Jump to content

Cryopreservation: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
m Fix PMC warnings
consistent citation formatting; templated cites; added missing PMIDs to cites
Line 4: Line 4:
[[File:Cryopreservation USDA Gene Bank.jpg|thumb|300px|Cryogenically preserved samples being removed from a liquid nitrogen dewar.]]
[[File:Cryopreservation USDA Gene Bank.jpg|thumb|300px|Cryogenically preserved samples being removed from a liquid nitrogen dewar.]]


'''Cryo-preservation''' or '''cryo-conservation''' is a process where [[organelle]]s, [[cell (biology)|cells]], [[Biological tissue|tissues]], [[extracellular matrix]], [[Organ (anatomy)|organs]], or any other biological constructs susceptible to damage caused by unregulated [[chemical kinetics]] are preserved by cooling to very low [[temperature]]s<ref>{{Cite journal |last=Pegg |first=David E. |date=January 1, 2007 |title=Principles of cryopreservation|journal=Methods in Molecular Biology |location=Clifton, N.J. |volume=368 |pages=39–57 |doi=10.1007/978-1-59745-362-2_3 |issn=1064-3745 |pmid=18080461|isbn=978-1-58829-377-0 }}</ref> (typically −80&nbsp;°C using solid [[carbon dioxide]] or −196&nbsp;°C using [[liquid nitrogen]]). At low enough temperatures, any [[Enzyme|enzymatic]] or chemical activity which might cause damage to the biological material in question is effectively stopped. Cryopreservation methods seek to reach low temperatures without causing additional damage caused by the formation of ice crystals during freezing. Traditional cryopreservation has relied on coating the material to be frozen with a class of molecules termed [[cryoprotectants]]. New methods are constantly being investigated due to the inherent [[toxicity]] of many cryoprotectants.<ref>{{Cite journal |last=Sambu |first=Sammy |date=June 25, 2015 |title=A Bayesian approach to optimizing cryopreservation protocols |journal=PeerJ |language=en |volume=3 |doi=10.7717/peerj.1039 |issn=2167-8359 |pmc=4485240 |pmid=26131379 |pages=e1039}}</ref> By default it should be considered that cryopreservation alters or compromises [[Cell (biology)#Cellular processes|the structure and function of cells]] unless it is proven otherwise for a particular cell population. [[Cryoconservation of animal genetic resources]] is the process in which animal genetic material is collected and stored with the intention of conservation of the breed.
'''Cryo-preservation''' or '''cryo-conservation''' is a process where [[organelle]]s, [[cell (biology)|cells]], [[Biological tissue|tissues]], [[extracellular matrix]], [[Organ (anatomy)|organs]], or any other biological constructs susceptible to damage caused by unregulated [[chemical kinetics]] are preserved by cooling to very low [[temperature]]s<ref>{{cite journal | vauthors = Pegg DE | title = Principles of cryopreservation | journal = Methods in Molecular Biology | volume = 368 | pages = 39–57 | date = January 1, 2007 | pmid = 18080461 | doi = 10.1007/978-1-59745-362-2_3 | isbn = 978-1-58829-377-0 }}</ref> (typically −80&nbsp;°C using solid [[carbon dioxide]] or −196&nbsp;°C using [[liquid nitrogen]]). At low enough temperatures, any [[Enzyme|enzymatic]] or chemical activity which might cause damage to the biological material in question is effectively stopped. Cryopreservation methods seek to reach low temperatures without causing additional damage caused by the formation of ice crystals during freezing. Traditional cryopreservation has relied on coating the material to be frozen with a class of molecules termed [[cryoprotectants]]. New methods are constantly being investigated due to the inherent [[toxicity]] of many cryoprotectants.<ref>{{cite journal | vauthors = Sambu S | title = A Bayesian approach to optimizing cryopreservation protocols | journal = PeerJ | volume = 3 | pages = e1039 | date = June 25, 2015 | pmid = 26131379 | pmc = 4485240 | doi = 10.7717/peerj.1039 }}</ref> By default it should be considered that cryopreservation alters or compromises [[Cell (biology)#Cellular processes|the structure and function of cells]] unless it is proven otherwise for a particular cell population. [[Cryoconservation of animal genetic resources]] is the process in which animal genetic material is collected and stored with the intention of conservation of the breed.


== Natural cryopreservation ==
== Natural cryopreservation ==
Line 10: Line 10:
[[Water bear|Water-bears]] (''[[Tardigrada]]''), microscopic multicellular organisms, can survive [[freezing]] by replacing most of their internal water with the [[sugar]] [[trehalose]], preventing it from crystallization that otherwise damages [[cell membranes]]. Mixtures of solutes can achieve similar effects. Some solutes, including salts, have the disadvantage that they may be toxic at intense concentrations. In addition to the water-bear, wood frogs can tolerate the freezing of their blood and other tissues. Urea is accumulated in tissues in preparation for overwintering, and liver glycogen is converted in large quantities to glucose in response to internal ice formation. Both urea and glucose act as "cryoprotectants" to limit the amount of ice that forms and to reduce [[osmotic]] shrinkage of cells. Frogs can survive many freeze/thaw events during winter if no more than about 65% of the total body water freezes. Research exploring the phenomenon of "freezing frogs" has been performed primarily by the Canadian researcher, Dr. [[Kenneth B. Storey]].{{citation needed|date=May 2014}}
[[Water bear|Water-bears]] (''[[Tardigrada]]''), microscopic multicellular organisms, can survive [[freezing]] by replacing most of their internal water with the [[sugar]] [[trehalose]], preventing it from crystallization that otherwise damages [[cell membranes]]. Mixtures of solutes can achieve similar effects. Some solutes, including salts, have the disadvantage that they may be toxic at intense concentrations. In addition to the water-bear, wood frogs can tolerate the freezing of their blood and other tissues. Urea is accumulated in tissues in preparation for overwintering, and liver glycogen is converted in large quantities to glucose in response to internal ice formation. Both urea and glucose act as "cryoprotectants" to limit the amount of ice that forms and to reduce [[osmotic]] shrinkage of cells. Frogs can survive many freeze/thaw events during winter if no more than about 65% of the total body water freezes. Research exploring the phenomenon of "freezing frogs" has been performed primarily by the Canadian researcher, Dr. [[Kenneth B. Storey]].{{citation needed|date=May 2014}}


'''Freeze tolerance''', in which organisms survive the winter by freezing solid and ceasing life functions, is known in a few vertebrates: five species of frogs (''[[Rana sylvatica]]'', ''[[Pseudacris triseriata]]'', ''[[Hyla crucifer]]'', ''[[Hyla versicolor]]'', ''[[Hyla chrysoscelis]]''), one of salamanders (''[[Hynobius]] keyserlingi''), one of snakes (''[[Thamnophis sirtalis]]'') and three of turtles (''[[Chrysemys picta]]'', ''[[Terrapene carolina]]'', ''[[Terrapene ornata]]'').<ref name=AJP1991 /> Snapping turtles ''[[Chelydra serpentina]]'' and wall lizards ''[[Podarcis muralis]]'' also survive nominal freezing but it has not been established to be adaptive for overwintering. In the case of ''Rana sylvatica'' one cryopreservant is ordinary glucose, which increases in concentration by approximately 19&nbsp;mmol/l when the frogs are cooled slowly.<ref name=AJP1991>{{cite journal|url=http://www.units.muohio.edu/cryolab/publications/documents/CostanzoLeeWright91AJP.pdf|title=Glucose loading prevents freezing injury in rapidly cooled wood frogs|author=Jon P. Costanzo|author2=Richard E. Lee|author3=Michael F. Wright|date=1991|journal=American Journal of Physiology|pages=R1549–R1553}}</ref>
'''Freeze tolerance''', in which organisms survive the winter by freezing solid and ceasing life functions, is known in a few vertebrates: five species of frogs (''[[Rana sylvatica]]'', ''[[Pseudacris triseriata]]'', ''[[Hyla crucifer]]'', ''[[Hyla versicolor]]'', ''[[Hyla chrysoscelis]]''), one of salamanders (''[[Hynobius]] keyserlingi''), one of snakes (''[[Thamnophis sirtalis]]'') and three of turtles (''[[Chrysemys picta]]'', ''[[Terrapene carolina]]'', ''[[Terrapene ornata]]'').<ref name=AJP1991 /> Snapping turtles ''[[Chelydra serpentina]]'' and wall lizards ''[[Podarcis muralis]]'' also survive nominal freezing but it has not been established to be adaptive for overwintering. In the case of ''Rana sylvatica'' one cryopreservant is ordinary glucose, which increases in concentration by approximately 19&nbsp;mmol/l when the frogs are cooled slowly.<ref name="AJP1991">{{cite journal | vauthors = Costanzo JP, Lee RE, Wright MF | title = Glucose loading prevents freezing injury in rapidly cooled wood frogs | journal = The American Journal of Physiology | volume = 261 | issue = 6 Pt 2 | pages = R1549–53 | date = December 1991 | pmid = 1750578 | doi = 10.1152/ajpregu.1991.261.6.R1549 | url = http://www.units.muohio.edu/cryolab/publications/documents/CostanzoLeeWright91AJP.pdf }}</ref>


== History ==
== History ==
{{See also|Cryonics#History}}
{{See also|Cryonics#History}}


One of the most important early theoreticians of cryopreservation was [[James Lovelock]]. In 1953, he suggested that damage to [[red blood cell]]s during freezing was due to [[osmosis|osmotic]] stress,<ref>{{cite journal |last=Lovelock |first=James |title=The haemolysis of human red blood-cells by freezing and thawing |journal=[[Biochimica et Biophysica Acta]] |date= 1953 |pages=414–26 |volume=10 |issue=3 |pmid= 13058999 |doi=10.1016/0006-3002(53)90273-X}}</ref> and that increasing the salt concentration in a dehydrating cell might damage it.<ref>{{cite book|editor-last1=Fuller|editor-first1=Barry J.|editor-last2=Lane |editor-first2=Nick|editor-last3=Benson|editor-first3=Erica E.|title=Life in the Frozen State|year=2004|publisher=[[CRC Press]]|url=https://books.google.com/books?id=PPven_q2xiQC&pg=PP7|page=7|isbn=978-0203647073}}</ref><ref>{{cite journal |last=Mazur |first=Peter |title=Cryobiology: the freezing of biological systems |journal=Science |date=1970 |pages=939–49 |volume=168 |issue=3934 |pmid=5462399 |doi=10.1126/science.168.3934.939 |bibcode=1970Sci...168..939M}}</ref> In the mid-1950s, he experimented with the cryopreservation of rodents, determining that hamsters could be frozen with 60% of the water in the brain crystallized into ice with no adverse effects; other organs were shown to be susceptible to damage.<ref>{{cite magazine|title=The Cryobiological Case for Cryonics|magazine=Cryonics|page=27|date=March 1988|volume=9(3)|issue=92|publisher=[[Alcor Life Extension Foundation]]|url=https://www.alcor.org/cryonics/cryonics8803.pdf}}</ref> This work led other scientists to attempt the short-term freezing of rats by 1955, which were fully active 4 to 7 days after being revived.<ref>{{cite journal|last1=Andjus|first1=R. K.|last2=Smith|first2=Audrey U.|author2-link=Audrey Smith|title=Reanimation of adult rats from body temperatures between 0 and +2° C|journal=[[The Journal of Physiology]]|date=1955|pages=446–72|volume=128|issue=3|pmid=13243342|pmc=1365897|doi=10.1113/jphysiol.1955.sp005318}}</ref>
One of the most important early theoreticians of cryopreservation was [[James Lovelock]]. In 1953, he suggested that damage to [[red blood cell]]s during freezing was due to [[osmosis|osmotic]] stress,<ref>{{cite journal | vauthors = Lovelock JE | title = The haemolysis of human red blood-cells by freezing and thawing | journal = Biochimica et Biophysica Acta | volume = 10 | issue = 3 | pages = 414–26 | date = March 1953 | pmid = 13058999 | doi = 10.1016/0006-3002(53)90273-X }}</ref> and that increasing the salt concentration in a dehydrating cell might damage it.<ref>{{cite book|editor-last1=Fuller|editor-first1=Barry J.|editor-last2=Lane |editor-first2=Nick|editor-last3=Benson|editor-first3=Erica E. | name-list-format = vanc |title=Life in the Frozen State|year=2004|publisher=[[CRC Press]]|url=https://books.google.com/books?id=PPven_q2xiQC&pg=PP7|page=7|isbn=978-0203647073}}</ref><ref>{{cite journal | vauthors = Mazur P | title = Cryobiology: the freezing of biological systems | journal = Science | volume = 168 | issue = 3934 | pages = 939–49 | date = May 1970 | pmid = 5462399 | doi = 10.1126/science.168.3934.939 | bibcode = 1970Sci...168..939M }}</ref> In the mid-1950s, he experimented with the cryopreservation of rodents, determining that hamsters could be frozen with 60% of the water in the brain crystallized into ice with no adverse effects; other organs were shown to be susceptible to damage.<ref>{{cite magazine|title=The Cryobiological Case for Cryonics|magazine=Cryonics|page=27|date=March 1988|volume=9(3)|issue=92|publisher=[[Alcor Life Extension Foundation]]|url=https://www.alcor.org/cryonics/cryonics8803.pdf}}</ref> This work led other scientists to attempt the short-term freezing of rats by 1955, which were fully active 4 to 7 days after being revived.<ref>{{cite journal | vauthors = Andjus RK, Smith AU | title = Reanimation of adult rats from body temperatures between 0 and + 2 degrees C | journal = The Journal of Physiology | volume = 128 | issue = 3 | pages = 446–72 | date = June 1955 | pmid = 13243342 | pmc = 1365897 | doi = 10.1113/jphysiol.1955.sp005318 | author2-link = Audrey Smith }}</ref>


