Jump to content

Sonoporation: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
Bmaghen (talk | contribs)
Added content to Microbubble agents section from my sandbox User:Your Bmaghen/sandbox
Bmaghen (talk | contribs)
Added the Membrane translocation mechanism section from my sandbox User:Bmaghen/sandbox
Line 13: Line 13:


Microbubbles have a gas core with high compressibility relative to their liquid environment, making them highly responsive to acoustic application. As a result of ultrasound stimulation, microbubbles undergo rapid expansion and contraction, otherwise known as [[cavitation]]. When a cell is attached to the cell membrane, the cavitation oscillations produced by ultrasound stimulation will push and pull on the membrane to create a membrane opening. These rapid oscillations are also responsible for adjacent fluid flow called microstreaming which increases pressure on surrounding cells producing further sonoporation to whole cell populations.<ref>{{Cite journal|last=Fan|first=Zhenzhen|last2=Kumon|first2=Ronald E|last3=Deng|first3=Cheri X|date=2014-04|title=Mechanisms of microbubble-facilitated sonoporation for drug and gene delivery|url=http://dx.doi.org/10.4155/tde.14.10|journal=Therapeutic Delivery|volume=5|issue=4|pages=467–486|doi=10.4155/tde.14.10|issn=2041-5990}}</ref>
Microbubbles have a gas core with high compressibility relative to their liquid environment, making them highly responsive to acoustic application. As a result of ultrasound stimulation, microbubbles undergo rapid expansion and contraction, otherwise known as [[cavitation]]. When a cell is attached to the cell membrane, the cavitation oscillations produced by ultrasound stimulation will push and pull on the membrane to create a membrane opening. These rapid oscillations are also responsible for adjacent fluid flow called microstreaming which increases pressure on surrounding cells producing further sonoporation to whole cell populations.<ref>{{Cite journal|last=Fan|first=Zhenzhen|last2=Kumon|first2=Ronald E|last3=Deng|first3=Cheri X|date=2014-04|title=Mechanisms of microbubble-facilitated sonoporation for drug and gene delivery|url=http://dx.doi.org/10.4155/tde.14.10|journal=Therapeutic Delivery|volume=5|issue=4|pages=467–486|doi=10.4155/tde.14.10|issn=2041-5990}}</ref>

== Membrane translocation mechanism ==
While sonoporation has been validated in preclinical studies, the way in which molecules cross cellular membrane barriers remains unclear. Different theories exist that may potentially explain barrier permeabilization and molecular delivery. The dominant hypotheses include pore formation, [[Endocytosis|endocytosis,]] and membrane wounds.
[[File:Endocytosis.png|thumb|417x417px|Schematic representation of molecular translocation via endocytosis. The second representation from the left illustrates the endocytotic mechanism involving clathrin-coated pits.]]
Pore formation following ultrasound application was first reported in 1999 in a study that observed cell membrane craters following ultrasound application at 255 kHz.<ref name=":0">{{Citation|last=Bouakaz|first=Ayache|title=Sonoporation: Concept and Mechanisms|date=2016|url=https://doi.org/10.1007/978-3-319-22536-4_10|work=Therapeutic Ultrasound|pages=175–189|editor-last=Escoffre|editor-first=Jean-Michel|series=Advances in Experimental Medicine and Biology|place=Cham|publisher=Springer International Publishing|language=en|doi=10.1007/978-3-319-22536-4_10|isbn=978-3-319-22536-4|access-date=2021-10-27|last2=Zeghimi|first2=Aya|last3=Doinikov|first3=Alexander A.|editor2-last=Bouakaz|editor2-first=Ayache}}</ref> Later, sonoporation mediated [[microinjection]] of [[dextran]] molecules showed that membrane permeability mechanisms differ depending on the size of dextran molecules. Microinjection of dextran molecules from 3 to 70 kDa was reported to have crossed the cellular membrane via transient pores. In contrast, dextran molecules of 155 and 500 kDa were predominantly found in vesicle-like structures, likely indicating the mechanism of endocytosis.<ref>{{Cite journal|last=Meijering|first=Bernadet D.M.|last2=Juffermans|first2=Lynda J.M.|last3=van Wamel|first3=Annemieke|last4=Henning|first4=Rob H.|last5=Zuhorn|first5=Inge S.|last6=Emmer|first6=Marcia|last7=Versteilen|first7=Amanda M.G.|last8=Paulus|first8=Walter J.|last9=van Gilst|first9=Wiek H.|last10=Kooiman|first10=Klazina|last11=de Jong|first11=Nico|date=2009-03-13|title=Ultrasound and Microbubble-Targeted Delivery of Macromolecules Is Regulated by Induction of Endocytosis and Pore Formation|url=https://www.ahajournals.org/doi/10.1161/CIRCRESAHA.108.183806|journal=Circulation Research|volume=104|issue=5|pages=679–687|doi=10.1161/CIRCRESAHA.108.183806}}</ref> This variability in membrane behavior has led to other studies investigating membrane rupture and resealing characteristics depending on ultrasound amplitude and duration.

