Biomimetics - Biologically Inspired Technologies - Yoseph Bar Cohen Episode 2 Part 7 pps

30 318 0
Biomimetics - Biologically Inspired Technologies - Yoseph Bar Cohen Episode 2 Part 7 pps

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

the recent years, and implants with textured surfaces have been developed in an effort to allow bone to grow into the implant, this theoretically has the advantage of allowing much stronger biological cementing. One of the long-term problems after hip replacement is loosening of the components, which can result in bone loss and pain. This restricts the use of total hip replacement among younger patients. This happens due to very small plastic particles produced by the wearing of the cup. Recently metal on metal joints have regained popularity and are particularly suited for the hip joint replacement in middle age patients since it gives a much longer lasting results compared to the other hip replacements (Dorr et al., 2000). Parallel developments allowed the development of total knee replacement. Initial attempts were to replace the joint cavity with hinges which can cover the joint space to reduce friction. But problems with loosening and infection frequently occurred. Frank Gunston developed a metal on plastic knee replacement joint in 1968 (Gunston, 1971). A three component knee-joint prosthesis was proposed by John Insall in 1972 which covered the femur, tibia, and the patella, and were held in place using cement (Ranawat et al., 1975). This has resulted in the development of the modern knee-joint prosthesis. Currently more than 150,000 knee-joint replacements are undertaken in United States alone (Noble et al., 2005). Similar to the hip prosthesis, attempts have been underway in recent years to achieve a cementless joint replacement, using biological ability to glue these components together by allowing new bone growth in the roughened surfaces of these devices, which then can give strength and eliminate the need for artificial gluing materials that could come loose. 18.10 BIO-ARTIFICIAL PANCREAS Long standing diabetes mellitus (types I and II) results due to the inability of the pancreas to secrete insulin. Therapy has been focused at administering the insulin exogenously to achieve acceptable blood sugar levels, however, it is often difficult to manage. Transplantation of the isolated islet cells (which secrete insulin) although promising is limited due to the associated need for immunosup- pression and limited organ supply. Devices such as microencapsulated islets (small diameter spherical chamber), and microencap- sulated islets (including hollow fiber, disk-shaped diffusion chambers and Millipore cellulose membranes) have been proposed (Lanza et al., 1992; Lim and Sun, 1980; Reach et al., 1981; Sullivan et al., 1991). Advancements in glucose sensing and insulin sensing technology have allowed developing automated closed loop insulin delivery systems that can deliver insulin in a more physiologic way. One such system currently undergoing clinical trials is a diffusion chamber for a bio-artificial endocrine pancreas (Bio-AEP), which is constructed by placing pancreatic islet cells, trapped in a scaffold; this is sandwiched between semipermeable membranes, and shielded by silicone (Hirotani et al., 1999). Although some of the results achieved in animal studies have been difficult to reproduce in large animal models, this therapy holds promise for the future treatment of diabetes mellitus. 18.11 VISUAL PROSTHESIS (ARTIFICIAL EYE) The understanding of the mammalian visual system has given impetus for conceptualizing an artificial visual prosthesis that can be used in the profoundly blind. The goal of these systems is to produce a visual perception to allow activities like reading, recognizing shapes and faces, negoti- ating complex spaces, and giving the perception of light surroundings. This is dealt with in greater detail in Chapters 11 and 17. Bar-Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 462 21.9.2005 3:40am 462 Biomimetics: Biologically Inspired Technologies 18.12 ARTIFICIAL SKIN SUBSTITUTES Successful application of skin substitutes has been applied widely in the clinical field for a few decades now. The development of skin substitutes or artificial skin began with growing sheets of cells in culture media and has progressed to developing complex structures with bi-layered skin that mimics the human skin. A deeper dermal element is constructed using synthetic epidermis. Currently there are three approaches used for manufacturing artificial skin, the gel approach where cells are grown in a gel of extracellular material like collagen; the scaffold approach where porous scaffolds created from collagen or synthetic material are used to allow cells to be seeded subse- quently (Jones et al., 2002); the third approach entails, self-assembly, it is still in animal testing stage and has to await clinical application. Some of the artificial skin substitutes available are, Alloderm 1 introduced in market in 1992 and is based on treating fresh cadaver skin in which the epidermal layer is removed and cellular components are destroyed (Bello et al., 2001). The freeze drying of this skin substitute renders it immunologically inert and hence is not rejected by the recipient (Losee et al., 2005; Terino, 2001). Integray approved in 1996 by FDA is another skin substitute available commercially and is made from cellular collagen and glycosaminoglycans matrix (Winfrey et al., 1999). The dermal compon- ent is made of collagen and the epidermal element is substituted by synthetic silicon. Dermagraft 1 is an allogenic dermal substitute, it comprises of a scaffold of polyglactin seeded with allogenic fibroblasts (Eaglstein, 1998). This is now used to treat skin ulcers and burn wounds. Another allogenic frozen dermal substitute is TransCyte 1 , which is used as a temporary replacement for wounds and burns (Noordenbos et al., 1999). It is created by seeding fibroblasts into a scaffold made from nylon mesh and silicone sheet. Bilayered substitutes are composed of allogenic keratinocytes seeded on a nonporous collagen gel and covered with a bovine collagen scaffold containing fibroblasts (OrCel 1 ). They offer the more biologically mimicking skin substitute (Still et al., 2003). 