Cryopreservation was applied to humans beginning in 1954 with three pregnancies resulting from the insemination of previously frozen sperm.<ref>{{cite news|title=Fatherhood After Death Has Now Been Proved Possible|newspaper=Cedar Rapids Gazette|date=April 9, 1954}}</ref> Fowl sperm was cryopreserved in 1957 by a team of scientists in the UK directed by [[Christopher Polge]].<ref>{{cite journal |doi=10.1098/rspb.1957.0068 |author=Polge C |title=Low-Temperature Storage of Mammalian Spermatozoa |journal= Proceedings of the Royal Society of London B|date=1957 |pages=498–508 |volume=147 |issue=929 |bibcode=1957RSPSB.147..498P}}</ref> <!--However, the rapid immersion of the samples in [[liquid nitrogen]] did not, for certain samples—such as some types of embryos, bone marrow and stem cells—produce the necessary viability to make them usable after thawing. Increased understanding of the mechanism of freezing injury to cells emphasised the importance of controlled or slow cooling to obtain maximum survival on thawing of the living cells. A controlled-rate cooling process, allowing biological samples to equilibrate to optimal physical parameters osmotically in a cryoprotectant (a form of anti-freeze) before cooling in a predetermined, controlled way proved necessary. The ability of cryoprotectants, in the early cases glycerol, to protect cells from freezing injury was discovered accidentally. Freezing injury has two aspects: direct damage from the ice crystals and secondary damage caused by the increase in concentration of solutes as progressively more ice is formed.--> During 1963, Peter Mazur, at [[Oak Ridge National Laboratory]] in the U.S., demonstrated that lethal intracellular freezing could be avoided if cooling was slow enough to permit sufficient water to leave the cell during progressive freezing of the extracellular fluid. That rate differs between cells of differing size and water permeability: a typical cooling rate around 1&nbsp;°C/minute is appropriate for many mammalian cells after treatment with cryoprotectants such as glycerol or dimethyl sulphoxide, but the rate is not a universal optimum.<ref>{{Cite journal |url=https://www.cell.com/biophysj/pdf/S0006-3495(63)86824-1.pdf |pmc=1366450|year=1963|last1=Mazur|first1=P.|title=Studies on Rapidly Frozen Suspensions of Yeast Cells by Differential Thermal Analysis and Conductometry|journal=Biophysical Journal|volume=3|issue=4|pages=323–353|pmid=13934216|bibcode=1963BpJ.....3..323M|doi=10.1016/S0006-3495(63)86824-1}}</ref>
Cryopreservation was applied to humans beginning in 1954 with three pregnancies resulting from the insemination of previously frozen sperm.<ref>{{cite news|title=Fatherhood After Death Has Now Been Proved Possible|newspaper=Cedar Rapids Gazette|date=April 9, 1954}}</ref> Fowl sperm was cryopreserved in 1957 by a team of scientists in the UK directed by [[Christopher Polge]].<ref>{{cite journal | vauthors = Polge C | title = Low-temperature storage of mammalian spermatozoa | journal = Proceedings of the Royal Society of London. Series B, Biological Sciences | volume = 147 | issue = 929 | pages = 498–508 | date = December 1957 | pmid = 13494462 | doi = 10.1098/rspb.1957.0068 | bibcode = 1957RSPSB.147..498P }}</ref> <!--However, the rapid immersion of the samples in [[liquid nitrogen]] did not, for certain samples—such as some types of embryos, bone marrow and stem cells—produce the necessary viability to make them usable after thawing. Increased understanding of the mechanism of freezing injury to cells emphasised the importance of controlled or slow cooling to obtain maximum survival on thawing of the living cells. A controlled-rate cooling process, allowing biological samples to equilibrate to optimal physical parameters osmotically in a cryoprotectant (a form of anti-freeze) before cooling in a predetermined, controlled way proved necessary. The ability of cryoprotectants, in the early cases glycerol, to protect cells from freezing injury was discovered accidentally. Freezing injury has two aspects: direct damage from the ice crystals and secondary damage caused by the increase in concentration of solutes as progressively more ice is formed.--> During 1963, Peter Mazur, at [[Oak Ridge National Laboratory]] in the U.S., demonstrated that lethal intracellular freezing could be avoided if cooling was slow enough to permit sufficient water to leave the cell during progressive freezing of the extracellular fluid. That rate differs between cells of differing size and water permeability: a typical cooling rate around 1&nbsp;°C/minute is appropriate for many mammalian cells after treatment with cryoprotectants such as glycerol or dimethyl sulphoxide, but the rate is not a universal optimum.<ref>{{cite journal | vauthors = Mazur P | title = Studies on rapidly frozen suspensions of yeast cells by differential thermal analysis and conductometry | journal = Biophysical Journal | volume = 3 | issue = 4 | pages = 323–53 | date = July 1963 | pmid = 13934216 | pmc = 1366450 | doi = 10.1016/S0006-3495(63)86824-1 | url = https://www.cell.com/biophysj/pdf/S0006-3495(63)86824-1.pdf | bibcode = 1963BpJ.....3..323M }}</ref>


The first human body to be frozen with the hope of future revival was [[James Bedford]]'s, a few hours after his cancer-caused death in 1967.<ref>{{cite web | title=Dear Dr. Bedford (and those who will care for you after I do) | publisher=Cryonics | date=July 1991 | url=http://www.alcor.org/Library/html/BedfordLetter.htm | access-date=2009-08-23 }}</ref> Bedford is the only [[cryonics]] patient frozen before 1974 still preserved today.<ref>{{cite web|url=https://alcor.org/Library/html/suspensionfailures.html|title=Suspension Failures – Lessons from the Early Days |last= Perry|first=R. Michael|website=ALCOR: Life Extension Foundation|date=October 2014|accessdate=August 29, 2018}}</ref>
The first human body to be frozen with the hope of future revival was [[James Bedford]]'s, a few hours after his cancer-caused death in 1967.<ref>{{cite web | title=Dear Dr. Bedford (and those who will care for you after I do) | publisher=Cryonics | date=July 1991 | url=http://www.alcor.org/Library/html/BedfordLetter.htm | access-date=2009-08-23 }}</ref> Bedford is the only [[cryonics]] patient frozen before 1974 still preserved today.<ref>{{cite web|url=https://alcor.org/Library/html/suspensionfailures.html|title=Suspension Failures – Lessons from the Early Days |last= Perry|first=R. Michael | name-list-format = vanc |website=ALCOR: Life Extension Foundation|date=October 2014|access-date=August 29, 2018}}</ref>


==Temperature==
==Temperature==
Line 27: Line 27:


[[Phenomena]] which can cause damage to cells during cryopreservation mainly occur during the freezing stage, and include: solution effects, [[extracellular]] ice formation, dehydration and [[intracellular]] ice formation. Many of these effects can be reduced by [[cryoprotectant]]s.
[[Phenomena]] which can cause damage to cells during cryopreservation mainly occur during the freezing stage, and include: solution effects, [[extracellular]] ice formation, dehydration and [[intracellular]] ice formation. Many of these effects can be reduced by [[cryoprotectant]]s.
Once the preserved material has become frozen, it is relatively safe from further damage. However, estimates based on the accumulation of radiation-induced DNA damage during cryonic storage have suggested a maximum storage period of 1000 years.<ref>{{cite journal |doi=10.1098/rspb.1957.0068 |author=Mazur P |title=Freezing of living cells: mechanisms and implications |journal=American Journal of Physiology |date=1984 |pages=C125-42 |volume=247 |issue=3 Pt 1 |bibcode=1957RSPSB.147..498P}}</ref>
Once the preserved material has become frozen, it is relatively safe from further damage. However, estimates based on the accumulation of radiation-induced DNA damage during cryonic storage have suggested a maximum storage period of 1000 years.<ref>{{cite journal | vauthors = Mazur P | title = Freezing of living cells: mechanisms and implications | journal = The American Journal of Physiology | volume = 247 | issue = 3 Pt 1 | pages = C125-42 | date = September 1984 | pmid = 6383068 | doi = 10.1098/rspb.1957.0068 | bibcode = 1957RSPSB.147..498P }}</ref>


; Solution effects: As ice crystals grow in freezing water, [[solutes]] are excluded, causing them to become concentrated in the remaining liquid water. High concentrations of some solutes can be very damaging.
; Solution effects: As ice crystals grow in freezing water, [[solutes]] are excluded, causing them to become concentrated in the remaining liquid water. High concentrations of some solutes can be very damaging.
Line 44: Line 44:
===Slow programmable freezing===
===Slow programmable freezing===
[[Image:Liquid nitrogen tank.JPG|thumb|150px|A tank of [[liquid nitrogen]], used to supply a cryogenic freezer (for storing laboratory samples at a temperature of about −150&nbsp;°C)]]
[[Image:Liquid nitrogen tank.JPG|thumb|150px|A tank of [[liquid nitrogen]], used to supply a cryogenic freezer (for storing laboratory samples at a temperature of about −150&nbsp;°C)]]
''Controlled-rate and slow freezing'', also known as ''slow programmable freezing (SPF)'',<ref>{{cite journal |author=Vutyavanich T|author2=Piromlertamorn W|author3=Nunta S |title=Rapid freezing versus slow programmable freezing of human spermatozoa |journal=Fertil. Steril. |volume=93 |issue=6 |pages=1921–8 |date=April 2010 |pmid=19243759 |doi=10.1016/j.fertnstert.2008.04.076}}</ref> is a set of well established techniques developed during the early 1970s which enabled the first human [[embryo]] frozen birth Zoe Leyland during 1984. Since then, machines that freeze biological samples using programmable sequences, or controlled rates, have been used all over the world for human, animal and cell biology – "freezing down" a sample to better preserve it for eventual thawing, before it is frozen, or cryopreserved, in liquid nitrogen. Such machines are used for freezing oocytes, skin, blood products, embryo, sperm, stem cells and general tissue preservation in hospitals, veterinary practices and research laboratories around the world. As an example, the number of live births from frozen embryos 'slow frozen' is estimated at some 300,000 to 400,000 or 20% of the estimated 3 million in vitro fertilisation ([[IVF]]) births.<ref>{{cite web |url=http://www.bionews.org.uk/commentary.lasso?storyid=4055 |archive-url=https://www.webarchive.org.uk/wayback/archive/20090526093911/http://www.bionews.org.uk/commentary.lasso?storyid=4055 |dead-url=yes |archive-date=2009-05-26 |title=dead link}}</ref>
''Controlled-rate and slow freezing'', also known as ''slow programmable freezing (SPF)'',<ref>{{cite journal | vauthors = Vutyavanich T, Piromlertamorn W, Nunta S | title = Rapid freezing versus slow programmable freezing of human spermatozoa | journal = Fertility and Sterility | volume = 93 | issue = 6 | pages = 1921–8 | date = April 2010 | pmid = 19243759 | doi = 10.1016/j.fertnstert.2008.04.076 }}</ref> is a set of well established techniques developed during the early 1970s which enabled the first human [[embryo]] frozen birth Zoe Leyland during 1984. Since then, machines that freeze biological samples using programmable sequences, or controlled rates, have been used all over the world for human, animal and cell biology – "freezing down" a sample to better preserve it for eventual thawing, before it is frozen, or cryopreserved, in liquid nitrogen. Such machines are used for freezing oocytes, skin, blood products, embryo, sperm, stem cells and general tissue preservation in hospitals, veterinary practices and research laboratories around the world. As an example, the number of live births from frozen embryos 'slow frozen' is estimated at some 300,000 to 400,000 or 20% of the estimated 3 million in vitro fertilisation ([[IVF]]) births.<ref>{{cite web |url=http://www.bionews.org.uk/commentary.lasso?storyid=4055 |archive-url=https://www.webarchive.org.uk/wayback/archive/20090526093911/http://www.bionews.org.uk/commentary.lasso?storyid=4055 |dead-url=yes |archive-date=2009-05-26 |title=dead link}}</ref>


