BMS354 Principles of Regenerative Medicine and Tissue Engineering Flashcards

1
Q

What is the difference between regenerative medicine and tissue engineering?

A
  • Regenerative medicine is an umbrella term that includes tissue engineering and cell therapy.
  • Tissue engineering differs from cell therapy through the use of biomaterials. Biomaterial can be used in both but in tissue engineering it is used more as a scaffold
  • Tissue engineering also has some applications that are distinct from regenerative medicine. For example, lab grown food
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Wy is there a clinical need for tissue engineering?

A

Organ failure is one of the most costly challenges faced in healthcare and the current treatments have many limitations

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

What are the available treatments for organ failure?

A
Surgical reconstruction  
Mechanical devices 
- Dialysis 
- Pace maker
- Hip replacements
Transplantation
- Skin grafts
- Organs from donors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are the limitations of surgical reconstruction as a treatment for organ failure?

A

Invasive, dangerous
Surgical complications
Morbidity at the donor sites

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

What are the limitations of mechanical devises as a treatment for organ failure?

A
  • Only mechanical support

- Do not grow with the tissue – children with these will need multiple surgeries to replace as they grow

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

What are the limitations of transplantation as a treatment for organ failure?

A

Rejection
- Must be on immunosuppressors
Limited supply
- Supply and demand – due to the limited supply, doctors are forced to use organs that may not be ideal e.g. from an elderly person
- E.g. in 2014, two patients who had a kidney transplant died sue to the kidney being infected with a parasitic worm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

When did the tissue engineering field emerge?

A
  • This research emerged in the 1970s and 1980s and was coined Tissue engineering in 1987
  • In the 1990s research accelerates and industry begins to emerge - In 1998, human embryonic stem cells were isolated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are the building blocks of tissue engineering?

A
Cells
Biomaterial scaffold (provide physical support)
Bioactive molecules (direct cellular behaviour)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Regenerative medicine and tissue engineering are multidisciplinary research fields. What other disciplines participate?

A
  • Material science
  • Cell, physiology, anatomy, molecular, computational biology
  • Robotics
  • Engineering
  • Biochemistry
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What tissues form an organ?

A

Epithelial, connective, muscle and nerve tissue form an organ

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

What is the role of the different tissues that make up an organ?

A

Epithelial tissue carry out the function while muscle and connective tissue provides structure

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

What are the three stages of wound healing?

A

Inflammatory phase
Proliferative phase
Remodelling phase

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

Explain the inflammatory phase of wound healing

A
  • Clears out dead cells from the injury and limiting the extent of tissue damage to prepare the environment for healing
  • Arterioles and venules near the site of injury constrict briefly and then dilate. This increases capillary permeability which moves fluid into the effected tissue. Blood clots occur due to increased blood viscosity
  • Leukocytes engulf bacteria and cellular debris through phagocytosis to clean the wound and secrete growth factors which recruits fibroblasts
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Explain the proliferative phase of wound healing

A
  • New tissue formation occurs to fill the wound space
  • Fibroblasts secrete collagen and growth factors which promote angiogenesis and endothelial cell proliferation and migration.
  • Granulation tissue is formed by fibroblasts and endothelial cells. It forms 2-4 days post the wound. Microscopically looks very granular due to newly formed blood vessels and forms the scaffold for remodelling
  • The newly formed blood vessels are leaky and allow blood cells and plasma cells to leak into the tissues
  • Epithelisation – when there is proliferation and migration of epithelial cells to form the new surface
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Explain the remodelling phase of wound healing

A
  • Begins after three weeks
  • Remodels the new connective tissue. This can take months and sometimes years
  • Final scar tissue formed by collagen synthesis and becomes avascular
  • Scar tissue can achieve 70-80% tensile strength by the end of three months
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What is the difference between regeneration and repair?

A

Regeneration
- If the injury is only mild and superficial then the epithelium will be regenerated and there will be no scar
Repair
- If the injury is severe then a scar will form

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What decides whether an injury will result in regeneration or repair?

A

The outcome of the injury is dependent on the severity, tissue damaged and length of injury

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

What is fibrosis and how is it different to repair?

A

Persistent tissue damage that results in a tissue scar

The process of scar formation is the same as repair but this term refers to when a stimulus is persistent

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

Give an example of fibrosis

A

Pulmonary fibrosis

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

What is meant by Fibrous encapsulation?

A

The body will try to protect itself from biomaterials by producing lots of collagen leading to its isolation from the bodies environment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What cell sources are used in regenerative medicine?

A

]Autologous: From same person
Allogenic: Same species but different people
Xenogeneic: Different species
Syngeinc or isogenic: Genetically identical but different person (twins)

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

What different cell types can be used in regenerative medicine?

A

Differentiated mature cells
Mixture of differentiated cells
Stem cells

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

What are the advantages and disadvantages of using differentiated mature cells in regenerative medicine?

A
  • These cells cannot self-renew meaning they can’t expand into large numbers
  • However, these cells are already functional. This is an advantage over stem cells as differentiation of stem cells is sometimes difficult to get mature functional stem cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What kind of differentiated mature cells are used in regenerative medicine?

A

Fibroblasts, keratinocytes, osteoblasts, endothelial cells, chondrocytes ect.

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

What kind of stem cells are used in regenerative medicine?

A

Adult stem cells
- Can be taken from the patient so will be no immune response
- If person has genetic disease this doesn’t help
Embryonic stem cells
- Can differentiate into any cell type
Induced pluripotent stem cells
- Can still be taken from the same person –allogenic
- Can differentiate into any cell type

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

How are cells cultured in a lab?

A
  • Cells are cultured in growth medium which contains the necessary requirements for cell growth e.g. nutrients, growth factors
  • Laminar hood keeps aseptic
  • Incubated at 37 degrees
  • Lids of the petri dish are not sealed as there needs to be gaseous exchange
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What ensures that the cells cultured for patient use are of a good quality?

A

Good manufacturing practice (GMP) ensures that medicinal products are consistently produced and controlled to the quality standards appropriate to their intended use
Must be free from animal products

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

What is the role of the ECM?

A

ECM provides support for cells and contributes to the mechanical properties of the cells. It also provides bioactive cues for cells, acts as the reservoirs of growth factors and potentiates their actions and acts as a scaffold for orderly tissue renewal

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

What is the composition of ECM?

A

Fibrous structural proteins – Collagen, elastins
Water hydrated gels – proteoglycans, hyaluronan
Adhesive glycoproteins – fibronectin, laminin

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

How does the structure of collagen allow its function?

A
  • Unusually high abundance of proline, hydroxyproline and glycine. They twist around each other to form a triple helix.
  • These triple helices are aligned side by side to form collagen fibrils.
  • These then create collagen fibres which have an incredible tensile strength
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Give the structure of proteoglycans?

A
  • Composed of glycosaminoglycan chains linked to a specific protein core
  • Very hydrophilic and form hydrated compressible gels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Where are proteoglycans found?

A

Joints

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

What is the role of fibronectin?

A

Allow the attachment of the cell to the ECM

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

What is a RDG sequence?

A

arginine, glycine, aspartic acid sequence in fibronectin

- what integrins on the cell surface bind to

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

How do cells adhere to ECM or biomaterials?

A

When cells adhere to the ECM there are no chemical bonds but the initial contact with the cell surface or material can activate signalling pathways in the cell through stress activated mechanical channels in the cell

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

What is the structure of an integrin?

A
  • Form heterodimers and are made up of 19 alpha and 8 beta subunits
  • They have extracellular, transmembrane and intracellular domain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Why is the structure of a integrin important?

A

The structure of the integrins determine which ECM proteins are recognised. Most recognise multiple ECM proteins – there is a lot of redundancy

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

What are the types of integrin signalling?

A
  • In inside out signalling, there is a ligand which is bound to cytoplasmic tail which leads to conformational change in the integrin and changes its ability to bind a ligand outside the cell
  • In outside in signalling, they bind an intrinsic ligand which transduces the signal inside the cell which leads to further assembly of the actin cytoskeleton and activation of signalling pathways
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What signalling pathways does integrin binding lead to?

A

The signalling pathways used by them are mediated through focal adhesion kinase (FAK). This auto phosphorylates and recruits downstream proteins including the focal adhesion complex (a macromolecular assembly of proteins at the site of cell adhesion). This can then activate proliferation and cell signalling

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

What is mechanotransduction?

A

The process by which external mechanical stimuli are transmitted into the nucleus

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

What are biomatierals?

A

Non-viable materials used in a medical device indented to interact with biological systems. They can be used to develop scaffolds for tissue engineering

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

What is the main problem. with biomaterials?

A

The main issue with biomaterials is that the body rejects them through the immune response

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

When was the first biomaterial that did not lead to rejection discovered?

A

In the second world war, a man examined the eyes of pilots and noticed that they had splinters in their eyes that did not induce an immune response. He concluded that this material (used to make the cockpits) could be used to create intraocular

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

Define biocompatibility

A

Biocompatibility of materials is the ability of a material to perform with an appropriate host response in a specific application

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What is meant by biomaterials leading to an appropriate host response?

A
  • Resistance to blood clotting
  • Resistance to bacterial colonisation
  • Normal healing
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What were the first generation of biomatierals?

A

Called bioinertness

- The body fluid did not interact with the materials and were stable to not induce any major adverse responses

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

When were biomaterial considered biocompatible?

A

If it was non-toxic, non-carcinogenic and had adequate mechanical properties

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

What was the second generation of biomatierals?

A

Bioactive

  • This era started with the bioglass: the first material to seamlessly seal with bone
  • Biodegradable materials
  • However, non of these materials response to the needs of the body
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What was the third generation of biomatierals?

A

Functional tissue

- Polymers

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

What are polymers?

A

Large molecules made up of linked monomeric units
They have a large molecular weight and can be presented in different structures e.g. linear, branched and network. A car tyre is a network polymer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What are the most common monomeric used units in biomaterials?

A

PE, PTFE, PVC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What is a homopolymer?

A

In the simplest form, there is one monomeric unit repeated

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

What is a copolymer?

A

Multiple different monomeric units in the same polymer

- Can manipulate the monomeric unit arrangement which will determine the property of the materials

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

What are hydrogels?

A

In natural state, polymers are brital and hard but some are naturally hydrophilic so will swell up in an aqueous medium. These are known as hydogels

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

Give an example for the use of hydrogels

A

Soft contact lenses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What are the classifications of biomaterials

A
  • Natural
  • Synthetic
  • Semi-synthetic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

Give examples of natural biomaterials

A

Protein based natural polymers
- E.g. Collagen (found commonly in animals and plants), gelatine, silk (low degradation time), Fibrin (component in blood clots)
Polysaccharides
- E.g. Chitosan, alginates, hyaluronan

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

Give examples of synthetic biomaterials

A

Polyactic acid, polygycolic acid

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What are semi-synthetic materials?

A

Hybrid molecules made by the incorporation of biologically active macromolecules onto the backbone of synthetic polymers

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

Give examples of semi- synthetic biomaterials

A

Semi synthetic PEG-fibrinogen

  • PEG controls the density stiffness and biodegradability
  • Fibrinogen presents biofunctional domains
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What are the advantages and disadvantages of natural biomaterials?

A

Advantages
- Built in bioactivity: Integrin recognise RDG domain so cells recognise it
Disadvantages
- Purification cost
- Immunogenicity – induce immune response
- Lack of mechanical properties
- Batch to batch variability – difficult to isolate from same source

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

What are the advantages and disadvantages of synthetic biomaterials?