18.13 ARTIFICIAL BLOOD Inadequate oxygen delivery to the tissues is common sequelae when significant blood loss occurs due to trauma or surgery. This is commonly treated in clinical practice by administering donated human blood. However, the availability of donors and the risk of transmission of infections limit this approach. Fatal reactions can occur due to a mismatch or presence of antibodies in the blood of the recipient; in addition, repeated blood transfusions can depress the immune function in the host. This is one of the reasons why an artificial blood substitute is highly desirable since it can avoid these complications. Two main approaches are used for achieving an artificial blood substitute, bio- artificial oxygen carriers and totally synthetic oxygen carriers. Bio-artificial oxygen carriers are hemoglobin-based oxygen carriers and use human, animal, or recombinant hemoglobin. Synthetic oxygen carriers use metal chelates that mimic the hemoglobin’s oxygen binding capacity. Artificial fluorinated organic compounds can physically dissolve large amounts of oxygen, perflurocarbon- based oxygen carriers are commonly employed for this purpose. However, in a strict sense they constitute oxygen carrier substitutes and not blood substitutes since they lack the coagulation factors and immune cells fighting infection that are essential in aiding coagulation and clot formation and fighting infection, which can be vital in the patients receiving these therapies. Examples of bio-artificial oxygen carriers include modified human or animal hemoglobin-based carriers, stabilized hemoglobin tetramers, polymerized hemoglobin, conjugated hemoglobin, and liposome encapsulated hemoglobin. Other carriers also include recombinant hemoglobin or from transgenic studies. Synthetic oxygen carriers include lipid–heme vesicles, hemoglobin aquasoms, and perflurocarbonbased carriers. More detailed reviewispresented elsewhere (Kim and Greenburg, 2004). Bar-Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 463 21.9.2005 3:40am Artificial Support and Replacement of Human Organs 463 18.14 OTHER SUBSTITUTES The last few decades have seen an explosive growth in the development of various implants such as pacemakers, stents for the arteries, cochlear implants (Rubinstein, 2004) to improve hearing, apheresis, small joints for the fingers and other joints, etc., the list is quite long and a brief review like this is unable to cover these areas in detail. Another field that is currently undergoing intense research is the field of xenotransplantation. Theoretically, this should allow transplantation of organs from animals to humans; however, there are several issues which need to be addressed include the risk of transmission of animal diseases to humans, the altered immune response that may accompany the species specific difference (Hammer, 2004; Schmidt et al., 2004). 18.15 LIMITATIONS OF THE CURRENT ORGAN REPLACEMENT SYSTEMS In spite of the significant advances made in the development of the artificial organs, some common problems plague all the systems. Biocompatibility (Hernandez et al., 2004; Jalan et al., 2004) is still a major problem, necessitating heparinization to avoid thrombosis. The use of heparin, to combat thrombosis, puts the patient at risk of bleeding-associated complications (Boyle et al., 2004; Minami et al., 2000; Rose et al., 2001). The organ systems do not truly replace the organs except in the case of total artificial heart. Most of the systems work on the principle of passive transport as in artificial kidney, artificial liver, and lung and hence fail to mimic the physiological functions of these individual organs. Most of these systems expose the body to increased infection risk due to the various lines and ports used for access (Rose et al., 2001; Tobin and Bambauer, 2003). This risk of infection can be serious in an already sick group of patients (El-Banayosy et al., 2001; Minami et al., 2000). Other limitations include nonphysiological support; for example, the organ support in case of kidneys need not be continuous as is the case of normal kidney which carries out the work 24 h a day, and this can disturb the delicate physiological balance necessary for the optimum biological function- ing. Mobility is restricted in all types of the support devices. The issue of energy supply is very important in the case of artificial ventricular assist devices and the artificial heart; these devices need to work continuously and lack of back-up systems can be catastrophic (Portner, 2001). Mechanical failure is an important issue if long term support is envisaged. 18.15.1 Impact of Other Technologies Technological advances are rapidly taking place around us and it is natural that these will significantly affect future organ support systems. The current organ replacement systems were designed in the 1960s and 1970s; it is a natural evolutionary step that new technology will replace the older systems. In concluding this section, we will take a glimpse at current developmental research in related fields and how it will impact the future of organ replacement systems. 