Lethal intracellular freezing can be avoided if cooling is slow enough to permit sufficient water to leave the cell during progressive freezing of the extracellular fluid. To minimize the growth of extracellular ice crystal growth and recrystallization,<ref>{{Cite journal |last=Deller |first=Robert C. |last2=Vatish |first2=Manu |last3=Mitchell |first3=Daniel A. |last4=Gibson |first4=Matthew I. |date=February 3, 2014 |title=Synthetic polymers enable non-vitreous cellular cryopreservation by reducing ice crystal growth during thawing |journal=Nature Communications |language=en |volume=5 |pages=3244 |doi=10.1038/ncomms4244 |pmid=24488146|bibcode=2014NatCo...5E3244D }}</ref> [[biomaterial]]s such as [[alginate]]s, [[polyvinyl alcohol]] or [[chitosan]] can be used to impede ice crystal growth along with traditional small molecule cryoprotectants.<ref>{{Cite journal |last=Sambu |first=Sammy |date=June 25, 2015 |title=A Bayesian approach to optimizing cryopreservation protocols |journal=PeerJ |language=en |volume=3 |doi=10.7717/peerj.1039 |issn=2167-8359 |pmc=4485240 |pmid=26131379 |pages=e1039}}</ref> That rate differs between cells of differing size and water [[Semipermeable membrane|permeability]]: a typical cooling rate of about 1&nbsp;°C/minute is appropriate for many mammalian cells after treatment with [[cryoprotectant]]s such as [[glycerol]] or [[dimethyl sulfoxide]], but the rate is not a universal optimum. The 1&nbsp;°C / minute rate can be achieved by using devices such as a rate-controlled freezer or a benchtop portable freezing container.<ref>{{cite journal |author=Thompson M|author2=Nemits M|author3=Ehrhardt R |title=Rate-controlled Cryopreservation and Thawing of Mammalian Cells |journal=Protocol Exchange |date=May 2011 |doi=10.1038/protex.2011.224 |url=http://www.nature.com/protocolexchange/protocols/2085}}</ref>
Lethal intracellular freezing can be avoided if cooling is slow enough to permit sufficient water to leave the cell during progressive freezing of the extracellular fluid. To minimize the growth of extracellular ice crystal growth and recrystallization,<ref>{{cite journal | vauthors = Deller RC, Vatish M, Mitchell DA, Gibson MI | title = Synthetic polymers enable non-vitreous cellular cryopreservation by reducing ice crystal growth during thawing | journal = Nature Communications | volume = 5 | pages = 3244 | date = February 3, 2014 | pmid = 24488146 | doi = 10.1038/ncomms4244 | bibcode = 2014NatCo...5E3244D }}</ref> [[biomaterial]]s such as [[alginate]]s, [[polyvinyl alcohol]] or [[chitosan]] can be used to impede ice crystal growth along with traditional small molecule cryoprotectants.<ref>{{cite journal | vauthors = Sambu S | title = A Bayesian approach to optimizing cryopreservation protocols | journal = PeerJ | volume = 3 | pages = e1039 | date = June 25, 2015 | pmid = 26131379 | pmc = 4485240 | doi = 10.7717/peerj.1039 }}</ref> That rate differs between cells of differing size and water [[Semipermeable membrane|permeability]]: a typical cooling rate of about 1&nbsp;°C/minute is appropriate for many mammalian cells after treatment with [[cryoprotectant]]s such as [[glycerol]] or [[dimethyl sulfoxide]], but the rate is not a universal optimum. The 1&nbsp;°C / minute rate can be achieved by using devices such as a rate-controlled freezer or a benchtop portable freezing container.<ref>{{cite journal | vauthors = Thompson M, Nemits M, Ehrhardt R |title=Rate-controlled Cryopreservation and Thawing of Mammalian Cells |journal=Protocol Exchange |date=May 2011 |doi=10.1038/protex.2011.224 |url=http://www.nature.com/protocolexchange/protocols/2085}}</ref>


Several independent studies have provided evidence that frozen embryos stored using slow-freezing techniques may in some ways be 'better' than fresh in IVF. The studies indicate that using frozen embryos and eggs rather than fresh embryos and eggs reduced the risk of stillbirth and premature delivery though the exact reasons are still being explored.
Several independent studies have provided evidence that frozen embryos stored using slow-freezing techniques may in some ways be 'better' than fresh in IVF. The studies indicate that using frozen embryos and eggs rather than fresh embryos and eggs reduced the risk of stillbirth and premature delivery though the exact reasons are still being explored.


===Vitrification===<!--Section linked from [[vitrification]]-->
===Vitrification===<!--Section linked from [[vitrification]]-->
Researchers [[Greg Fahy]] and William F. Rall helped to introduce vitrification to reproductive cryopreservation in the mid-1980s.<ref name="vitrification origination">{{cite journal |last=Rall |first=WF |author2=Fahy, GM |title=Ice-free cryopreservation of mouse embryos at −196 degrees C by vitrification |journal=Nature |date=February 14–20, 1985 |volume=313 |issue=6003 |pages=573–5 |pmid=3969158 |bibcode=1985Natur.313..573R |doi=10.1038/313573a0}}<!--|accessdate=24 October 2012--></ref> As of 2000, researchers claim vitrification provides the benefits of cryopreservation without damage due to ice crystal formation.<ref name="cryonics 2000">{{cite news|url=http://www.alcor.org/cryonics/cryonics2000-4.pdf |title=Alcor: The Origin of Our Name |accessdate=August 25, 2009 |date=Winter 2000 |publisher=Alcor Life Extension Foundation}}</ref> The situation became more complex with the development of tissue engineering as both cells and biomaterials need to remain ice-free to preserve high cell viability and functions, integrity of constructs and structure of biomaterials. Vitrification of tissue engineered constructs was first reported by Lilia Kuleshova,<ref name=Kuleshova>{{cite journal |last1=Kuleshova |first1=L L |last2=Wang |first2=XW |last3=Wu |first3=YN |last4=Zhou |first4=Y |last5=Yu |first5=H |title=Vitrification of encapsulated hepatocytes with reduced cooling and warming rates |journal=Cryo Letters |volume=25 |issue=4 |pages=241–254 |year=2004}}</ref> who also was the first scientist to achieve vitrification of [[oocyte]]s, which resulted in live birth in 1999.<ref name="Kuleshova1999">{{cite journal |last1=Kuleshova |first1=Lilia |last2=Gianoroli |first2=Luca |last3=Magli |first3=Cristina |last4=Ferraretti |first4=Anna |last5=Trounson |first5=Alan |doi=10.1093/humrep/14.12.3077 |pmid=10601099 |title=Birth following vitrification of small number of human oocytes |journal=Human Reproduction |volume=14 |issue=12 |pages=3077–3079 |year=1999}}</ref> For clinical cryopreservation, vitrification usually requires the addition of [[cryoprotectant]]s prior to cooling. The cryoprotectants act like [[antifreeze]]: they decrease the freezing temperature. They also increase the viscosity. Instead of [[crystal]]lizing, the syrupy solution becomes an [[amorphous ice]]—it ''vitrifies''. Rather than a phase change from liquid to solid by crystallization, the amorphous state is like a "solid liquid", and the transformation is over a small temperature range described as the "[[glass transition]]" temperature.
Researchers [[Greg Fahy]] and William F. Rall helped to introduce vitrification to reproductive cryopreservation in the mid-1980s.<ref name="vitrification origination">{{cite journal | vauthors = Rall WF, Fahy GM | title = Ice-free cryopreservation of mouse embryos at -196 degrees C by vitrification | journal = Nature | volume = 313 | issue = 6003 | pages = 573–5 | date = February 14–20, 1985 | pmid = 3969158 | doi = 10.1038/313573a0 | bibcode = 1985Natur.313..573R }}<!--|access-date=24 October 2012--></ref> As of 2000, researchers claim vitrification provides the benefits of cryopreservation without damage due to ice crystal formation.<ref name="cryonics 2000">{{cite news|url=http://www.alcor.org/cryonics/cryonics2000-4.pdf |title=Alcor: The Origin of Our Name |access-date=August 25, 2009 |date=Winter 2000 |publisher=Alcor Life Extension Foundation}}</ref> The situation became more complex with the development of tissue engineering as both cells and biomaterials need to remain ice-free to preserve high cell viability and functions, integrity of constructs and structure of biomaterials. Vitrification of tissue engineered constructs was first reported by Lilia Kuleshova,<ref name=Kuleshova>{{cite journal | vauthors = Kuleshova LL, Wang XW, Wu YN, Zhou Y, Yu H | title = Vitrification of encapsulated hepatocytes with reduced cooling and warming rates | journal = Cryo Letters | volume = 25 | issue = 4 | pages = 241–54 | year = 2004 | pmid = 15375435 }}</ref> who also was the first scientist to achieve vitrification of [[oocyte]]s, which resulted in live birth in 1999.<ref name="Kuleshova1999">{{cite journal | vauthors = Kuleshova L, Gianaroli L, Magli C, Ferraretti A, Trounson A | title = Birth following vitrification of a small number of human oocytes: case report | journal = Human Reproduction | volume = 14 | issue = 12 | pages = 3077–9 | date = December 1999 | pmid = 10601099 | doi = 10.1093/humrep/14.12.3077 }}</ref> For clinical cryopreservation, vitrification usually requires the addition of [[cryoprotectant]]s prior to cooling. The cryoprotectants act like [[antifreeze]]: they decrease the freezing temperature. They also increase the viscosity. Instead of [[crystal]]lizing, the syrupy solution becomes an [[amorphous ice]]—it ''vitrifies''. Rather than a phase change from liquid to solid by crystallization, the amorphous state is like a "solid liquid", and the transformation is over a small temperature range described as the "[[glass transition]]" temperature.


Vitrification of water is promoted by rapid cooling, and can be achieved without cryoprotectants by an extremely rapid decrease of temperature (megakelvins per second). The rate that is required to attain glassy state in pure water was considered to be impossible until 2005.<ref>{{cite journal |author=Bhat SN |author2=Sharma A |author3=Bhat SV |title=Vitrification and glass transition of water: insights from spin probe ESR |journal=Phys Rev Lett |date=2005 |pages=235702 |volume=95 |issue=23 |pmid=16384318 |doi=10.1103/PhysRevLett.95.235702 |bibcode=2005PhRvL..95w5702B |arxiv=cond-mat/0409440}}</ref>
Vitrification of water is promoted by rapid cooling, and can be achieved without cryoprotectants by an extremely rapid decrease of temperature (megakelvins per second). The rate that is required to attain glassy state in pure water was considered to be impossible until 2005.<ref>{{cite journal | vauthors = Bhat SN, Sharma A, Bhat SV | title = Vitrification and glass transition of water: insights from spin probe ESR | journal = Physical Review Letters | volume = 95 | issue = 23 | pages = 235702 | date = December 2005 | pmid = 16384318 | doi = 10.1103/PhysRevLett.95.235702 | arxiv = cond-mat/0409440 | bibcode = 2005PhRvL..95w5702B }}</ref>


Two conditions usually required to allow vitrification are an increase of the viscosity and a decrease of the freezing temperature. Many solutes do both, but larger molecules generally have a larger effect, particularly on viscosity. Rapid cooling also promotes vitrification.
Two conditions usually required to allow vitrification are an increase of the viscosity and a decrease of the freezing temperature. Many solutes do both, but larger molecules generally have a larger effect, particularly on viscosity. Rapid cooling also promotes vitrification.


