit

Off the shelf: ‘Avance’

191
Q

What can be used to make a nerve conduit?

A
  • Decellularised nerve conduits

- Fabricated (biological, semi-synthetic, synthetic)

192
Q

How can nerve conduits be bioengineered?

A

“Bottom up approach”

Use biomaterials to fabricate scaffold. We are in control of production, more flexibility to change biodegradability/porosity, biochemical signals, incorporation of support cells, electrical activity, intraluminnal channels and oriented nerve substratum.

193
Q

What natural and synthetic materials have been used to engineer nerve conduits?

A

Many natural materials have been used e.g. chitosan, collagen, fibrin, fibronectin, gelatin, keratin and silk fibroin (biodegradable, biocompatible, structural integrity).

Synthetic materials can also be used.
Biodegradable materials like poly(lactic) acid, poly(lactic-co-glycolic) acid, poly(caprolactone), poly(ethylene glycol). Electrically active materials which can contribute to the function of the nerve like Piezoelectric.
Non-biodegradable like silicone and Gore-tex can be used but must be removed from the body.

194
Q

What are length limitations of conduits?

A

The chance of successful regeneration with nerve guides is reduced once an injury gap reaches a certain value.
At short gap lengths, the fibrin cable is robust enough to provide a platform for regeneration.
At longer lengths thinning restricts regeneration.
No fibrin cable at large lengths.

195
Q

What is the critical gap length?

A

Length at which regeneration occurs 50% of the time.
Mouse 5mm
Rat 10mm

196
Q

What are the different approaches to try and increase the critical gap length?

A
Aim is to bridge this gap so it is no longer an issue using...
-ECM components
-Intraluminal support
-Cell grafts
-Neurotrophic factors
Or combinatorial approach
197
Q

How are ECM components being used to increase the critical gap length?

A
  • Matrices generally increase the critical gap length
  • Effective materials are weak, visoelastic hydrogels with a high water content (high concentrations would prevent axonal penetration).
  • Matrigel promotes nerve regeneration but not suitable for clinic because it comes from mouse sarcoma cells
  • Laminin, fibronectin and collagen involved
198
Q

How is intraluminal support being used to increase the critical gap length?

A

Relies on scaffold generation of conduit to have intraluminal guidance, micro-grooved luminal design, electrospun fibrous outer conduit, variations in conduit design, surface functionalisation and combinatorial approaches.

199
Q

How are neurotrophic factors being used to increase the critical gap length?

A

Neurotrophic factors can be placed within the tube.
-Support axonal growth
-Migration and proliferation of Schwann cells
-Increase neuroprotection modulation of intrinsic signalling pathways
Nerve growth factors and Neurotrophin-3 show potential.

200
Q

How can gradients be used to provide neurotrophic factors to increase the critical gap length?

A

Challenge is to release neurotrophic factors in a controlled way to avoid dissipation. Could use a polymer to release them which will then biodegrade.
Means of delivery could include diffusion-based release, suspension, affinity-based delivery, microsphere encapsulation.
Gradients rather than isotropic presence.
Regenerative medicine is trying to recrete gradient of laminin 1 and growth factors across the nerve conduits to guide axon growth.

201
Q

How can cell grafts be used to increase the critical gap length?

A

Can deliver cell grafts with the nerve conduits.
May be neurotrophic factors, schwann cells, differentiated or undifferentiated stem cells or geneticallt modified cells. All experimental.
Schwann cells are critical for sucessful nerve regeneration could be delivered along with nerve conduits. They create bands of bungner, secrete neurotrophic factors and proliferate (4-7x cells).

202
Q

What special nerve conduit have Yeh et al (2017) created?

A

Development of biometric micro-patterned device incorporated with neurotrophic gradient and schwann cells.

  • Used a micro-patterned surface that can directionally guide the axon as physical cue.
  • Neurotrophic gradient membrane that can continually attract axon outgrowth from proximal to distal stump as chemical cue.
  • Schwann cells that can support the growth of neurite and form myelin sheath around axon as biological cue.
203
Q

What are some examples of more complex tissue constructs and how they are created?

A

Blood vessels.
Upper airway tubes like trachea and larynx.
Urogenital tissues.
Most will have a layer of endothelium, a layer of muscle, connective tissue.
Best approach so far has been to used existing organs, decellularise them and then recellularise with patient’s autologous cells, and then transplant back in.

204
Q

What are the bottlenecks hindering the success of tissue construct transplants in vivo clinically?

A

Biological bottlenecks
Vascularization
-In vitro: nutrients and oxygen flow through the media
-In vivo: must have an immediate blood supply or necrosis
Host response: inflammatory and immune response

Regulation and commercialisation.

205
Q

What is the role of vascularization in tissue engineering?

A

Most tissues in the body contain a vasculatory network.
Most cells are located 100-200um from capillaries but there are differences in cells’ sensitivity to oxygenation. Islet cells are particularly sensitive whereas cartilage cells can maintain viability even in 1mm TE construct.
In vitro this is overcome by using bioreactors.
-Avoid graft necrosis
-Generate thicker tissues
-Help graft innveration
-Improve graft function

206
Q

Why is vascularization required in tissues?

A

Folkman (1971)
Hypothesis: Tumour growth is angiogenesis-dependant.
Patient with retinoblastoma with a highly vascularized tumour. Tumour had a number of metastases with about 1mm diameter. Metastases only contained a rim of viable cells on the outside but central cells were necrotic due to a lack of vascularization.
Therefore tissues require vascularization when diffusion is no longer sufficient.

207
Q

How are blood vessels structured?

A

There are macrovessels (arteries and veins), microvessels (arterioles and venules) and capillaries which mediate the exchange of nutrients due to differences between blood pressure and interstitial fluid.

Thick layer of connective tissue surrounding the vessel, several layers of smooth muscle. Inner layer is single layer of endothelium, lining the lumen separated from the SM by basal lamina.
Exact composition depends on anatomy and function.
e.g. capillaries only contain endothelium surrounded by pericytes.

208
Q

What are the different types of blood vessel formation?

A
  • Vasculogenesis: de novo blood vessel formation that occurs in embryogenesis, formation from progenitor cells.
  • Angiogenesis: new blood vessel formation via extension/remodelling of existing blood vessels.
  • Arterogenesis: maturation of blood vessels via increasing in the lumen.
209
Q

What is vasculogenesis?

A

Embryo is small so oxygen and nutrients can diffuse to support the life of cells. When embryo reaches a certain size it requires vascularization. Mesoderm produces precursors called Hemangioblasts. These differentiate and create tubes which create a primary capillary plexus.

210
Q

What is angiogenesis?

A

Sprouting new blood vessels from the existing ones. The major drive of this process is hypoxia.
Hypoxia-induced factors (TF) will initiate cascade of evenets to lead to new blood vessels sprouting.
Angiogenesis stimulus VEGF is produced and signals from hypoxic area to endothelial cells. Endothelial cells become active and secrete enzymes like matrix metalloproteinases. These enzymes digest the basal lamina and create a hole. Endothelial cells will start moving through the holes and create vascular sprouts. These will proliferate and moce towards VEGF signal. Lumen forms and recruitment of pericytes and SMC allows it to mature. Blood starts flowing through and oxygen is delivered.

Physiologically takes place in wound healing and ovarian cycle.

211
Q

What are some of the most important factors in angiogenesis?

A
  • Vascular Endothelial Growth Factor (VEGF) promotes endothelial cell proliferation, migration and differentiation.
  • Hypoxia inducible factor (HIF) stimulates transcription of VEGF, PDGFR, TGF-a, EGF and erythropoietin.
  • Platelet derived growth factor (PDGF) recruits and encourages proliferation of pericytes and SMCs.
  • Angiopoietin regulates endothelial cell survival, sprouting, pericyte recruitment.
  • Matrix metalloproteinases. Basal lamina degrades, ECM remodelling.
212
Q

What is VEGF?

A

Family of growth factors with preferences for different receptors.
VEGF-A, VEGF-B, PlGF will preferentially bind to VEGFR-1 and are involved in vasculogenesis, Whereas VEGF-C and VEGF-D will bind to VEGFR2 and important for angiogenesis.

213
Q

What is aterogenesis?

A

Blood vessels remodelling as a response to fluid shear stress (can be caused by occlusion).
Increase in shear stress causes endothelial cells to release GFs (like TGF-B).
Causes proliferation of endothelial and smooth muscle cells and matrix remodelling.

214
Q

What are the two main approaches for vascularization of 3D contructs?

A
  • Strategies for facilitating vascular ingrowth (scaffold design or functionalization)
  • Prevascularization (in vitro or in vivo)
215
Q

How did Richardson et al (2003) initate angiogenesis?

A

Background: angiogenesis is a multistep process. VEGF is the initiator of angiogenesis but not sufficient to induce mature vessels. PDGF promotes maturation of blood vessels.
Hypothesis: Dual delivery of VEGF and PDGF can direct the formation of a mature vasculature.
VEGF is mixed into a scaffold and released to stimulate the growth of immature vessels.
PDGF is encapsulated into PLG microspheres to facilitate maturation of nascent vessels.
SUCCESSFULLY creates a mature vascular network.

216
Q

What are the disadvantages of using dual growth factor delivery using polymers?

A

Time-consuming process because microvessel growth rate i about 5um/h.
May not be sufficient to prevent necrosis in 3D contructs after implantation.

217
Q

What occurs in in vitro prevascularisation?

A

The TE constructs is cultured in vitro to build prevascularized structure with endothelial cells. The perivascular network can create a connection with the existing blood vessels in the tissue (transplant site) by anastomising. Endothelial cells spontaneously self-assemble into capillary-like structures.
This is faster than new blood vessels formation but is not as mature or well-spaced as would like. Also issues with where the cells will be sourced from.

218
Q

What occurs in in vivo prevascularisation?

A

A scaffold is implanted into easily accessible and well vascularized tissue. Subcutaneous tissue or muscles pouches for example.
Microvessels ingrow from the host.
After vascularization the implant is transferred to the defect site.
Requires 3 surgeries.

219
Q

What is the flap technique?

A

Prevascularization method.
A scaffold is implanted into a muscle flap.
Microvessels ingrowth from the host.
After vascularization the entire flap is transferred to the site in need of repair. The vascular pedicle of the flap is surgically anastomosed to host vessels.
Major morbidity to the donor site.

220
Q

What is the AV loop technique?

A

Arteriovenous loop technique.
Uses a vein or synthetic graft to form a shunt loop between an artery and vein. Protected with a plastic chamber.
AV loop leads to spontaneous sprouting of vessels. When vascularized it can be transferred to the repair site. Allows tissue vascularization but the tissue is not embedded in the surrounded muscle tissue. No major morbidity at the donor site.

221
Q

What are the advantages and disadvantages of scaffold design in vascularization?

A
  • Versatile strength, easy to develop, easy to translocate to multiple tissues.
  • Relies on vessel ingrowth, limited result if used alone, can introduce cell seeding problems.
222
Q

What are the advantages and disadvantages of in vitro prevascularization?

A
  • Doesn’t rely on ingrowth, no extra surgery

- Complex strategy, varies from tissue to tissue, vessel maturation in vitro needs attention, anastomosis is slower.

223
Q

What are the advantages and disadvantages of in vivo prevascularization?

A
  • Direct perfusion after microsurgery, mature and organised vasculature
  • Extra implantation/surgery, identification of location with vascular axis, scaffold might be filled with fibrosis tissue.
224
Q

What are the advantages and disadvantages of angiogenic factor delivery for vascularization?

A
  • Angiogenic factors have been effective, possibility to tune vascularization
  • Still relies on vessel ingrowth, factors might have a negative effect, release profile of these factors is critical.
225
Q

What is the host response?

A

Host response is very complex and occurs at various levels.
Local interactions: how the TE construct interacts with the local environment.
Systemic interactions: through blood and immune system, it can communicate with the rest of the body.
Outcome of both of these determines the functional integration or complications of the TE construct.

Body responds to biomaterial via the inflammatory response and to cells via the immune response. The is known because of the response to medical devices and cell replacement strategies.
There is cross-talk between them.

226
Q

What are the ways in which the body and the biomaterial can interact with each other?

A

The effects of biomaterial on body include changes to wound healing, infection, toxicity, tumourigenicity, particle embolization, hypersensitivity reactions.
Effects of body on biomaterial include enzymatic degradation, calcification, abrasion and corrosion.

227
Q

What is the wound healing response?

A

3 major phases: inflammatory, proliferative, remodelling.
Cute blood vessels bleed into the wound, blood clot forms and leukocytes clean the wound. Blood vessels regrow and granulation tissue forms, epithelium regnerate and scar forms. If the stimulus is removed (acute) cell death occurs but regeneration and repair can occur. If the wound is persistent (chronic) fibrosis occurs leading to a tissue scar.
In TE construct there is a response similar to wound healing.

228
Q

What is the foreign body response?

A

Similar to wound healing.
Implantation of TE construct injures the tissue causing a flow of blood to enter. Proteins from the blood will coat the surface of biomaterial. This creates a transient matrix, which serves as an attachment site, upon which neutrophils attach first. Monocytes also attach and differentiate into macrophages. Macrophages cannot clear the debris because biomaterial is too large. Cue frustrated phagocytosis

229
Q

What is the foreign body response?

A

Similar to wound healing.
Implantation of TE construct injures the tissue causing a flow of blood to enter. Proteins from the blood will coat the surface of biomaterial. This creates a transient matrix, which serves as an attachment site, upon which neutrophils attach first. Monocytes also attach and differentiate into macrophages. Macrophages cannot clear the debris because biomaterial is too large. Cue frustrated phagocytosis. This leads to macrophages fusing creating foreign body giants. If the material is still too big, they can secrete ROS and other enzymes which can degrade the surface of the material.

230
Q

What is fibrous encapsulation?

A

Tissue response to implanted biomaterials.
Abundant deposition of extracellular matrix/collagen.
Isolation of biomaterial from local tissue environment. This hinders tissue construct integration - cannot interact with surrounding tissues.

231
Q

How can fibrous encapsulation effect TE devices?

A

Implanted microelectrodes are implanted to intiate transmission of electrical signals. Encapsulation prevent this transmission.
Vascularization cannot penetrate fibrous capsule.
Diffusion barrier - some devices rely on their interaction with blood to measure levels but encapsulation prevents this.

232
Q

How can TE devices cause infection?

A

Very common when related to medical devices. Joint prostheses have fewer infections.
Initially localized to the implant site, but may cause sepsis.
When the skin barrier it’s easy to see how bacterial infection can occur. But in urinary catheters, the skin barrier is not broken but bacterial infections still occur. When coated with antibiotics (didn’t reach systemic) the bacterial infections decreased. Suggested bacterial biofilms.

233
Q

What are bacterial biofilms?

A

Communities of bacteria grown on surfaces of abiotic materials and host tissues. The bacteria embed themselves in a matrix known as ‘extracellular polymeric substance’ or aka slime. Ancient adaptation. Allows for coordinated behaviour and enhanced survival at the population level.

234
Q

How do bacterial biofilms develop?

A

Planktonic bacteria begin to attach to surfaces, secrete slime and proliferate. The adhesion becomes irreversible. They also maturate, causing some of the bacteria to leave the colony, become planktonic and establishing new attachments.

235
Q

What are some examples of biofilm infections?

A

Staphylococci are particularly proven to create biofilms. Worldwide there is an increas ein multidrug-resistant methicillin-resistant strains. S. aureus (MRSA) and S.epidermis (MRSE).
Biofilm infections can occur on surgical repair mechanisms (staples, sutures, mashes).
Can occur on orthopedic prosthetics. May take months to develop and difficult to access.

236
Q

How do cells interact with the surface of biomaterials? What is ‘the race to the surface’?

A

Cells do not interact directly with materials. A layer of protein (from growth media or plasma) adheres to the surface. Protein adsorption is affected by the surface properties of the material.
The race to the surface refers to whether the cells or the bacteria can make it to the surface first. If there are cells already attached to the surface there is less room for the bacterial cells to attach or create biofilms.

237
Q

What are non-fouling surfaces?

A

Surfaces that resist adsorption of proteins and/pr adhesion of cells. Poly(ethyl glycol) and zwitterionic polymers. In medical devices, inhibits bacterial colonization.
To functionalize, add RGD so cells can adhere.

238
Q

What are differentially instructive matrices?

A

Using peptides that can self-assemble into 3D network. These matrices allow attachement of mammalian cells but not bacterial cells due to topology. Labelled bacterial cells have been shown not to ‘SaNet’.

239
Q

What are systemic and remote effects of medical devices?

A

By-products of physical/chemical wear can enter the bloodstream and cause side-effects in other parts of the body.
Example: metal-on-metal hip replacements, releasing colbalt and chromium in the blood. Can bind to proteins and present to the immune system.

240
Q

What are thromboembolic complications?

A

Exposure of blood to an artificial material can cause thrombosis, embolization and consumption of platelets and plasma coagulation.
e.g. stent thrombosis
Blood in contact with metal stent. Rare but serious complication. Death (20-48%) // MI (60-70%)
Combatted with anticoagulant drugs clinically.

241
Q

How can tumours be linked to TE implants?

A

Has not been experimentally proven but it may be the case that you can say that tumours developing in the region of the medical device may be causal, althought no evidence. Mechanisms not clearly understood.

242
Q

What are the mechanisms of immune response?

A
  • Humoral immunity: mediated by soluble antibodies produced by B lymphocytes.
  • Cellular immunity: mediated by T lymphocytes
243
Q

What are strategies for immunomodulation?

A
  • Topography: some will attach others won’t
  • Stiffness: influences cell behaviour
  • Non-fouling coatings
  • Functionalization with biomimetic coatings
  • Delivery of anti-inflammatory agents
  • Encapsulation of anti-inflammatory agents
  • Gene delivery systems: modifying cells to secrete certain factors
  • The use of embedded immune cells secreting pro-angiogenic and pro-regenerative cytokines
244
Q

How can immunoisolation as a TE stragery for treatment of type 1 diabetes?

A

Type 1 diabetes is an autoimmune disease where there is a loss of insulin-secreting islet cells. The Edmonton protocol shows sucessful islet transplantation from cadavers, but there is a need for immunosuppression. Immunoisolation of islet cells could give the secretion of insulin using a selectively permeable membrane, wouldn’t require immunosuppression.

245
Q

What is Viacyte doing?

A

Performing clinical trials using immunoisolation of islet cells to treat type 1 diabetes. Using human embryonic stem cells.
Active area of research with lots of different approaches.

246
Q

What are other applications of immunoisolation?

A
  • Hypopituitarism
  • Geneticallt engineering cells to secrete neurotrophic or angiogenic factors
  • Enhancing tumour-specific cellular immunity
247
Q

Which incidences contributed to the establishing of a regulatory framework?

A
  • Elixir Sulfanilamide incident (1937): Used for a Staphylococcus infection. Developed for children and added raspberry flavour but was toxic to those who were given the drug (aka antifreeze).
  • The Nuremberg Code (1946): Volunteers must consent to testing.
  • The Softenon (thalidomide) incident (1956). Mothers given drug for morning sickness but babies born with deformed limbs. Need for analysis of long-term.

This led to the Declaration of Helsinki (1964): A statement of ethical principles for medical research involving human subjects, including research on identifiable human material and data.

248
Q

What is the process of conventional drug development?

A
  • Discovery: in research lab in academia/pharma shows safety and promise of drug
  • Pre-clinical: done in vitro in suitable assays, then animal models
  • CT Phase I: small group of healthy people. Looking at safety.
  • CT Phase II: larger group. Looking at efficiency and safety.
  • CT Phase III: much larger group. If there is an existing drug will involve comparison to this. Risk/benefit ratios.
  • Authorization
249
Q

What are the regulatory agencies?

A
  • European Medicines Agency (EMA) which includes the EMA Committee for Advanced Therapies
  • Food and Drug Administration (FDA) in USA
  • Ministry of Health, Labour and Welfare in Japan

To unit these: The International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for human use.

250
Q

What are ATMP?

A

Advanced Therapies Medicinal Products
Which is either:
-A gene therapy medicinal product
-A somatic cell therapy medicinal product
-A tissue engineered product (also a combination product because it contains cells and scaffold)

251
Q

What are the major concerns related to the use of cells in a TE product?

A
  • May survive in the tissue potentially for the lifetime of the recipient
  • May respond to their environment and interact with other cells and humoral factors in ways which are not encounted with other medicines
  • May have the capacity to replicate or mature in vivo thus evolving their functionality after they have been administered to the patient
  • May migrate and distribute in tissues or organs other than the one intended
  • Cells must be free from microbiological contamination and be of sufficient identity, purity and potency
  • Difficult to sterilize
  • Must have good storage protocols (master/working cell banks)
252
Q

Why are the regulatory requirements for TE products so complex?

A

No standard set of preclinical tests.
The diversity and inherent properties of TE products require a case by case consideration.
An overarching set of general considerations available.

253
Q

What are the issues surrounding product consistency of TE products?

A
Since the product has been regulated safely you must adhere to this protocol each time it is made to avoid risks. Since TE is biological, cells can change, age and have genetic changes.
In scaffolds, must consider:
-Material sourcing
Adventitious agents
-Toxicity of process additives
-Sterility and Sterilization
254
Q

Give an example of a TE products and its journey through clinical trials?

A

Using embryonic stem cell-derived retinal pigment epithelium patches in Macular degeneration.

  • Engineering of RPE patch: pluripotent stem cell culture allowed to spontaneously differentiate. RPE cells are exised and expanded and placed on a scaffold. At every stage there are tests of appearance and the karotype.
  • Preclinical trials: Considerations of safety, functionality, engraftment, immune response in animal models. Was tested in pigs due to human eye similarity.
  • Clinical trials: Phase I not appropriate to test on healthy volunteers due to surgery, instead this is a very small exploratory trial.
255
Q

Why is randomisation important in clinical trials?

A
  • A tool ensuring that the situation at the start of the trial is identical in different groups studied (the treatment group and comparator)
  • Every participant has the same chance of receiving the treatment studied
  • Standardized and indepdent of the investigator
  • Issues/solutions for TE products - double blind trials are very difficult when administration involves surgery
256
Q

How does standardization of patient care occur following a clinical trial?

A

Important for determining the efficacy.
Either one person follows up all the patients or all care givers should be appropriately trained.
The trial should be designed to minimize the effect of the procedure on patient care.

257
Q

What is hospital exemption?

A

When treatment is given before it has been clinical trials. ATMPs exempt from the centralized marketing authorization procedure. Individual case studies based on risk-benefit assessment.

258
Q

Why do few TE products make it to the clinic?

A

The valley of death is the gap between discoveries in the lab and therapeutic leads that enter clinical trials. Research often doesn’t leave academia because big pharma companies are adverse to the big risk.

  • Financial rewards are not guaranteed
  • Case by case regulatory pathway
  • Manufacturing and scale-up is complex and expensive
  • Distribution and storage are complicated by the biological nature of the products
  • No unique business model
259
Q

Give an example of a TE company that tried to launch its products into the market?

A

1990s Advanced Tissue Sciences made products such as Transcyte and Dermagraft. Funded by enthusiastic researchers, promising market for burns victims and products were praised by doctors. But ATS never made a profit and went bankrupt in 2002.
Started clinical trials for Dermagraft in 1991 and applied for FDA approval as a medical devise. Expected approval in 1995 but unanticipated delays meant that FDA did not approve until 2001 - could not recover.

260
Q

What mistakes and lessons can we learn from ATS?

A

Mistakes:
-Regulatory approval and caused delay in releasing product to market
-Manufacturing was challenging
-Funding was raised to build a manufacturing facility which could not be paid off
Lessons:
-Management: financial planning, R and D costs, clinical trials costs, strategic focus
-Regulators: importance of communication
-Investors: reg. med. therapies are high risk investment

261
Q

What are the bottlenecks for tissue engineering becoming a mainstream product?

A
  • Biological bottlenecks: vascularization and host response, difficult to become functionally integrated
  • Complex regulatory/commercialisation landscape, less well-established, expensive, risky, lengthy
262
Q

Why might we need to engineer food?

A

World’s population is rapidly increasing - expected to reach 10 billion by 2050.
Meat is the most valuable livestock product since it contains all the essential amino acids, highly biodegradable minerals and vitamins. Incredibly resource-dependent and environmentally taxing. 24% greenhouse gases, 37% landmass, 70% water withdrawal.
Animals are farmed close together, giving a breeding ground for diseases and antibiotic resistance.
Current meat production is unsustainable - need for engineered food.

263
Q

What is the cultured meat concept?

A

Churchill in 1932 proposed that we would be able to grow parts of a chicken rather than growing whole ones and wasting most of it.
NASA cultured muscle tissues from the common goldfish in 2002 (Benjamin et al).
First lab-grown burger in 2013, PR opened the world’s eyes to this possibility.

264
Q

What can we learn from muscle that could be manipulated in TE food?

A

Myofibres bundle into muscle fibers which bundle into fascicles which are attached to bone via ligaments. Satellite cells are quiescent cells (low-turnover) and regenerate muscles upon injury. In this instance they become activated and proliferate. Give rise to myoblasts which develop into myotubes leading to de novo myofibers.
Satellite cells are used in TE because they have the ability to proliferate and have well-established differentiation cues.

265
Q

How is TE muscle for food created?

A

Take an adult biopsy from an animal and isolate satellite cells using enzymatic treatment to remove other things.
Culture the cells in a proliferative phase. 30 doublings max (not infinite). The medium is not chemically defined and you’re still using serum from animals.
Then use known signals/factors/transcription factors to induce differentiation into appropriate cell.

266
Q

What are some alternative cell sources for cultured meat production?

A
  • Embryonic or induced pluripotent stem cells: unlimited proliferation but need for differentiation optimization.
  • Alternative adult stem cells e.g. mesenchymal stem cells
267
Q

What must the scaffolds for cultured meat be like?

A
  • Edible and non-toxic material
  • Non-allogenic biomaterial
  • Supportive of full differentiation/maturation

Porous collagen microspheres could be a potential scaffold and can be placed in bioreactors for scale-up.
But this does not produce a highly organised structure. 3D printing could solve this problem.

268
Q

What are the specific challenges for food TE?

A
  • Scale: global meat production is 293 million tons/year. Requires 1 bioreactor per 10 humans (significant footprint) and culture medium would be vast.
  • Efficiency: optimisation of scale-up and differentiation. Needs to be cost-effective.
  • Customer acceptancy: Needs to be as good as current meat. Plus addition of ‘healthier’ ‘reduced fat’ ‘increased vitamins/linoleic acid’ (anti-inflammatory and anti-tumourogenic).
269
Q

What other animal products could you replace using TE?

A

Fat from pre-adipocytes to give taste.
Milk from mammary gland cells?!
Leather from skin samples taken from animal, induced collagen production, layered sheets fused together, finished, dyed and conditioned (good that it won’t have hair).

270
Q

Why was organ-on-a-chip developed?

A

Severe limitations of current culture systems.

  • Many cells in vitro lose their in vivo phenotype in 2D culture.
  • Model organisms are more relevant but don’t have single cell analysis tools

So organ-on-a-chip technology was developed as a middle ground between experimental tractability and physiological relevance.

271
Q

What are organ-on-a-chips?

A
Device for culturing cells in continuously perfused micro-metersized chambers.
Incorporates minimal units that mimic tissue and organ level functions.
Incorporates microfabrication (creating the chips themselves) and microfluidics (how fluids behave at a micro-metersized level).
Can be made in the lab using polydimethylsiloxane whic begins as a liquid but sets as a solid due to the high temperature, leaving a channel down the middle (silicon mould can be 3D printed for specific structure). Can use channels to create gradients of different molecules.
272
Q

How has/could organ-on-a-chip be used?

A

Lung on a chip: already established. Porous membrane with a channel whereby lung cells are fixed and the air flows above them and below is a capillary cell layer and blood flow. The vacuum surrounding the membrane changes the pressure, giving a stretching motion. Mimics the basic functional unit of the alveolus.

  • Bacteria can be introduced and WBCs move from the blood layer to attack
  • Pulmonary oedema can be modelled by introducing IL-2 and analysing water retention.

Body-on-a-chip systems holds promise to mimic multiple tissues and organs and how they link together and communicate.