18.15.1.1 Tissue Engineering Tissue engineering is a science that uses living cells combined with biomaterials for diagnostic and therapeutic purposes. This involves generation of cells, tissues, and complex organoid structures in the laboratory to replace natural organ function partially or completely (Fuchs et al., 2001). Application of tissue engineering has resulted in the development of bio-artificial kidney (Aebischer et al., 1987), liver (Chamuleau, 2002; Kulig et al., 2004), tissue-engineered heart valves (Hoerstrup et al., 2000a,b; Stock et al., 2002) and generation of myocardial cells to treat Bar-Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 464 21.9.2005 3:40am 464 Biomimetics: Biologically Inspired Technologies heart failure (Thompson et al., 2003). From the initial euphoria in 1990 to disappointment in 2004, tissue engineering has been put to test; a number of products have not shown benefit in clinical trials, that in turn is reflected in the lack of market interest in these products (Lysaght and Reyes, 2001; Lysaght and Hazlehurst, 2004). Tissue engineering has the necessary potential of seeding appropriate scaffolding with cells of interest as in the case of tubule cells used in artificial kidney (Fey-Lamprecht et al., 2003; Humes, 2000; Ozgen et al., 2004). As our understanding increases in terms of cell growth characteristics in relation to biomaterials, we are likely to move towards bio-artificial organ replacement systems. Normal organs, however, are composed of many different cell types with complex messaging and interactions. Using a single cell type may not necessarily guarantee adequate functioning of such systems. The importance of developing appropriate scaffolds for the blood vessels to grow can be key to future development of solid organs (Kaihara et al., 2000; MacNeill et al., 2002). The current systems use altered cancerous cells or cells from animal origin, which raises the likelihood of risk of cancerous transformation and transmission of animal originated diseases (van de Kerkhove et al., 2004). However, using adult stem cells from the patients’ own bone marrow may be the solution which will be more widely applied in the future. 18.15.1.2 Stem Cell Technology Stem cells are the precursor cells from which any type of cell differentiation is possible (Jain, 2002). There are two types of stem cell sources that can be used, one from the embryonic stage and another from the adult stem cells within the bone marrow. Stem cells from the embryonic stage offer the characteristic of differentiating into any possible cell type (Kakinuma et al., 2003; Sukhikh and Shtil, 2002); but recent findings, however, of increasing plasticity shown by the human hematopoietic stem cells to differentiate into different cell types has led to interest in developing them as a cell therapy for organ failure (Liu et al., 2004a–c; Schuster et al., 2004; Strom et al., 2004; Yokoo et al., 2003). 18.15.1.3 Impact of Understanding the Human Genome The human genome sequence now has been decoded (Venter et al., 2001). This offers the potential of synthetic DNA which can create proteins of interest. Theoretically, this can be used to develop synthetic organ systems and conceivably a complete organism. However, there are several limita- tions to this concept since we still do not have the insight into the function and role of all the human genes. Early indications suggest a possibility of tailor-made treatment based on the individual patient’s genomic characteristics; how this will apply to the treatment and replacement of organ systems remains to be fully explored. 18.15.1.4 Microelectromechanical Systems Microdevices have been applied for certain diagnostic, therapeutic, and selected surgical proced- ures (Evans et al., 2003; Polla et al., 2000; Richards Grayson et al., 2004). Microelectromechanical systems (MEMS) employ the same manufacturing methods as silicone chips for computer industry. They can be a useful tool for rapid screening of diseases, measurement of blood levels of hormones and drugs, targeted drug delivery, and novel micro-stimulators in neurosciences (Evans et al., 2003; Huang et al., 2002; Liu et al., 2004a–c; Polla et al., 2000; Roy et al., 2001). What makes MEMS more promising is the building of small rotors capable of running on miniscule energy (Epstein and Senturia, 1997; Miki et al., 2003). These have enormous potential to provide the energy source for organ replacement systems. In addition, they can provide the capability to detect the minute changes in hormones and endorphins on which the response of the organ support system can be tailored. Bar-Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 465 21.9.2005 3:40am Artificial Support and Replacement of Human Organs 465 18.15.2 Nanotechnology and Biomimetics Living organisms are the perfect example of the advanced nanotechnological manufacturing by nature. What could be more interesting than trying to build artificial organs from the beginning and mimicking nature? Advances in the development of artificial organs to date have relied mainly on supplanting the function of an organ with an alternative process. As in the case of heart, it is the pumping mechanism, in lungs the oxygenation of the blood, and in liver and kidney, the removal of harmful wastes. But biological organs play even more complex and dynamic role in the physiological mileu in terms of metabolic and other organ function. The biofeedback in these natural organs takes place at nano-dimensions, which the present replacement systems are unable to mimic precisely enough to bring about changes in the functionality of the devices. Nanotechnology can provide molecularly manipulated nanostructured materials which will mimic the natural surfaces. Sensing and control can be achieved in these systems using microelec- tronics and novel interface technologies (Lee et al., 2004). Drug delivery systems at nanoscale can maintain the function of normal cells (Prokop, 2001). Molecular self-assembly can simulate the surface geometry by polymeric patterning; since this has immense importance in the behavior of the individual cell and cell to cell communication, adhesion and migration (Chaikof et al., 2002; Hilt, 2004). Current cell and tissue culture systems fail to mimic the natural processes that provide extracellular matrix. Extracellular matrix plays an important role in the repair processes and thus influences cell behavior and survival. Scaffolds at a micro-level can be created using nanotechnol- ogy, and can incorporate the extracellular matrix containing glycosaminoglycans and glycoproteins supporting cell growth and proliferation (Bouhadir et al., 2001; Chaikof et al., 2002). The advances in nanotechnology allow us to synthesize novel materials, fabricate them in two or three-dimensional forms as scaffolds and allow the growth of new cells and ultimately whole organs (Chaikof et al., 2002; Karlsson et al., 2004; Moldovan and Ferrari, 2002). The National Institute of Health (NIH) has taken a big initiative in funding nanomedicine- related research and development. The NIH roadmap aims to have applications in drug delivery, cell repair, anticancer methodologies, and biomachines that could remove and replace a damaged cell or tissue. The biggest advantage of nanotechnology will be in understanding the organ function at minute levels and creating bio-engineered cells and tissues capable of replacing human organs. Structural and functional creation of artificial organs using nanotechnology will need precise understanding of the structure and function of the organ; the current knowledge of anatomical structures can greatly help in this regard. This can allow bioengineers to create exact scaffolds for the blood vessels and cells to grow. The issue of energy source can only be solved, however, if micro-machines are built which can derive energy from oxygen, glucose and other substances that are easily available in the body. 18.16 SUMMARY The current emphasis on replacement by mechanical systems is already profoundly affected by newer technologies. In the future, bio-compatible surfaces will be designed keeping in mind the precise interactions at atomic and molecular levels rather than the trial-and-error approach that was adopted several decades ago. These newer technologies will definitely have an impact on future artificial medical implants, be it artificial heart valves, vascular conduits, or artificial organ systems. Design and technology will certainly move to center stage in the coming years. Unique problems will be posed for the today’s scientists, physicians, and engineers, who are slow to adjust to collaborative research. Current funding structure is limited in supporting such collaborations; the cost of such design and manufacturing will be prohibitive for one group or individual organizations Bar-Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 466 21.9.2005 3:40am 466 Biomimetics: Biologically Inspired Technologies to sustain. Answers to these problems will hopefully be addressed in the future federal funding mechanism as outlined in the initiative by NIH on nanotechnology. One of the questions that is frequently debated is whether future organ replacement technology will involve miniaturizing the current systems or building newer organ replacement systems from scratch. As outlined above, current organ replacement systems have several disadvantages which will be difficult to overcome even if they are miniaturized. Miniaturization will certainly play an important role in devising therapeutic interventions such as drug delivery. Devising organ replacement systems from scratch will help address the current problems of biocompati- bility and better mimic the organ function at cellular level. This will involve creating novel anatomical models of scaffoldings which are biocompatible and bioactive to allow cell growth and differentiation so that complex organs can be developed. Such new organ systems will need to produce energy from oxygen, glucose, and other substances freely available in the blood and be self-sufficient. How far we are from the reality of buying off-the-shelf artificial organs? May be in next 10 years? As the pace of developments in the fields of nanotechnology, tissue engineering, and others is accelerating, the reality of having a self-sustaining artificial organ replacement system is a possible reality in the upcoming years. REFERENCES Aebischer, P, Ip, TK, Panol, G, et al. The bioartificial kidney: progress towards an ultrafiltration device with renal epithelial cells processing, Life Support Syst, 5, 2, 1987, 159–68. AHA. Heart Disease and Stroke Statistics: 2004 Update, American Heart Association, Dallas, TX, 2003. Akustu, T and Kolff, WJ. Permanent substitute for valves and hearts, Trans Am Soc Artif Intern Organs,4, 1958. Anderson, R and Smith, B. Deaths: leading causes for 2001, National Vital Statistics Report, 52, 9, 2003, National Center for Health Statistics, Hyattsville, Maryland. Bartlett, RH, Gazzaniga, AB, Jefferies, MR, et al. Extracorporeal membrane oxygenation (ECMO) cardiopul- monary support in infancy, Trans Am Soc Artif Intern Organs, 22, 1976, 80–93. Baumgartner, W, Reitz, B, Kasper, E, et al. Heart and Lung Transplantation, second edition, WB Saunders, Philadelphia, PA, 2002. Bello, YM, Falabella, AF and Eaglstein, WH. Tissue-engineered skin. Current status in wound healing, Am J Clin Dermatol, 2, 5, 2001, 305–13. Bing, RJ. Lindbergh and the biological sciences (a personal reminiscence), Tex Heart Inst J, 14, 3, 1987, 230–7. Boettcher, W, Merkle, F and Weitkemper, HH. History of extracorporeal circulation: the invention and modification of blood pumps, J Extra Corpor Technol, 35, 3, 2003, 184–91. Bouhadir, KH and Mooney, DJ. Promoting angiogenesis in engineered tissues, J Drug Target, 9, 6, 2001, 397– 406. Boyle, M, Kurtovic, J, Bihari, D, et al. Equipment review: the molecular adsorbents recirculating system (MARS(R)), Crit Care, 8, 4, 2004, 280–6. Burke, DJ, Burke, E, Parsaie, F, et al. The Heartmate II: design