A

Advantages
- Controlled mechanical properties and degradation kinetics
- Easy processability into custom shapes and structures
- Minimal batch to batch variability – synthesised on an industrial scale
- Cost effective
Disadvantages
- Biocompatibility may be difficult to predict
- Immune rejection

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

What are degradable biomaterials?

A
  • Used as a scaffold and overtime degrades and replaced with tissue. Therefore, biodegradability can be a positive thing.
  • This means that there is a hydrolysis of a covalent bond. This results in the formation of by-products which also need to be non-toxic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What are resorbable biomaterials?

A

These materials can be reabsorbed into the body and removed so that there is total elimination of the initial foreign material and its by-products

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

What properties should a biomaterial possess to be used for internal sutures?

A
  • Do not want another surgery to remove so they should be biodegradable and the length of biodegradability should depend on the organ as it needs to hold the wound together and heal but if in there to long it will induce a host response
  • Used sheep gut but now synthetic used
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What properties should a biomaterial possess to be used for soft contact lenses?

A
  • Required to correct material so must be transparent and hold their shape to maintain refractory angle
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What properties should a biomaterial possess to be used for artificial hip joints?

A
  • Need to have high mechanical strength
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
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
69
Q

How did Engler et al, 2006 investigate how mechanical properties of biomaterials can influence how cells behave?

A

Mesenchymal stem cells placed on three different materials. High (represent bone), medium and low (represent blood) stiffness materials. They all lead to different responses on the stem cells showing they can sense their location and respond accordingly

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

What are surface modifications of biomatierals?

A

Alter the surface of the biomaterial to achieve the desired properties
- The layer modifying should not be too thick as this would change the bulk properties and prevents delamination resistance

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

How are surface modifications achieved?

A

This can be done by

  • Chemically altering the atoms in the existing surface
  • Over coating the existing surface with a material of a different composition
  • Creating surface textures or patterns
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

How do cells interact with biomaterials?

A
  • The proteins from serum/growth medium adhere to the surface of the glass and the cells adhere to this protein
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What are non fouling materials?

A

Materials that do not allow protein absorption

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

How can non fouling materials be an advantage?

A

This is an advantage sometimes in medicine as it will result in no bacteria colonisation

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

How are non fouling materials a disadvantage?

A

In tissue engineering it is a disadvantage as want cells to adhere

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

How can non fouling materials be functionalised?

A

By chemically adding RDG domains to the surface

- Can also use this to decide where the cells attach to the surface

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

How can RDG domains be added to non fouling materials?

A

Using micro contact printing

  • A prepolymer is poured on - It is then cured and the stamp peeled off the master
  • The stamp is then cut into smaller places and Inked by soaking in ink solution
  • The ink is printed bb contacting an inked stamp with a suitable surface
  • A patterned substrate is obtained
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What did Chen et al, 1997 hypothesise about the role of cell shape?

A
  • Hypothesised that cell shape controlled cell survival
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

How did Chen at al, 1997 test that cell shape controlled survival?

A
  • Used micro stamping of fibronectin islands. Cells attached to these islands and assumes the exact shape of the islands. They then looked at the survival of these shapes. The shape alone affected whether cells proliferated or entered apoptosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What properties must a scaffold have?

A
  • Provide mechanical and biological functions of ECM
  • Porous to allow vasculature bring the oxygen and nutrients needed for the cells covering the scaffold
  • Degradable so that they do not need to be removed by surgery.
  • The degradation products need to be non-toxic.
  • The degradation kinetics need to be carefully designed based on the tissue it is being used in
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What are acellular tissue matrices?

A
  • Another material for scaffold fabrication
  • Could be considered naturally derived. They are obtained by decellularisaing tissues and organs – remove all cells and antigens resulting a white tissue matrix. This matrix will therefore not induce an immune response so can be used without immunosuppressans.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

How are tissues decellularised?

A
  • Usually involves mechanical scraping, acids and bases and enzymes to digest the cellular and nuclear components
  • If process not complete or bacterial contamination then it may result in scar tissue formation and an immune response
  • Cross linking of ECM can be problematic as the body cannot remodel it so again can induce scar tissue formation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What are the of advantages acellular tissue matrices?

A

The process preserves intricate structures that are difficult to replicate outside the body but are important for function

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

What are the of disadvantages acellular tissue matrices?

A

Accessibility – use the dermis of the skin, submucosa of small intestine, pericardium. Some are now commercially available (Alloderm)

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

Why is the architecture of scaffolds important?

A
  • The final shape of the tissue engineered structure is defined by the scaffold – can use MRI to find the exact shape needed
  • Microscopic architecture also very important. Need to be porous to allow vasculature. When trying to recreate this in vitro need to keep this in mind
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What is a pore?

A

A pore is the could space within a scaffold

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

What is porosity?

A

The porosity is the number of pores and how well they are connected

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

Why is the porosity of a scaffold important?

A

Some scaffold has 100% accessibility and 100% interconnecting meaning can get to any pore though multiple roots but some are not. This is important because if too porous then there will be a lower mechanical strength

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

What are the properties of an ideal scaffold?

A
  • Adequate mechanical properties
  • Biodegradable
  • Allows cell attachment and function
  • Accessible
  • Allows tissue and integration and vascularisation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What are the methods of scaffold fabrication?

A
  • Porogen leaching
  • Phase separation
  • Electrospinning
  • Additive manufacturing
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

Outline Porogen leaching

A
  • Uses a polymer dissolved in a solvent. The polymer is mixed with salt particle of a certain size
  • This mixture is poured into a mold. Then evaporate the solvent and the polymer will solidify. The salt particles are still dispersed inside it.
  • Put this solid polymer into water and the salt will leach out leaving with a porous scaffold where the salt was present in the polymer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

Evaluate porogen leaching as a method of scaffold fabrication

A

Can change the size if the pores by changing the size of the salt particles but the way they are distributed cannot be changed

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

Outline phase separation

A
  • Polymer is dissolved in a suitable solvent and subsequently placed in a mold that will be rapidly cooled until the solvent freezes
  • Solvent is then removed by freeze-drying and pores will be left behind in the polymer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

Outline electrospinning

A
  • Commonly used due to its ability to provides very fine fibres (nanofibers).
  • Polymer solution in syringe which is attached to a high voltage power supply. This creates an electromagnetic field between the needle and a grounded plate.
  • The polymer is slowly injected and due to the electromagnetic field it causes the polymer fibres to spray and the solvent will evaporate and the fibres are collected on the grounded plate
  • The parameters (e.g. voltage, distance from plate ect) can be changed which will influence the types of fibres get at the end
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What is additive manufacturing?

A

The process of joining materials to make objects from 3D model date, usually layer upon layer

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

What is the most common type of additive manufacturing?

A

3D printing

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

Outline 3D printing

A
  • Uses a roller which delivers a layer of polymer onto the surface.
  • Have cartridges which are filled with adhesive. This is controlled by a computer and the adhesive is only added to certain areas.
  • Once one layer is done, the platform moves down and the roller adds another polymer layer and the process is repeated.
  • When complete, the adhesive is cured by heating and unwanted polymer is removed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What are the advantages and limitations of 3D printing?

A

Advantages
- Production of scaffolds with precise morphologies
- Combines medical imaging to fabricate anatomically shaped implants
Limitations
- Limited number of biomaterials can be used

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

What is the problem with 3D printing as a method of scaffold fabrication?

A

Material is printed and then the cells are put on the scaffold. This has problems because the cells sometimes do not populate the scaffold properly and may sit in the pores which are needed to be open for vasculature to go through.

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

What is Cell encapsulation?

A

A variation on 3D printing where cells are mixed with the polymer and then printed out

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

What did Kang et al, 2016 do to further the use of 3D printing?

A

Looked at 3D printing system to produce human scale tissue with structural integrity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What did Kang et al, 2016 use to produce human scale tissue?

A
  • A new 3D printer called an integrated Tissue-Organ Printer (ITOP).
  • Common problem faced was killing the cells during in printing. To overcome the issues they were having, this printer had several cartridges which dispense different things.
  • Cells were encapsulated in a polymer already (with polymer that is not toxic to cells).
  • They used a sacrificial material that allowed them to keep the mechanical structure of the construct while it was being printed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What sacrificial material did Kang et al, 2016 use?

A

Used two types of sacrificial materials: PCL and pluronics (only in place during printing). Printed Cell-laden hydrogels and sacrificial scaffold to provide initial mechanical integrity to the cells

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

What did Kang et al, 2016 achieve using cell encapsulation?

A
  • Made ear, bone and muscle
    Able to show that after removing the scaffold, muscle fibres maintained integrity and would mature in differentiation medium
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

What biomolecules have been explored for inducing tissue regeneration?

A
  • Small molecules
  • Proteins and peptides
  • Oligonucleotides
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What are BMPs?

A
  • Abundant in bone matrix and made by osteoblasts (bone forming cells)
  • Members of the TGFbeta family
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

What is meant by BMPs being osteoinductive?

A

If ectopically implant BMPs then it will form ectopic bone. This ability is believed to be due to their ability to recruit osteoblasts and cause tissue specific stromal cells to differentiate into osteoblasts

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

How did Lutolf et al, 2003 investigate the role of bioactive molecules in scaffolds?

A
  • It was clear that BMPs were only osteoinductive if they are locally delivered and retained to have the correct concentration
  • They hypothesised that they could use scaffolds to module the effect of BMPs and to enable them to be delivered locally and maintained at the correct concentration to have the osteoinductive affect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

How did Lutolf et al, 2003 prepare the gel for the scaffold they used?

A
  • They prepared gel formed of the non-fouling PEG which was then functionalised using RGD domains. They then crosslinked PEG and added a site for cleavage by matrix metalloproteinases.
  • These are enzymes that the cells use to remodel the matrix. They entrapped BMP2 inside these gels.
  • The pore sizes are large enough to hold BMP but small enough that it doesn’t just diffuse through the gel
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

How did Lutolf et al, 2003 show that the gel they produced could be cleaved and degraded?

A
  • Did functional studies to show gel worked. Used 3D fibroblasts and see if they can degrade the matrix using matrix metalloproteinases which they could. The scaffold could therefore be degraded and the cleavage sites could be cut by cells
  • They then checked if BMP2 can be released from scaffolds. Saw a gradual release of BMP over time
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

How did Lutolf et al, 2003 show that their scaffolds with BMP entrapped could promote bone regeneration?

A
  • Tested this in vivo. The idea is that if can bring BMP to the site of injury then it should induce tissue regeneration in the bone.
  • Took rats with critical size bone defect. Used a control scaffold with no BMP which showed no repair as well as a control that had BMP but no cleavage sites added.
  • In scaffolds with BMP and cleavage sites resulted in bone regeneration in the rat
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

How could the scaffold from Lutolf et al, 2003 result in bone regeneration?

A

This occurred as responding cells adhere to RGDs in matrix. Cells secrete MMPs that degrade MMP cleavable bonds serving as crosslinks for the matrix. BMP liberated from matrix diffuses from site, signaling osteoblast precursor cells. Osteoblasts secrete bone matrix and bone is regenerated

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

What are the limitations of the conventional scaffold fabrication techniques?

A
  • E.g. salt leaching, gas forming, phase separation and freeze drying
  • They do not enable precise control of internal scaffold architecture or the fabrication of complex architectures
  • Using toxic solvents in these techniques can lead to cell death if not correctly removed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

What are the limitations of phase separation?

A

Limitations include the addition of organic solvents such as ethanol inhibits the incorporation of bioactive molecules or cells during scaffold fabrication and the small pore sizes obtained

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

How can the pore size in scaffolds be altered in electorspinning?

A

Pharm et al showed that the average pore size of electrospun scaffolds increased with increasing fibre diameter

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What is a disadvantage of electrospinning?

A

Disadvantages include involving toxic organic solvents which can be harmful to cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

How can the disadvantages of electrospinning be overcome?

A
  • This can be overcome using melt electrospinning which doesn’t involve organic solvents and instead heats the polymer to its melting temperature.
  • However, as the polymer melts it has a lower charge density meaning that the fibres produced are thicker than those produced from electrospinning
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

Why is silk better then industrial produced replacements?

A

Industry makes polymeric fibres such as Kevlar which are high performing and take a lot of energy to break. Nature makes natural fibres such as silk from spiders/snails and due to natural selection, it is much stronger, cheaper, lower energy and biodegradable then anything we can make in industry

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

Define silk

A

They are structural proteins which are spun into fibres for use outside the bod

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

What is the primary structure of silk?

A
  • Silks are fibrous proteins and usually 100s kDa long

- Contains high proportions of glycine and alanine which are biologically cheap amino acids

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

How does the primary structure of silk make they tough?

A
  • The fact that the chains can move slightly due to the prolines producing kinks meaning that they are tough and can dissipate energy
  • Protein hydration and hydrogen bonds are key to structure and strength
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What is the secondary structure of silk?

A

The prolines make the protein feedback on its self to form beta sheets

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What is the structure of silk at a nanoscale?

A

Formed of a combination of order and disordered parts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What is the structure of silk at a hacroscale?

A

Formed many fibres which form layers to produce silk

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

Is kevlar (commerically produced) or silk stronger?

A

Kevlar is stiffer then spider silk but silk is stronger, it doesn’t break with more force - it bends

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

How can we change the mechanical properties of the silk produced?

A

Silks are spun to move at the natural speed of the animal. Spiders can slightly change this speed. Can take silk and change the speeding conditions and can change the mechanical properties of the silk. Can therefore replicate any of the fibres in a better and environmental way

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

Why is silk useful in biomedical research?

A

Can be turned into any structure so can be used to recreate any apart of the human body e.g. neural implants, collagen, lasers. Silk will degrade in the body and the rate of degradation can be controlled by controlling the beta sheets

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

Give examples of what silk has been used to in regenerative medicine?

A

Peripheral nerve repair
- Put fibres inside a straw to form a nerve conduit
Vascular repair
- Vascular graft for haemodialysis – stop vein collapse
Orthopaedic implants
- Treatments of meniscus and cartilage damage in the knee – this is a common
- Can use an artificial meniscus which can be put in between the cartilage on the knee. The mechanical properties work due to its biological basis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What are bioreactors?

A

Tanks full of medium where can grow cells to a high density under a tight control of conditions. This can maximise the yield produced

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What are the current challenges in tissue engineering?

A
  • Trying to create not just the cells but a 3D structure with cells in an appropriate spatial orientation. Growing to relative clinical sizes is difficult
  • The growth and 3D assembly of multiple cell types that are required for more complex functional tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What is the problem with normal tissue culture?

A
  • The media is static on top of the cells in the cell culture dish. This can result in a concentration gradient across the flask changing the cell density and the behaviour of the cells
  • Only know the pH at the time of feeding, after that have no control
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What are dynamic culture systems?

A
  • Designed to mix the media and give control over the environment
  • A medium tank is connected to a peristaltic pump which feeds the media to the cells. Can also have a control of gas pH to monitor or control
  • This results in homogeneous concentrations of nutrients, toxins and other components
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What are the three roles of bioreactors in tissue engineering?

A
  • To establish spatially uniform cell distributions on 3D scaffolds
  • To overcome mass transport limitations in 3D culture
  • To expose the developing tissue to physical stimuli
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

Why is the way that a scaffold is seeded important?

A

The way the cells attach and distribute will determine the mechanical and functional properties of the construct

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What is needed when seeding cells on a scaffold?

A
  • Need a high seeding efficiency to stop the loss of rare and important cells. There may not be huge amounts of cells which can be used so cannot waste them. This process needs to be maximised
  • Want to achieve a uniform distribution of cells across the scaffold
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What is the problem with seeding cells in a static way?

A

Can deliver the cells in a static way by pipetting the cells into the scaffold and by gravity they will attach to the scaffold. This can lead to cells just attaching to the top and not moving through the scaffold

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

How can the problem of static cell seeding be overcome?

A

Can flow media through the construct meaning that the cells can access entire scaffold leading to a more even distribution

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

Describe the assay used to see how the seeded cells are distributed on a scaffold?

A
  • MTT assay – indicated were cells are and if they are alive. It is a purple die that will turn purple from yellow if high metabolic activity meaning the cells are dying
  • Statically seeded cells accumulate at the top. High density can lead to negative effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

What are the transport limitations of 3D cultures?

A
  • Once the cells are inside the scaffold, they need oxygen
  • Mass transfer of external and internal mass and removal of metabolites and CO2
  • There is a maximum distance between cells and capillaries is 200micrometres. This is the same in vitro meaning that cells need to be close to the medium
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

How did Wendt et al, 2008 overcome the transport limitations of 3D culture?

A
  • Chondrocytes seeded using perfusion cell seeding
  • Cultured the cells statically and by perfusion
  • In the statically cultured cells, the cells only survive at the edges as that is where the nutrients could get to the cells
  • In the perfusion cultured scaffolds, the cells were well nourished and created well organised structures
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

Why do developing tissues have to be exposed to physical stimuli?

A
  • Tissues and organs in the body are subject to complex biomechanical environment
  • There are many forces inside the body including hydrostatic, mechanical and electrical
  • To engineer something in vitro, the cells need these pressures – they have evolved in a way to need this
  • Bioreactors allow this
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

What are the design considerations that need to be taken into account when bioreactors?

A
  • Diversity in bioreactor design reflects the range of signals needed for formation of various tissues
  • They need to be biocompatible and sterile to stop bacterial infections
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

What are the types of bioreactor?

A
  • Spinner flask bioreactor
  • Rotating wall bioreactor
  • Perfusion bioreactor
  • Compression bioreactor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

What are spinner flask bioreactors?

A
  • The magnetic stirrer stirs the solution to help with the mass transfer of medium to the cells. This helps with the external transfer
  • Very accessible and cheap
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

What are rotating wall bioreactor

A
  • Flask spins around and the scaffold are suspended. Gravity pulls the scaffolds down and the forces of the flask rotating are pulling in the opposite direction. These forces cancel each other out. The scaffold is suspended in medium
  • As the cells expand, the forces need to be increased to counterbalance gravity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

What is a perfusion bioreactor?

A
  • The culture medium continually circulates through the construct
  • Most mass transfer limitations are mitigated
  • The effects of direct perfusion and be high dependant on the medium flow rate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

What is compression bioreactor?

A

Apply mechanical stimulus to the construct

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

What type of bioreactor would be used to decellularise organs?

A
  • Complex organs are difficult to decellularise

Can use a perfusion bioreactor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

What type of bioreactor would be used to engineer articular cartilage?

A
  • Compression bioreactor

- Cartilage is a load baring tissue so is under mechanical stress in differentiation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

What type of bioreactor would be used to study the effects if near 0 gravity?

A
  • Rotating wall bioreactors
  • These were designed by Nasa and there is evidence that in space peoples bone mass decreases. Can therefore study bone density in a 0-gravity environment using these rotating wall bioreactors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

Give a study into investigating the effects of 0 gravity?

A

Tamma et al, 2009

  • Culture osteoblasts
  • There are more osteoblasts produced in near 0 gravity environment which could explain the decreases in gravity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

What is a common problem with heart valves?

A
  • Valves may not open or close entirely. This is a huge problem world-wide but options are limited. Often involves surgical repair or valve replacement
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What are the problem with heart valve replacements?

A

Patients receiving them need ant-thrombotic drugs which can predispose them to lack of blood clots

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What is the problem with creating heart valve replacements that grow with the developing body?

A
  • Autologous leads to no rejection but unlikely to get the right cells needed.
  • To differentiate these cells appropriately, they need to withstand forces from the moment they enter the body. Animal model studies have shown that they can’t do this unless they have been exposed to the forces outside the body.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

How did Engelmayr et al, 2008 use a bioreactor to design a construct for a specific function?

A

Wanted to mimic the mechanical stimulation and perfusion of heart valves in vitro to improve engineered tissues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What did Engelmayr et al, 2008 do to mimic mechanical stimulation of heart valves?

A

Made a flex stretch bioreactor
- Can drive movable post from a flex to a stretch position. This can mimic the role of a heart valve in the body
- To mimic blood flow over the valves, they have the medium flow over the scaffold. This exposes the cells to hydrostatic pressure
Used mesenchymal cells derived from sheep bone marrow and cultured in PGA/PLA (synthetic) scaffolds. Then treated with and without the bioreactor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

What did Engelmayr et al, 2008 conclude?

A

Concluded that the forces were necessary to mimic what happens in the body to promote mesenchymal stem cell differentiation and tissue formation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

How have bioreactors helped to engineer vocal folds?

A
  • The vocal folds has specific physical stimuli in vivo

- This bioreactor mimics the sound by speakers to create vocal folds

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

What are the challenges in bioreactor design?

A

Mimic native cell behaviour
- Further understanding of tissue development and regeneration
Scale up
- All small scale equipment so to generate on a large scale for clinical application may be difficult

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

Why was skin the first organ to be fabricated?

A

Is the first organ to be fabricated due to its relatively simple structure and the huge clinical need. It is also because skin cells were the first cells to be cultured outside the body

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

What is the main problem with skin replacements?

A

The skin that is replaced still does not look or function the same

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

What percentage of body mass does the skin account for?

A

10%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

What is the function of the skin?

A

It is needed for protection, regulation and sensation

- The main function is protection of the body from UV light and microbes

164
Q

What are the three layers of the skin?

A

Epidermis
Dermis
Hypodermis

165
Q

How thick is the epidermis?

A

Varies from 0.5 mm on eyelids to 1.55mm on soles of feet

166
Q

What is the role of the epidermis?

A

Protect from environmental factors

167
Q

What is the structure of the epidermis?

A
  • No blood vessels, gets from nutrients from the dermis
  • Mainly Keratinocytes which secrete keratin which gives skin its toughness and creates the water proof layer. Contributes 90% of epidermis
  • Melanocytes which secrete melanin to protect from the UV light
  • Merkel cells – involved in sensation of touch
  • Langerhans cells are antigen presenting cells and are involved in the immune response
  • Forms a stratified epithelium
168
Q

What is the basement membrane?

A

Mitotic cells are found in the bottom basal layer and differentiate as they move up the layers.
Basal layer also secretes ECM which produces basement membrane which separates the epidermis from the dermis

169
Q

Outline the dermis

A
  • Makes the bulk of the skin
  • Composed of collagen with some elastin and glycosaminoglycans
  • Cells found are mainly fibroblasts which lay down collagen as well as blood vessels, sweat glands and hair follicles
170
Q

Outline the hypodermis

A

The bottom layer which functions as a thermal insulator and stores fat as a major source of energy

171
Q

What is meant by contracture of the skin?

A

During wound repair, fibroblasts are recruited and lay down new collagen. Myofiberblasts are similar to fibroblasts and smooth muscle cells and their role is to contract the wound. If they are over stimulated it can lead to over contraction and cannot move the skin

172
Q

What injuries often require skin replacement?

A

Acute trauma e.g. burns
Chronic wounds
Surgery
Genetic disorders

173
Q

What are the types of skin wounds?