Various cellular reactions to ultrasound indicate the mechanism of molecular uptake via endocytosis. These observed reactionary phenomena include [[ion exchange]], hydrogen peroxide, and cell intracellular calcium concentration. Studies have used patch clamping techniques to monitor membrane potential ion exchange for the role of endocytosis in sonoporation. Ultrasound application to cells and adjacent microbubbles was shown to produce marked cell membrane hyperpolarization along with progressive intracellular calcium increase, which is believed to be a consequence of calcium channels opening in response to microbubble oscillations. These findings act as support for ultrasound application inducing calcium-mediated uncoating of clathrin-coated pits seen in traditional endocytosis pathways.<ref>{{Cite journal|last=Hauser|first=Joerg|last2=Ellisman|first2=Mark|last3=Steinau|first3=Hans-Ulrich|last4=Stefan|first4=Esenwein|last5=Dudda|first5=Marcel|last6=Hauser|first6=Manfred|date=2009-12|title=Ultrasound Enhanced Endocytotic Activity of Human Fibroblasts|url=http://dx.doi.org/10.1016/j.ultrasmedbio.2009.06.1090|journal=Ultrasound in Medicine & Biology|volume=35|issue=12|pages=2084–2092|doi=10.1016/j.ultrasmedbio.2009.06.1090|issn=0301-5629}}</ref><ref>{{Cite journal|last=Tran|first=T.A.|last2=Roger|first2=S.|last3=Le Guennec|first3=J.Y.|last4=Tranquart|first4=F.|last5=Bouakaz|first5=A.|date=2007-01|title=Effect of ultrasound-activated microbubbles on the cell electrophysiological properties|url=http://dx.doi.org/10.1016/j.ultrasmedbio.2006.07.029|journal=Ultrasound in Medicine & Biology|volume=33|issue=1|pages=158–163|doi=10.1016/j.ultrasmedbio.2006.07.029|issn=0301-5629}}</ref> Other work reported sonoporation induced the formation of hydrogen peroxide, a cellular reaction that is also known to be involved with endocytosis.<ref name=":0" />

Mechanically created wounds in the plasma membrane have been observed as a result of sonoporation-produced shear forces. The nature of these wounds may vary based on the degree of acoustic cavitation leading to a spectrum of cell behavior, from membrane [[Bleb (cell biology)|blebbing]] to instant [[Lysis|cell lysis]]. Multiple studies examining membrane wounds note observing resealing behavior, a process dependent on recruitment of ATP and intracellular vesicles. <ref name=":0" />

Following sonoporation-mediated membrane permeabilization, cells can automatically repair the membrane openings through a phenomenon called "reparable sonoporation."<ref name=":1">{{Cite journal|last=Wu|first=Junru|date=2018-12-18|title=Acoustic Streaming and Its Applications|url=http://dx.doi.org/10.3390/fluids3040108|journal=Fluids|volume=3|issue=4|pages=108|doi=10.3390/fluids3040108|issn=2311-5521}}</ref>


==References==
==References==

Revision as of 00:40, 7 November 2021

Schematic of Sonoporation Mechanism. This figure depicts the general understanding of sonoporation where a dedicated sonoporator applies ultrasound to induce microbubble cavitation and eventually pore formation. The therapeutic gene or drug of interest thus may translocate within the cell.

Sonoporation, or cellular sonication, is the use of sound (typically ultrasonic frequencies) for modifying the permeability of the cell plasma membrane. This technique is usually used in molecular biology and non-viral gene therapy in order to allow uptake of large molecules such as DNA into the cell, in a cell disruption process called transfection or transformation. Sonoporation employs the acoustic cavitation of microbubbles to enhance delivery of these large molecules.[1] The bioactivity of this technique is similar to, and in some cases found superior to, electroporation. Extended exposure to low-frequency (<MHz) ultrasound has been demonstrated to result in complete cellular death (rupturing), thus cellular viability must also be accounted for when employing this technique. Through a phenomenon called "reparable sonoporation", cells can automatically repair the formed pores if they are small enough.[2]

Sonoporation is under active study for the introduction of foreign genes in tissue culture cells, especially mammalian cells. Sonoporation is also being studied for use in targeted Gene therapy in vivo, in a medical treatment scenario whereby a patient is given modified DNA, and an ultrasonic transducer might target this modified DNA into specific regions of the patient's body.[3]

Equipment

Sonoporation is performed with a dedicated sonoporator. Sonoporation may also be performed with custom-built piezoelectric transducers connected to bench-top function generators and acoustic amplifiers. Standard ultrasound medical devices may also be used in some applications.

Measurement of the acoustics used in sonoporation is listed in terms of mechanical index, which quantifies the likelihood that exposure to diagnostic ultrasound will produce an adverse biological effect by a non-thermal action based on pressure.[4]

Microbubble agents

Sonoporation uses microbubbles for significantly enhancing transfection, and in some cases is required for DNA uptake.[5] These microbubble agents include Optison, manufactured by General Electric Healthcare.

Microbubbles have a gas core with high compressibility relative to their liquid environment, making them highly responsive to acoustic application. As a result of ultrasound stimulation, microbubbles undergo rapid expansion and contraction, otherwise known as cavitation. When a cell is attached to the cell membrane, the cavitation oscillations produced by ultrasound stimulation will push and pull on the membrane to create a membrane opening. These rapid oscillations are also responsible for adjacent fluid flow called microstreaming which increases pressure on surrounding cells producing further sonoporation to whole cell populations.[6]

Membrane translocation mechanism

While sonoporation has been validated in preclinical studies, the way in which molecules cross cellular membrane barriers remains unclear. Different theories exist that may potentially explain barrier permeabilization and molecular delivery. The dominant hypotheses include pore formation, endocytosis, and membrane wounds.

Schematic representation of molecular translocation via endocytosis. The second representation from the left illustrates the endocytotic mechanism involving clathrin-coated pits.

Pore formation following ultrasound application was first reported in 1999 in a study that observed cell membrane craters following ultrasound application at 255 kHz.[7] Later, sonoporation mediated microinjection of dextran molecules showed that membrane permeability mechanisms differ depending on the size of dextran molecules. Microinjection of dextran molecules from 3 to 70 kDa was reported to have crossed the cellular membrane via transient pores. In contrast, dextran molecules of 155 and 500 kDa were predominantly found in vesicle-like structures, likely indicating the mechanism of endocytosis.[8] This variability in membrane behavior has led to other studies investigating membrane rupture and resealing characteristics depending on ultrasound amplitude and duration.

Various cellular reactions to ultrasound indicate the mechanism of molecular uptake via endocytosis. These observed reactionary phenomena include ion exchange, hydrogen peroxide, and cell intracellular calcium concentration. Studies have used patch clamping techniques to monitor membrane potential ion exchange for the role of endocytosis in sonoporation. Ultrasound application to cells and adjacent microbubbles was shown to produce marked cell membrane hyperpolarization along with progressive intracellular calcium increase, which is believed to be a consequence of calcium channels opening in response to microbubble oscillations. These findings act as support for ultrasound application inducing calcium-mediated uncoating of clathrin-coated pits seen in traditional endocytosis pathways.[9][10] Other work reported sonoporation induced the formation of hydrogen peroxide, a cellular reaction that is also known to be involved with endocytosis.[7]

Mechanically created wounds in the plasma membrane have been observed as a result of sonoporation-produced shear forces. The nature of these wounds may vary based on the degree of acoustic cavitation leading to a spectrum of cell behavior, from membrane blebbing to instant cell lysis. Multiple studies examining membrane wounds note observing resealing behavior, a process dependent on recruitment of ATP and intracellular vesicles. [7]

Following sonoporation-mediated membrane permeabilization, cells can automatically repair the membrane openings through a phenomenon called "reparable sonoporation."[11]

References

  1. ^ Yizhi Song (2007). "Ultrasound-mediated DNA transfer for bacteria". Nucleic Acids Res. 35 (19): e129. doi:10.1093/nar/gkm710. PMC 2095817. PMID 17890732.
  2. ^ Wu, Junru (December 2018). "Acoustic Streaming and Its Applications". Fluids. 3 (4): 108. doi:10.3390/fluids3040108.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  3. ^ Junru Wu; Wesley Le Mars Nyborg (2006). Emerging Therapeutic Ultrasound. ISBN 978-981-256-685-0.
  4. ^ Charles C. Church (2005). "Frequency, pulse length, and the mechanical index". Acoustics Research Letters Online. 6 (3): 162–168. doi:10.1121/1.1901757.
  5. ^ P.A Dijkmans; Juffermans, LJ; Musters, RJ; Van Wamel, A; Ten Cate, FJ; Van Gilst, W; Visser, CA; De Jong, N; Kamp, O (2004). "Microbubbles and ultrasound: from diagnosis to therapy". European Journal of Echocardiography. 5 (4): 245–246. doi:10.1016/j.euje.2004.02.001. PMID 15219539.
  6. ^ Fan, Zhenzhen; Kumon, Ronald E; Deng, Cheri X (2014-04). "Mechanisms of microbubble-facilitated sonoporation for drug and gene delivery". Therapeutic Delivery. 5 (4): 467–486. doi:10.4155/tde.14.10. ISSN 2041-5990. {{cite journal}}: Check date values in: |date= (help)
  7. ^ a b c Bouakaz, Ayache; Zeghimi, Aya; Doinikov, Alexander A. (2016), Escoffre, Jean-Michel; Bouakaz, Ayache (eds.), "Sonoporation: Concept and Mechanisms", Therapeutic Ultrasound, Advances in Experimental Medicine and Biology, Cham: Springer International Publishing, pp. 175–189, doi:10.1007/978-3-319-22536-4_10, ISBN 978-3-319-22536-4, retrieved 2021-10-27
  8. ^ Meijering, Bernadet D.M.; Juffermans, Lynda J.M.; van Wamel, Annemieke; Henning, Rob H.; Zuhorn, Inge S.; Emmer, Marcia; Versteilen, Amanda M.G.; Paulus, Walter J.; van Gilst, Wiek H.; Kooiman, Klazina; de Jong, Nico (2009-03-13). "Ultrasound and Microbubble-Targeted Delivery of Macromolecules Is Regulated by Induction of Endocytosis and Pore Formation". Circulation Research. 104 (5): 679–687. doi:10.1161/CIRCRESAHA.108.183806.
  9. ^ Hauser, Joerg; Ellisman, Mark; Steinau, Hans-Ulrich; Stefan, Esenwein; Dudda, Marcel; Hauser, Manfred (2009-12). "Ultrasound Enhanced Endocytotic Activity of Human Fibroblasts". Ultrasound in Medicine & Biology. 35 (12): 2084–2092. doi:10.1016/j.ultrasmedbio.2009.06.1090. ISSN 0301-5629. {{cite journal}}: Check date values in: |date= (help)
  10. ^ Tran, T.A.; Roger, S.; Le Guennec, J.Y.; Tranquart, F.; Bouakaz, A. (2007-01). "Effect of ultrasound-activated microbubbles on the cell electrophysiological properties". Ultrasound in Medicine & Biology. 33 (1): 158–163. doi:10.1016/j.ultrasmedbio.2006.07.029. ISSN 0301-5629. {{cite journal}}: Check date values in: |date= (help)
  11. ^ Wu, Junru (2018-12-18). "Acoustic Streaming and Its Applications". Fluids. 3 (4): 108. doi:10.3390/fluids3040108. ISSN 2311-5521.{{cite journal}}: CS1 maint: unflagged free DOI (link)