and development of a fully sealed axial flow left ventricular assist system, Artif Organs, 25, 5, 2001, 380–5. Chaikof, EL, Matthew, H, Kohn, J, et al. Biomaterials and scaffolds in reparative medicine, Ann N Y Acad Sci, 961, 2002, 96–105. Chamuleau, RA. Bioartificial liver support anno 2001, Metab Brain Dis, 17, 4, 2002, 485–91. Chen, C, Paden, B, Antaki, J, et al. A magnetic suspension theory and its application to the HeartQuest ventricular assist device, Artif Organs, 26, 11, 2002, 947–51. Clowes, GJ, Hopkins, A and Kolobow, T. Oxygen diffusion through plastic films, TASAIO, 1, 1955, 23–4. Cook, LN. Update on extracorporeal membrane oxygenation, Paediatr Respir Rev, 5 Suppl A, 2004, S329–37. Cooley, DA, Liotta, D, Hallman, GL, et al. Orthotopic cardiac prosthesis for two-staged cardiac replacement, Am J Cardiol, 24, 5, 1969, 723–30. Bar-Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 467 21.9.2005 3:40am Artificial Support and Replacement of Human Organs 467 Copeland, JG. Mechanical assist device; my choice: the CardioWest total artificial heart, Transplant Proc, 32, 7, 2000, 1523–4. Copeland, JG, III, Smith, RG, Arabia, FA, et al. Comparison of the CardioWest total artificial heart, the Novacor left ventricular assist system and the Thoratec ventricular assist system in bridge to trans- plantation, Ann Thorac Surg, 71, 90030, 2001, 92S–97. Curtis, JJ, Boley, TM, Walls, JT, et al. Frequency of seal disruption with the sarns centrifugal pump in postcardiotomy circulatory assist, Artif Organs, 18, 3, 1994, 235–7. Curtis, JJ, Wagner-Mann, CC, Mann, F, et al. Subchronic use of the St. Jude centrifugal pump as a mechanical assist device in calves, Artif Organs, 20, 6, 1996, 662–5. Curtis, JJ, Walls, JT, Wagner-Mann, CC, et al. Centrifugal pumps: description of devices and surgical techniques, Ann Thoracic Surg, 68, 2, 1999, 666–71. DeBakey, ME. Left ventricular bypass pump for cardiac assistance 1: clinical experience, Am J Cardiol, 27, 1, 1971, 3–11. Dekkers, RJ, FitzGerald, DJ and Couper, GS. Five-year clinical experience with Abiomed BVS 5000 as a ventricular assist device for cardiac failure, Perfusion, 16, 1, 2001, 13–8. Demetriou, AA, Brown, RS, Jr., Busuttil, RW, et al. Prospective, randomized, multicenter, controlled trial of a bioartificial liver in treating acute liver failure, Ann Surg, 239, 5, 2004, 660–7; discussion 667–70. Deng, L, El-Banayosy, A, et al. Mechanical circulatory support for advanced heart failure: effect of patient selection on outcome, Circulation, 103, 2, 2001, 231–7. DeVries, W, Anderson, J, Joyce, L, et al. Clinical use of the total artificial heart, N Engl J Med, 310, 5, 1984, 273–8. Di Bella, I, Pagani, F, Banfi, C, et al. Results with the Novacor assist system and evaluation of long-term assistance, Eur J Cardiothorac Surg, 18, 1, 2000, 112–6. Doi, K, Golding, LA, Massiello, AL, et al. Preclinical readiness testing of the arrow international CorAide left ventricular assist system, Ann Thorac Surg, 77, 6, 2004, 2103–10. Dorr, LD, Wan, Z, Longjohn, DB, et al. Total hip arthroplasty with use of the Metasul metal-on-metal articulation. Four to seven-year results, J Bone Joint Surg Am, 82, 6, 2000, 789–98. Dowling, RD, Etoch, SW, Stevens, KA, et al. Current status of the AbioCor implantable replacement heart, Ann Thorac Surg, 71, 90030, 2001, 147S–149. Dowling, RD, Gray, LA, Jr., Etoch, SW, et al. The AbioCor implantable replacement heart, Ann Thorac Surg, 75, 6 Suppl, 2003, S93–9. Eaglstein, WH. Dermagraft treatment of diabetic ulcers, J Dermatol, 25, 12, 1998, 803–4. El-Banayosy, A, Korfer, R, Arusoglu, L, et al. Device and patient management in a bridge-to-transplant setting, Ann Thorac Surg, 71, 90030, 2001, 98S–102. El-Banayosy, A, Arusoglu, L, Kizner, L, et al. Preliminary experience with the LionHeart left ventricular assist device in patients with end-stage heart failure, Ann Thorac Surg, 75, 5, 2003, 1469–75. Epstein, AH and Senturia, SD. Macro power from micro machinery, Science, 276, 5316, 1997, 1211. Evans, M, Sewter, C and Hill, E. An encoded particle array tool for multiplex bioassays, Assay Drug Dev Technol, 1, 1 Pt 2, 2003, 199–207. Farrar, DJ. The thoratec ventricular assist device: a paracorporeal pump for treating acute and chronic heart failure, Semin Thorac Cardiovasc Surg, 12, 3, 2000, 243–50. Farrar, DJ, Holman, WR, McBride, LR, et al. Long-term follow-up of Thoratec ventricular assist device bridge-to-recovery patients successfully removed from support after recovery of ventricular function, J Heart Lung Transplant, 21, 5, 2002, 516–21. FC Spencer, BE, Trinkle, JK, et al. Assisted circulation for cardiac failure following intracardiac surgery with cardiorespiratory bypass, J Thorac Cardiovasc Surg, 49, 56, 1959. Fey-Lamprecht, F, Albrecht, W, Groth, T, et al. Morphological studies on the culture of kidney epithelial cells in a fiber-in-fiber bioreactor design with hollow fiber membranes, J Biomed Mater Res, 65A, 2, 2003, 144–57. Frazier, O. First use of an untethered, vented electric left ventricular assist device for long-term support (published erratum appears in Circulation 1995 June 15, 91, 12, 3026), Circulation, 89, 6, 1994, 2908–14. Frazier, OH, Akustu, T and Cooley, DA. Total artificial heart (TAH) utilization in man, Trans Am Soc Artif Intern Organs, 23, 1982. Bar-Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 468 21.9.2005 3:40am 468 Biomimetics: Biologically Inspired Technologies Frazier, OH, Rose, EA, Oz, MC, et al. Multicenter clinical evaluation of the HeartMate vented electric left ventricular assist system in patients awaiting heart transplantation, J Thorac Cardiovasc Surg, 122, 6, 2001, 1186–95. Frazier, OH, Shah, NA, Myers, TJ, et al. Use of the Flowmaker (Jarvik 2000) left ventricular assist device for destination therapy and bridging to transplantation, Cardiology, 101, 1–3, 2004, 