A

Epidermal
Superficial partial thickness wounds
Deep partial thickness wounds
Full thickness wounds

174
Q

Outline epidermal skin wounds

A
  • Affect only epidermis and are characterised by erythema and minor pain
  • They do not require surgical treatment
  • Do not lead to scarring
175
Q

Outline superficial partial thickness wounds skin wounds

A
  • Affect the epidermis and superficial part of the dermis
  • The wounds are wet and weeping with red to pink blisters
  • Very painful sue to the exposure of sensory nerves
  • Heal spontaneously
176
Q

Outline deep partial thickness wounds

A
  • Involves greater dermal damage
  • Moist and white/red in appearance
  • Result in fewer skin appendages remaining
  • Scarring is more pronounced
177
Q

Outline full thickness wounds

A
  • Complete destruction of epithelial regenerative elements
  • Wound appearance, dry leathery and rigid
  • No spontaneous healing
178
Q

How are skin injuries treated?

A

Early excision of dry scab (ESCHAR)
- Remove denatured proteins as they can trigger inflammatory response and can harbour microbes. The patients are already immune repressed so this can cause problems
Wound closure
- reduces scarring and increase mortality
Skin grafts
- A skin graft is a tissue of epidermis and varying amounts of dermis that is detached from its own blood supply and placed in a new area

179
Q

How are skin grafts categorised?

A

Categorised based on how much dermis is taken

  • Split thickness: the epidermis and part of the dermis is taken
  • Full thickness: both epidermis and dermis is taken
180
Q

What is an autograft?

A

Removing skin from uninjured part to injured part

Use meshing of skin grafts to cover a larger area

181
Q

What is meant by graft take?

A

The new cells need to get nutrients from the wound bed. This can keep the cells alive for 2-3 days
In the meantime, there is vascular growth within the wound bed. The graft will only ‘take’ if this occurs

182
Q

Why does the wound bed need to be prepared?

A

For the graft to take, the wound must be prepared properly

  • The graft must adhere to the wound bed (no bleeding, infection or movement)
  • The graft needs a thin layer of connective tissue. Cannot graft skin directly onto bone or tendon
183
Q

What is a akin allograft?

A
  • If the trauma is very extensive then sometimes there is not enough skin autologous skin to use for the graft. Then consider skin allografts
  • The use of cadaveric skin for temporary prevention of fluid loss or wound contamination. However, this can lead to pathogen transmission and immunogenic rejection
184
Q

What would an ideal skin substitute have?

A
  • Readily available
  • Cause no immune response
  • Cover and protect the wound
  • Enhance the healing
  • Lessen the pain
  • Leave no scars
  • However, there are no skin substitutes so far which do all of these things
185
Q

How are skin substitutes classified?

A
Layer to be substitute 
- Epidermis 
- Dermis 
- Compound
Durability 
- Temporary 
- Permanent 
Product origin
- Biologic 
- Biosynthetic 
- Synthetic
186
Q

How are epidermal substitutes made?

A
  • A key step is isolation of keratinocytes from a skin biopsy and expanding in culture
  • Cultured keratinocytes are delivered on the wound and form a new epidermal layer
187
Q

What are cultured epithelial sheets?

A

Used as an epidermal substitute
- Skin biopsy from the patient, isolate the epidermis and extract the keratinocytes - The keratinocytes are cultured and form colonies which will merge and create a stratified epithelium. This forms cultured epithelial sheets

188
Q

What is the problem with cultured epithelial sheets?

A

The graft takes of these vary a lot (from 15-80%). It is difficult to work with something with such a varied success rate

189
Q

Why do the great takes of cultured epithelial sheets vary so much?

A

Studies indicated that when the keratinocytes are cultured, if they differentiate so that they don’t produce the correct integrins anymore then when they are placed on the dermis then they will not attach. The cells need to be at the right stage of differentiation

190
Q

What are single cell suspensions?

A

Used as an epidermal substitute

  • Take a tissue biopsy, culture the cells and deliver back to the patient
  • Autologous cells used so results in no immune response
191
Q

What is MySkin?

A

Used as an epidermal substitute

  • Developed by Celltran ltd which was part of the University of Sheffield
  • Sub confluent autologous keratinocytes. The cells did not fill the entire flask which kept them in the right differentiation stage
  • They also developed a synthetic silicone delivery membrane which allowed successful delivery to the wound bed
  • Was successful in treating chronic wounds. Required repeated delivery
192
Q

What are the advantages of epidermal substitutes?

A
  • Keratinocytes can be easily expanded in vitro

- Can be autologous

193
Q

What are the disadvantages of epidermal substitutes?

A

Only contain keratinocytes so can only replace the epidermal layer. If there are deeper injuries then also require a dermal substitute

194
Q

What are dermal substitutes?

A

Needed for deeper skin wounds
- Most products are acellular. Idea is to provide a scaffold that will be infiltrated by cells of the host and remodel the matrix

195
Q

What does the integral dermal regenerative template consist of?

A
  • Dermal layer: made from bovine type 1 collagen and shark chondroitin-6-sulfate. This is xenogeneic (not from a human)
  • Epidermal layer: made from silicone. This regulated heat and fluid loss.
  • The collagen-chondroitin matric allows in growth of cells from the wound bed and the artificial silicone epidermis regulates heat and fluid loss
196
Q

Why is the integral dermal regenerative template semi-permeant?

A

Once the cells infiltrate, the silicone membrane can be removed and replaces with epidermal substitute

197
Q

What is Alloderm?

A

Dermal substitue

  • Acellular human allogenic dermal matrix (decellularised skin) preserved by freeze drying
  • Full thickness skin burns alloplastic breast reconstruction, abdominal wall reconstruction, rhinoplasty
198
Q

How are derma substitutes used?

A

Entails a two-step process

  • Put dermal substitute into the wound bed
  • Add epidermal substitute which delivers the cells of the epidermis
199
Q

What are compound substitutes?

A

A types of skin graft

  • Aims to mimic the histological structure to normal skin – contain both epidermis and dermis
  • Most of these products are based on allogenic skin incorporated (different person) into a dermal scaffold
  • Enable production of large batches of the product because from a different pattern
200
Q

How are compound substitutes used as a temporary bioactive dressing?

A

Composite skin which consists of bovine type 1 collagen cultures with allogenic male neonatal fibroblasts and keratinocytes
- There are no skin appendages, melanocytes, hair cells
Allcell: includes cultured allogenic fibroblasts and keratinocytes obtained from the same neonatal foreskin
- Fibroblasts are seeded into a bovine type 1 collagen sponge with keratinocytes on top
- Cytokines and growth factors from the product promote host cell migration and wound healing

201
Q

What are the limitations of current skin replacement strategies?

A
  • The average wait time ranges from 3 to 12 weeks after the biopsy is taken. If want to produce autologous skin, it can only happen once the patient arrives at the clinic. It is difficult to produce quick autologous treatments. This is why many use allogenic cells
  • Only use keratinocytes and fibroblasts. There are many other cell types in the skin which are required for normal function and look.
  • Some products are considered clinical success but economic failure.
202
Q

How did Hirsch et al, 2017 use gene therapy?

A

Used gene therapy to genetically modify 7 year olds cells who was suffering with Epidermolysis bullosa (EB) and provide a life saving treatment

203
Q

What is Epidermolysis bullosa (EB)?

A
  • Causes spontaneous skin blistering and is very painful with life threatening complications (skin is a protective barrier so without it can be prone to infections and sepsis).
  • It causes a high risk of skin cancer (probably due to the skin constantly trying to heal) and affects 1 in 50000
204
Q

How does skin blistering EB occur?

A
  • Epidermis and dermis are separated by the basement membrane and blisters occur in-between these layers
  • Normally the basement membrane allows the epithelial cells in the basal layer to adhere to the dermis.
  • This is allowed to happen due to the macromolecular protein complex in the basement membrane.
  • Keratin and other intermediate filaments such as laminin interact with Alpha6beta4 integrins.
  • Laminin also interacts with collagen of the dermal layer. The basement membrane is a structured and ordered structure.
  • If there is a mutation that affects any of these interactions then the dermis and epidermis will come apart
205
Q

What mutation did the boy with EB have in Hirsch et al, 2017?

A

This patient has a mutation in laminin 332 and this results in dysfunctional laminin stopping integrin interaction
- Mutation that affects the splicing of laminin 332 within intron 4

206
Q

What was the phenotype of the boy with EB?

A
  • Had denuded and blistering skin and very few areas of non-blistering skin
  • Treated with antibiotics to stop sepsis and attempted skin transplants to replace denuded skin but this was unsuccessful. Thought that there was no treatment and would have to be put on palliative care
207
Q

How did Mavillo et al, 2006 provide a basework for Hirsch to treat EB?

A

Corrected EB by genetically modifying epidermal stem cells and transplanted into the patient. This was only done on a very small area of skin so was not a life-saving treatment

208
Q

How did Mavillo et al, 2006 allow for the treatment of boy with EB in Hirsch et al, 2017?

A

This study was taken as a proof of concept that this could work and allowed a one off compassionate treatment for this boy as had not been through clinical trials

209
Q

Why would autologous cell therapy alone not work for the boy with EB?

A

Taking a skin biopsy and culturing keratinocytes to put back in the patient would not work in this case as it is a genetic disease so this would happen again.
- There also is not enough skin

210
Q

How was the treatment in Hirsch et al, 2017 carried out?

A
  • Took the correct sequence of the mutated gene and put in viral DNA and delivered to the patient cells. They did not correct the gene itself but did deliver a correct version
  • This was done ex vivo. They took a small skin biopsy and cultured the keratinocytes on feeder layers and expanded.
  • They were then transduced with the viral vectors. This should result in corrected cells.
  • Prepared the skin substitutes. They cultured on fibrin cultured grafts
  • There were 3 transplantations targeting different areas
211
Q

Why did Hirsch et al, 2017 carry out next generation sequencing on the corrected cells?

A
  • To look where in the DNA did the viral genome integrate to ensure that the gene integrated into a safe place and not genes essential for cell function (these would die off before insertion so shouldn’t affect treatment) or into an oncogene (which could lead to cancer if inserted into the patient).
  • Most of the genes were inserted into introns rather than exons and appeared to be safe to proceed
212
Q

There was extensive follow up in Hirsch et al, 2017 over 21 months, what were they looking for?

A
  • Wanted to establish that the regenerated epidermis came from the modified cells.
  • To ensure that the epidermis had adhered to the dermis
  • Look at the expression of laminin
213
Q

How did Hirsch et al, 2017 establish that the regenerated epidermis came from the modified cells?

A
  • Probed for cadherin to act as a control and for the viral promotor (t-LAMB3).
  • In normal skin, cadherin was present but no viral promotor and then after 4 months can see the viral promotor is the epidermis of the arm showing it has regenerated from corrected cells.
214
Q

How did Hirsch et al, 2017 establish that the epidermis had adhered to the dermis?

A

o . They looked at histological sections of the skin. Follow ups showed no blisters and a clear epidermal-dermal junction

215
Q

How did Hirsch et al, 2017 look at the expression of laminin?

A

Using immunocytochemistry
- After treatment saw clear expression of laminin between the epidermis and dermis which could not be seen before treatment.

216
Q

How could the phenotype of EB be seen when culturing the cells?

A

The phenotype of EB could be seen when culturing the cells. The mutated cells could not adhere to the flask but the genetically correct keratinocytes were able to stick to the cells

217
Q

What future work could follow on from Hirsch et al, 2017?

A
  • Would this work on other patients – clinical trials
  • Alternative gene editing strategies (CRIPSR)
  • Patient age, the boy was young so keratinocytes would have high proliferative ability. Would this work in older patients when regeneration has decreased?
218
Q

Why was the skin decolourised in Hirsch et al, 2017?

A

Because the colour of the skin is given my melanocytes and the treatment was given with keratinocytes

219
Q

Why is it difficult ti have a skin substitute that would be able to recapitulate all the properties of the skin?

A

Developmentally the epidermis and dermis come from different germ layers
- Could use ES or IPS cells

220
Q

How did Takagi et al, 2016 attempt to improve on skin grafts?

A

Attempted to recreate the skin structures (integumentary organ)
- They hypothesised that pluripotent stem cells can be sued to mimic the developmental patterning

221
Q

What experimental approach did Takagi et al, 2016 use?

A

Produce IPS cells. They then created embryoid bodies which start to differentiate randomly. They then aggregated inside collagen and transplanted under the skin of a SCID mouse

222
Q

What was the problem with the used by Takagi et al, 2016?

A

This is not a very controlled system

- the IPS cells differentiate randomly in embryo bodies

223
Q

What did Takagi et al, 2016 find despite the uncontrolled experimental approach?

A
  • The approach worked. Hair grown from SCID mice and originated from the IPS cells (due to the hair colour).
  • Wnt3b improved the hair creating
  • Took sections of the hair shafts and saw that there were also additional structures of the skin from the IPS cells
  • Transplanted this into nude mice which lead to outgrowth
224
Q

How did Takagi et al, 2016 show that the hair cells can from the IPS cells?

A

Proved that these cells came from the original IPS cells as they were taken from a male mouse and the nude and SCID mice were female. Therefore, could just look for the presence of the Y chromosome

225
Q

What other uses of skin substitutes are there?

A

Disease modelling
- To reduce the use of animals in experiments
- This can lead to drug discovery
- For many illnesses such as EB, Psoriasis, Skin cancer and Allergic contact dermatitis
Chemical testing
- Test environmental factors in the skin
- Cosmetic industry

226
Q

Give an example of skin substitutes being used to model diseases

A

EB has been modelled in vitro (Itoh et al, 2011)

  • Can take cells and reprogrammed to IPS cells and differentiated back to keratinocytes to see what is wrong with them
  • Can probe them to see the mutation. In this case, there was no collagen
  • Creating skin equivalents, we can model which approaches will help alleviate the phenotype
227
Q

How many people suffer from vision loss due to corneal disease or injury?

A

10 million

228
Q

How many corneal transplants are performed in the US?

A

40000

229
Q

What is the cornea?

A
  • The cornea is the transparent convex tissue which together with the Sclera forms the outer shell of the eyeball. The sclera covers 5/6 and the cornea 1/6
  • The cornea covers the iris
230
Q

What is the structure of the cornea?

A

There are three main layers: corneal epithelium, stroma and corneal endothelium. Separated by Bowman’s membrane and Descemet’s membrane

231
Q

What are the functions of the cornea?

A

Aid sight

Protect the eye

232
Q

How does the cornea aid sight?

A
  • Due to its transparency (99% of incidence light). The light comes through the cornea onto the lens and focus on the retina
  • It has a major refractive power (2/3s of eyes refractive power)
233
Q

What is the corneal epithelium?

A
  • Outer most layer of the cornea
  • Thickness of 50micrometres
  • Highly innervated
234
Q

What are the functions of the corneal epithelium?

A
  • Prevent fluid loss
  • Create a barrier to pathogens
  • Respond rapidly to wounding (needs to establish the permeable barrier)
235
Q

What is the structure of the corneal epithelium?

A
  • There are 2-3 layers of superficial cells which create tight junctions and generates the primary barrier in the epithelium. On the outer surface, there are microvilli which are covered in Glycocalyx which maintains the tear film of the eye
  • Under the superficial cells there are the wing cells and then basal cells. Basal cells communicate through desmosomes and are attached to Bowman’s membrane
236
Q

Does the corneal epithelium regenerate?

A

The whole layer regenerates every 5-7 days

237
Q

What is the storma?

A

Bulk of the cornea with 90% of the corneal thickness

Relatively acellular and contains mainly collagens, proteoglycans and glycoproteins

238
Q

What is the function of the stroma?

A

Functions include strength and transparency

239
Q

How does the structure of the stroma allow for transparency of the cornea?

A

At the microscopic level, the collagen fibrils are arranged in autologous layers. This is important because the geometric organisation of the collagen means that it transmits most the light without scattering so is transparent

240
Q

Does the stroma contain any cells?

A

Even though mainly acellular, does contain keratocytes which are long, thin and flattened cells which synthesise and maintain ECM

241
Q

What is the stromal endothelium?

A

A single layer of cells in the cornea which are metabolically active

242
Q

What is the function of the stromal endothelium?

A

Functions are to maintain stromal hydration

243
Q

How does the stromal endothelium allow for corneal transparency?

A

Maintains stromal hydration to maintain transparency. To do this it has evolved as a leaky pump

  • It is avascular so nutrients must be delivered from either the tear fluid or the aqueous humor
  • It allows solute and nutrients from the aqueous humor but has to maintain stromal hydration to stop the ECM from collapse
  • They are therefore very metabolic so that the solutes are constantly coming in and then water is being pumped out
244
Q

What is the most densely innervated body structure?

A

The cornea

245
Q

Why does the cornea need to be innervated?

A
  • Blink reflex
  • Wound healing
  • Tear production
246
Q

Is the cornea innervated?

A

Yes highly but avascular to maintain transparency

247
Q

How does the cornea get nutrition and oxygen?

A

Through tear fluid and aqueous humor

248
Q

What properties would an ideal corneal substitute have?

A
  • Transparent
  • Refractive
  • Prevent formation of blood vessels but allow for innervation
  • Low immunogenicity
  • Mechanical properties to allow nutrient delivery
249
Q

What layer of the cornea is most amenable to regenerative medicine therapies?

A

Epithelium as it regenerates normally every 5-7 days. This tells us that there are stem cells available which could be utilised for a treatment

250
Q

Why is corneal transplantation not a good enough solution to corneal injuries?

A

Only available to 7% of affected people world-wide so needs for alternative

251
Q

What is a Ketatoprosthesis?

A

A medical device to replace the cornea for people who have repeated failed grafts
- Optical polymethylmethacrylate to replace the cornea and then a titanium ring to keep it in place

252
Q

What is the problem with Ketatoprosthesis?

A
  • It is susceptible to inflammation and glaucoma

- Required a life-long range of antibiotics

253
Q

What type of stem cells are found in the limbus of the eye?

A

Limbal Epithelial Stem Cells

254
Q

Where is the niche for the limbal epithelium stem cells?

A

In the limbus of the eye

  • Limbus is sometimes quite pigmented so can see that it is undulated and it is here where the niches are.
  • It is located in the corneal rim at the border between the sclera and cornea
255
Q

How do gimbal cells regenerate the corneal epithelium?

A
  • The stem cells can divide asymmetrically and the daughter cells moves out and differentiates.
  • This gives rise to cells within the basal layer which are still proliferating.
  • Transit amplifying cells will give rise to wing cells which are post-mitotic.
  • Renewal of the whole tissue comes from the Limbal stem cells as eventually transit amplifying cells will die off
256
Q

What are Limbal stem cell deficiencies?

A

Creates a vascularised tissue as the cells migrate over the cornea – conjunctiva overgrowth
- Causes vison loss

257
Q

What causes Limbal stem cell deficiencies?

A

Most are due to injury bit there are some conditions such as Aniridia which cause it

258
Q

How are Limbal stem cell deficiencies treated?

A

Holoclar

  • Grafts from cultured cells from the non-diseased eye
  • Biopsy from non diseased eye. Can be culture in vitro to form epithelial cell sheets. Usually detach cells using trypsin but with sheets we want them to remain together so use temperature responsible dishes. At 37 degrees’ cells can attached and at 20 the dishes become hydrophobic so the cells can be lifted off the plate
259
Q

How was Holoclar developed?

A

There was a proof of concept study: Pellegrini et al, 1997

75 out of 104 patients showed improved vision

260
Q

What are the disadvantages of Holoclar

A
  • If someone had a genetic condition then the cells would still be injured.
  • There will also be a delay as won’t start the making the treatment until know the injury.
  • Have to take a graft from the healthy eye which could result in damage
261
Q

What other cells could be used to treat Limbal stem cell deficiencies treated?

A

Mucosal cells

  • These cells don’t scar so won’t harm the healthy eye
  • However, there is a risk of neovascularisation of the cornea
  • Doesn’t do the same thing as Limbal stem cells so may need to investigate reprogramming
262
Q

How do stromal injuries affect vision?

A

If damaged can create myofiberblasts which would usually apoptose after wound healing. In some pathologies, they continue to secrete ECM which will be an irregular shape and stop the transparency of the cornea

263
Q

What are the two regenerative approaches to treat stromal injuries?

A

Biomaterials

Cell-based

264
Q

Give a study that used biomaterials to treat a stromal injury?

A

Fagerholm et al, 2014
- Used recombinant human collagen as a cell free scaffold
- Upon implantation, there was endogenous cell recruitment and began to remodel the matrix and lay down new ECM. A new cornea was formed
o Because it is acellular, there is no need for immune suppression
o There was innervation of the scaffold
o The visual acuity wasn’t great so the scaffold didn’t have the correct mechanical properties

265
Q

How did Islam et al, 2018 improve upon Fagerholm et al, 2014 treatment for stromal injuries?

A
  • Cell free corneal implants consisting of recombinant human collagen and a synthetic lipid were grafted by keratoplasty into corneas of unilaterally blind patients at high risk pf rejecting donor allografts
  • Followed for 24 months. Patients with acute disease were relieved of pain 2 weeks post operation. Corneas with immune or degenerative conditions showed no visual improvement
  • 3 out of 6 patients showed significant vision improvement
  • Further testing is needed but this study showed that cell free implants are potential safe efficacious options for treating high risk patients
266
Q

How can cell based therapies be used to treat stromal injuries?

A

Found that in the limbus there are also Limbal stromal cells which remodel stromal scarring in model animals and supress fibrotic scar formation

267
Q

What are the remaining challenges when treating corneal defects?

A

Endothelium does not regenerate

  • Could potentially transform them with oncogenes but this could be problematic in therapies
  • Could use pluripotent stem cells
268
Q

Give an example of the clinical need for tissue engineering?

A
  • Osteochondral defects affect both the articular cartilage and the underlying subchondral bone and leads to mechanical instability of the joint and the risk of inducing osteoarthritic degenerative changes
  • To repair these large defects, there must be transplantation of osteochondral units. However, the problems with these transplants include limited material available, donor site morbidity and difficulty in matching the topology of the grafts with the injured sites
  • 3D tissue grafts of pre-defined size and shaped can be engineered by combining the patient’s own cells with 3D porous biomaterials and can overcome the limitations of transplanting osteochondral units
269
Q

Why is the cell seeding of scaffolds so important?

A
  • Cell seeding is the dissemination of isolated cells within a scaffold. It is the first step in 3D culture and plays a crucial role in determining the progression of tissue formation
  • The initial distribution of cells within the scaffold after seeding has been related to the distribution of tissue subsequently formed within engineered constructs suggesting that uniform cell seeding could establish the basis for uniform tissue generation
270
Q

What is the problem with static cell seeding?

A

Static loading of cells by micro pipetting is the most common cell seeding method. However, it is often associated with low seeding efficiencies and non-uniform distributions within scaffolds.

271
Q

What bioreactors can be used to overcome the limitations of static cell seeding?

A
  • Spinner flask bioreactors can be used which will uniformly distribute cells into scaffold with high porosity and high interconnectivity. If the scaffold is thick or less porous then spinner flask bioreactors also lead to non-uniform cell distribution.
  • Can also use perfusion bioreactors which enhances the convective transport of the cells throughout the entire scaffold volume. Using perfusion bioreactors for seeding is useful because culturing of the construct can also be performed under perfusion within a single unit
272
Q

How can the limitations of mass transport of nutrients in scaffolds be overcome?

A

Rotating wall bioreactors can also enhance mass transfer of nutrients. Culturing in a rotating wall bioreactor resulted in cartilaginous constructs having better biochemical and biomechanical properties than static or spinner flask cultures

273
Q

What is the major challenge of using engineered grafts in clinic?

A

One major challenge is to translate research scale production models into clinically applicable designs that are reproducible, economically acceptable and maintains good manufacturing practice

274
Q

What was the aim of Takagi et al, 2016?

A
  • The integumentary organ system has important roles in protecting, waterproofing and thermal regulation and regenerative therapies could provide important treatments for burns and scars as well as models for drug screening
  • They therefore generated a 3D bioengineered integumentary organ system from IPS cells which included appendage organs such as hair follicles. They used a novel in vivo transplantation model called clustering-dependant embryoid body transplantation
275
Q

How did Takagi et al, 2016 produce a bioengineered skin replacement?

A
  • Epithelial germ layers were induced from iPS cells. Saw that both epithelial and mesenchymal cells were generated in the embryoid bodies through the presence of integumentary markers Sox2 and p63 and neural crest markers Snail and Twist.
  • Transplanted the EBs into nude mice. Formed a teratoma consisting of all three germ layers. When EBs were transplanted with collagen gel, which contained EBs cultured for 7 days under non-adhesive conditions, organ formation occurred in 4 out of 9 explants
276
Q

Was Takagi et al, 2016 bioengineered skin replacement successful?

A
  • The bioengineered integumentary organ system was fully functional in nude mice and can be properly connected to surrounding host tissues without tumorigenesis
  • The bioengineered hair follicles show the proper hair eruption and cycle
  • Potential uses of this bioengineered organ system include in vitro assay system, animal model alternative and organ replacement therapy
277
Q

How common are peripheral nerve injuries?

A

Significant problem in society with 9000 cases in the UK each year

278
Q

Why are peripheral nerve injuries a major burden on society?

A

Occurs mainly in the young population

  • This is unlike other injuries. This is because it occurs mainly in car accidents
  • Therefore can’t work and become a burden
279
Q

Give the structure of a peripheral nerve

A
  • Axons are surrounded by myelinating Schwann cells and are enclosed by endoneurium (connective tissue)
  • They are bundled together into fascicles which are bundles together by Perineurium
  • Epineurium then groups the fascicles creating nerve cable
280
Q

What are the three types of peripheral nerve injuries?

A

Elongation
- Connective tissues allow 10-20% elongation before structural damage occurs
- Severe lesions that disrupt the axon
Laceration
- 30% of nerve injuries
Compression
- External mechanical pressure in the conductive membrane

281
Q

What are the three grades of nerve injuries?

A

Neuropraxia
Axontmesis
Neurotmesis

282
Q

What is neuropraxia?

A
  • No/little structural damage and no loss of nerve continuity
  • Symptoms are transient and reversible
  • E.g. carpal tunnel syndrome
283
Q

What is axontmesis?

A

Complete interruption of the axon and its myelin sheath

Perineurium and Epineurium intact

284
Q

What is neurotmesis?

A

Nerve and the surrounding stroma are completely disconnected
No spontaneity recovery
Weakness and atrophy

285
Q

What occurs in the PNS to allow nerve regeneration?

A

Wallerian degeneration

286
Q

Outline Wallerian degeneration

A
  • Distal axons come away from the body and start to degenerate to produce debris
  • Debris is phagocytosed by macrophages
  • Schwann cells will start to clear up the debris. They proliferate and align themselves in tracts called the bands of bunger to create a scaffold for regenerating axons guiding them to the target site
  • They produce neurotrophic factors which attract axons
287
Q

Why is there more success in PNS regeneration than in CNS

A

No wallerian degneration

  • Macrophages infiltrate much more slowly due to the BBB meaning they cannot prevent the inhibitory action of myelin.
  • Reactive astrocytes also produce glial scars that inhibit regeneration in the CNS
288
Q

What are the main approaches to neural tissue repair in the PNS?

A

Surgical reconstruction
Grafts
Nerve conduits

289
Q

How does surgical reconstruction allow for PNS nerve regeneration?

A
  • The severed nerve stumps will suture the individual fascicles back together. This is only possible if the never stumps are close to each other.
  • If the gap is too large then pulling them together will reduce the blood flow. Stretching by 8% reduced blood flow by 50%.
290
Q

What are the types of grafts that can be used to aid the PNS nerve regeneration?

A

Autologous
- A nerve segment from elsewhere in patient’s body
- Advantage is that there is a low risk of immune rejection
- Disadvantages include loss of function at the donor site, two surgeries required, limited size and type
Allogenic
- Same species donor
- Advantages include no secondary surgery and no loss of function of donor site
- Disadvantages include higher risk of rejection and limit in availability

291
Q

What are nerve conduits and how do they aid PNS nerve regeneration?

A

Hollow structures that nerves are placed in to guide the regenerative nerves. This will increase the concentration of intraluminal proteins (growth factors) as they are trapped within the structure

292
Q

Outline nerve regeneration in the presence of nerve conduit

A
  • When the stumps are placed inside the conduits, the conduit fills with plasma which is full of ECM proteins and fibrin. Fibrin form fibrin cables which from a scaffold allowing Schwann cells to migrate and guide axons. This will allow regeneration. The result will be thinner than the original tissue but regeneration does occur
  • Whole regeneration process takes years but begins within hours after implantation
293
Q

What properties would an ideal a nerve conduit possess?

A
  • Permeable (porous) – provide nutrients to Schwann cells
  • Long term degradable – Need regeneration to take place but not have to remove by another surgery
  • Biocompatible – low immunogenicity
  • The material needs to be suturable to attach the nerve stumps
294
Q

Which classes of biomaterials could be used for nerve conduits?

A

Fabricated

Decellularised nerve conduits

295
Q

What type of fabricated materials can be used for nerve conduits?

A
Natural
- Collagen
- Silk
Synthetic 
- Biodegradable materials 
- Electrically active materials 
Non-biodegradable materials 
- Semi-synthetic
296
Q

What is the advantage of using fabricated materials for nerve conduits?

A

More in control of their structure

297
Q

How can decellularised nerves be used for nerve conduits?

A
  • Retains the ECM architecture and removes antigens (stops immune response)
  • It is hollow so not in the way of regenerative axons and less immunogenic than allogenic approaches
298
Q

What are the limitations of nerve conduits to aid PNS nerve regeneration?

A

The chance of successful regeneration with nerve guides is reduced, once an injury gap reaches a certain value (critical gap length)

  • At short gap lengths, the fibrin cable is robust enough to provide a platform for regeneration
  • At longer lengths, thinning restricts regeneration
299
Q

What is the critical gap length of nerve injures?

A

This is the length at which regeneration occurs 50% of the time

300
Q

What approaches are being used to try increase the critical gap length?

A
  • Creating conduits that are no longer hollow but instead contain ECM components, intraluminal supports, neurotrophic factors and cells which promote regeneration
  • Approaches use one or more of these (combinatorial approaches)
301
Q

How can ECM in nerve conduits increase the critical gap length?

A
  • Effective matrices are weak, viscoelastic hydrogels with a high-water content (high concentrations would prevent axonal penetration)
  • Matrigel (ECM proteins produced by mouse sarcoma cells) promotes nerve regeneration. Not very robust as there as a large batch to batch variably as come from mouse sarcoma cells. Therefore, not ideal for clinical use
302
Q

How can Intraluminal support in nerve conduits increase the critical gap length?

A
  • Tunnels within hollow tube to recreate the natural scaffolding of the nerve
  • This has been used successfully in clinic. Post-operational tests showed that nerve functionality increased
303
Q

How can neurotrophic factors in nerve conduits increase the critical gap length?

A
  • Support axonal growth
  • Migration and proliferation of Schwann cells
  • Increase neuroprotection modulation of intrinsic signalling pathways
  • Need for controlled release of these factors
  • NGFs have little influence over motor neurons
304
Q

How can different cells in nerve conduits increase the critical gap length?

A
Schwann cells
- Critical for successful nerve regeneration 
- Bands of Bungner 
- Secrete neurotrophic factors 
- Proliferate 
Stem cells 
- Used for grafting
305
Q

Give evidence for how neutrophil factors increase the effectiveness of nerve conduits?

A
  • NGF plays an important role in the survival of sensory neurons and in the extension of the axons. It is therefore being used in nerve conduits
  • Incorporating NGF results in an increase in functional recovery compared to control groups
  • It influences the proliferation and migration of Schwann cells and facilitates regeneration
306
Q

Give a study into the importance of vasculature in the body?

A

Folkman et al, 1971

  • Tumour growth dependants on the angiogenesis
  • Patient had a tumour of the eye which was well vascularised. Around it there were several small metastases which had a periphery of alive cells but the centre was necrotic. These metastases did not have vasculature meaning the centre cells could not be nourished
  • Small tumours can be fed by diffusion but when it grows it requires vasculature to keep the cells alive
  • This knowledge could offer a potential treatment for tumours – block the nourishment of the cells
307
Q

Why is vascularisation needed in tissue engineering constructs?

A
  • Avoid graft necrosis
  • Generate thicker tissues
  • Help graft innervation
  • Improve graft function
308
Q

What are the types of blood vessels?

A
  • Macro vessels: Arteries and veins
  • Micro vessels: arterioles and venules
  • Capillaries
309
Q

What is the role of capillaries?

A

Responsible for the transfer of the nutrients. This is achieved through the difference of pressure in the capillaries and the interstitial fluid

310
Q

Give a brief structure of capillaries?

A

Single layer of endothelial cells surrounding the lumen which secrete anti-thrombotic factors to stop blood clotting

311
Q

What are the three types of blood vessel formation?

A
  • Vasculogenesis
  • Angiogenesis
  • Arteriogenesis
312
Q

What is Vasculogenesis?

A
  • De novo blood vessel formation from progenitor cells
  • The embryo becomes large enough to need vasculature. Formed mesoderm precursors which differentiate into hemangioblasts which move to certain sites in the embryo to form tubes and then the primary capillary plexus
313
Q

What is Angiogenesis?

A

New blood vessel formation via extension of existing blood vessels

314
Q

Outline angiogenesis

A
  • When there is a lack of oxygen (hypoxia). VEGF signals to endothelial cells which secrete enzymes, matrix metalloproteinases, which act on the basement membrane and create punctures.
  • The endothelial cells proliferate and create vasculature sprouts which elongate towards the hypoxic signal and form a new lumen
  • Mature by the recruitment of smooth muscle cells and pericytes
  • This will result in the place being less hypoxic meaning that the hypoxia inducible factor decreases then angiogenesis is turned off (negative feedback loop)
315
Q

Where does angiogenesis occur?

A
  • Occurs during wound healing and ovarian cycle
316
Q

What is VEGF?

A

Vascular endothelial growth factor (VEGF) – Involved in endothelial cells proliferation, migration and differentiation
angiogenesis factor

317
Q

What is HIF?

A

Hypoxia inducible factor (HIF) – stimulation of transcription of VEGF, PDGFR, TFG-alpha, EGF and erythropoietin in response to hypoxia

318
Q

What is PDGF?

A

Platelet-derived growth factor (PDGF) – Recruitment and proliferation of smooth muscle and pericytes - this matures developing vessels

319
Q

What is Angiopoietin?

A

Involved in angiogenesis

- Regulation of endothelial cell survival, sprouting, pericyte recruitment

320
Q

What are matrix metalloproteinases?

A

Secreted in response to hypoxia and involved in angiogenesis
- Basal lamina degradation, ECM remodelling

321
Q

What are the types of VEGF signalling?

A
  • VEGFR2 involved in angiogenesis, VEGFR1 involved in Vasculogenesis and VEGFR3 in lymphangiogenesis
  • VEGFR2 activation results in endothelial cell proliferation, migration and survival
322
Q

What is Arteriogenesis?

A

Maturation of blood vessels via increasing the lumen

323
Q

Outline Arteriogenesis

A

Occlusion of an artery which prevents blood flow from an artery which will exert certain amounts of sheer stress onto its endothelial cells. This releases TGFbeta which causes degradation of ECM and promotes remodelling through the proliferation of endothelial and smooth muscle cells. This increases the lumen sizes and forms an artery

324
Q

What are the main differences between the three processes that form blood vessels?

A
  • Vasculogenesis doesn’t require any pre-vasculature
  • The key signals are different: Vasculogenesis – growth factors in the embryo, Angiogenesis – hypoxia, Arteriogenesis – sheer stress of the vessel
325
Q

What are the two main strategies used to create scaffolds that facilitate vascular ingrowth?

A

Scaffold design
- Different ways to make porous scaffolds which doesn’t compromise mechanical properties
Scaffold functionalisation
- Can use different molecules to promote vascularisation e.g. VEGF, PDGF, bFGF
- It is crucial to have controlled release of growth factors – the timing is important

326
Q

How did Richardson et al, 2001 create a mature vasculature network?

A

They created a polymeric system for dual growth factor delivery

  • VEGF is the key initiator of angiogenesis but the vessels would not be mature without PDGF. They hypothesised that both factors are required and should be delivered in a certain way
  • Delivered by two stage release of growth factors
327
Q

What did Richardson et al, 2001 find?

A

Results showed the formation of a mature vascular network when there was the dual release of VEGF and PDGF. This was not seen in just one of the growth factors

  • The distinct kinetics of delivery allowed mimicking of the natural process
  • Highlighted the importance of multiple growth factor delivery for engineering artificial tissues
328
Q

What are the disadvantages of facilitating vasculature ingrowth of a scaffold?

A
  • Time consuming
  • Slow growth rate
  • May not be sufficient to prevent necrosis in 3D constructs
329
Q

What other strategies can be used to ensure scaffolds have vasculature?

A

Prevascularisation strategies

  • In vitro prevascularisation
  • In vivo prevascularisation
330
Q

Outline In vitro prevascularisation

A
  • The tissue engineering construct is cultured in vitro to build prevascularised structure
  • The network can create a connection with the existing vessel in tissue (anastomose). This is faster than the new blood vessel formation
331
Q

What are the problems with in vitro prevascularisation?

A

There is a problem with how cells would be sources in the clinic and issues with using mature endothelial cells

332
Q

What techniques can be used for in vivo prevascularisation?

A
  • Angiogenic ingrowth
  • Flap technique
  • Arteriovenous (AV) – loop technique
333
Q

Outline angiogenic ingrowth as a technique for in vivo prevascularisation?

A
  • Place scaffold in enriched vascularised network and micro vessels will ingrowth from the host. After this occurs, can remove and transfer to the defect site
  • This method involves three surgeries so is not likely to be used
334
Q

Outline the flap technique as a technique for in vivo prevascularisation?

A
  • A scaffold is implanted into a muscle flap which is then vascularised from the host
  • The whole flap is then transferred to the site in need of repair. This allows larger vessels to be transferred
  • Problem is that you are sacrificing the while muscle flap
335
Q

Outline the arteriovenous (AV) – loop technique as a technique for in vivo prevascularisation?

A
  • Uses a vein or synthetic graft to form a shunt loop between an artery and vein which leads to spontaneous sprouting of vessels
  • The loop is placed in a protected chamber and can then be transferred to repair site
  • This doesn’t have donor site morbidity
336
Q

What happens in the body upon injury?

A

Injury of the body triggers reaction that begins with local interactions and then becomes systemic due to the immune cells spreading around the entire body. The systemic interactions determine the success of the transplant and can lead to rejection

337
Q

What responses occurs upon implantation of tissue engineered structure?

A

The knowledge of how the body may react comes from medical devices for biomaterials and the inflammatory response. Medical devices usually contain living cells. The way the body responds to living cells is based on the immune response

338
Q

What are the two main things that occur when biomaterials are implanted in the body?

A

Effect of biomaterial on the body

Effects of body on biomaterial

339
Q

What effect does the biomaterial have on the body?

A
  • Changes wound healing
  • Infection
  • Toxicity
  • Tumourigenicity
  • Particle embolization
  • Hypersensitivity reactions
340
Q

What effect does the body have on the biomaterial?

A
  • Enzyme degradation
  • Calcification
  • Abrasion
  • Corrosion
341
Q

What occurs to the healing response when biomaterials are transplanted?

A

The foreign body response

342
Q

What is the foreign body response?

A
  • Transplantation results in tissue injury
  • Blood vessels damage and blood will flow out. ECM proteins such as fibrin lay down a scaffold on the material which enable cells such as monocytes to adhere to the material
  • Macrophages are phagocytic and clear up the debris
  • Biomaterial is so large that the macrophages can’t digest then they undergo frustrated phagocytosis. It attracts other macrophages to the site.
  • Macrophages fuse with other macrophages (granuloma) and forms a foreign body cell and release agents such a super oxides and free radicles which damage the surface of the material
  • Fibrous encapsulation then occurs
343
Q

What is fibrous encapsulation?

A
  • Fibroblasts lay down collagen and ECM components which leads to isolation from biomaterial to the local tissue environment.
  • The granuloma ages and increases vascularisation. Endothelial cells form tubes which form into capillaries.
  • This is an evolutionary response to protect the body but now causes problems with medical devices
344
Q

When is a material too large to be phagocytes by macrophages?

A

Larger than 10 micrometres

345
Q

How does the foreign body response affect construct integration?

A
  • The way the body responds is hard to predict but some biomaterials induce larger responses
  • Minimal foreign body reaction leads to optimal healing so need to identify the appropriate biomaterial for each application to reduce this
346
Q

How common is infection when constructs are implanted?

A
  • Implant infections are common and are sometimes located to the implant site but can be more serious and lead to sepsis
  • There are however some materials that rarely cause infection
347
Q

What are bacterial biofilms?

A
  • Communities of bacteria grown on abiotic materials. This is an issue for infection
  • Thought to be an ancient evolutionary response that allow bacteria to behave like multicellular organism by embedding themselves in extracellular polymeric substance (slime) and allows them to increase survival
  • They adhere to the material and secrete slime allowing them to adhere to each other and irreversibly adhere to the material.
  • They proliferate and form multi-layer structures. They can become planktonic again and an colonies other sites so that the film is propagated.
348
Q

Can bacterial biofilms be removed with antibiotics?

A

Not usually

349
Q

What bacteria are prone to creating bacterial biofilms?

A
  • Staphylococci (lives on the skin) are particularly prone to creating biofilms. This is an issue when the skin is damaged and can then access the biomaterial
  • This problem is enhanced due to the development of the anti-biotic resistant strains
350
Q

What medical devices are prone to bacterial biofilms?

A

Orthopaedic prosthetic joint infections also have this problem and it is difficult to diagnose. Typically treated with systemic antibiotics or remove the biomaterial

351
Q

How do either bacterial biofilms or endogenous cells colonise the biomaterial?

A

There is a kind of race between the bacteria and host cells to reach the surface and whichever reaches first populates the surface

352
Q

How can we ensure thee cells adhere before the bacteria?

A

Cells do not interact directly interact with materials, instead a layer of protein adheres to the surface and is absorbed by the material. Could therefore use non-fouling surfaces which are resistant to protein absorption to stop all cells adhering. Then micro stamp it with RGD domain to allow cells to attach

353
Q

Explain a study that created a biomaterial which repels bacteria colonisation while allowing mammalian cell adherence?

A

Differentially instructive extracellular protein micro-nets (Farugui et al, 2014)

  • Used peptide assembled into structures (SaNet) with differential instructive matrix which enhance mammalian cell adhesion but resist bacteria adhesion
  • This is achieved due to its similarities to native extracellular matrices. This similar protein expression allows it to maintain the biofunctional characteristics: anti-microbial carpets and filopodia recognised adhesion points. This therefore blocks bacteria colonisation and enhances mammalian cell binding
  • This was shown using SaNet in comparison to collagen and fibronectin. Bacteria cells did not adhere at all
354
Q

How can biomaterials be toxic?

A
  • Wear and tear of the biomaterial can lead to the release of by-products. In metal-on-metal hip replacements caused the release of cobalt and chromium into the blood.
  • This doesn’t cause an immune reaction but bind to other proteins in the blood which causes them to be perceived as foreign by immune response. This results in failure of the replacement
355
Q

How can thromboembolic complications occur with biomaterials?

A

Stents
- An artery clot will be cured using a stent insertion. The stent is covered in anti-coagulant cells. This is rare with severe clinical consequences

356
Q

Are biomaterial transplants related to tumour formation?

A

Whether a tumour is caused by an implant is difficult to prove. Animal models have not been useful and the mechanism are not clearly understood

357
Q

Why is better to use autologous cells in biomaterials?

A
  • Autologous cells to stop the immune response and remove the need for immunosuppressants.
  • This is sometimes not possible due to genetic diseases in the cells or lack of supply. Sometimes necessary to use allogenic strategies
358
Q

What is the Edmonton protocol?

A

Treatment for diabetes

- Showed islet cells can be transplanted obtained from cadavers. This requires immunosuppression otherwise rejection

359
Q

How else can diabetes be treated that doesn’t involve rejection?

A
  • Do not need a structure just the cells so in theory it doesn’t matter where the cells are placed as long as it leads to the release of insulin.
  • Therefore, investigated the immunoisolation approach
360
Q

How is the immunoisolation approach being investigated as a treatment for diabetes?

A
  • Islet cells are placed in a device which has a semi-permeable membrane
  • Can receive nutrients through the membrane and secrete insulin but T cells and antibodies cannot enter and kill the cells
  • This appears like a viable approach that is in clinical trials
361
Q

What company is currently in clinical trials for the immunoisolation approach?

A

Viacyte is using hES cells to derive the beta cells and placing them in a semi-permeable membrane

362
Q

What needs to be prevented for the immunoisolation approach to successfully treat diabetes?

A
  • For this to be viable, fibrous encapsulation must be prevented as insulin would not reach the blood stream and the cells in the device could not be nourished. There are some allogenic materials that prevent this form occurring.
  • Could use genetically modified cells
363
Q

What are the typical causes of acute peripheral nerve injury?

A

Road traffic accidents, work accidents and domestic accidents

364
Q

What percentage of people show permanent sensory or motor deficit following nerve repair?

A

80-90%

365
Q

Why can’t peripheral nerves be replaced with stem cells?

A

A single neuron can be very long (metre) – makes it difficult to treat

366
Q

What are the two current ways nerve gap injuries are repaired?

A

Suturing the proximal and distal ends

Autografting

367
Q

How can a nerve guidance channel be made precisely?

A

Micro-sterolithography

  • Used UV light to produce a photocatalytic reaction and give a sharp point for which to direct a beam. Can move this through a material
  • Take a liquid version of a material that is biocompatible (PEG, PLA, PCL) which are inert. Use a photo catalyst.
  • Move the stage with the liquid through the UV beam to create a solid – this gives precise controls over the physical dimensions – can get down to 50micrometres wall width.
368
Q

How were the nerve guidance channels improved upon?

A

Added Guidance scaffolds

- Used aligned polycaprolactone fibres made by electrospinning to create fibres of different diameters

369
Q

Did adding aligned polycaprolactone fibres to nerve guidance channels improve neurone growth in vitro?

A

Took some neuronal cells and grew on the fibres and looked for neurites that grew from the cells. Found that there was a correlation between the length of the neurite and the diameter of the fibre

370
Q

Did adding aligned polycaprolactone fibres to nerve guidance channels improve neurone growth in vivo?

A

Dorsal root ganglion cultures

  • Coated in laminin
  • Label for nuclei, axons and Schwann cells. The Schwann cells almost always physically contact the axons.
  • Took the fibres and looked if axons and Schwann cells grow along them. They did and Schwann cells were in contact with the axons
371
Q

Why was adding schwann cells to the nerve guidance channel difficult even though it has been shown to increase regeneration?

A
  • Need to make sure the conduit is not to expensive so must limit the complexity but still make it clinically viable
  • Problem of cell sourcing: can’t be from a donor so then it must be autologous. This is the same problem as the start (grafting)
  • Took long to culture and often became containated
372
Q

How were they able to produce Schwann cells in culture that could be used in the nerve guidance channel?

A
  • They express the enzyme D-amino acid oxidase which secretes an amino acid from the valine form. Contaminating fibroblasts do not have this enzyme so if culture in a medium with D-Valine then they will die.
  • Removed many of the components of the medium making it fit the clinical need.
373
Q

How has a regulatory framework evolved over time?

A

Evolved over time as a result of incidents
- Elixir sulphanilamide incident (1937): Treat streptococcal infections for children. They dissolved safinamide in anti-freeze and raspberry flavour and caused the death of over 100 children
- The Nuremberg Code (1946): esting should be done only with informed consent
- The thalidomide incident (1956): Caused severe developmental defects of the pregnant women’s children so have to look at long term safety now
Result of this is Good Clinical Practice which ensures that the data from clinical trials will be reliable and robust

374
Q

What are the stages in drug development?

A
Lab discovery 
Pre-clinical studies 
- In vitro: Cell lines
- In vivo: Animal models
Clinical trials 
- Phase 1: Done on healthy volunteers to ensure safety
- Phase 2: Small group of people with the disease
- Phase 3: Larger group
375
Q

What regulatory agencies ensure good clinical practice?

A

EMA, FDA
‘The international conference on harmonisation of technical requirements for registration of pharmaceuticals for human use’ ensures that there are not major differences between different countries so the whole population can benefit

376
Q

What category does tissue engineering fall into in the legislative framework?

A

ATMP is a medicinal product which is either

  • A gene therapy medicinal product
  • A somatic cell therapy medicinal product
  • A tissue engineered product
377
Q

Why do tissue engineering therapies need special regulation?

A

Use of cells
- Survival: The cells can survive for the life of the patient and could acquire genetic and epigenetic changes
- Evolving functionality: Cells may evolve to continue proliferation and form a tumour
- Migration: Cells can bio distribute and end up in the wrong environment. This would interact differently and could have adverse effects
No standard set of preclinical tests
- Each application is very different and has never been done before
- Need case by case considerations

378
Q

What are the overarching regulatory requirements for new treatments?

A

The establishment of manufacturing process and control
- Product consistency: Needs to be reproducible so trained in the same way with the same tools and products used
Evidence for safety and efficacy in preclinical models
- Involves testing in animal models to model human conditions and to consider safety and functionality.
- Aware of limitations of the model used
Clinical trials on human participants

379
Q

How do clinical trials for tissue engineering differ from drug trials?

A

Exploratory trial
- Application requires surgery so using healthy volunteers is not appropriate
- Primary concern is safety but is carried out on actual patients
Dosage
- Not as straight forward as in small molecule drugs
Defining the comparator
- Usually compare to the gold standard drug – is it better
- Harder in regenerative medicine as it requires surgery which can be done differently depending on the skill of the surgeon making comparison difficult
Randomisation
- Important that it is independent from the investigator so that the results are objective
- Cannot do a blind study where some get treatment and some get control in this aspect

380
Q

What is meant by the risk benefit assessment?

A
  • Depends on the health and age of the patient and other treatments available. Has to occur on a case to case bases
  • Hospital exemption – experimental therapies can be used with consent of the hospital and patients
381
Q

What is meant by the valley of death when trying to commercialising a tissue engineering product?

A
  • The gap between discoveries of therapeutic leads in the labs that never meet the patient
  • Early phase therapeutic development is carried out in universities but they are not equipped to carry out the preclinical or clinical trials. Usually investors or pharmaceutical companies do this e.g. Big pharma.
  • However, some companies do not invest in tissue engineering products as there is no guarantee that it will be successful and will therefore not make any money
382
Q

What are the key bottlenecks in tissue engineering commercialisation?

A
  • Financial rewards are not guaranteed
  • The government is the highest payer so the industry is heavily influenced by politics
  • 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
383
Q

What is the problem with model organisms?

A
  • Need to use model systems to model diseases and processes but it is difficult to find models that are physiologically relevant and can be used
  • As the physiologically relevance increases, the experimental tractability decreases
384
Q

How would a more appropriate model improve drug development?

A
  • Companies screen 5-10000 compounds. Usually on one of these found to be approved. This translates a huge time to lab to the market and a huge cost in development
  • Drugs end up being withdrawal from market e.g. VIOXX lead to 88 thousand people having heart attacks and 38K died
  • The idea would be that the models used would be more predictive to the outcomes in humans
385
Q

What are the disadvantages of the current models used?

A

In vitro models
- Primary cells: Not representative to the entire body and difficult to source them and every source will have batch-batch variability
- Immortalised cell lines: May change the phenotype of the cells and change their behaviour – may test drugs on something that doesn’t reflect in vivo
In vivo models
Animal models: Even primates do not necessarily give the correct outcome E.g. Monoclonal antibody – 6 patients died in phase 1. It was tested in primates but in humans it caused major organ failure.

386
Q

What is the problem with organoids?

A

There isn’t a good control on spatial relationship between cells – organoids self-organise so we are not on control. Organs on chips were brought in to overcome this

387
Q

What are organs-on-chips?

A
  • Not aiming to replace an organ but instead to be used in vitro. They mimic the minimal units to model the organ function
388
Q

How are organs-on-chips fabricated?

A
  • Polymers (PDMS) mixed with crosslinking agent and pour onto the mould and heat up to solidify
  • Can then peal this off and place on a glass slide
  • PDMS is see through meaning can minor cells grown under the microscope and is biocompatible
  • Puncture several holes to allow the liquid in and out –perfusion system
389
Q

What are the microfluidics of organs-on-chips?

A
  • How fluid behaves in very small volumes

- They follow laminar flow rather than turbulent flow. This gives more control over the fluid and create gradients

390
Q

What are the typical components of an organ on a chip?

A
  • Geometrical confinement and patterning (Control over where the cells are places)
  • Presence of flow
  • Environmental control (pH and temperature)
  • Sensors and physiological readouts
391
Q

What important features of lungs must be mimicked when making a lung on a chip?

A
  • Lungs are used for gas exchange and must have a large surface area to volume ratio. This achieved by branching structure and alveolar epithelium.
  • The alveolus is lined by alveolar epithelium.
  • Due to breathing, there is constant cyclical stretching of the alveolus. This is important because cells need to encounter these in vivo functions to develop
392
Q

Outline the lung-on-a-chip

A
  • Epithelium cells of alveoli and placed on ones side of the membrane and on the other site endothelium cells to represent capillaries. This produces the alveoli capillary interphase and flow air in the top chamber and media in the bottom (represent blood). This creates the air blood interphase
  • Modelled cyclical stretching by having vacuum chambers are on either side of the two layers
393
Q

How did they test the viability of lung-on-a-chip?

A

When air flows through the top chamber – this increased surfactant from epithelial cells – mimicking in vivo

394
Q

What occurs during pulmonary inflammation?

A
  • Can be a life threatening disease and acute or chronic
  • Cytokines TNF-alpha released form epithelium and activates endothelium which start to express cell adhesion antigens (ICAM-1). When this happens, the leukocytes starts to attach to the endothelial cells and allow neutrophils to enter the alveoli to digest the invaded bacteria
395
Q

How can pulmonary inflammation be modelled using lung-on-a-chip?

A
  • Put TNF-alpha in upper chamber and measure the effects of the endothelium to them. Can measure using immunohistochemistry
  • Add leukocytes in the lower chamber and can see if it passes the membrane
  • They then used E.coli labelled with GFP instead of TNF-alpha to see bacterial infection and found that neutrophils invade them
396
Q

What is pulmonary oedema?

A
  • Excess build up of fluid in the lungs and causes respiratory failure
  • Can occur as a side effect of cancer therapies e.g. systemic administration of IL-2
397
Q

How did Huh et al, 2012 mimic pulmonary oedema?

A

Added IL-2 to the blood compartment

  • Noticed that cells become more permeable and were letting liquid enter the air chamber
  • This gets worse and worse overtime until the whole chop is filled with liquid as breathing (mimicked) increased
  • Also caused blood clots
398
Q

How did Huh et al, 2012 use lungs-on-chips to screen drugs for pulmonary odoema??

A
  • Drug Ang-1 previously shown to stabilise junctions between endothelial cells and Ang-2 is an antagonist and destabilises the endothelial barrier so was used of a control
  • Found that Ang-1 reduced permeability
399
Q

What are the other uses of organs-on-chips?

A

Body-on-a-chip
- Linking all the relative organs for drug development
Personalised medicine
- Can take other things than just cells – breathing patterns, exercise patterns, plasma biomarkers. Can add environmental markers relevant for the patient and can use to accurately predict the response to drugs

400
Q

Why should we engineer food?

A

By the year 2050 expect to have human population of 10 billion so need a massive increase to cover this
Meat production
- Responsible for more greenhouse gas emissions then the entire transportation system and is the biggest use of water
- Antibiotic use is also a massive issue – develop antibiotic resistance
- Current meat production is not sustainable

401
Q

How can lab grown food be produced?

A

Follows the basic principles of creating any tissue: the building blocks of tissue engineering put together in a bioreactor to create skeletal muscle

402
Q

What cells could be used to produce lab based meat?

A

Satellite cells
- Take a small biopsy and isolate satellite cells and put them in culture (not immortal)
- Proliferation and differentiation phase which can lead to myoblast and myotube formation
IPSCs
- Unlimited proliferation ability
- Need to optimise differentiation
- Can include multiple cell types

403
Q

What properties should the biomaterials/scaffold for TE meat possess?

A
  • Edible
  • Non-allergenic
  • Inert
  • Needs to support full differentiation and maturation
404
Q

What fabrication strategies could be used for lab grown meat?

A
Bioreactors 
- Grow large numbers of cells
- Put on porous collagen microspheres 
- For muscles to form in vitro they need mechanical and electrical stimulation – could design bioreactors 
3D printing 
- Allow a much finer structure
405
Q

What are the technical challenges of food in Tissue engineering?

A

Lead to commercial companies
- They patent their discovery= can’t build upon research
Scale
- Global meat production is 293 million tons/year.
- Need 1 bioreactor per 10 humans – own carbon footprint
Efficiency
- Optimisation of scale up
- Optimisation of differentiation
Customer acceptancy of food tissue engineering
- Need taste and texture
- Ethics and attitude

406
Q

What are benefits of cultured meat?

A
Healthier meat 
- Added vitamins 
- Reducing fat
- Increasing levels of conjugated linoleic acid
Control over texture and flavour
Creation of new products
407
Q

What animal products could be substituted through TE?

A
  • Meat
  • Dairy
  • Leather (skin culture)