For established methods of cryopreservation, the solute must penetrate the cell membrane in order to achieve increased viscosity and decrease freezing temperature inside the cell. Sugars do not readily permeate through the membrane. Those solutes that do, such as [[dimethyl sulfoxide]], a common cryoprotectant, are often toxic in intense concentration. One of the difficult compromises of vitrifying cryopreservation concerns limiting the damage produced by the cryoprotectant itself due to cryoprotectant toxicity. Mixtures of cryoprotectants and the use of ice blockers have enabled the [[Twenty-First Century Medicine]] company to vitrify a [[rabbit]] [[kidney]] to −135&nbsp;°C with their proprietary vitrification mixture. Upon rewarming, the kidney was transplanted successfully into a rabbit, with complete functionality and viability, able to sustain the rabbit indefinitely as the sole functioning kidney.<ref>{{cite journal |doi=10.4161/org.5.3.9974 |display-authors=4 |author=Fahy GM |author2=Wowk B |author3=Pagotan R |author4=Chang A |author5=Phan J |author6=Thomson B |author7=Phan L |title=Physical and biological aspects of renal vitrification |journal=Organogenesis |volume=5 |issue=3 |date=2009 |pages=167–175 |pmc=2781097 |pmid=20046680}}</ref>
For established methods of cryopreservation, the solute must penetrate the cell membrane in order to achieve increased viscosity and decrease freezing temperature inside the cell. Sugars do not readily permeate through the membrane. Those solutes that do, such as [[dimethyl sulfoxide]], a common cryoprotectant, are often toxic in intense concentration. One of the difficult compromises of vitrifying cryopreservation concerns limiting the damage produced by the cryoprotectant itself due to cryoprotectant toxicity. Mixtures of cryoprotectants and the use of ice blockers have enabled the [[Twenty-First Century Medicine]] company to vitrify a [[rabbit]] [[kidney]] to −135&nbsp;°C with their proprietary vitrification mixture. Upon rewarming, the kidney was transplanted successfully into a rabbit, with complete functionality and viability, able to sustain the rabbit indefinitely as the sole functioning kidney.<ref>{{cite journal | vauthors = Fahy GM, Wowk B, Pagotan R, Chang A, Phan J, Thomson B, Phan L | title = Physical and biological aspects of renal vitrification | journal = Organogenesis | volume = 5 | issue = 3 | pages = 167–75 | date = July 2009 | pmid = 20046680 | pmc = 2781097 | doi = 10.4161/org.5.3.9974 }}</ref>


===Persufflation===
===Persufflation===
Blood can be replaced with inert [[noble gases]] and/or metabolically vital gases like [[Dioxygen_in_biological_reactions | oxygen]], so that organs can cool more quickly and less antifreeze is needed. Since regions of tissue are separated by gas, small expansions do not accumulate, thereby protecting against shattering.<ref>{{cite journal | journal=New Scientist | title= Heart of glass could be key to banking organs | author=Linda Geddes |date= Sep 11, 2013}}</ref> A small company, Arigos Biomedical, "has already recovered pig hearts from the 120 degrees below zero",<ref>{{cite journal | journal=BOSS Magazine | author=Matthew Flynn | date=Oct 10, 2018 | title= Heart of Ice |url=https://thebossmagazine.com/freezing-and-thawing-organs/}}</ref> although the definition of "recovered" is not clear. Pressures of 60 atm can help increase heat exchange rates.<ref>{{cite patent | number=US9314015B2 | title=Method and apparatus for prevention of thermo-mechanical fracturing in vitrified tissue using rapid cooling and warming by persufflation | inventor=Stephen Van Sickle, Tanya Jones | fdate= Jul 5, 2013 | pubdate=Apr 19, 2013}}</ref> Gaseous oxygen perfusion/persufflation can enhance organ preservation relative to static cold storage or hypothermic machine perfusion, since the lower viscosity of gases, may help reach more regions of preserved organs and deliver more oxygen per gram tissue.<ref>{{cite journal | journal=Cryobiology | volume=64 | issue=3 |pages= 125–143| date= Jan 26, 2012 | pmc= 3519283 | title= Persufflation (or Gaseous Oxygen Perfusion) as a Method of Organ Preservation | author= Thomas M. Suszynski, Michael D. Rizzari, William E. Scott, Linda A. Tempelman, Michael J. Taylor, Klearchos K. Papas}}</ref>
Blood can be replaced with inert [[noble gases]] and/or metabolically vital gases like [[Dioxygen_in_biological_reactions | oxygen]], so that organs can cool more quickly and less antifreeze is needed. Since regions of tissue are separated by gas, small expansions do not accumulate, thereby protecting against shattering.<ref>{{cite journal | journal=New Scientist | title= Heart of glass could be key to banking organs | first = Linda | last = Geddes | name-list-format = vanc |date= Sep 11, 2013}}</ref> A small company, Arigos Biomedical, "has already recovered pig hearts from the 120 degrees below zero",<ref>{{cite journal | journal=BOSS Magazine | first = Matthew | last = Flynn | name-list-format = vanc | date=Oct 10, 2018 | title= Heart of Ice |url=https://thebossmagazine.com/freezing-and-thawing-organs/}}</ref> although the definition of "recovered" is not clear. Pressures of 60 atm can help increase heat exchange rates.<ref>{{cite patent | number=US9314015B2 | title=Method and apparatus for prevention of thermo-mechanical fracturing in vitrified tissue using rapid cooling and warming by persufflation | inventor=Stephen Van Sickle, Tanya Jones | fdate= Jul 5, 2013 | pubdate=Apr 19, 2013}}</ref> Gaseous oxygen perfusion/persufflation can enhance organ preservation relative to static cold storage or hypothermic machine perfusion, since the lower viscosity of gases, may help reach more regions of preserved organs and deliver more oxygen per gram tissue.<ref name="pmid22301419">{{cite journal | vauthors = Suszynski TM, Rizzari MD, Scott WE, Tempelman LA, Taylor MJ, Papas KK | title = Persufflation (or gaseous oxygen perfusion) as a method of organ preservation | journal = Cryobiology | volume = 64 | issue = 3 | pages = 125–43 | date = June 2012 | pmid = 22301419 | pmc = 3519283 | doi = 10.1016/j.cryobiol.2012.01.007 }}</ref>


== Freezable tissues ==
== Freezable tissues ==
Line 70: Line 70:
* [[Blood]]
* [[Blood]]
** Special cells for transfusion
** Special cells for transfusion
** [[Stem cell]]s. It is optimal in high concentration of synthetic serum, stepwise equilibration and slow cooling.<ref>{{cite journal |display-authors=4|author=Lee JY|author2=Lee JE|author3=Kim DK|author4=Yoon TK|author5=Chung HM|author6=Lee DR |title=High concentration of synthetic serum, stepwise equilibration and slow cooling as an efficient technique for large-scale cryopreservation of human embryonic stem cells |journal=Fertil. Steril. |volume=93 |issue=3 |pages=976–85 |date=November 2008 |pmid=19022437 |doi=10.1016/j.fertnstert.2008.10.017}}</ref>
** [[Stem cell]]s. It is optimal in high concentration of synthetic serum, stepwise equilibration and slow cooling.<ref>{{cite journal | vauthors = Lee JY, Lee JE, Kim DK, Yoon TK, Chung HM, Lee DR | title = High concentration of synthetic serum, stepwise equilibration and slow cooling as an efficient technique for large-scale cryopreservation of human embryonic stem cells | journal = Fertility and Sterility | volume = 93 | issue = 3 | pages = 976–85 | date = February 2010 | pmid = 19022437 | doi = 10.1016/j.fertnstert.2008.10.017 }}</ref>
** [[Umbilical cord blood]] ''Further information: [[Cord blood bank#Cryopreservation]]''
** [[Umbilical cord blood]] ''Further information: [[Cord blood bank#Cryopreservation]]''
* Tissue samples like [[tumor]]s and [[Histology|histological cross sections]]
* Tissue samples like [[tumor]]s and [[Histology|histological cross sections]]
Line 82: Line 82:
Proponents of cryonics claim that cryopreservation using present technology, particularly vitrification of the brain, may be sufficient to preserve people in an "[[information theoretical death|information theoretic]]" sense so that they could be revived and made whole by hypothetical vastly advanced future technology. Not only is there no guarantee of its success, many people{{who|date=February 2019}} argue that human cryopreservation is unethical. According to certain views{{which|date=February 2019}} of the mind body problem, some philosophers{{who|date=February 2019}} believe that the mind, which contains thoughts, memories, and personality, is separate from the brain. When someone dies, their mind leaves the body. If a cryopreserved patient gets successfully resuscitated, no one knows if they would be the same person that they once were or if they would be an empty shell of the memory of who they once were.{{synth|date=February 2019}}
Proponents of cryonics claim that cryopreservation using present technology, particularly vitrification of the brain, may be sufficient to preserve people in an "[[information theoretical death|information theoretic]]" sense so that they could be revived and made whole by hypothetical vastly advanced future technology. Not only is there no guarantee of its success, many people{{who|date=February 2019}} argue that human cryopreservation is unethical. According to certain views{{which|date=February 2019}} of the mind body problem, some philosophers{{who|date=February 2019}} believe that the mind, which contains thoughts, memories, and personality, is separate from the brain. When someone dies, their mind leaves the body. If a cryopreserved patient gets successfully resuscitated, no one knows if they would be the same person that they once were or if they would be an empty shell of the memory of who they once were.{{synth|date=February 2019}}


Right now scientists are trying to see if transplanting cryopreserved human organs for transplantation is viable, if so this would be a major step forward for the possibility of reviving a cryopreserved human.<ref>{{cite web|last1=Devlin|first1=Hannah|title=Cryonics: Does It Offer Humanity a Chance to Return from the Dead?|url=https://www.theguardian.com/science/2016/nov/18/cryogenics-does-it-offer-humanity-a-chance-to-return-from-the-dead|website=thegaurdian.com|publisher=The Guardian|accessdate=17 April 2017}}</ref>
Right now scientists are trying to see if transplanting cryopreserved human organs for transplantation is viable, if so this would be a major step forward for the possibility of reviving a cryopreserved human.<ref>{{cite web|last1=Devlin|first1=Hannah | name-list-format = vanc |title=Cryonics: Does It Offer Humanity a Chance to Return from the Dead?|url=https://www.theguardian.com/science/2016/nov/18/cryogenics-does-it-offer-humanity-a-chance-to-return-from-the-dead|website=thegaurdian.com|publisher=The Guardian|access-date=17 April 2017}}</ref>


===Embryos=== <!--Embryo storage redirects here-->
===Embryos=== <!--Embryo storage redirects here-->
Line 88: Line 88:
Cryopreservation for embryos is used for embryo storage, e.g., when [[in vitro fertilization]] (IVF) has resulted in more embryos than is currently needed.
Cryopreservation for embryos is used for embryo storage, e.g., when [[in vitro fertilization]] (IVF) has resulted in more embryos than is currently needed.


Pregnancies have been reported from embryos stored for 16 years.<ref>[http://www.planer.com/PlanerComWebsite.nsf/7dc62b5d9e44dd40802575460048fc4e/76a07726303dac468025757d005b004d?OpenDocument Planer NEWS and Press Releases > 'Twins' born 16 years apart.]{{dead link|date=August 2017 |bot=InternetArchiveBot |fix-attempted=yes }} January 6, 2006.</ref> Many studies have evaluated the children born from frozen embryos, or “frosties”. The result has been uniformly positive with no increase in birth defects or development abnormalities.<ref>{{cite web|url=http://www.givf.com/fertility/embryofreezing.cfm |archiveurl=https://archive.is/20121206043620/http://www.givf.com/fertility/embryofreezing.shtml |archivedate=December 6, 2012 |title=Genetics & IVF Institute |publisher=Givf.com |accessdate=July 27, 2009 |deadurl=yes |df= }}</ref> A study of more than 11,000 cryopreserved human embryos showed no significant effect of storage time on post-thaw survival for IVF or oocyte donation cycles, or for embryos frozen at the pronuclear or cleavage stages.<ref name=riggs>{{cite journal |display-authors=4 |author=Riggs R |author2=Mayer J |author3=Dowling-Lacey D |author4=Chi TF |author5=Jones E |author6=Oehninger S |title=Does storage time influence postthaw survival and pregnancy outcome? An analysis of 11,768 cryopreserved human embryos |journal=Fertil. Steril. |volume=93 |issue=1 |pages=109–15 |date=November 2008 |pmid=19027110 |doi=10.1016/j.fertnstert.2008.09.084}}</ref> Additionally, the duration of storage did not have any significant effect on clinical pregnancy, miscarriage, implantation, or live birth rate, whether from IVF or oocyte donation cycles.<ref name=riggs/> Rather, oocyte age, survival proportion, and number of transferred embryos are predictors of pregnancy outcome.<ref name=riggs/>
Pregnancies have been reported from embryos stored for 16 years.<ref>[http://www.planer.com/PlanerComWebsite.nsf/7dc62b5d9e44dd40802575460048fc4e/76a07726303dac468025757d005b004d?OpenDocument Planer NEWS and Press Releases > 'Twins' born 16 years apart.]{{dead link|date=August 2017 |bot=InternetArchiveBot |fix-attempted=yes }} January 6, 2006.</ref> Many studies have evaluated the children born from frozen embryos, or “frosties”. The result has been uniformly positive with no increase in birth defects or development abnormalities.<ref>{{cite web|url=http://www.givf.com/fertility/embryofreezing.cfm |archive-url=https://archive.is/20121206043620/http://www.givf.com/fertility/embryofreezing.shtml |archive-date=December 6, 2012 |title=Genetics & IVF Institute |publisher=Givf.com |access-date=July 27, 2009 |deadurl=yes |df= }}</ref> A study of more than 11,000 cryopreserved human embryos showed no significant effect of storage time on post-thaw survival for IVF or oocyte donation cycles, or for embryos frozen at the pronuclear or cleavage stages.<ref name=riggs>{{cite journal | vauthors = Riggs R, Mayer J, Dowling-Lacey D, Chi TF, Jones E, Oehninger S | title = Does storage time influence postthaw survival and pregnancy outcome? An analysis of 11,768 cryopreserved human embryos | journal = Fertility and Sterility | volume = 93 | issue = 1 | pages = 109–15 | date = January 2010 | pmid = 19027110 | doi = 10.1016/j.fertnstert.2008.09.084 }}</ref> Additionally, the duration of storage did not have any significant effect on clinical pregnancy, miscarriage, implantation, or live birth rate, whether from IVF or oocyte donation cycles.<ref name=riggs/> Rather, oocyte age, survival proportion, and number of transferred embryos are predictors of pregnancy outcome.<ref name=riggs/>


=== Ovarian tissue ===
=== Ovarian tissue ===
{{Main|Ovarian tissue cryopreservation}}
{{Main|Ovarian tissue cryopreservation}}
Cryopreservation of ovarian tissue is of interest to women who want to preserve their reproductive function beyond the natural limit, or whose reproductive potential is threatened by cancer therapy,<ref>{{cite journal |author=Isachenko V |author2=Lapidus I |author3=Isachenko E |title=Human ovarian tissue vitrification versus conventional freezing: morphological, endocrinological, and molecular biological evaluation |journal=Reproduction |date=2009 |pages=319–27 |volume=138 |pmid=19439559 |issue=2 |doi=10.1530/REP-09-0039 |display-authors=etal}}</ref> for example in hematologic malignancies or breast cancer.<ref name=Oktay>{{cite journal |author=Oktay K |author2=Oktem O |title=Ovarian cryopreservation and transplantation for fertility preservation for medical indications: report of an ongoing experience |journal=Fertil. Steril. |volume=93 |issue=3 |pages=762–8 |date=November 2008 |pmid=19013568 |doi=10.1016/j.fertnstert.2008.10.006}}</ref> The procedure is to take a part of the ovary and perform slow freezing before storing it in liquid nitrogen whilst therapy is undertaken. Tissue can then be thawed and implanted near the fallopian, either orthotopic (on the natural location) or heterotopic (on the abdominal wall),<ref name=Oktay/> where it starts to produce new eggs, allowing normal conception to occur.<ref>[http://www.saintluc.be/actualites/presse/2004/2004-tamara-lancet-complet.pdf Livebirth after orthotopic transplantation of cryopreserved ovarian tissue]{{Dead link|date=November 2018 |bot=InternetArchiveBot |fix-attempted=yes }} The Lancet, September 24, 2004</ref> The ovarian tissue may also be transplanted into mice that are immunocompromised ([[SCID mice]]) to avoid [[graft rejection]], and tissue can be harvested later when mature follicles have developed.<ref>{{cite journal |display-authors=4|author=Lan C|author2=Xiao W|author3=Xiao-Hui D|author4=Chun-Yan H|author5=Hong-Ling Y |title=Tissue culture before transplantation of frozen-thawed human fetal ovarian tissue into immunodeficient mice |journal=Fertil. Steril. |volume=93 |issue=3 |pages=913–9 |date=December 2008 |pmid=19108826 |doi=10.1016/j.fertnstert.2008.10.020}}</ref>
Cryopreservation of ovarian tissue is of interest to women who want to preserve their reproductive function beyond the natural limit, or whose reproductive potential is threatened by cancer therapy,<ref>{{cite journal | vauthors = Isachenko V, Lapidus I, Isachenko E, Krivokharchenko A, Kreienberg R, Woriedh M, Bader M, Weiss JM | display-authors = 6 | title = Human ovarian tissue vitrification versus conventional freezing: morphological, endocrinological, and molecular biological evaluation | journal = Reproduction | volume = 138 | issue = 2 | pages = 319–27 | date = August 2009 | pmid = 19439559 | doi = 10.1530/REP-09-0039 }}</ref> for example in hematologic malignancies or breast cancer.<ref name=Oktay>{{cite journal | vauthors = Oktay K, Oktem O | title = Ovarian cryopreservation and transplantation for fertility preservation for medical indications: report of an ongoing experience | journal = Fertility and Sterility | volume = 93 | issue = 3 | pages = 762–8 | date = February 2010 | pmid = 19013568 | doi = 10.1016/j.fertnstert.2008.10.006 }}</ref> The procedure is to take a part of the ovary and perform slow freezing before storing it in liquid nitrogen whilst therapy is undertaken. Tissue can then be thawed and implanted near the fallopian, either orthotopic (on the natural location) or heterotopic (on the abdominal wall),<ref name=Oktay/> where it starts to produce new eggs, allowing normal conception to occur.<ref>[http://www.saintluc.be/actualites/presse/2004/2004-tamara-lancet-complet.pdf Livebirth after orthotopic transplantation of cryopreserved ovarian tissue]{{Dead link|date=November 2018 |bot=InternetArchiveBot |fix-attempted=yes }} The Lancet, September 24, 2004</ref> The ovarian tissue may also be transplanted into mice that are immunocompromised ([[SCID mice]]) to avoid [[graft rejection]], and tissue can be harvested later when mature follicles have developed.<ref>{{cite journal | vauthors = Lan C, Xiao W, Xiao-Hui D, Chun-Yan H, Hong-Ling Y | title = Tissue culture before transplantation of frozen-thawed human fetal ovarian tissue into immunodeficient mice | journal = Fertility and Sterility | volume = 93 | issue = 3 | pages = 913–9 | date = February 2010 | pmid = 19108826 | doi = 10.1016/j.fertnstert.2008.10.020 }}</ref>


===Oocytes===
===Oocytes===
Line 104: Line 104:


===Testicular tissue===
===Testicular tissue===
Cryopreservation of immature testicular tissue is a developing method to avail reproduction to young boys who need to have gonadotoxic therapy. Animal data are promising, since healthy offspring have been obtained after transplantation of frozen testicular cell suspensions or tissue pieces. However, none of the fertility restoration options from frozen tissue, i.e. cell suspension transplantation, [[tissue grafting]] and [[in vitro maturation]] (IVM) has proved efficient and safe in humans as yet.<ref>{{cite journal |display-authors=4 |author=Wyns C |author2=Curaba M |author3=Vanabelle B |author4=Van Langendonckt A |author5=Donnez J |title=Options for fertility preservation in prepubertal boys |journal=Hum. Reprod. Update |volume=16 |issue=3 |pages=312–28 |date=2010 |pmid=20047952 |doi=10.1093/humupd/dmp054}}</ref>
Cryopreservation of immature testicular tissue is a developing method to avail reproduction to young boys who need to have gonadotoxic therapy. Animal data are promising, since healthy offspring have been obtained after transplantation of frozen testicular cell suspensions or tissue pieces. However, none of the fertility restoration options from frozen tissue, i.e. cell suspension transplantation, [[tissue grafting]] and [[in vitro maturation]] (IVM) has proved efficient and safe in humans as yet.<ref>{{cite journal | vauthors = Wyns C, Curaba M, Vanabelle B, Van Langendonckt A, Donnez J | title = Options for fertility preservation in prepubertal boys | journal = Human Reproduction Update | volume = 16 | issue = 3 | pages = 312–28 | date = 2010 | pmid = 20047952 | doi = 10.1093/humupd/dmp054 }}</ref>


===Moss===
===Moss===
[[Image:Ecotypes of Physcomitrella patens.JPG|thumb|200px|Four different [[ecotypes]] of ''[[Physcomitrella patens]]'' stored at the [[International Moss Stock Center|IMSC]].]]
[[Image:Ecotypes of Physcomitrella patens.JPG|thumb|200px|Four different [[ecotypes]] of ''[[Physcomitrella patens]]'' stored at the [[International Moss Stock Center|IMSC]].]]
Cryopreservation of whole [[moss]] [[plant]]s, especially [[Physcomitrella patens]], has been developed by [[Ralf Reski]] and coworkers<ref>Schulte, J., Ralf Reski (2004): High-throughput cryopreservation of 140000 Physcomitrella patens mutants. Plant Biol. 6, 119-127. {{cite journal |title=High throughput cryopreservation of 140,000 Physcomitrella patens mutants |author=Schulte J. |author2=Reski R. |publisher=Plant Biotechnology, Freiburg University, Freiburg, Germany |pmid=15045662 |doi=10.1055/s-2004-817796 |volume=6 |issue=2 |journal=Plant Biol (Stuttg) |pages=119–27 |year=2004}}</ref> and is performed at the [[International Moss Stock Center]]. This [[biobank]] collects, preserves, and distributes moss [[mutant]]s and moss [[ecotype]]s.<ref>ScienceDaily: Mosses, deep frozen. {{cite web |title=Mosses, deep-frozen |url=https://www.sciencedaily.com/releases/2010/02/100224134325.htm}}</ref>
Cryopreservation of whole [[moss]] [[plant]]s, especially [[Physcomitrella patens]], has been developed by [[Ralf Reski]] and coworkers<ref>Schulte, J., Ralf Reski (2004): High-throughput cryopreservation of 140000 Physcomitrella patens mutants. Plant Biol. 6, 119-127. {{cite journal | vauthors = Schulte J, Reski R | title = High throughput cryopreservation of 140,000 Physcomitrella patens mutants | journal = Plant Biology | volume = 6 | issue = 2 | pages = 119–27 | year = 2004 | pmid = 15045662 | doi = 10.1055/s-2004-817796 | publisher = Plant Biotechnology, Freiburg University, Freiburg, Germany }}</ref> and is performed at the [[International Moss Stock Center]]. This [[biobank]] collects, preserves, and distributes moss [[mutant]]s and moss [[ecotype]]s.<ref>ScienceDaily: Mosses, deep frozen. {{cite web |title=Mosses, deep-frozen |url=https://www.sciencedaily.com/releases/2010/02/100224134325.htm}}</ref>


===Mesenchymal stromal cells (MSCs)===
===Mesenchymal stromal cells (MSCs)===
MSCs, when transfused immediately within a few hours post-thawing, may show reduced function or show decreased efficacy in treating diseases as compared to those MSCs which are in log phase of cell growth (fresh). As a result, cryopreserved MSCs should be brought back into log phase of cell growth in ''in vitro'' culture before these are administered for clinical trials or experimental therapies. Re-culturing of MSCs will help in recovering from the shock the cells get during freezing and thawing. Various clinical trials on MSCs have failed which used cryopreserved products immediately post-thaw as compared to those clinical trials which used fresh MSCs.<ref>{{cite journal|last1=Francois|first1=M|display-authors=etal|journal=Cytotherapy|date=2012|volume=14|issue=2|pages=147–152|pmc=3279133|title=Cryopreserved mesenchymal stromal cells display impaired immunosuppressive properties as a result of heat-shock response and impaired interferon-γ licensing|doi=10.3109/14653249.2011.623691|pmid=22029655}}</ref>
MSCs, when transfused immediately within a few hours post-thawing, may show reduced function or show decreased efficacy in treating diseases as compared to those MSCs which are in log phase of cell growth (fresh). As a result, cryopreserved MSCs should be brought back into log phase of cell growth in ''in vitro'' culture before these are administered for clinical trials or experimental therapies. Re-culturing of MSCs will help in recovering from the shock the cells get during freezing and thawing. Various clinical trials on MSCs have failed which used cryopreserved products immediately post-thaw as compared to those clinical trials which used fresh MSCs.<ref>{{cite journal | vauthors = François M, Copland IB, Yuan S, Romieu-Mourez R, Waller EK, Galipeau J | title = Cryopreserved mesenchymal stromal cells display impaired immunosuppressive properties as a result of heat-shock response and impaired interferon-γ licensing | journal = Cytotherapy | volume = 14 | issue = 2 | pages = 147–52 | date = February 2012 | pmid = 22029655 | pmc = 3279133 | doi = 10.3109/14653249.2011.623691 }}</ref>


==Preservation of microbiology cultures==
==Preservation of microbiology cultures==
Line 120: Line 120:


===Bacteria===
===Bacteria===
Many common culturable laboratory strains are deep-frozen to preserve genetically and phenotypically stable, long-term stocks. Sub-culturing and prolonged refrigerated samples may lead to loss of plasmid(s) or mutations. Common final glycerol percentages are 15, 20 and 25. From a fresh culture plate, one single colony of interest is chosen and liquid culture is made. From the liquid culture, the medium is directly mixed with equal amount of glycerol; the colony should be checked for any defects like mutations. All antibiotics should be washed from the culture before long-term storage. Methods vary, but mixing can be done gently by inversion or rapidly by vortex and cooling can vary by either placing the cryotube directly at −50 to −95&nbsp;°C, shock-freezing in liquid nitrogen or gradually cooling and then storing at −80&nbsp;°C or cooler (liquid nitrogen or liquid nitrogen vapor). Recovery of bacteria can also vary, namely if beads are stored within the tube then the few beads can be used to plate or the frozen stock can be scraped with a loop and then plated, however, since only little stock is needed the entire tube should never be completely thawed and repeated freeze-thaw should be avoided. 100% recovery is not feasible regardless of methodology.<ref>Freeze-Drying and Cryopreservation of Bacteria</ref><ref>{{cite web|url=http://www.addgene.org/plasmid_protocols/create_glycerol_stock/|title=Addgene: Protocol - How to Create a Bacterial Glycerol Stock|website=Addgene.org|accessdate=9 September 2015}}</ref><ref>{{Cite web |url=http://www.qiagen.com/knowledge-and-support/spotlight/plasmid-resource-center/growth%20of%20bacterial%20cultures/ |title=Archived copy |access-date=2014-05-15 |archive-url=https://web.archive.org/web/20130907031938/http://www.qiagen.com/Knowledge-and-Support/Spotlight/Plasmid-Resource-Center/Growth%20of%20bacterial%20cultures |archive-date=2013-09-07 |dead-url=yes |df= }}</ref>
Many common culturable laboratory strains are deep-frozen to preserve genetically and phenotypically stable, long-term stocks. Sub-culturing and prolonged refrigerated samples may lead to loss of plasmid(s) or mutations. Common final glycerol percentages are 15, 20 and 25. From a fresh culture plate, one single colony of interest is chosen and liquid culture is made. From the liquid culture, the medium is directly mixed with equal amount of glycerol; the colony should be checked for any defects like mutations. All antibiotics should be washed from the culture before long-term storage. Methods vary, but mixing can be done gently by inversion or rapidly by vortex and cooling can vary by either placing the cryotube directly at −50 to −95&nbsp;°C, shock-freezing in liquid nitrogen or gradually cooling and then storing at −80&nbsp;°C or cooler (liquid nitrogen or liquid nitrogen vapor). Recovery of bacteria can also vary, namely if beads are stored within the tube then the few beads can be used to plate or the frozen stock can be scraped with a loop and then plated, however, since only little stock is needed the entire tube should never be completely thawed and repeated freeze-thaw should be avoided. 100% recovery is not feasible regardless of methodology.<ref>Freeze-Drying and Cryopreservation of Bacteria</ref><ref>{{cite web|url=http://www.addgene.org/plasmid_protocols/create_glycerol_stock/|title=Addgene: Protocol - How to Create a Bacterial Glycerol Stock|website=Addgene.org|access-date=9 September 2015}}</ref><ref>{{Cite web |url=http://www.qiagen.com/knowledge-and-support/spotlight/plasmid-resource-center/growth%20of%20bacterial%20cultures/ |title=Archived copy |access-date=2014-05-15 |archive-url=https://web.archive.org/web/20130907031938/http://www.qiagen.com/Knowledge-and-Support/Spotlight/Plasmid-Resource-Center/Growth%20of%20bacterial%20cultures |archive-date=2013-09-07 |dead-url=yes |df= }}</ref>


=== Worms ===
=== Worms ===
Line 139: Line 139:
{{Reflist}}
{{Reflist}}


== Sources ==
== Further reading ==
{{refbegin}}
{{refbegin}}
* {{ cite book |editor=Engelmann, F. |editor2=M. E. Dulloo |editor3=C. Astorga |editor4=S. Dussert |editor5=F. Anthony |date=2007 |title=Conserving coffee genetic resources |publisher=Bioversity International, CATIE, IRD |url=http://www.bioversityinternational.org/Publications/pubfile.asp?ID_PUB=1244 |pages=61}}
* {{ cite book | veditors = Engelmann F, Dulloo ME, Astorga C, Dussert S, Anthony F |date=2007 |title=Conserving coffee genetic resources |publisher=Bioversity International, CATIE, IRD |url=http://www.bioversityinternational.org/Publications/pubfile.asp?ID_PUB=1244 |pages=61}}
* {{ cite book |author=Panis, B |author2=Tien Thinh, N. |last-author-amp=yes |date=2001 |title=Cryopreservation of Musa germplasm |publisher=INIBAP (now Bioversity International) |url=http://www.bioversityinternational.org/Publications/pubfile.asp?ID_PUB=722 |pages=45}}
* {{ cite book | vauthors = Panis B, Tien Thinh N |date=2001 |title=Cryopreservation of Musa germplasm |publisher=INIBAP (now Bioversity International) |url=http://www.bioversityinternational.org/Publications/pubfile.asp?ID_PUB=722 |pages=45}}
* {{cite web |author=ReproTech Limited |date=2012 |title=Fertility Preservation |publisher=ReproTech Limited |url=http://reprotech.com/cryostorage/fertility-preservation/about-fertility-preservation.html |deadurl=yes |archive-url=https://web.archive.org/web/20120904082833/http://reprotech.com/cryostorage/fertility-preservation/about-fertility-preservation.html |archive-date=2012-09-04 |df= }}
* {{cite book | vauthors = Nakasone KK, Peterson SW, Jong SC | chapter = Preservation and distribution of fungal cultures. | title = Biodiversity of fungi: inventory and monitoring methods. | location = Amsterdam | publisher = Elsevier Academic Press | date = 2004 | pages = 37–47 }}
* {{cite journal | vauthors = Perry SF | title = Freeze-drying and cryopreservation of bacteria | journal = Methods in Molecular Biology | location = Clifton, N.J. | volume = 38 | issue = | pages = 21–30 | date = 1995 | pmid = 7647859 | doi = 10.1385/0-89603-296-5:21 }}
{{refend}}
{{refend}}
* {{cite web |author=ReproTech Limited |date=2012 |title=Fertility Preservation |publisher=ReproTech Limited |url=http://reprotech.com/cryostorage/fertility-preservation/about-fertility-preservation.html |deadurl=yes |archiveurl=https://web.archive.org/web/20120904082833/http://reprotech.com/cryostorage/fertility-preservation/about-fertility-preservation.html |archivedate=2012-09-04 |df= }}
{{refend}}
* Nakasone, Karen K, et al., 2004, PRESERVATION AND DISTRIBUTION OF FUNGAL CULTURES.
* Stephen, F., 1995, Freeze-Drying and Cryopreservation of Bacteria


== External links ==
== External links ==

Revision as of 05:59, 25 June 2019

Tubes of biological samples being placed in liquid nitrogen.
Cryogenically preserved samples being removed from a liquid nitrogen dewar.

Cryo-preservation or cryo-conservation is a process where organelles, cells, tissues, extracellular matrix, organs, or any other biological constructs susceptible to damage caused by unregulated chemical kinetics are preserved by cooling to very low temperatures[1] (typically −80 °C using solid carbon dioxide or −196 °C using liquid nitrogen). At low enough temperatures, any enzymatic or chemical activity which might cause damage to the biological material in question is effectively stopped. Cryopreservation methods seek to reach low temperatures without causing additional damage caused by the formation of ice crystals during freezing. Traditional cryopreservation has relied on coating the material to be frozen with a class of molecules termed cryoprotectants. New methods are constantly being investigated due to the inherent toxicity of many cryoprotectants.[2] By default it should be considered that cryopreservation alters or compromises the structure and function of cells unless it is proven otherwise for a particular cell population. Cryoconservation of animal genetic resources is the process in which animal genetic material is collected and stored with the intention of conservation of the breed.

Natural cryopreservation

Water-bears (Tardigrada), microscopic multicellular organisms, can survive freezing by replacing most of their internal water with the sugar trehalose, preventing it from crystallization that otherwise damages cell membranes. Mixtures of solutes can achieve similar effects. Some solutes, including salts, have the disadvantage that they may be toxic at intense concentrations. In addition to the water-bear, wood frogs can tolerate the freezing of their blood and other tissues. Urea is accumulated in tissues in preparation for overwintering, and liver glycogen is converted in large quantities to glucose in response to internal ice formation. Both urea and glucose act as "cryoprotectants" to limit the amount of ice that forms and to reduce osmotic shrinkage of cells. Frogs can survive many freeze/thaw events during winter if no more than about 65% of the total body water freezes. Research exploring the phenomenon of "freezing frogs" has been performed primarily by the Canadian researcher, Dr. Kenneth B. Storey.[citation needed]

Freeze tolerance, in which organisms survive the winter by freezing solid and ceasing life functions, is known in a few vertebrates: five species of frogs (Rana sylvatica, Pseudacris triseriata, Hyla crucifer, Hyla versicolor, Hyla chrysoscelis), one of salamanders (Hynobius keyserlingi), one of snakes (Thamnophis sirtalis) and three of turtles (Chrysemys picta, Terrapene carolina, Terrapene ornata).[3] Snapping turtles Chelydra serpentina and wall lizards Podarcis muralis also survive nominal freezing but it has not been established to be adaptive for overwintering. In the case of Rana sylvatica one cryopreservant is ordinary glucose, which increases in concentration by approximately 19 mmol/l when the frogs are cooled slowly.[3]

History

One of the most important early theoreticians of cryopreservation was James Lovelock. In 1953, he suggested that damage to red blood cells during freezing was due to osmotic stress,[4] and that increasing the salt concentration in a dehydrating cell might damage it.[5][6] In the mid-1950s, he experimented with the cryopreservation of rodents, determining that hamsters could be frozen with 60% of the water in the brain crystallized into ice with no adverse effects; other organs were shown to be susceptible to damage.[7] This work led other scientists to attempt the short-term freezing of rats by 1955, which were fully active 4 to 7 days after being revived.[8]

Cryopreservation was applied to humans beginning in 1954 with three pregnancies resulting from the insemination of previously frozen sperm.[9] Fowl sperm was cryopreserved in 1957 by a team of scientists in the UK directed by Christopher Polge.[10] During 1963, Peter Mazur, at Oak Ridge National Laboratory in the U.S., demonstrated that lethal intracellular freezing could be avoided if cooling was slow enough to permit sufficient water to leave the cell during progressive freezing of the extracellular fluid. That rate differs between cells of differing size and water permeability: a typical cooling rate around 1 °C/minute is appropriate for many mammalian cells after treatment with cryoprotectants such as glycerol or dimethyl sulphoxide, but the rate is not a universal optimum.[11]

The first human body to be frozen with the hope of future revival was James Bedford's, a few hours after his cancer-caused death in 1967.[12] Bedford is the only cryonics patient frozen before 1974 still preserved today.[13]

Temperature

Storage at very low temperatures is presumed to provide an indefinite longevity to cells, although the actual effective life is rather difficult to prove. Researchers experimenting with dried seeds found that there was noticeable variability of deterioration when samples were kept at different temperatures – even ultra-cold temperatures. Temperatures less than the glass transition point (Tg) of polyol's water solutions, around −136 °C (137 K; −213 °F), seem to be accepted as the range where biological activity very substantially slows, and −196 °C (77 K; −321 °F), the boiling point of liquid nitrogen, is the preferred temperature for storing important specimens. While refrigerators, freezers and extra-cold freezers are used for many items, generally the ultra-cold of liquid nitrogen is required for successful preservation of the more complex biological structures to virtually stop all biological activity.

Risks

Phenomena which can cause damage to cells during cryopreservation mainly occur during the freezing stage, and include: solution effects, extracellular ice formation, dehydration and intracellular ice formation. Many of these effects can be reduced by cryoprotectants. Once the preserved material has become frozen, it is relatively safe from further damage. However, estimates based on the accumulation of radiation-induced DNA damage during cryonic storage have suggested a maximum storage period of 1000 years.[14]

Solution effects
As ice crystals grow in freezing water, solutes are excluded, causing them to become concentrated in the remaining liquid water. High concentrations of some solutes can be very damaging.
Extracellular ice formation
When tissues are cooled slowly, water migrates out of cells and ice forms in the extracellular space. Too much extracellular ice can cause mechanical damage to the cell membrane due to crushing.
Dehydration
Migration of water, causing extracellular ice formation, can also cause cellular dehydration. The associated stresses on the cell can cause damage directly.
Intracellular ice formation
While some organisms and tissues can tolerate some extracellular ice, any appreciable intracellular ice is almost always fatal to cells.

Main methods to prevent risks

The main techniques to prevent cryopreservation damages are a well established combination of controlled rate and slow freezing and a newer flash-freezing process known as vitrification.

Slow programmable freezing

A tank of liquid nitrogen, used to supply a cryogenic freezer (for storing laboratory samples at a temperature of about −150 °C)

Controlled-rate and slow freezing, also known as slow programmable freezing (SPF),[15] is a set of well established techniques developed during the early 1970s which enabled the first human embryo frozen birth Zoe Leyland during 1984. Since then, machines that freeze biological samples using programmable sequences, or controlled rates, have been used all over the world for human, animal and cell biology – "freezing down" a sample to better preserve it for eventual thawing, before it is frozen, or cryopreserved, in liquid nitrogen. Such machines are used for freezing oocytes, skin, blood products, embryo, sperm, stem cells and general tissue preservation in hospitals, veterinary practices and research laboratories around the world. As an example, the number of live births from frozen embryos 'slow frozen' is estimated at some 300,000 to 400,000 or 20% of the estimated 3 million in vitro fertilisation (IVF) births.[16]

Lethal intracellular freezing can be avoided if cooling is slow enough to permit sufficient water to leave the cell during progressive freezing of the extracellular fluid. To minimize the growth of extracellular ice crystal growth and recrystallization,[17] biomaterials such as alginates, polyvinyl alcohol or chitosan can be used to impede ice crystal growth along with traditional small molecule cryoprotectants.[18] That rate differs between cells of differing size and water permeability: a typical cooling rate of about 1 °C/minute is appropriate for many mammalian cells after treatment with cryoprotectants such as glycerol or dimethyl sulfoxide, but the rate is not a universal optimum. The 1 °C / minute rate can be achieved by using devices such as a rate-controlled freezer or a benchtop portable freezing container.[19]

Several independent studies have provided evidence that frozen embryos stored using slow-freezing techniques may in some ways be 'better' than fresh in IVF. The studies indicate that using frozen embryos and eggs rather than fresh embryos and eggs reduced the risk of stillbirth and premature delivery though the exact reasons are still being explored.

Vitrification

Researchers Greg Fahy and William F. Rall helped to introduce vitrification to reproductive cryopreservation in the mid-1980s.[20] As of 2000, researchers claim vitrification provides the benefits of cryopreservation without damage due to ice crystal formation.[21] The situation became more complex with the development of tissue engineering as both cells and biomaterials need to remain ice-free to preserve high cell viability and functions, integrity of constructs and structure of biomaterials. Vitrification of tissue engineered constructs was first reported by Lilia Kuleshova,[22] who also was the first scientist to achieve vitrification of oocytes, which resulted in live birth in 1999.[23] For clinical cryopreservation, vitrification usually requires the addition of cryoprotectants prior to cooling. The cryoprotectants act like antifreeze: they decrease the freezing temperature. They also increase the viscosity. Instead of crystallizing, the syrupy solution becomes an amorphous ice—it vitrifies. Rather than a phase change from liquid to solid by crystallization, the amorphous state is like a "solid liquid", and the transformation is over a small temperature range described as the "glass transition" temperature.

Vitrification of water is promoted by rapid cooling, and can be achieved without cryoprotectants by an extremely rapid decrease of temperature (megakelvins per second). The rate that is required to attain glassy state in pure water was considered to be impossible until 2005.[24]

Two conditions usually required to allow vitrification are an increase of the viscosity and a decrease of the freezing temperature. Many solutes do both, but larger molecules generally have a larger effect, particularly on viscosity. Rapid cooling also promotes vitrification.

For established methods of cryopreservation, the solute must penetrate the cell membrane in order to achieve increased viscosity and decrease freezing temperature inside the cell. Sugars do not readily permeate through the membrane. Those solutes that do, such as dimethyl sulfoxide, a common cryoprotectant, are often toxic in intense concentration. One of the difficult compromises of vitrifying cryopreservation concerns limiting the damage produced by the cryoprotectant itself due to cryoprotectant toxicity. Mixtures of cryoprotectants and the use of ice blockers have enabled the Twenty-First Century Medicine company to vitrify a rabbit kidney to −135 °C with their proprietary vitrification mixture. Upon rewarming, the kidney was transplanted successfully into a rabbit, with complete functionality and viability, able to sustain the rabbit indefinitely as the sole functioning kidney.[25]

Persufflation

Blood can be replaced with inert noble gases and/or metabolically vital gases like oxygen, so that organs can cool more quickly and less antifreeze is needed. Since regions of tissue are separated by gas, small expansions do not accumulate, thereby protecting against shattering.[26] A small company, Arigos Biomedical, "has already recovered pig hearts from the 120 degrees below zero",[27] although the definition of "recovered" is not clear. Pressures of 60 atm can help increase heat exchange rates.[28] Gaseous oxygen perfusion/persufflation can enhance organ preservation relative to static cold storage or hypothermic machine perfusion, since the lower viscosity of gases, may help reach more regions of preserved organs and deliver more oxygen per gram tissue.[29]

Freezable tissues

Generally, cryopreservation is easier for thin samples and small clumps of individual cells, because these can be cooled more quickly and so require lesser doses of toxic cryoprotectants. Therefore, cryopreservation of human livers and hearts for storage and transplant is still impractical.

Nevertheless, suitable combinations of cryoprotectants and regimes of cooling and rinsing during warming often allow the successful cryopreservation of biological materials, particularly cell suspensions or thin tissue samples. Examples include:

Additionally, efforts are underway to preserve humans cryogenically, known as cryonics. For such efforts either the brain within the head or the entire body may experience the above process. Cryonics is in a different category from the aforementioned examples, however: while countless cryopreserved cells, vaccines, tissue and other biological samples have been thawed and used successfully, this has not yet been the case at all for cryopreserved brains or bodies. At issue are the criteria for defining "success".

Proponents of cryonics claim that cryopreservation using present technology, particularly vitrification of the brain, may be sufficient to preserve people in an "information theoretic" sense so that they could be revived and made whole by hypothetical vastly advanced future technology. Not only is there no guarantee of its success, many people[who?] argue that human cryopreservation is unethical. According to certain views[which?] of the mind body problem, some philosophers[who?] believe that the mind, which contains thoughts, memories, and personality, is separate from the brain. When someone dies, their mind leaves the body. If a cryopreserved patient gets successfully resuscitated, no one knows if they would be the same person that they once were or if they would be an empty shell of the memory of who they once were.[improper synthesis?]

Right now scientists are trying to see if transplanting cryopreserved human organs for transplantation is viable, if so this would be a major step forward for the possibility of reviving a cryopreserved human.[31]

Embryos

Cryopreservation for embryos is used for embryo storage, e.g., when in vitro fertilization (IVF) has resulted in more embryos than is currently needed.

Pregnancies have been reported from embryos stored for 16 years.[32] Many studies have evaluated the children born from frozen embryos, or “frosties”. The result has been uniformly positive with no increase in birth defects or development abnormalities.[33] A study of more than 11,000 cryopreserved human embryos showed no significant effect of storage time on post-thaw survival for IVF or oocyte donation cycles, or for embryos frozen at the pronuclear or cleavage stages.[34] Additionally, the duration of storage did not have any significant effect on clinical pregnancy, miscarriage, implantation, or live birth rate, whether from IVF or oocyte donation cycles.[34] Rather, oocyte age, survival proportion, and number of transferred embryos are predictors of pregnancy outcome.[34]

Ovarian tissue

Cryopreservation of ovarian tissue is of interest to women who want to preserve their reproductive function beyond the natural limit, or whose reproductive potential is threatened by cancer therapy,[35] for example in hematologic malignancies or breast cancer.[36] The procedure is to take a part of the ovary and perform slow freezing before storing it in liquid nitrogen whilst therapy is undertaken. Tissue can then be thawed and implanted near the fallopian, either orthotopic (on the natural location) or heterotopic (on the abdominal wall),[36] where it starts to produce new eggs, allowing normal conception to occur.[37] The ovarian tissue may also be transplanted into mice that are immunocompromised (SCID mice) to avoid graft rejection, and tissue can be harvested later when mature follicles have developed.[38]

Oocytes

Human oocyte cryopreservation is a new technology in which a woman’s eggs (oocytes) are extracted, frozen and stored. Later, when she is ready to become pregnant, the eggs can be thawed, fertilized, and transferred to the uterus as embryos. Since 1999, when the birth of the first baby from an embryo derived from vitrified-warmed woman’s eggs was reported by Kuleshova and co-workers in the journal of Human Reproduction,[22] this concept has been recognized and widespread. This break-through in achieving vitrification of woman’s oocytes made an important advance in our knowledge and practice of the IVF process, as clinical pregnancy rate is four times higher after oocyte vitrification than after slow freezing.[39] Oocyte vitrification is vital for preservation fertility in young oncology patients and for individuals undergoing IVF who object, either for religious or ethical reasons, to the practice of freezing embryos.

Semen

Semen can be used successfully almost indefinitely after cryopreservation. The longest reported successful storage is 22 years.[40] It can be used for sperm donation where the recipient wants the treatment in a different time or place, or as a means of preserving fertility for men undergoing vasectomy or treatments that may compromise their fertility, such as chemotherapy, radiation therapy or surgery.

Testicular tissue

Cryopreservation of immature testicular tissue is a developing method to avail reproduction to young boys who need to have gonadotoxic therapy. Animal data are promising, since healthy offspring have been obtained after transplantation of frozen testicular cell suspensions or tissue pieces. However, none of the fertility restoration options from frozen tissue, i.e. cell suspension transplantation, tissue grafting and in vitro maturation (IVM) has proved efficient and safe in humans as yet.[41]

Moss

Four different ecotypes of Physcomitrella patens stored at the IMSC.

Cryopreservation of whole moss plants, especially Physcomitrella patens, has been developed by Ralf Reski and coworkers[42] and is performed at the International Moss Stock Center. This biobank collects, preserves, and distributes moss mutants and moss ecotypes.[43]

Mesenchymal stromal cells (MSCs)

MSCs, when transfused immediately within a few hours post-thawing, may show reduced function or show decreased efficacy in treating diseases as compared to those MSCs which are in log phase of cell growth (fresh). As a result, cryopreserved MSCs should be brought back into log phase of cell growth in in vitro culture before these are administered for clinical trials or experimental therapies. Re-culturing of MSCs will help in recovering from the shock the cells get during freezing and thawing. Various clinical trials on MSCs have failed which used cryopreserved products immediately post-thaw as compared to those clinical trials which used fresh MSCs.[44]

Preservation of microbiology cultures

Bacteria and fungi can be kept short-term (months to about a year, depending) refrigerated, however, cell division and metabolism is not completely arrested and thus is not an optimal option for long-term storage (years) or to preserve cultures genetically or phenotypically, as cell divisions can lead to mutations or sub-culturing can cause phenotypic changes. A preferred option, species-dependent, is cryopreservation. Nematode worms are the only multicellular eukaryotes that have been shown to survive cryopreservation. [45][46]

Fungi

Fungi, notably zygomycetes, ascomycetes and higher basidiomycetes, regardless of sporulation, are able to be stored in liquid nitrogen or deep-frozen. Crypreservation is a hallmark method for fungi that do not sporulate (otherwise other preservation methods for spores can be used at lower costs and ease), sporulate but have delicate spores (large or freeze-dry sensitive), are pathogenic (dangerous to keep metabolically active fungus) or are to be used for genetic stocks (ideally to have identical composition as the original deposit). As with many other organisms, cryoprotectants like DMSO or glycerol (e.g. filamentous fungi 10% glycerol or yeast 20% glycerol) are used. Differences between choosing cryoprotectants are species (or class) dependent, but generally for fungi penetrating cryoprotectants like DMSO, glycerol or polyethylene glycol are most effective (other non-penetrating ones include sugars mannitol, sorbitol, dextran, etc.). Freeze-thaw repetition is not recommended as it can decrease viability. Back-up deep-freezers or liquid nitrogen storage sites are recommended. Multiple protocols for freezing are summarized below (each uses screw-cap polypropylene cryotubes):[47]

Bacteria

Many common culturable laboratory strains are deep-frozen to preserve genetically and phenotypically stable, long-term stocks. Sub-culturing and prolonged refrigerated samples may lead to loss of plasmid(s) or mutations. Common final glycerol percentages are 15, 20 and 25. From a fresh culture plate, one single colony of interest is chosen and liquid culture is made. From the liquid culture, the medium is directly mixed with equal amount of glycerol; the colony should be checked for any defects like mutations. All antibiotics should be washed from the culture before long-term storage. Methods vary, but mixing can be done gently by inversion or rapidly by vortex and cooling can vary by either placing the cryotube directly at −50 to −95 °C, shock-freezing in liquid nitrogen or gradually cooling and then storing at −80 °C or cooler (liquid nitrogen or liquid nitrogen vapor). Recovery of bacteria can also vary, namely if beads are stored within the tube then the few beads can be used to plate or the frozen stock can be scraped with a loop and then plated, however, since only little stock is needed the entire tube should never be completely thawed and repeated freeze-thaw should be avoided. 100% recovery is not feasible regardless of methodology.[48][49][50]

Worms

The microscopic soil-dwelling nematode roundworms Panagrolaimus detritophagus and Plectus parvus are the only eukaryotic organisms that have been proven to be viable after long-term cryopreservation to date. In this case, the preservation was natural rather than artificial, due to permafrost.

See also

References

  1. ^ Pegg DE (January 1, 2007). "Principles of cryopreservation". Methods in Molecular Biology. 368: 39–57. doi:10.1007/978-1-59745-362-2_3. ISBN 978-1-58829-377-0. PMID 18080461.
  2. ^ Sambu S (June 25, 2015). "A Bayesian approach to optimizing cryopreservation protocols". PeerJ. 3: e1039. doi:10.7717/peerj.1039. PMC 4485240. PMID 26131379.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  3. ^ a b Costanzo JP, Lee RE, Wright MF (December 1991). "Glucose loading prevents freezing injury in rapidly cooled wood frogs" (PDF). The American Journal of Physiology. 261 (6 Pt 2): R1549–53. doi:10.1152/ajpregu.1991.261.6.R1549. PMID 1750578.
  4. ^ Lovelock JE (March 1953). "The haemolysis of human red blood-cells by freezing and thawing". Biochimica et Biophysica Acta. 10 (3): 414–26. doi:10.1016/0006-3002(53)90273-X. PMID 13058999.
  5. ^ Fuller, Barry J.; Lane, Nick; Benson, Erica E., eds. (2004). Life in the Frozen State. CRC Press. p. 7. ISBN 978-0203647073. {{cite book}}: Unknown parameter |name-list-format= ignored (|name-list-style= suggested) (help)
  6. ^ Mazur P (May 1970). "Cryobiology: the freezing of biological systems". Science. 168 (3934): 939–49. Bibcode:1970Sci...168..939M. doi:10.1126/science.168.3934.939. PMID 5462399.
  7. ^ "The Cryobiological Case for Cryonics" (PDF). Cryonics. Vol. 9(3), no. 92. Alcor Life Extension Foundation. March 1988. p. 27.
  8. ^ Andjus RK, Smith AU (June 1955). "Reanimation of adult rats from body temperatures between 0 and + 2 degrees C". The Journal of Physiology. 128 (3): 446–72. doi:10.1113/jphysiol.1955.sp005318. PMC 1365897. PMID 13243342.
  9. ^ "Fatherhood After Death Has Now Been Proved Possible". Cedar Rapids Gazette. April 9, 1954.
  10. ^ Polge C (December 1957). "Low-temperature storage of mammalian spermatozoa". Proceedings of the Royal Society of London. Series B, Biological Sciences. 147 (929): 498–508. Bibcode:1957RSPSB.147..498P. doi:10.1098/rspb.1957.0068. PMID 13494462.
  11. ^ Mazur P (July 1963). "Studies on rapidly frozen suspensions of yeast cells by differential thermal analysis and conductometry" (PDF). Biophysical Journal. 3 (4): 323–53. Bibcode:1963BpJ.....3..323M. doi:10.1016/S0006-3495(63)86824-1. PMC 1366450. PMID 13934216.
  12. ^ "Dear Dr. Bedford (and those who will care for you after I do)". Cryonics. July 1991. Retrieved 2009-08-23.
  13. ^ Perry, R. Michael (October 2014). "Suspension Failures – Lessons from the Early Days". ALCOR: Life Extension Foundation. Retrieved August 29, 2018. {{cite web}}: Unknown parameter |name-list-format= ignored (|name-list-style= suggested) (help)
  14. ^ Mazur P (September 1984). "Freezing of living cells: mechanisms and implications". The American Journal of Physiology. 247 (3 Pt 1): C125-42. Bibcode:1957RSPSB.147..498P. doi:10.1098/rspb.1957.0068. PMID 6383068.
  15. ^ Vutyavanich T, Piromlertamorn W, Nunta S (April 2010). "Rapid freezing versus slow programmable freezing of human spermatozoa". Fertility and Sterility. 93 (6): 1921–8. doi:10.1016/j.fertnstert.2008.04.076. PMID 19243759.
  16. ^ "dead link". Archived from the original on 2009-05-26. {{cite web}}: Unknown parameter |dead-url= ignored (|url-status= suggested) (help)
  17. ^ Deller RC, Vatish M, Mitchell DA, Gibson MI (February 3, 2014). "Synthetic polymers enable non-vitreous cellular cryopreservation by reducing ice crystal growth during thawing". Nature Communications. 5: 3244. Bibcode:2014NatCo...5E3244D. doi:10.1038/ncomms4244. PMID 24488146.
  18. ^ Sambu S (June 25, 2015). "A Bayesian approach to optimizing cryopreservation protocols". PeerJ. 3: e1039. doi:10.7717/peerj.1039. PMC 4485240. PMID 26131379.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  19. ^ Thompson M, Nemits M, Ehrhardt R (May 2011). "Rate-controlled Cryopreservation and Thawing of Mammalian Cells". Protocol Exchange. doi:10.1038/protex.2011.224.
  20. ^ Rall WF, Fahy GM (February 14–20, 1985). "Ice-free cryopreservation of mouse embryos at -196 degrees C by vitrification". Nature. 313 (6003): 573–5. Bibcode:1985Natur.313..573R. doi:10.1038/313573a0. PMID 3969158.
  21. ^ "Alcor: The Origin of Our Name" (PDF). Alcor Life Extension Foundation. Winter 2000. Retrieved August 25, 2009.
  22. ^ a b Kuleshova LL, Wang XW, Wu YN, Zhou Y, Yu H (2004). "Vitrification of encapsulated hepatocytes with reduced cooling and warming rates". Cryo Letters. 25 (4): 241–54. PMID 15375435.
  23. ^ Kuleshova L, Gianaroli L, Magli C, Ferraretti A, Trounson A (December 1999). "Birth following vitrification of a small number of human oocytes: case report". Human Reproduction. 14 (12): 3077–9. doi:10.1093/humrep/14.12.3077. PMID 10601099.
  24. ^ Bhat SN, Sharma A, Bhat SV (December 2005). "Vitrification and glass transition of water: insights from spin probe ESR". Physical Review Letters. 95 (23): 235702. arXiv:cond-mat/0409440. Bibcode:2005PhRvL..95w5702B. doi:10.1103/PhysRevLett.95.235702. PMID 16384318.
  25. ^ Fahy GM, Wowk B, Pagotan R, Chang A, Phan J, Thomson B, Phan L (July 2009). "Physical and biological aspects of renal vitrification". Organogenesis. 5 (3): 167–75. doi:10.4161/org.5.3.9974. PMC 2781097. PMID 20046680.
  26. ^ Geddes, Linda (Sep 11, 2013). "Heart of glass could be key to banking organs". New Scientist. {{cite journal}}: Unknown parameter |name-list-format= ignored (|name-list-style= suggested) (help)
  27. ^ Flynn, Matthew (Oct 10, 2018). "Heart of Ice". BOSS Magazine. {{cite journal}}: Unknown parameter |name-list-format= ignored (|name-list-style= suggested) (help)
  28. ^ US9314015B2, Stephen Van Sickle, Tanya Jones, "Method and apparatus for prevention of thermo-mechanical fracturing in vitrified tissue using rapid cooling and warming by persufflation", published Apr 19, 2013 
  29. ^ Suszynski TM, Rizzari MD, Scott WE, Tempelman LA, Taylor MJ, Papas KK (June 2012). "Persufflation (or gaseous oxygen perfusion) as a method of organ preservation". Cryobiology. 64 (3): 125–43. doi:10.1016/j.cryobiol.2012.01.007. PMC 3519283. PMID 22301419.
  30. ^ Lee JY, Lee JE, Kim DK, Yoon TK, Chung HM, Lee DR (February 2010). "High concentration of synthetic serum, stepwise equilibration and slow cooling as an efficient technique for large-scale cryopreservation of human embryonic stem cells". Fertility and Sterility. 93 (3): 976–85. doi:10.1016/j.fertnstert.2008.10.017. PMID 19022437.
  31. ^ Devlin, Hannah. "Cryonics: Does It Offer Humanity a Chance to Return from the Dead?". thegaurdian.com. The Guardian. Retrieved 17 April 2017. {{cite web}}: Unknown parameter |name-list-format= ignored (|name-list-style= suggested) (help)
  32. ^ Planer NEWS and Press Releases > 'Twins' born 16 years apart.[permanent dead link] January 6, 2006.
  33. ^ "Genetics & IVF Institute". Givf.com. Archived from the original on December 6, 2012. Retrieved July 27, 2009. {{cite web}}: Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  34. ^ a b c Riggs R, Mayer J, Dowling-Lacey D, Chi TF, Jones E, Oehninger S (January 2010). "Does storage time influence postthaw survival and pregnancy outcome? An analysis of 11,768 cryopreserved human embryos". Fertility and Sterility. 93 (1): 109–15. doi:10.1016/j.fertnstert.2008.09.084. PMID 19027110.
  35. ^ Isachenko V, Lapidus I, Isachenko E, Krivokharchenko A, Kreienberg R, Woriedh M, et al. (August 2009). "Human ovarian tissue vitrification versus conventional freezing: morphological, endocrinological, and molecular biological evaluation". Reproduction. 138 (2): 319–27. doi:10.1530/REP-09-0039. PMID 19439559.
  36. ^ a b Oktay K, Oktem O (February 2010). "Ovarian cryopreservation and transplantation for fertility preservation for medical indications: report of an ongoing experience". Fertility and Sterility. 93 (3): 762–8. doi:10.1016/j.fertnstert.2008.10.006. PMID 19013568.
  37. ^ Livebirth after orthotopic transplantation of cryopreserved ovarian tissue[permanent dead link] The Lancet, September 24, 2004
  38. ^ Lan C, Xiao W, Xiao-Hui D, Chun-Yan H, Hong-Ling Y (February 2010). "Tissue culture before transplantation of frozen-thawed human fetal ovarian tissue into immunodeficient mice". Fertility and Sterility. 93 (3): 913–9. doi:10.1016/j.fertnstert.2008.10.020. PMID 19108826.
  39. ^ Glujovsky D, Riestra B, Sueldo C, Fiszbajn G, Repping S, Nodar F, Papier S, Ciapponi A. Vitrification versus slow freezing for women undergoing oocyte cryopreservation" Cochrane Database of Systematic Reviews 2014, Issue 8. Art. No.: CD010047. doi:10.1002/14651858.CD010047.pub2
  40. ^ Planer NEWS and Press Releases > Child born after 22 year semen storage using Planer controlled rate freezer Archived 2012-09-08 at archive.today 14/10/2004
  41. ^ Wyns C, Curaba M, Vanabelle B, Van Langendonckt A, Donnez J (2010). "Options for fertility preservation in prepubertal boys". Human Reproduction Update. 16 (3): 312–28. doi:10.1093/humupd/dmp054. PMID 20047952.
  42. ^ Schulte, J., Ralf Reski (2004): High-throughput cryopreservation of 140000 Physcomitrella patens mutants. Plant Biol. 6, 119-127. Schulte J, Reski R (2004). "High throughput cryopreservation of 140,000 Physcomitrella patens mutants". Plant Biology. 6 (2). Plant Biotechnology, Freiburg University, Freiburg, Germany: 119–27. doi:10.1055/s-2004-817796. PMID 15045662.
  43. ^ ScienceDaily: Mosses, deep frozen. "Mosses, deep-frozen".
  44. ^ François M, Copland IB, Yuan S, Romieu-Mourez R, Waller EK, Galipeau J (February 2012). "Cryopreserved mesenchymal stromal cells display impaired immunosuppressive properties as a result of heat-shock response and impaired interferon-γ licensing". Cytotherapy. 14 (2): 147–52. doi:10.3109/14653249.2011.623691. PMC 3279133. PMID 22029655.
  45. ^ Weisberger, M. (2018). Worms Frozen for 42,000 Years in Siberian Permafrost Wriggle to Life. Retrieved from https://www.livescience.com/63187-siberian-permafrost-worms-revive.html
  46. ^ Shatilovich, A.V., Tchesunov, A.V., Neretina, T.V. et al. Dokl Biol Sci (2018) 480: 100. https://doi.org/10.1134/S0012496618030079
  47. ^ "Archived copy" (PDF). Archived from the original (PDF) on 2014-05-17. Retrieved 2014-05-15. {{cite web}}: Unknown parameter |dead-url= ignored (|url-status= suggested) (help)CS1 maint: archived copy as title (link)
  48. ^ Freeze-Drying and Cryopreservation of Bacteria
  49. ^ "Addgene: Protocol - How to Create a Bacterial Glycerol Stock". Addgene.org. Retrieved 9 September 2015.
  50. ^ "Archived copy". Archived from the original on 2013-09-07. Retrieved 2014-05-15. {{cite web}}: Unknown parameter |dead-url= ignored (|url-status= suggested) (help)CS1 maint: archived copy as title (link)

Further reading

  • Engelmann F, Dulloo ME, Astorga C, Dussert S, Anthony F, eds. (2007). Conserving coffee genetic resources. Bioversity International, CATIE, IRD. p. 61.
  • Panis B, Tien Thinh N (2001). Cryopreservation of Musa germplasm. INIBAP (now Bioversity International). p. 45.
  • ReproTech Limited (2012). "Fertility Preservation". ReproTech Limited. Archived from the original on 2012-09-04. {{cite web}}: Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  • Nakasone KK, Peterson SW, Jong SC (2004). "Preservation and distribution of fungal cultures.". Biodiversity of fungi: inventory and monitoring methods. Amsterdam: Elsevier Academic Press. pp. 37–47.
  • Perry SF (1995). "Freeze-drying and cryopreservation of bacteria". Methods in Molecular Biology. 38. Clifton, N.J.: 21–30. doi:10.1385/0-89603-296-5:21. PMID 7647859.

External links