111–6. Fruhauf, NR, Oldhafer, KJ, Holtje, M, et al. A bioartificial liver support system using primary hepatocytes: a preclinical study in a new porcine hepatectomy model, Surgery, 136, 1, 2004, 47–56. Fuchs, JR, Nasseri, BA and Vacanti, JP. Tissue engineering: a 21st century solution to surgical reconstruction, Ann Thorac Surg, 72, 2, 2001, 577–91. Gibbon, J. Application of a mechanical heart and lung apparatus to cardiac surgery, Minn Med, 37, 1954, 171. Goldstein, DJ. Worldwide experience with the MicroMed DeBakey ventricular assist device(R) as a bridge to transplantation, Circulation, 108, 90101, 2003, 272II–277. Gott, VL, Alejo, DE and Cameron, DE. Mechanical heart valves: 50 years of evolution, Ann Thorac Surg, 76, 6, 2003, S2230–9. Gottschalk, CW and Fellner, SK. History of the science of dialysis, Am J Nephrol, 17, 3–4, 1997, 289–98. Griffith, BP, Kormos, RL, Borovetz, HS, et al. HeartMate II left ventricular assist system: from concept to first clinical use, Ann Thorac Surg, 71, 90030, 2001, 116S–120. Gunston, FH. Polycentric knee arthroplasty. Prosthetic simulation of normal knee movement, J Bone Joint Surg Br, 53, 2, 1971, 272–7. Hall, CW, Liotta, D, Henly, WS, et al. Development of artificial intrathoracic circulatory pumps*1, *2, Am J Surg, 108, 5, 1964, 685–92. Hammer, C. Xenotransplantation — will it bring the solution to organ shortage? Ann Transplant, 9, 1, 2004, 7–10. Hansell, DR. Extracorporeal membrane oxygenation for perinatal and pediatric patients, Respir Care, 48, 4, 2003, 352–62; discussion 363–6. Hernandez, MR, Galan, AM, Cases, A, et al. Biocompatibility of cellulosic and synthetic membranes assessed by leukocyte activation, Am J Nephrol, 24, 2, 2004, 235–41. Hilt, JZ. Nanotechnology and biomimetic methods in therapeutics: molecular scale control with some help from nature, Adv Drug Deliv Rev, 56, 11, 2004, 1533–6. Hirotani, S, Eda, R, Kawabata, T, et al. Bioartificial endocrine pancreas (Bio-AEP) for treatment of diabetes: effect of implantation of Bio-AEP on the pancreas, Cell Transplant, 8, 4, 1999, 399–404. Hoerstrup, SP, Sodian, R, Daebritz, S, et al. Functional living trileaflet heart valves grown in vitro, Circulation, 102, 19 Suppl 3, 2000a, III44–9. Hoerstrup, SP, Sodian, R, Sperling, JS, et al. New pulsatile bioreactor for in vitro formation of tissue engineered heart valves, Tissue Eng, 6, 1, 2000b, 75–9. Hoy, FBY, Mueller, DK, Geiss, DM, et al. Bridge to recovery for postcardiotomy failure: is there still a role for centrifugal pumps? Ann Thoracic Surg, 70, 4, 2000, 1259–63. Huang, Y, Mather, EL, Bell, JL, et al. MEMS-based sample preparation for molecular diagnostics, Anal Bioanal Chem, 372, 1, 2002, 49–65. Humes, HD. Bioartificial kidney for full renal replacement therapy, Semin Nephrol, 20, 1, 2000, 71–82. Humes, HD, MacKay, SM, Funke, AJ, et al. The bioartificial renal tubule assist device to enhance CRRT in acute renal failure, Am J Kidney Dis, 30, 5 Suppl 4, 1997, S28–31. Jain, KK. Stem cell technologies in regenerative medicine, Expert Opin Biol Ther, 2, 7, 2002, 771–3. Jalan, R, Sen, S and Williams, R. Prospects for extracorporeal liver support, Gut, 53, 6, 2004, 890–8. Jones, I, Currie, L and Martin, R. A guide to biological skin substitutes, Br J Plast Surg, 55, 3, 2002, 185–93. Kaihara, S, Borenstein, J, Koka, R, et al. Silicon micromachining to tissue engineer branched vascular channels for liver fabrication, Tissue Eng , 6, 2, 2000, 105–17. Kakinuma, S, Tanaka, Y, Chinzei, R, et al. Human umbilical cord blood as a source of transplantable hepatic progenitor cells, Stem Cells, 21, 2, 2003, 217–27. Kaplon, RJ, Oz, MC, Kwiatkowski, PA, et al. Miniature axial flow pump for ventricular assistance in children and small adults, J Thorac Cardiovasc Surg, 111, 1, 1996, 13–8. Karlsson, M, Davidson, M, Karlsson, R, et al. Biomimetic nanoscale reactors and networks, Annu Rev Phys Chem, 55, 2004, 613–49. Kim, HW and Greenburg, AG. Artificial oxygen carriers as red blood cell substitutes: a selected review and current status, Artif Organs, 28, 9, 2004, 813–28. Bar-Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 469 21.9.2005 3:40am Artificial Support and Replacement of Human Organs 469 Kobayashi, N, Okitsu, T, Nakaji, S, et al. Hybrid bioartificial liver: establishing a reversibly immortalized human hepatocyte line and developing a bioartificial liver for practical use, J Artif Organs, 6, 4, 2003, 236–44. Kolff, WJ. Lasker Clinical Medical Research Award. The artificial kidney and its effect on the development of other artificial organs, Nat Med, 8, 10, 2002, 1063–5. Kolobow, T and Bowman, R. Construction and evaluation of an alveolar membrane heart lung, Trans Am Soc Artif Intern Organs, 9, 1963, 238–45. Kulig, KM and Vacanti, JP. Hepatic tissue engineering, Transpl Immunol, 12, 3–4, 2004, 303–10. Kung, RT and Hart, RM. Design considerations for bearingless rotary pumps, Artif Organs, 21, 7, 1997, 645–50. Lanza, RP, Borland, KM, Lodge, P, et al. Treatment of severely diabetic pancreatectomized dogs using a diffusion-based hybrid pancreas, Diabetes, 41, 7, 1992, 886–9. Lawson, DS, Walczak, R, Lawson, AF, et al. North American neonatal extracorporeal membrane oxygenation (ECMO) devices: 2002 survey results, J Extra Corpor Technol, 36, 1, 2004, 16–21. Lee, SC, Bhalerao, K and Ferrari, M. Object-oriented design tools for supramolecular devices and biomedical nanotechnology, Ann N Y Acad Sci, 1013, 2004, 110–23. LeGallois, C. Experiences on the Principle of Life, Thomas, Philadelphia, 1813. Leprince, P, Bonnet, N, Rama, A, et al. Bridge to transplantation with the Jarvik–7 (CardioWest) total artificial heart: a single-center 15-year experience, J Heart Lung Transplant, 22, 12, 2003, 1296–303. Lick, SD, Zwischenberger, JB, Alpard, SK, et al. Development of an ambulatory artificial lung in an ovine survival model, Asaio J, 47, 5, 2001, 486–91. Lim, F and Sun, AM. Microencapsulated islets as bioartificial endocrine pancreas, Science, 210, 4472, 1980, 908–10. Liu, JP, Gluud, LL, Als-Nielsen, B, et al. Artificial and bioartificial support systems for liver failure, Cochrane Database Syst Rev, 1, 2004a, CD003628. Liu, J, Hu, Q, Wang, Z, et al. Autologous stem cell transplantation for myocardial repair, Am J Physiol Heart Circ Physiol, 287, 2, 2004b, H501–11. Liu, R, Wang, X and Zhou, Z. Application of MEMS microneedles array in biomedicine, Sheng Wu Yi Xue Gong Cheng Xue Za Zhi, 21, 3, 2004c, 482–5. Losee, JE, Fox, I, Hua, LB, et al. Transfusion-free pediatric burn surgery: techniques and strategies, Ann Plast Surg, 54, 2, 2005, 165–71. Lukes, J and Merckelbach, FM. [Experiences with arthroplasty of the hip joint according to the method of the Judet brothers]. Arthroplastics of the hip joint; consideration of the Judet and Smith-Petersen surgical methods, Acta Chir Orthop Traumatol Cech, 25, 2, 1958, 127–33. Lysaght, MJ and Hazlehurst, AL. Tissue engineering: the end of the beginning, Tissue Eng, 10, 1–2, 2004, 309–20. Lysaght, MJ and Reyes, J. The growth of tissue engineering, Tissue Eng, 7, 5, 2001, 485–93. MacNeill, BD, Pomerantseva, I, Lowe, HC, et al. Toward a new blood vessel, Vasc Med, 7, 3, 2002, 241–6. Magovern, GJ, Jr. The biopump and postoperative circulatory support, Ann Thoracic Surg, 55, 1, 1993, 245–9. Malchesky, PS. Artificial organs and vanishing boundaries, Artif Organs, 25, 2, 2001, 75–88. Malinin, TI. Remembering Alexis Carrel and Charles A. Lindbergh, Tex Heart Inst J, 23, 1, 1996, 28–35. Mallory, TH. John Charnley remembered: regaining our bearings, Orthopedics, 27, 9, 2004, 921–2. Mehta, SM, Pae, WE, Jr., Rosenberg, G, et al. The LionHeart LVD–2000: a completely implanted left ventricular assist device for chronic circulatory support, Ann Thorac Surg, 71, 90030, 2001, 156S–161. Mesana, TG. Rotary blood pumps for cardiac assistance: a ‘‘must?’’ Artif Organs, 28, 2, 2004, 218–25. Miki, N, Teo, CJ, Ho, LC, et al. Enhancement of rotordynamic performance of high-speed micro-rotors for power MEMS applications by precision deep reactive ion etching, Sensors and Actuators A: Physical, 104, 3, 2003, 263–7. Minami, K, El-Banayosy, A, Sezai, A, et al. Morbidity and outcome after mechanical ventricular support using Thoratec, Novacor, and HeartMate for bridging to heart transplantation, Artif Organs, 24, 6, 2000, 421–6. Moldovan, NI and Ferrari, M. Prospects for microtechnology and nanotechnology in bioengineering of replacement microvessels, Arch Pathol Lab Med, 126, 3, 2002, 320–4. Morgan, JA, John, R, Rao, V, et al. Bridging to transplant with the HeartMate left ventricular assist device: the Columbia Presbyterian 12-year experience, J Thoracic Cardiovasc Surg, 127, 5, 2004, 1309–16. Bar-Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 470 21.9.2005 3:40am 470 Biomimetics: Biologically Inspired Technologies Moussy, Y. Bioartificial kidney. I. Theoretical analysis of convective flow in hollow fiber modules: application to a bioartificial hemofilter, Biotechnol Bioeng, 68, 2, 2000, 142–52. Mullin, EJ, Metcalfe, MS and Maddern, GJ. Artificial liver support: potential to retard regeneration? Arch Surg, 139, 6, 2004, 670–7. Neff, G. Hip arthroplasty according to Smith-Peterson or Judet, Helv Chir Acta, 21, 5–6, 1954, 380–3. Nikolovski, J, Gulari, E and Humes, HD. Design engineering of a bioartificial renal tubule cell therapy device, Cell Transplant, 8, 4, 1999, 351–64. Noble, PC, Gordon, MJ, Weiss, JM, et al. Does total knee replacement restore normal knee function? Clin Orthop, 431, 2005, 157–65. Nojiri, C. Left ventricular assist system with a magnetically levitated impeller technology, Nippon Geka Gakkai Zasshi, 103, 9, 2002, 607–10. Noon, GP, Ball, JW, Jr. and Papaconstantinou, HT. Clinical experience with BioMedicus centrifugal ven- tricular support in 172 patients, Artif Organs, 19, 7, 1995, 756–60. Noon, GP, Morley, DL, Irwin, S, et al. Clinical experience with the MicroMed DeBakey ventricular assist device, Ann Thorac Surg, 71, 90030, 2001, 133S–138. Noordenbos, J, Dore, C and Hansbrough, JF. Safety and efficacy of TransCyte for the treatment of partial- thickness burns, J Burn Care Rehabil, 20, 4, 1999, 275–81. Nose, Y. Design and development strategy for the rotary blood pump, Artif Organs, 22, 6, 1998, 438–46. Nose, Y, Yoshikawa, M, Murabayashi, S, et al. Development of rotary blood pump technology: past, present, and future, Artif Organs, 24, 6, 2000, 412–20. Okada, Y, Ueno, S, Ohishi, T, et al. Magnetically levitated motor for rotary blood pumps, Artif Organs, 21, 7, 1997, 739–45. Ozgen, N, Terashima, M, Aung, T, et al. Evaluation of long-term transport ability of a bioartificial renal tubule device using LLC-PK1 cells, Nephrol Dial Transplant, 19, 9, 2004, 2198–207. Petrou, S and Edwards, L. Cost effectiveness analysis of neonatal extracorporeal membrane oxygenation based on four year results from the UK Collaborative ECMO Trial, Arch Dis Child Fetal Neonatal Ed, 89, 3, 2004, F263–8. Pierce, WS. Permanent heart substitution: better solutions lie ahead, Jama, 259, 6, 1988, 891. Polla, DL, Erdman, AG, Robbins, WP, et al. Microdevices in medicine, Annu Rev Biomed Eng, 2, 2000, 551–76. Portner, PM. Permanent Mechanical Circulatory Assistance, Heart and Lung Transplantation, second edition, 2001. Prokop, A. Bioartificial organs in the twenty-first century: nanobiological devices, Ann N Y Acad Sci, 944, 2001, 472–90. Ranawat, CS, Insall, J and Shine, J. Duo-condylar knee replacement, Curr Pract Orthop Surg, 6, 1975, 28–35. Reach, G, Poussier, P, Sausse, A, et al. Functional evaluation of a bioartificial pancreas using isolated islets perifused with blood ultrafiltrate, Diabetes, 30, 4, 1981, 296–301. Remadi, JP, Marticho, P, Butoi, I, et al. Clinical experience with the mini-extracorporeal circulation system: an evolution or a revolution? Ann Thorac Surg, 77, 6, 2004, 2172–5; discussion 2176. Richards Grayson, AC, Scheidt Shawgo, R, Li, Y, et al. Electronic MEMS for triggered delivery, Adv Drug Deliv Rev, 56, 2, 2004, 173–84. Robbins, RC, Kown, MH, Portner, PM, et al. The totally implantable Novacor left ventricular assist system, Ann Thorac Surg, 71, 90030, 2001, 162S–165. Rose, EA, Gelijns, AC, Moskowitz, AJ, et al. Long-term mechanical left ventricular assist device for end-stage heart failure, N Engl J Med, 345, 20, 2001, 1435–43. Roy, S, Ferrara, LA, Fleischman, AJ, et al. Microelectromechanical systems and neurosurgery: a new era in a new millennium, Neurosurgery, 49, 4, 2001, 779–97; discussion 797–8. Rubinstein, JT. How cochlear implants encode speech, Curr Opin Otolaryngol Head Neck Surg, 12, 5, 2004, 444–8. Schmidt, P, Andersson, G, Blomberg, J, et al. Possible transmission of zoonoses in xenotransplantation: porcine endogenous retroviruses (PERVs) from an immunological point of view, Acta Vet Scand Suppl, 99, 2004, 27–34. Schuster, MD, Kocher, AA, Seki, T, et al. Myocardial neovascularization by bone marrow angioblasts results in cardiomyocyte regeneration, Am J Physiol Heart Circ Physiol, 287, 2, 2004, H525–32. Bar-Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 471 21.9.2005 3:40am Artificial Support and Replacement of Human Organs 471 [...].. .Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 4 72 21.9 .20 05 3:40am 4 72 Biomimetics: Biologically Inspired Technologies Still, J, Glat, P, Silverstein, P, et al The use of a collagen sponge/living cell composite material to treat donor sites in burn patients, Burns, 29 , 8, 20 03, 8 37 41 Stock, UA, Vacanti, JP, Mayer, JE,... corn grits (B, grits are particles of broken seed endosperm) The final volume increase is about 150% in split pea seeds and 20 0% in corn seed particles Note that the smaller grit particles hydrate much faster than the larger peas Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c019 Final Proof page 478 21 .9 .20 05 8:02pm 478 Biomimetics: Biologically Inspired Technologies teacher’s demonstration... regrowing root and root-like haustoria However, translocation of individuals occurs frequently in some developmental stages of lower plants and in motile single-celled and multicellular algae Botanists call this type of individual 473 Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c019 Final Proof page 474 21 .9 .20 05 8:02pm 474 Biomimetics: Biologically Inspired Technologies movement... Ther, 3, 5, 20 03, 3 87 94 Zimmer, HG Perfusion of isolated organs and the first heart–lung machine, Can J Cardiol, 17, 9, 20 01, 963–9 Zwischenberger, JB, Anderson, CM, Cook, KE, et al Development of an implantable artificial lung: challenges and progress, Asaio J, 47, 4, 20 01, 31 6 -2 0 Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c019 Final Proof page 473 21 .9 .20 05 8:02pm 19 Nastic... 1953) As in human-made inflatable structures (e.g., sleeping bags) pressurization of the cells leads to the expansion as well as stiffening and hydrostatic stabilization of the cells, tissues, and entire structures (e.g., Niklas, Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c019 Final Proof page 476 21 .9 .20 05 8:02pm 476 Biomimetics: Biologically Inspired Technologies 19 92) To prevent... 473 19 .2 Motors in Nature’s Nastic Designs 474 19 .2. 1 Osmotic Motors 475 19 .2. 2 Colloid-Based Motors 476 19 .2. 2.1 Macroscopic Swelling Bodies 477 19 .2. 3 Fibrous Motors 479 19.3 Nastic Structures in Plants 481 19.3.1 Hydrostat Motor Cells — Source and Location of Movements 4 82 19.3 .2 From Isotropic Cell... (e.g., P-proteins, mucilage, and colloid vacuoles), dead cell walls (Figure 19.5), one or more living cells (guard cell of stomatal complex, leaf-rolling bulliform cells), organs (trap closure, shoot bending) to entire organisms (e.g., emergence of mescal cactus Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c019 Final Proof page 4 82 21.9 .20 05 8:02pm 4 82 Biomimetics: Biologically Inspired. .. cover into the light and open air (Figure 19. 17) Although smaller-sized volume oscillations occur also in fruits, leaves, and stems, they are most prominent in the strange, leafless Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c019 Final Proof page 490 21 .9 .20 05 8:02pm 490 Biomimetics: Biologically Inspired Technologies Figure 19. 17 Picture shows the almost completely buried shoot... as Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c019 Final Proof page 484 21 .9 .20 05 8:02pm 484 Biomimetics: Biologically Inspired Technologies Figure 19.9 (See color insert following page 3 02) A stable, erect sunflower stem (left panel) depends on the pressure of internal, easily expandable hydrostat tissue (pith ¼ transparent cells in center panel) that tensions the stronger-walled... (Walter, 19 57) This high degree of hydration is not osmotic but due to the presence of adsorptive forces (called adhesion or imbibition) that can equal and exceed the pressure of osmotic systems by reaching values of up to Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c019 Final Proof page 477 21 .9 .20 05 8:02pm Nastic Structures: The Enacting and Mimicking of Plant Movements 477 100 MPa . progress, Asaio J, 47, 4, 20 01, 31 6 -2 0. Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 4 72 21.9 .20 05 3:40am 4 72 Biomimetics: Biologically Inspired Technologies 19 Nastic. Presbyterian 1 2- year experience, J Thoracic Cardiovasc Surg, 1 27 , 5, 20 04, 1309–16. Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c018 Final Proof page 470 21 .9 .20 05 3:40am 470 Biomimetics: . relevant bone levers. Bar- Cohen : Biomimetics: Biologically Inspired Technologies DK3163_c019 Final Proof page 474 21 .9 .20 05 8:02pm 474 Biomimetics: Biologically Inspired Technologies To increase

Ngày đăng: 10/08/2014, 01:22

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan