Skip to main content
Erschienen in: Reproductive Biology and Endocrinology 1/2003

Open Access 01.12.2003 | Review

Tissue engineering and cell based therapies, from the bench to the clinic: The potential to replace, repair and regenerate

Erschienen in: Reproductive Biology and Endocrinology | Ausgabe 1/2003

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

The field of Regenerative Biology as it applies to Regenerative Medicine is an increasingly expanding area of research with hopes of providing therapeutic treatments for diseases and/or injuries that conventional medicines and even new biologic drug therapies cannot effectively treat. Extensive research in the area of Regenerative Medicine is focused on the development of cells, tissues and organs for the purpose of restoring function through transplantation. The general belief is that replacement, repair and restoration of function is best accomplished by cells, tissues or organs that can perform the appropriate physiologic/metabolic duties better than any mechanical device, recombinant protein therapeutic or chemical compound. Several strategies are currently being investigated and include, cell therapies derived from autologous primary cell isolates, cell therapies derived from established cell lines, cell therapies derived from a variety of stem cells, including bone marrow/mesenchymal stem cells, cord blood stem cells, embryonic stem cells, as well as cells tissues and organs from genetically modified animals. This mini-review is not meant to be exhaustive, but aims to highlight clinical applications for the four areas of research listed above and will address a few key advances and a few of the hurdles yet to be overcome as the technology and science improve the likelihood that Regenerative Medicine will become clinically routine.

Introduction

Many diseases and or physical defects due to injury result in the loss of specialized cells within organ systems and lead to organ system dysfunction. For example, Parkinson's disease results in the progressive loss of dopaminergic neurons within the substantia nigra region of the brain leading to motor deficits that result in dystonia and dyskinesia. Injuries, such as meniscal tears and spinal cord injury, can also result in the degeneration and loss of tissue leading to physical defects that can affect normal behavior. Additionally, insulin dependant diabetes mellitus (IDDM), multiple sclerosis (MS) and other autoimmune disorders lead to a loss of tissue that disrupts normal metabolism and bodily functions. The potential to treat these conditions with cell-based therapies holds promise for tissue/organ repair with the ultimate goal to regenerate and restore normal function. Several cell types will be discussed and are defined as; tissue specific differentiated cells, such as chondrocytes; progenitor cells isolated from specific tissues, such as bone marrow stem cells or neural stem cells; and embryonic stem cells that are derived from the inner cell mass of the developing blastomere.

Autologous Cell Therapy

Tissue specific differentiated autologous cells (as opposed to autologous progenitor cells, see below) harvested from an individual, cultured ex vivo to expand, and reintroduced into a second site for repairing damaged tissue with "self" is ideal from an immunologic perspective. Several pre-clinical models as well as clinical applications are currently being explored and include chondrocytes for cartilage repair [112], keratinocytes and/or dermal fibroblasts for burn and wound repair [1317], myocytes for myocardial repair [1823], retinal pigment epithelial cells for age related macular degeneration [2427] and Schwann cell transplantation to restore myelin in CNS lesions. [2832]. The two most developed autologous cell therapies that have advanced from the laboratory to the clinic involve the repair of cartilage using autologous chondrocytes and the treatment of burns with autologous cultured keratinocytes.
Grande et. al., first demonstrated that autologous chondrocyte cultures could be utilized to repair articular cartilage defects in the rabbit knee [11, 12]. Subsequently, this technique has been applied to the clinical treatment of articular cartilage defects [1, 4, 7, 33] and has now evolved into an FDA approved therapy supplied by Genzyme Biosurgery http://​www.​fda.​gov/​cber/​approvltr/​autogen082297L.​htm. Genzyme Biosurgery has also developed an autologous keratinocyte culture procedure and currently markets Epicel® as a treatment for burn victims http://​www.​genzymebiosurger​y.​com/​prod/​burn/​gzbx_​p_​pt_​burn.​asp. Although repairing "self" with "self" is attractive and doesn't require immunosuppressive drug therapy for graft maintenance, there are limitations related to the harvesting of tissue and expanded tissue culture. Typically, harvesting the original source tissue from the patient requires a surgical procedure which minimally is a biopsy, but could also require a large resection of the tissue. The concern is causing a second site defect that leads to pain, discomfort or a deficit that effects behavior, hopefully to a lesser extent than the original. The ability of the adult tissue to expand in tissue culture to generate sufficient numbers of cells is also a potential limitation. Additionally, primary cell cultures can become senescent or dedifferentiate during the culture period. Another limitation is that this type of therapy is only amenable to tissues that can sustain surgical harvesting and ex vivo culturing, emphasized by the fact that only two autologous cell therapies have achieved FDA approval for the medical market.
Autologous progenitor cells harvested from an individual and used for "self" tissue repair is also immunologically ideal. The most widely used source of adult progenitor cells are derived from bone marrow. The mesenchymal compartment within the bone marrow has the capacity to differentiate into many cell and tissue types given the appropriate growth conditions [34]. Early studies using bone marrow stromal cells for tissue repair focused on the repair of bone defects [35] however, more recent studies have applied bone marrow progenitor cells to repair a variety damaged tissue types, including cartilage [3638], myocardium [39, 40], liver [41], spinal cord injury [4244] and most recently diabetes [45]. However, these differentiation studies are still in the experimental stage. The potential to differentiate "self" progenitor cells into a variety of tissues is extremely promising for the field of Regenerative Medicine, however continued experimentation is necessary to understand the differentiation processes and to be able to reproducibly guide these cells into the appropriate tissue, prior to clinical application.
Autologous peripheral blood or autologous bone marrow stem cells are currently used clinically, but not from a commercial stand point. A current search of the FDA web site http://​clinicaltrials.​gov resulted in 108 studies involving autologous peripheral blood or autologous bone marrow stem cells as a treatment therapy, 96 of which were related to cancer treatments. None of the trials are related to stem cell differentiation followed by transplantation.

Allogeneic Tissues and Cell Lines

The use of allogeneic tissue for transplantation is clinically routine due to the development of immunosuppressive drug therapies, such as cyclosporine, FK506 and rapamycin. The use of engineered tissue and specialized cell lines for the treatment of disease and injury is more recent and will also require immunosuppression unless engineering strategies are utilized to make the tissue resistant to immune destruction or through tissue processing to reduce immunogenicity. As is the case for autologous cell therapies, the furthest advances are in the area of connective tissue replacement, cartilage and skin. [4648] Currently, Apligraft® (Organogenesis, Inc) is used as a dermal replacement for chronic wounds and is composed of neonatal foreskin kerotinocytes and dermal fibroblasts [49]. Although earlier studies demonstrated that Langerhan's cell-free epidermal skin cultures were rejected following transplantation [50], Apligraft tissue appears to be uniquely non-immunogenic due to the processing of the tissue [51, 52] and represents an exception to the need for immunosuppression during allogeneic transplantation. A similar product, Dermagraft® is also available from Smith & Nephew.
Another interesting allogeneic cell type harvested from cadaveric sources for the treatment of Parkinson's disease are allogeneic cultured retinal pigment epithelial cells that are encapsulated to provide an immune barrier [53]. Titan Pharmaceuticals, Inc has advanced this research into the clinic and is currently conducting a Phase IIb clinical trial with initial positive results [54].
Allogeneic cell lines are also being developed as a source of cells for regenerating damaged tissue due to disease. The human NT2 cell line was shown to differentiate and develop into neurons in rodent stroke models [55, 56] and is currently being tested in clinical trials for the treatment of stroke by Layton Biosciences, Inc [57, 58]. Patients receiving the NT2 cell grafts do receive immunosuppression to inhibit immune rejection of the graft [57, 58].
The development of allogeneic engineered tissues for commercial purposes has similar limitations as commercial autologous cell therapies with the added complication of immune rejection. Encapsulation, tissue processing, tolerance induction and/or genetic modification will be necessary unless the patient population is receptive to and the severity of the disease warrants the use of immunosuppressive drugs.

Allogeneic stem cells

Allogeneic bone marrow transplantation is used clinically to treat hematologic disorders and cancer, but as is the case for autologous bone marrow transplantation, not from a commercial, tissue engineering standpoint. New clinical trials are focusing on the use of peripheral blood stem cells and specific subsets of bone marrow stem cells for these indications (for example [59, 60]). Searching the FDA clinical trial data base identified 117 trials that utilize allogeneic stem cells or bone marrow in the treatment regime. Sorting through those trials, 99 were specific for stem cell therapy. As for autologous bone marrow or peripheral blood stem cell therapies, most were for cancer indications, 84/99. The data base included 9 trials utilizing allogeneic stem cells for anemia/hematologic disorders, 2 trials are designed to treat metabolic storage diseases and there are single trials for each of the following; Granulomatous Disease, HIV patients not responsive to highly active antiretroviral therapy, Mycosis Fungoides and Sezary Syndrome and allogeneic bone marrow rejection http://​clinicaltrials.​gov. Virtually all of these therapies are in combination with some form of immunosuppression and none of the trials are related to stem cell differentiation followed by transplantation.
The discovery and isolation of neural stem cells from fetal [6164] and adult human brain [65, 66] is a significant development in the area of neural cell differentiation that has led to the possibility of producing specialized cells for the treatment of neurologic disorders, such as Parkinson's disease and spinal cord injury. Many groups have studied the differentiation of neurospheres into neuron, glial, and astrocyte lineages, however the work from Goldman's group [66, 67] sets a president for selection strategies that are most relevant to the field of tissue engineering. Nunes, et. al., and Keyoung et. al., showed that transfected/transduced specific promoter constructs driving green fluorescent protein (GFP) can be used to select specific neural progenitor cells by flow cytometry [66, 67]. Expanding on these techniques could lead to the eventual development of a commercial application of neural stem cells by reproducibly selecting the desired neural phenotype. Demonstrating that specific lineages can be selected genetically with drug selectable or fluorescent expression plasmids lays the ground work for further selection schemes and further genetic modifications, such as modifying the immune response leading to a universally accepted source of human neural tissue for transplantation.
Although stem cells from adult tissues have more plasticity than originally thought, they typically are limited in their capacity to generate all possible tissue and cell types. Stem cells derived from the inner cell mass of the early embryonic blastocyst (ES cells) can proliferate indefinitely [68] and can give rise to virtually any cell type [69]. The development of human embryonic stem cells [70] has raised the possibility that an unlimited supply of human tissue could be generated from ES cells and that these tissues could be used to replace and repair damaged tissue in any organ system. It should be noted that although these human cells are referred to as ES cells, they cannot be qualified as a true "ES cell" which is defined by the ability to contribute to the germ line during embryonic development. ES cells from other species are tested in this manner, however it is ethically unfeasible with human ES cells. Having qualified the definition of a human ES cell, several studies have demonstrated that human ES cells retain the capacity to differentiate into a variety of tissues, including neuronal cells, myocytes, adipocytes and hematopoietic cells [7177]. The challenge now is to be able to direct differentiation or select for the desired phenotype and to develop these therapies in the absence of immunosuppression, similar to the strategies taken by the field of xenotransplantation. The ability to genetically modify ES cells is a great advantage and could be used to overcome both the directed differentiation/selection hurdle as well as the immune response hurdle.

Xenotransplantation

In the ongoing search for a reliable source of tissue to replace lost cells, tissues and organs, research in the area of xenotransplantation (cross species transplantation) has grown tremendously in the last 20 years. Overcoming the immunologic hurdle of cross species transplantation as well as the problem of cross-species pathogen infectivity, i.e., xenozoonosis, are the scientific challenges facing the field [78, 79]. The ability to genetically modify species such as the pig through transgenesis and nuclear transfer, to express human genes and to mutate detrimental genes expressed in pig cells [8086] still holds promise for engineered tissues and organs for human transplantation. The production of galα1,3gal transferase null transgenic pigs [84] represents a significant development towards eliminating both hyperacute and acute vascular rejection and may lead to extended survival of pig organs in old world primates, including humans, in combination with standard triple drug immunosuppressive therapy.
Interestingly, there have been a series of pig-to-human xenotransplantation clinical experiments for the treatment of diabetes [87, 88] and FDA approved clinical trials for the treatment of neurologic disorders using outbred pig tissue [8991]. Although there was some evidence of cell engraftment in both indications [87, 90], no efficacy was established due to the transplant [87, 89]. To date, a Phase I clinical trial was completed using transgenically engineered pig livers to detoxify the blood of fulminant hepatic failure (FHF) patients via extracoporial perfusion [92], however there is yet to be an FDA approved transgenic animal tissue for use in human transplantation.
Although the theoretical risk of xenozoonosis is a risk and represents a significant psychosocial issue, several studies investigating the possibility of cross species infectivity, including a retrospective analysis of 160 human transplant recipients exposed to porcine tissues have yet to reveal transmission of porcine viruses to humans or primates in vivo [9396]. The prospect of xenotransplantation is still relevant to solid organ and islet transplantation and with FDA oversight, animal as well as patient monitoring, the risks associated with xenozoonosis will be overcome.

The Future

The challenges associated with stem cell differentiation, tissue engineering, and xenotransplantation are many-fold in each of the respective fields, however one of the biggest challenges beyond the science and biology is the development of an FDA approved product from any of the developmental areas discussed above. The regulatory issues and points to consider documents are in the early stages of development by the FDA in collaboration with expert review panels and will continue to change as the technology advances into clinical applications. The experiences of Genzyme Biosurgery, Organogenesis, Diacrin, Layton Biosciences, Titan Pharmaceuticals and other tissue engineering companies will benefit the field as a whole.
Literatur
1.
Zurück zum Zitat Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, Peterson L: Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N Engl J Med. 1994, 331: 889-895. 10.1056/NEJM199410063311401.CrossRefPubMed Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, Peterson L: Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N Engl J Med. 1994, 331: 889-895. 10.1056/NEJM199410063311401.CrossRefPubMed
2.
Zurück zum Zitat Brittberg M, Nilsson A, Lindahl A, Ohlsson C, Peterson L: Rabbit articular cartilage defects treated with autologous cultured chondrocytes. Clin Orthop. 1996, 270-283. 10.1097/00003086-199605000-00034. Brittberg M, Nilsson A, Lindahl A, Ohlsson C, Peterson L: Rabbit articular cartilage defects treated with autologous cultured chondrocytes. Clin Orthop. 1996, 270-283. 10.1097/00003086-199605000-00034.
3.
Zurück zum Zitat Minas T: Chondrocyte implantation in the repair of chondral lesions of the knee: economics and quality of life. Am J Orthop. 1998, 27: 739-744.PubMed Minas T: Chondrocyte implantation in the repair of chondral lesions of the knee: economics and quality of life. Am J Orthop. 1998, 27: 739-744.PubMed
4.
Zurück zum Zitat Brittberg M: Autologous chondrocyte transplantation. Clin Orthop. 1999, S147-55. 10.1097/00003086-199910001-00016. Brittberg M: Autologous chondrocyte transplantation. Clin Orthop. 1999, S147-55. 10.1097/00003086-199910001-00016.
5.
Zurück zum Zitat Minas T, Peterson L: Advanced techniques in autologous chondrocyte transplantation. Clin Sports Med. 1999, 18: 13-44, v-vi.CrossRefPubMed Minas T, Peterson L: Advanced techniques in autologous chondrocyte transplantation. Clin Sports Med. 1999, 18: 13-44, v-vi.CrossRefPubMed
6.
Zurück zum Zitat Robinson D, Ash H, Aviezer D, Agar G, Halperin N, Nevo Z: Autologous chondrocyte transplantation for reconstruction of isolated joint defects: the Assaf Harofeh experience. Isr Med Assoc J. 2000, 2: 290-295.PubMed Robinson D, Ash H, Aviezer D, Agar G, Halperin N, Nevo Z: Autologous chondrocyte transplantation for reconstruction of isolated joint defects: the Assaf Harofeh experience. Isr Med Assoc J. 2000, 2: 290-295.PubMed
7.
Zurück zum Zitat Brittberg M, Tallheden T, Sjogren-Jansson B, Lindahl A, Peterson L: Autologous chondrocytes used for articular cartilage repair: an update. Clin Orthop. 2001, S337-48. Brittberg M, Tallheden T, Sjogren-Jansson B, Lindahl A, Peterson L: Autologous chondrocytes used for articular cartilage repair: an update. Clin Orthop. 2001, S337-48.
8.
Zurück zum Zitat King PJ, Bryant T, Minas T: Autologous chondrocyte implantation for chondral defects of the knee: indications and technique. J Knee Surg. 2002, 15: 177-184.PubMed King PJ, Bryant T, Minas T: Autologous chondrocyte implantation for chondral defects of the knee: indications and technique. J Knee Surg. 2002, 15: 177-184.PubMed
9.
Zurück zum Zitat Peterson L, Brittberg M, Kiviranta I, Akerlund EL, Lindahl A: Autologous chondrocyte transplantation. Biomechanics and long-term durability. Am J Sports Med. 2002, 30: 2-12.PubMed Peterson L, Brittberg M, Kiviranta I, Akerlund EL, Lindahl A: Autologous chondrocyte transplantation. Biomechanics and long-term durability. Am J Sports Med. 2002, 30: 2-12.PubMed
10.
Zurück zum Zitat Peterson L, Minas T, Brittberg M, Lindahl A: Treatment of osteochondritis dissecans of the knee with autologous chondrocyte transplantation: results at two to ten years. J Bone Joint Surg Am. 2003, 85-A Suppl 2: 17-24.PubMed Peterson L, Minas T, Brittberg M, Lindahl A: Treatment of osteochondritis dissecans of the knee with autologous chondrocyte transplantation: results at two to ten years. J Bone Joint Surg Am. 2003, 85-A Suppl 2: 17-24.PubMed
11.
Zurück zum Zitat Grande DA, Singh IJ, Pugh J: Healing of experimentally produced lesions in articular cartilage following chondrocyte transplantation. Anat Rec. 1987, 218: 142-148.CrossRefPubMed Grande DA, Singh IJ, Pugh J: Healing of experimentally produced lesions in articular cartilage following chondrocyte transplantation. Anat Rec. 1987, 218: 142-148.CrossRefPubMed
12.
Zurück zum Zitat Grande DA, Pitman MI, Peterson L, Menche D, Klein M: The repair of experimentally produced defects in rabbit articular cartilage by autologous chondrocyte transplantation. J Orthop Res. 1989, 7: 208-218.CrossRefPubMed Grande DA, Pitman MI, Peterson L, Menche D, Klein M: The repair of experimentally produced defects in rabbit articular cartilage by autologous chondrocyte transplantation. J Orthop Res. 1989, 7: 208-218.CrossRefPubMed
13.
Zurück zum Zitat Navsaria HA, Myers SR, Leigh IM, McKay IA: Culturing skin in vitro for wound therapy. Trends Biotechnol. 1995, 13: 91-100. 10.1016/S0167-7799(00)88913-1.CrossRefPubMed Navsaria HA, Myers SR, Leigh IM, McKay IA: Culturing skin in vitro for wound therapy. Trends Biotechnol. 1995, 13: 91-100. 10.1016/S0167-7799(00)88913-1.CrossRefPubMed
14.
Zurück zum Zitat Kuroyanagi Y, Kenmochi M, Ishihara S, Takeda A, Shiraishi A, Ootake N, Uchinuma E, Torikai K, Shioya N: A cultured skin substitute composed of fibroblasts and keratinocytes with a collagen matrix: preliminary results of clinical trials. Ann Plast Surg. 1993, 31: 340-9; discussion 349-51.CrossRefPubMed Kuroyanagi Y, Kenmochi M, Ishihara S, Takeda A, Shiraishi A, Ootake N, Uchinuma E, Torikai K, Shioya N: A cultured skin substitute composed of fibroblasts and keratinocytes with a collagen matrix: preliminary results of clinical trials. Ann Plast Surg. 1993, 31: 340-9; discussion 349-51.CrossRefPubMed
15.
Zurück zum Zitat Lam PK, Chan ES, Liew CT, Yen RS, Lau HC, King WW: Dermal fibroblasts do not enhance the graft take rate of autologous, cultured keratinocyte suspension on full-thickness wounds in rats. Ann Plast Surg. 2001, 46: 146-149. 10.1097/00000637-200102000-00010.CrossRefPubMed Lam PK, Chan ES, Liew CT, Yen RS, Lau HC, King WW: Dermal fibroblasts do not enhance the graft take rate of autologous, cultured keratinocyte suspension on full-thickness wounds in rats. Ann Plast Surg. 2001, 46: 146-149. 10.1097/00000637-200102000-00010.CrossRefPubMed
16.
Zurück zum Zitat Boyce ST, Greenhalgh DG, Kagan RJ, Housinger T, Sorrell JM, Childress CP, Rieman M, Warden GD: Skin anatomy and antigen expression after burn wound closure with composite grafts of cultured skin cells and biopolymers. Plast Reconstr Surg. 1993, 91: 632-641.CrossRefPubMed Boyce ST, Greenhalgh DG, Kagan RJ, Housinger T, Sorrell JM, Childress CP, Rieman M, Warden GD: Skin anatomy and antigen expression after burn wound closure with composite grafts of cultured skin cells and biopolymers. Plast Reconstr Surg. 1993, 91: 632-641.CrossRefPubMed
17.
Zurück zum Zitat Carsin H, Ainaud P, Le Bever H, Rives J, Lakhel A, Stephanazzi J, Lambert F, Perrot J: Cultured epithelial autografts in extensive burn coverage of severely traumatized patients: a five year single-center experience with 30 patients. Burns. 2000, 26: 379-387. 10.1016/S0305-4179(99)00143-6.CrossRefPubMed Carsin H, Ainaud P, Le Bever H, Rives J, Lakhel A, Stephanazzi J, Lambert F, Perrot J: Cultured epithelial autografts in extensive burn coverage of severely traumatized patients: a five year single-center experience with 30 patients. Burns. 2000, 26: 379-387. 10.1016/S0305-4179(99)00143-6.CrossRefPubMed
18.
Zurück zum Zitat Penn MS, Francis GS, Ellis SG, Young JB, McCarthy PM, Topol EJ: Autologous cell transplantation for the treatment of damaged myocardium. Prog Cardiovasc Dis. 2002, 45: 21-32. 10.1053/pcad.2002.123466.CrossRefPubMed Penn MS, Francis GS, Ellis SG, Young JB, McCarthy PM, Topol EJ: Autologous cell transplantation for the treatment of damaged myocardium. Prog Cardiovasc Dis. 2002, 45: 21-32. 10.1053/pcad.2002.123466.CrossRefPubMed
19.
Zurück zum Zitat Yoo KJ, Li RK, Weisel RD, Mickle DA, Li G, Yau TM: Autologous smooth muscle cell transplantation improved heart function in dilated cardiomyopathy. Ann Thorac Surg. 2000, 70: 859-865. 10.1016/S0003-4975(00)01630-1.CrossRefPubMed Yoo KJ, Li RK, Weisel RD, Mickle DA, Li G, Yau TM: Autologous smooth muscle cell transplantation improved heart function in dilated cardiomyopathy. Ann Thorac Surg. 2000, 70: 859-865. 10.1016/S0003-4975(00)01630-1.CrossRefPubMed
20.
Zurück zum Zitat Dorfman J, Duong M, Zibaitis A, Pelletier MP, Shum-Tim D, Li C, Chiu RC: Myocardial tissue engineering with autologous myoblast implantation. J Thorac Cardiovasc Surg. 1998, 116: 744-751.CrossRefPubMed Dorfman J, Duong M, Zibaitis A, Pelletier MP, Shum-Tim D, Li C, Chiu RC: Myocardial tissue engineering with autologous myoblast implantation. J Thorac Cardiovasc Surg. 1998, 116: 744-751.CrossRefPubMed
21.
Zurück zum Zitat Al Attar N, Carrion C, Ghostine S, Garcin I, Vilquin JT, Hagege AA, Menasche P: Long-term (1 year) functional and histological results of autologous skeletal muscle cells transplantation in rat. Cardiovasc Res. 2003, 58: 142-148. 10.1016/S0008-6363(02)00790-3.CrossRefPubMed Al Attar N, Carrion C, Ghostine S, Garcin I, Vilquin JT, Hagege AA, Menasche P: Long-term (1 year) functional and histological results of autologous skeletal muscle cells transplantation in rat. Cardiovasc Res. 2003, 58: 142-148. 10.1016/S0008-6363(02)00790-3.CrossRefPubMed
22.
Zurück zum Zitat Tran N, Li Y, Bertrand S, Bangratz S, Carteaux JP, Stoltz JF, Villemot JP: Autologous cell transplantation and cardiac tissue engineering: potential applications in heart failure. Biorheology. 2003, 40: 411-415.PubMed Tran N, Li Y, Bertrand S, Bangratz S, Carteaux JP, Stoltz JF, Villemot JP: Autologous cell transplantation and cardiac tissue engineering: potential applications in heart failure. Biorheology. 2003, 40: 411-415.PubMed
23.
Zurück zum Zitat Gulbins H, Meiser BM, Reichenspurner H, Reichart B: Cell transplantation--a potential therapy for cardiac repair in the future?. Heart Surg Forum. 2002, 5: E28-34.PubMed Gulbins H, Meiser BM, Reichenspurner H, Reichart B: Cell transplantation--a potential therapy for cardiac repair in the future?. Heart Surg Forum. 2002, 5: E28-34.PubMed
24.
Zurück zum Zitat Peyman GA, Blinder KJ, Paris CL, Alturki W, Nelson N. C., Jr., Desai U: A technique for retinal pigment epithelium transplantation for age-related macular degeneration secondary to extensive subfoveal scarring. Ophthalmic Surg. 1991, 22: 102-108.PubMed Peyman GA, Blinder KJ, Paris CL, Alturki W, Nelson N. C., Jr., Desai U: A technique for retinal pigment epithelium transplantation for age-related macular degeneration secondary to extensive subfoveal scarring. Ophthalmic Surg. 1991, 22: 102-108.PubMed
25.
Zurück zum Zitat Ishida M, Lui GM, Yamani A, Sugino IK, Zarbin MA: Culture of human retinal pigment epithelial cells from peripheral scleral flap biopsies. Curr Eye Res. 1998, 17: 392-402.CrossRefPubMed Ishida M, Lui GM, Yamani A, Sugino IK, Zarbin MA: Culture of human retinal pigment epithelial cells from peripheral scleral flap biopsies. Curr Eye Res. 1998, 17: 392-402.CrossRefPubMed
26.
Zurück zum Zitat Binder S, Stolba U, Krebs I, Kellner L, Jahn C, Feichtinger H, Povelka M, Frohner U, Kruger A, Hilgers RD, Krugluger W: Transplantation of autologous retinal pigment epithelium in eyes with foveal neovascularization resulting from age-related macular degeneration: a pilot study. Am J Ophthalmol. 2002, 133: 215-225. 10.1016/S0002-9394(01)01373-3.CrossRefPubMed Binder S, Stolba U, Krebs I, Kellner L, Jahn C, Feichtinger H, Povelka M, Frohner U, Kruger A, Hilgers RD, Krugluger W: Transplantation of autologous retinal pigment epithelium in eyes with foveal neovascularization resulting from age-related macular degeneration: a pilot study. Am J Ophthalmol. 2002, 133: 215-225. 10.1016/S0002-9394(01)01373-3.CrossRefPubMed
27.
Zurück zum Zitat Semkova I, Kreppel F, Welsandt G, Luther T, Kozlowski J, Janicki H, Kochanek S, Schraermeyer U: Autologous transplantation of genetically modified iris pigment epithelial cells: a promising concept for the treatment of age-related macular degeneration and other disorders of the eye. Proc Natl Acad Sci U S A. 2002, 99: 13090-13095. 10.1073/pnas.202486199.PubMedCentralCrossRefPubMed Semkova I, Kreppel F, Welsandt G, Luther T, Kozlowski J, Janicki H, Kochanek S, Schraermeyer U: Autologous transplantation of genetically modified iris pigment epithelial cells: a promising concept for the treatment of age-related macular degeneration and other disorders of the eye. Proc Natl Acad Sci U S A. 2002, 99: 13090-13095. 10.1073/pnas.202486199.PubMedCentralCrossRefPubMed
28.
Zurück zum Zitat Baron-Van Evercooren A, Avellana-Adalid V, Lachapelle F, Liblau R: Schwann cell transplantation and myelin repair of the CNS. Mult Scler. 1997, 3: 157-161.CrossRefPubMed Baron-Van Evercooren A, Avellana-Adalid V, Lachapelle F, Liblau R: Schwann cell transplantation and myelin repair of the CNS. Mult Scler. 1997, 3: 157-161.CrossRefPubMed
29.
Zurück zum Zitat Blakemore WF, Crang AJ: The use of cultured autologous Schwann cells to remyelinate areas of persistent demyelination in the central nervous system. J Neurol Sci. 1985, 70: 207-223. 10.1016/0022-510X(85)90088-7.CrossRefPubMed Blakemore WF, Crang AJ: The use of cultured autologous Schwann cells to remyelinate areas of persistent demyelination in the central nervous system. J Neurol Sci. 1985, 70: 207-223. 10.1016/0022-510X(85)90088-7.CrossRefPubMed
30.
Zurück zum Zitat Stangel M, Hartung HP: Remyelinating strategies for the treatment of multiple sclerosis. Prog Neurobiol. 2002, 68: 361-376. 10.1016/S0301-0082(02)00105-3.CrossRefPubMed Stangel M, Hartung HP: Remyelinating strategies for the treatment of multiple sclerosis. Prog Neurobiol. 2002, 68: 361-376. 10.1016/S0301-0082(02)00105-3.CrossRefPubMed
31.
Zurück zum Zitat Wrathall JR, Kapoor V, Kao CC: Observation of cultured peripheral non-neuronal cells implanted into the transected spinal cord. Acta Neuropathol (Berl). 1984, 64: 203-212.CrossRef Wrathall JR, Kapoor V, Kao CC: Observation of cultured peripheral non-neuronal cells implanted into the transected spinal cord. Acta Neuropathol (Berl). 1984, 64: 203-212.CrossRef
32.
Zurück zum Zitat Cheng B, Chen Z: Fabricating autologous tissue to engineer artificial nerve. Microsurgery. 2002, 22: 133-137. 10.1002/micr.21740.CrossRefPubMed Cheng B, Chen Z: Fabricating autologous tissue to engineer artificial nerve. Microsurgery. 2002, 22: 133-137. 10.1002/micr.21740.CrossRefPubMed
33.
Zurück zum Zitat Bentley G, Biant LC, Carrington RW, Akmal M, Goldberg A, Williams AM, Skinner JA, Pringle J: A prospective, randomised comparison of autologous chondrocyte implantation versus mosaicplasty for osteochondral defects in the knee. J Bone Joint Surg Br. 2003, 85: 223-230. 10.1302/0301-620X.85B2.13543.CrossRefPubMed Bentley G, Biant LC, Carrington RW, Akmal M, Goldberg A, Williams AM, Skinner JA, Pringle J: A prospective, randomised comparison of autologous chondrocyte implantation versus mosaicplasty for osteochondral defects in the knee. J Bone Joint Surg Br. 2003, 85: 223-230. 10.1302/0301-620X.85B2.13543.CrossRefPubMed
34.
Zurück zum Zitat Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR: Multilineage potential of adult human mesenchymal stem cells. Science. 1999, 284: 143-147. 10.1126/science.284.5411.143.CrossRefPubMed Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR: Multilineage potential of adult human mesenchymal stem cells. Science. 1999, 284: 143-147. 10.1126/science.284.5411.143.CrossRefPubMed
35.
Zurück zum Zitat Takagi K, Urist MR: The role of bone marrow in bone morphogenetic protein-induced repair of femoral massive diaphyseal defects. Clin Orthop. 1982, 224-231. Takagi K, Urist MR: The role of bone marrow in bone morphogenetic protein-induced repair of femoral massive diaphyseal defects. Clin Orthop. 1982, 224-231.
36.
Zurück zum Zitat Wakitani S, Goto T, Pineda SJ, Young RG, Mansour JM, Caplan AI, Goldberg VM: Mesenchymal cell-based repair of large, full-thickness defects of articular cartilage. J Bone Joint Surg Am. 1994, 76: 579-592.PubMed Wakitani S, Goto T, Pineda SJ, Young RG, Mansour JM, Caplan AI, Goldberg VM: Mesenchymal cell-based repair of large, full-thickness defects of articular cartilage. J Bone Joint Surg Am. 1994, 76: 579-592.PubMed
37.
Zurück zum Zitat Im GI, Kim DY, Shin JH, Hyun CW, Cho WH: Repair of cartilage defect in the rabbit with cultured mesenchymal stem cells from bone marrow. J Bone Joint Surg Br. 2001, 83: 289-294. 10.1302/0301-620X.83B2.10495.CrossRefPubMed Im GI, Kim DY, Shin JH, Hyun CW, Cho WH: Repair of cartilage defect in the rabbit with cultured mesenchymal stem cells from bone marrow. J Bone Joint Surg Br. 2001, 83: 289-294. 10.1302/0301-620X.83B2.10495.CrossRefPubMed
38.
Zurück zum Zitat Wakitani S, Imoto K, Yamamoto T, Saito M, Murata N, Yoneda M: Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage. 2002, 10: 199-206. 10.1053/joca.2001.0504.CrossRefPubMed Wakitani S, Imoto K, Yamamoto T, Saito M, Murata N, Yoneda M: Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage. 2002, 10: 199-206. 10.1053/joca.2001.0504.CrossRefPubMed
39.
Zurück zum Zitat Orlic D, Kajstura J, Chimenti S, Bodine DM, Leri A, Anversa P: Transplanted adult bone marrow cells repair myocardial infarcts in mice. Ann N Y Acad Sci. 2001, 938: 221-9; discussion 229-30.CrossRefPubMed Orlic D, Kajstura J, Chimenti S, Bodine DM, Leri A, Anversa P: Transplanted adult bone marrow cells repair myocardial infarcts in mice. Ann N Y Acad Sci. 2001, 938: 221-9; discussion 229-30.CrossRefPubMed
40.
Zurück zum Zitat Orlic D, Kajstura J, Chimenti S, Bodine DM, Leri A, Anversa P: Bone marrow stem cells regenerate infarcted myocardium. Pediatr Transplant. 2003, 7 Suppl 3: 86-88. 10.1034/j.1399-3046.7.s3.13.x.CrossRefPubMed Orlic D, Kajstura J, Chimenti S, Bodine DM, Leri A, Anversa P: Bone marrow stem cells regenerate infarcted myocardium. Pediatr Transplant. 2003, 7 Suppl 3: 86-88. 10.1034/j.1399-3046.7.s3.13.x.CrossRefPubMed
41.
Zurück zum Zitat Terai S, Yamamoto N, Omori K, Sakaida I, Okita K: A new cell therapy using bone marrow cells to repair damaged liver. J Gastroenterol. 2002, 37 Suppl 14: 162-163.CrossRefPubMed Terai S, Yamamoto N, Omori K, Sakaida I, Okita K: A new cell therapy using bone marrow cells to repair damaged liver. J Gastroenterol. 2002, 37 Suppl 14: 162-163.CrossRefPubMed
42.
Zurück zum Zitat Chopp M, Zhang XH, Li Y, Wang L, Chen J, Lu D, Lu M, Rosenblum M: Spinal cord injury in rat: treatment with bone marrow stromal cell transplantation. Neuroreport. 2000, 11: 3001-3005.CrossRefPubMed Chopp M, Zhang XH, Li Y, Wang L, Chen J, Lu D, Lu M, Rosenblum M: Spinal cord injury in rat: treatment with bone marrow stromal cell transplantation. Neuroreport. 2000, 11: 3001-3005.CrossRefPubMed
43.
Zurück zum Zitat Akiyama Y, Radtke C, Honmou O, Kocsis JD: Remyelination of the spinal cord following intravenous delivery of bone marrow cells. Glia. 2002, 39: 229-236. 10.1002/glia.10102.PubMedCentralCrossRefPubMed Akiyama Y, Radtke C, Honmou O, Kocsis JD: Remyelination of the spinal cord following intravenous delivery of bone marrow cells. Glia. 2002, 39: 229-236. 10.1002/glia.10102.PubMedCentralCrossRefPubMed
44.
Zurück zum Zitat Sasaki M, Honmou O, Akiyama Y, Uede T, Hashi K, Kocsis JD: Transplantation of an acutely isolated bone marrow fraction repairs demyelinated adult rat spinal cord axons. Glia. 2001, 35: 26-34. 10.1002/glia.1067.PubMedCentralCrossRefPubMed Sasaki M, Honmou O, Akiyama Y, Uede T, Hashi K, Kocsis JD: Transplantation of an acutely isolated bone marrow fraction repairs demyelinated adult rat spinal cord axons. Glia. 2001, 35: 26-34. 10.1002/glia.1067.PubMedCentralCrossRefPubMed
45.
Zurück zum Zitat Ianus A, Holz GG, Theise ND, Hussain MA: In vivo derivation of glucose-competent pancreatic endocrine cells from bone marrow without evidence of cell fusion. J Clin Invest. 2003, 111: 843-850. 10.1172/JCI200316502.PubMedCentralCrossRefPubMed Ianus A, Holz GG, Theise ND, Hussain MA: In vivo derivation of glucose-competent pancreatic endocrine cells from bone marrow without evidence of cell fusion. J Clin Invest. 2003, 111: 843-850. 10.1172/JCI200316502.PubMedCentralCrossRefPubMed
46.
Zurück zum Zitat Peretti GM, Caruso EM, Randolph MA, Zaleske DJ: Meniscal repair using engineered tissue. J Orthop Res. 2001, 19: 278-285. 10.1016/S0736-0266(00)90010-X.CrossRefPubMed Peretti GM, Caruso EM, Randolph MA, Zaleske DJ: Meniscal repair using engineered tissue. J Orthop Res. 2001, 19: 278-285. 10.1016/S0736-0266(00)90010-X.CrossRefPubMed
47.
Zurück zum Zitat Eaglstein WH, Falanga V: Tissue engineering and the development of Apligraf a human skin equivalent. Adv Wound Care. 1998, 11: 1-8.PubMed Eaglstein WH, Falanga V: Tissue engineering and the development of Apligraf a human skin equivalent. Adv Wound Care. 1998, 11: 1-8.PubMed
48.
Zurück zum Zitat Chu CR, Coutts RD, Yoshioka M, Harwood FL, Monosov AZ, Amiel D: Articular cartilage repair using allogeneic perichondrocyte-seeded biodegradable porous polylactic acid (PLA): a tissue-engineering study. J Biomed Mater Res. 1995, 29: 1147-1154.CrossRefPubMed Chu CR, Coutts RD, Yoshioka M, Harwood FL, Monosov AZ, Amiel D: Articular cartilage repair using allogeneic perichondrocyte-seeded biodegradable porous polylactic acid (PLA): a tissue-engineering study. J Biomed Mater Res. 1995, 29: 1147-1154.CrossRefPubMed
49.
Zurück zum Zitat Parenteau NL, Bilbo P, Nolte CJ, Mason VS, Rosenberg M: The organotypic culture of human skin keratinocytes and fibroblasts to achieve form and function. Cytotechnology. 1992, 9: 163-171.CrossRefPubMed Parenteau NL, Bilbo P, Nolte CJ, Mason VS, Rosenberg M: The organotypic culture of human skin keratinocytes and fibroblasts to achieve form and function. Cytotechnology. 1992, 9: 163-171.CrossRefPubMed
50.
Zurück zum Zitat Aubock J, Irschick E, Romani N, Kompatscher P, Hopfl R, Herold M, Schuler G, Bauer M, Huber C, Fritsch P: Rejection, after a slightly prolonged survival time, of Langerhans cell-free allogeneic cultured epidermis used for wound coverage in humans. Transplantation. 1988, 45: 730-737.CrossRefPubMed Aubock J, Irschick E, Romani N, Kompatscher P, Hopfl R, Herold M, Schuler G, Bauer M, Huber C, Fritsch P: Rejection, after a slightly prolonged survival time, of Langerhans cell-free allogeneic cultured epidermis used for wound coverage in humans. Transplantation. 1988, 45: 730-737.CrossRefPubMed
51.
Zurück zum Zitat Sabolinski ML, Alvarez O, Auletta M, Mulder G, Parenteau NL: Cultured skin as a 'smart material' for healing wounds: experience in venous ulcers. Biomaterials. 1996, 17: 311-320. 10.1016/0142-9612(96)85569-4.CrossRefPubMed Sabolinski ML, Alvarez O, Auletta M, Mulder G, Parenteau NL: Cultured skin as a 'smart material' for healing wounds: experience in venous ulcers. Biomaterials. 1996, 17: 311-320. 10.1016/0142-9612(96)85569-4.CrossRefPubMed
52.
Zurück zum Zitat Briscoe DM, Dharnidharka VR, Isaacs C, Downing G, Prosky S, Shaw P, Parenteau NL, Hardin-Young J: The allogeneic response to cultured human skin equivalent in the hu-PBL-SCID mouse model of skin rejection. Transplantation. 1999, 67: 1590-1599. 10.1097/00007890-199906270-00014.CrossRefPubMed Briscoe DM, Dharnidharka VR, Isaacs C, Downing G, Prosky S, Shaw P, Parenteau NL, Hardin-Young J: The allogeneic response to cultured human skin equivalent in the hu-PBL-SCID mouse model of skin rejection. Transplantation. 1999, 67: 1590-1599. 10.1097/00007890-199906270-00014.CrossRefPubMed
53.
Zurück zum Zitat Subramanian T, Marchionini D, Potter EM, Cornfeldt ML: Striatal xenotransplantation of human retinal pigment epithelial cells attached to microcarriers in hemiparkinsonian rats ameliorates behavioral deficits without provoking a host immune response. Cell Transplant. 2002, 11: 207-214.PubMed Subramanian T, Marchionini D, Potter EM, Cornfeldt ML: Striatal xenotransplantation of human retinal pigment epithelial cells attached to microcarriers in hemiparkinsonian rats ameliorates behavioral deficits without provoking a host immune response. Cell Transplant. 2002, 11: 207-214.PubMed
54.
Zurück zum Zitat Watts RL, Raiser, CD, Stover, NP, Cornfeldt, ML, Schweikert, ML,, Allen RC, Somerville, NJ, Subramanian, T, Bakay, RAE: Stereotaxic Intrastriatal Implantation of Retinal Pigment Epithelial Cells Attached to Microcarriers in Six Advanced Parkinson Disease (PD) Patients: Two Year Follow-Up. American Academy of Neurology, 55th Annual Meeting, March 29 - April 5th, 2003. 2003, Honolulu, Hawaii Watts RL, Raiser, CD, Stover, NP, Cornfeldt, ML, Schweikert, ML,, Allen RC, Somerville, NJ, Subramanian, T, Bakay, RAE: Stereotaxic Intrastriatal Implantation of Retinal Pigment Epithelial Cells Attached to Microcarriers in Six Advanced Parkinson Disease (PD) Patients: Two Year Follow-Up. American Academy of Neurology, 55th Annual Meeting, March 29 - April 5th, 2003. 2003, Honolulu, Hawaii
55.
Zurück zum Zitat Philips MF, Muir JK, Saatman KE, Raghupathi R, Lee VM, Trojanowski JQ, McIntosh TK: Survival and integration of transplanted postmitotic human neurons following experimental brain injury in immunocompetent rats. J Neurosurg. 1999, 90: 116-124.CrossRefPubMed Philips MF, Muir JK, Saatman KE, Raghupathi R, Lee VM, Trojanowski JQ, McIntosh TK: Survival and integration of transplanted postmitotic human neurons following experimental brain injury in immunocompetent rats. J Neurosurg. 1999, 90: 116-124.CrossRefPubMed
56.
Zurück zum Zitat Borlongan CV, Tajima Y, Trojanowski JQ, Lee VM, Sanberg PR: Cerebral ischemia and CNS transplantation: differential effects of grafted fetal rat striatal cells and human neurons derived from a clonal cell line. Neuroreport. 1998, 9: 3703-3709.CrossRefPubMed Borlongan CV, Tajima Y, Trojanowski JQ, Lee VM, Sanberg PR: Cerebral ischemia and CNS transplantation: differential effects of grafted fetal rat striatal cells and human neurons derived from a clonal cell line. Neuroreport. 1998, 9: 3703-3709.CrossRefPubMed
57.
Zurück zum Zitat Kondziolka D, Wechsler L, Goldstein S, Meltzer C, Thulborn KR, Gebel J, Jannetta P, DeCesare S, Elder EM, McGrogan M, Reitman MA, Bynum L: Transplantation of cultured human neuronal cells for patients with stroke. Neurology. 2000, 55: 565-569.CrossRefPubMed Kondziolka D, Wechsler L, Goldstein S, Meltzer C, Thulborn KR, Gebel J, Jannetta P, DeCesare S, Elder EM, McGrogan M, Reitman MA, Bynum L: Transplantation of cultured human neuronal cells for patients with stroke. Neurology. 2000, 55: 565-569.CrossRefPubMed
58.
Zurück zum Zitat Nelson PT, Kondziolka D, Wechsler L, Goldstein S, Gebel J, DeCesare S, Elder EM, Zhang PJ, Jacobs A, McGrogan M, Lee VM, Trojanowski JQ: Clonal human (hNT) neuron grafts for stroke therapy: neuropathology in a patient 27 months after implantation. Am J Pathol. 2002, 160: 1201-1206.PubMedCentralCrossRefPubMed Nelson PT, Kondziolka D, Wechsler L, Goldstein S, Gebel J, DeCesare S, Elder EM, Zhang PJ, Jacobs A, McGrogan M, Lee VM, Trojanowski JQ: Clonal human (hNT) neuron grafts for stroke therapy: neuropathology in a patient 27 months after implantation. Am J Pathol. 2002, 160: 1201-1206.PubMedCentralCrossRefPubMed
59.
Zurück zum Zitat Watanabe T, Kawano Y, Watanabe A, Takaue Y: Autologous and allogeneic transplantation with peripheral blood CD34+ cells: a pediatric experience. Haematologica. 1999, 84: 167-176. 10.1159/000015248.PubMed Watanabe T, Kawano Y, Watanabe A, Takaue Y: Autologous and allogeneic transplantation with peripheral blood CD34+ cells: a pediatric experience. Haematologica. 1999, 84: 167-176. 10.1159/000015248.PubMed
60.
Zurück zum Zitat Baron F, Baudoux E, Fillet G, Beguin Y: Retrospective comparison of CD34-selected allogeneic peripheral blood stem cell transplantation followed by CD8-depleted donor lymphocyte infusions with unmanipulated bone marrow transplantation. Hematology. 2002, 7: 137-143.CrossRefPubMed Baron F, Baudoux E, Fillet G, Beguin Y: Retrospective comparison of CD34-selected allogeneic peripheral blood stem cell transplantation followed by CD8-depleted donor lymphocyte infusions with unmanipulated bone marrow transplantation. Hematology. 2002, 7: 137-143.CrossRefPubMed
61.
Zurück zum Zitat Uchida N, Buck DW, He D, Reitsma MJ, Masek M, Phan TV, Tsukamoto AS, Gage FH, Weissman IL: Direct isolation of human central nervous system stem cells. Proc Natl Acad Sci U S A. 2000, 97: 14720-14725. 10.1073/pnas.97.26.14720.PubMedCentralCrossRefPubMed Uchida N, Buck DW, He D, Reitsma MJ, Masek M, Phan TV, Tsukamoto AS, Gage FH, Weissman IL: Direct isolation of human central nervous system stem cells. Proc Natl Acad Sci U S A. 2000, 97: 14720-14725. 10.1073/pnas.97.26.14720.PubMedCentralCrossRefPubMed
62.
Zurück zum Zitat Svendsen CN, Caldwell MA, Shen J, ter Borg MG, Rosser AE, Tyers P, Karmiol S, Dunnett SB: Long-term survival of human central nervous system progenitor cells transplanted into a rat model of Parkinson's disease. Exp Neurol. 1997, 148: 135-146. 10.1006/exnr.1997.6634.CrossRefPubMed Svendsen CN, Caldwell MA, Shen J, ter Borg MG, Rosser AE, Tyers P, Karmiol S, Dunnett SB: Long-term survival of human central nervous system progenitor cells transplanted into a rat model of Parkinson's disease. Exp Neurol. 1997, 148: 135-146. 10.1006/exnr.1997.6634.CrossRefPubMed
63.
Zurück zum Zitat Svendsen CN, ter Borg MG, Armstrong RJ, Rosser AE, Chandran S, Ostenfeld T, Caldwell MA: A new method for the rapid and long term growth of human neural precursor cells. J Neurosci Methods. 1998, 85: 141-152. 10.1016/S0165-0270(98)00126-5.CrossRefPubMed Svendsen CN, ter Borg MG, Armstrong RJ, Rosser AE, Chandran S, Ostenfeld T, Caldwell MA: A new method for the rapid and long term growth of human neural precursor cells. J Neurosci Methods. 1998, 85: 141-152. 10.1016/S0165-0270(98)00126-5.CrossRefPubMed
64.
Zurück zum Zitat Svendsen CN, Clarke DJ, Rosser AE, Dunnett SB: Survival and differentiation of rat and human epidermal growth factor-responsive precursor cells following grafting into the lesioned adult central nervous system. Exp Neurol. 1996, 137: 376-388. 10.1006/exnr.1996.0039.CrossRefPubMed Svendsen CN, Clarke DJ, Rosser AE, Dunnett SB: Survival and differentiation of rat and human epidermal growth factor-responsive precursor cells following grafting into the lesioned adult central nervous system. Exp Neurol. 1996, 137: 376-388. 10.1006/exnr.1996.0039.CrossRefPubMed
65.
Zurück zum Zitat Svendsen CN, Caldwell MA, Ostenfeld T: Human neural stem cells: isolation, expansion and transplantation. Brain Pathol. 1999, 9: 499-513.CrossRefPubMed Svendsen CN, Caldwell MA, Ostenfeld T: Human neural stem cells: isolation, expansion and transplantation. Brain Pathol. 1999, 9: 499-513.CrossRefPubMed
66.
Zurück zum Zitat Nunes MC, Roy NS, Keyoung HM, Goodman RR, McKhann G., 2nd, Jiang L, Kang J, Nedergaard M, Goldman SA: Identification and isolation of multipotential neural progenitor cells from the subcortical white matter of the adult human brain. Nat Med. 2003, 9: 439-447. 10.1038/nm837.CrossRefPubMed Nunes MC, Roy NS, Keyoung HM, Goodman RR, McKhann G., 2nd, Jiang L, Kang J, Nedergaard M, Goldman SA: Identification and isolation of multipotential neural progenitor cells from the subcortical white matter of the adult human brain. Nat Med. 2003, 9: 439-447. 10.1038/nm837.CrossRefPubMed
67.
Zurück zum Zitat Keyoung HM, Roy NS, Benraiss A, Louissaint A., Jr., Suzuki A, Hashimoto M, Rashbaum WK, Okano H, Goldman SA: High-yield selection and extraction of two promoter-defined phenotypes of neural stem cells from the fetal human brain. Nat Biotechnol. 2001, 19: 843-850. 10.1038/nbt0901-843.CrossRefPubMed Keyoung HM, Roy NS, Benraiss A, Louissaint A., Jr., Suzuki A, Hashimoto M, Rashbaum WK, Okano H, Goldman SA: High-yield selection and extraction of two promoter-defined phenotypes of neural stem cells from the fetal human brain. Nat Biotechnol. 2001, 19: 843-850. 10.1038/nbt0901-843.CrossRefPubMed
68.
Zurück zum Zitat Evans MJ, Kaufman MH: Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981, 292: 154-156.CrossRefPubMed Evans MJ, Kaufman MH: Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981, 292: 154-156.CrossRefPubMed
69.
Zurück zum Zitat Nagy A, Gocza E, Diaz EM, Prideaux VR, Ivanyi E, Markkula M, Rossant J: Embryonic stem cells alone are able to support fetal development in the mouse. Development. 1990, 110: 815-821.PubMed Nagy A, Gocza E, Diaz EM, Prideaux VR, Ivanyi E, Markkula M, Rossant J: Embryonic stem cells alone are able to support fetal development in the mouse. Development. 1990, 110: 815-821.PubMed
70.
Zurück zum Zitat Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM: Embryonic stem cell lines derived from human blastocysts. Science. 1998, 282: 1145-1147. 10.1126/science.282.5391.1145.CrossRefPubMed Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM: Embryonic stem cell lines derived from human blastocysts. Science. 1998, 282: 1145-1147. 10.1126/science.282.5391.1145.CrossRefPubMed
71.
Zurück zum Zitat Guan K, Chang H, Rolletschek A, Wobus AM: Embryonic stem cell-derived neurogenesis. Retinoic acid induction and lineage selection of neuronal cells. Cell Tissue Res. 2001, 305: 171-176. 10.1007/s004410100416.CrossRefPubMed Guan K, Chang H, Rolletschek A, Wobus AM: Embryonic stem cell-derived neurogenesis. Retinoic acid induction and lineage selection of neuronal cells. Cell Tissue Res. 2001, 305: 171-176. 10.1007/s004410100416.CrossRefPubMed
72.
Zurück zum Zitat Kaufman DS, Hanson ET, Lewis RL, Auerbach R, Thomson JA: Hematopoietic colony-forming cells derived from human embryonic stem cells. Proc Natl Acad Sci U S A. 2001, 98: 10716-10721. 10.1073/pnas.191362598.PubMedCentralCrossRefPubMed Kaufman DS, Hanson ET, Lewis RL, Auerbach R, Thomson JA: Hematopoietic colony-forming cells derived from human embryonic stem cells. Proc Natl Acad Sci U S A. 2001, 98: 10716-10721. 10.1073/pnas.191362598.PubMedCentralCrossRefPubMed
73.
Zurück zum Zitat Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A, Livne E, Binah O, Itskovitz-Eldor J, Gepstein L: Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest. 2001, 108: 407-414. 10.1172/JCI200112131.PubMedCentralCrossRefPubMed Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A, Livne E, Binah O, Itskovitz-Eldor J, Gepstein L: Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest. 2001, 108: 407-414. 10.1172/JCI200112131.PubMedCentralCrossRefPubMed
74.
Zurück zum Zitat Dani C: Embryonic stem cell-derived adipogenesis. Cells Tissues Organs. 1999, 165: 173-180. 10.1159/000016697.CrossRefPubMed Dani C: Embryonic stem cell-derived adipogenesis. Cells Tissues Organs. 1999, 165: 173-180. 10.1159/000016697.CrossRefPubMed
75.
Zurück zum Zitat Trounson A: Human embryonic stem cells: mother of all cell and tissue types. Reprod Biomed Online. 2002, 4 Suppl 1: 58-63.CrossRefPubMed Trounson A: Human embryonic stem cells: mother of all cell and tissue types. Reprod Biomed Online. 2002, 4 Suppl 1: 58-63.CrossRefPubMed
76.
Zurück zum Zitat Odorico JS, Kaufman DS, Thomson JA: Multilineage differentiation from human embryonic stem cell lines. Stem Cells. 2001, 19: 193-204.CrossRefPubMed Odorico JS, Kaufman DS, Thomson JA: Multilineage differentiation from human embryonic stem cell lines. Stem Cells. 2001, 19: 193-204.CrossRefPubMed
77.
Zurück zum Zitat Carpenter MK, Rosler E, Rao MS: Characterization and differentiation of human embryonic stem cells. Cloning Stem Cells. 2003, 5: 79-88. 10.1089/153623003321512193.CrossRefPubMed Carpenter MK, Rosler E, Rao MS: Characterization and differentiation of human embryonic stem cells. Cloning Stem Cells. 2003, 5: 79-88. 10.1089/153623003321512193.CrossRefPubMed
78.
Zurück zum Zitat Appel J. Z., 3rd, Alwayn IP, Cooper DK: Xenotransplantation: the challenge to current psychosocial attitudes. Prog Transplant. 2000, 10: 217-225.CrossRefPubMed Appel J. Z., 3rd, Alwayn IP, Cooper DK: Xenotransplantation: the challenge to current psychosocial attitudes. Prog Transplant. 2000, 10: 217-225.CrossRefPubMed
79.
Zurück zum Zitat Platt JL: Immunobiology of xenotransplantation. Transpl Int. 2000, 13 Suppl 1: S7-10. 10.1007/s001470050265.CrossRefPubMed Platt JL: Immunobiology of xenotransplantation. Transpl Int. 2000, 13 Suppl 1: S7-10. 10.1007/s001470050265.CrossRefPubMed
80.
Zurück zum Zitat Fodor WL, Williams BL, Matis LA, Madri JA, Rollins SA, Knight JW, Velander W, Squinto SP: Expression of a functional human complement inhibitor in a transgenic pig as a model for the prevention of xenogeneic hyperacute organ rejection. Proc Natl Acad Sci U S A. 1994, 91: 11153-11157.PubMedCentralCrossRefPubMed Fodor WL, Williams BL, Matis LA, Madri JA, Rollins SA, Knight JW, Velander W, Squinto SP: Expression of a functional human complement inhibitor in a transgenic pig as a model for the prevention of xenogeneic hyperacute organ rejection. Proc Natl Acad Sci U S A. 1994, 91: 11153-11157.PubMedCentralCrossRefPubMed
81.
Zurück zum Zitat Ramsoondar JJ, Machaty Z, Costa C, Williams BL, Fodor WL, Bondioli KR: Production of {alpha}1,3-Galactosyltransferase-Knockout Cloned Pigs Expressing Human {alpha}1,2-Fucosylosyltransferase. Biol Reprod. 2003 Ramsoondar JJ, Machaty Z, Costa C, Williams BL, Fodor WL, Bondioli KR: Production of {alpha}1,3-Galactosyltransferase-Knockout Cloned Pigs Expressing Human {alpha}1,2-Fucosylosyltransferase. Biol Reprod. 2003
82.
Zurück zum Zitat Costa C, Zhao L, Burton WV, Bondioli KR, Williams BL, Hoagland TA, Ditullio PA, Ebert KM, Fodor WL: Expression of the human alpha1,2-fucosyltransferase in transgenic pigs modifies the cell surface carbohydrate phenotype and confers resistance to human serum-mediated cytolysis. Faseb J. 1999, 13: 1762-1773.PubMed Costa C, Zhao L, Burton WV, Bondioli KR, Williams BL, Hoagland TA, Ditullio PA, Ebert KM, Fodor WL: Expression of the human alpha1,2-fucosyltransferase in transgenic pigs modifies the cell surface carbohydrate phenotype and confers resistance to human serum-mediated cytolysis. Faseb J. 1999, 13: 1762-1773.PubMed
83.
Zurück zum Zitat Costa C, Zhao L, Burton WV, Rosas C, Bondioli KR, Williams BL, Hoagland TA, Dalmasso AP, Fodor WL: Transgenic pigs designed to express human CD59 and H-transferase to avoid humoral xenograft rejection. Xenotransplantation. 2002, 9: 45-57. 10.1034/j.1399-3089.2002.0o142.x.CrossRefPubMed Costa C, Zhao L, Burton WV, Rosas C, Bondioli KR, Williams BL, Hoagland TA, Dalmasso AP, Fodor WL: Transgenic pigs designed to express human CD59 and H-transferase to avoid humoral xenograft rejection. Xenotransplantation. 2002, 9: 45-57. 10.1034/j.1399-3089.2002.0o142.x.CrossRefPubMed
84.
Zurück zum Zitat Phelps CJ, Koike C, Vaught TD, Boone J, Wells KD, Chen SH, Ball S, Specht SM, Polejaeva IA, Monahan JA, Jobst PM, Sharma SB, Lamborn AE, Garst AS, Moore M, Demetris AJ, Rudert WA, Bottino R, Bertera S, Trucco M, Starzl TE, Dai Y, Ayares DL: Production of alpha 1,3-galactosyltransferase-deficient pigs. Science. 2003, 299: 411-414. 10.1126/science.1078942.PubMedCentralCrossRefPubMed Phelps CJ, Koike C, Vaught TD, Boone J, Wells KD, Chen SH, Ball S, Specht SM, Polejaeva IA, Monahan JA, Jobst PM, Sharma SB, Lamborn AE, Garst AS, Moore M, Demetris AJ, Rudert WA, Bottino R, Bertera S, Trucco M, Starzl TE, Dai Y, Ayares DL: Production of alpha 1,3-galactosyltransferase-deficient pigs. Science. 2003, 299: 411-414. 10.1126/science.1078942.PubMedCentralCrossRefPubMed
85.
Zurück zum Zitat Dai Y, Vaught TD, Boone J, Chen SH, Phelps CJ, Ball S, Monahan JA, Jobst PM, McCreath KJ, Lamborn AE, Cowell-Lucero JL, Wells KD, Colman A, Polejaeva IA, Ayares DL: Targeted disruption of the alpha1,3-galactosyltransferase gene in cloned pigs. Nat Biotechnol. 2002, 20: 251-255. 10.1038/nbt0302-251.CrossRefPubMed Dai Y, Vaught TD, Boone J, Chen SH, Phelps CJ, Ball S, Monahan JA, Jobst PM, McCreath KJ, Lamborn AE, Cowell-Lucero JL, Wells KD, Colman A, Polejaeva IA, Ayares DL: Targeted disruption of the alpha1,3-galactosyltransferase gene in cloned pigs. Nat Biotechnol. 2002, 20: 251-255. 10.1038/nbt0302-251.CrossRefPubMed
86.
Zurück zum Zitat Lai L, Kolber-Simonds D, Park KW, Cheong HT, Greenstein JL, Im GS, Samuel M, Bonk A, Rieke A, Day BN, Murphy CN, Carter DB, Hawley RJ, Prather RS: Production of alpha-1,3-galactosyltransferase knockout pigs by nuclear transfer cloning. Science. 2002, 295: 1089-1092. 10.1126/science.1068228.CrossRefPubMed Lai L, Kolber-Simonds D, Park KW, Cheong HT, Greenstein JL, Im GS, Samuel M, Bonk A, Rieke A, Day BN, Murphy CN, Carter DB, Hawley RJ, Prather RS: Production of alpha-1,3-galactosyltransferase knockout pigs by nuclear transfer cloning. Science. 2002, 295: 1089-1092. 10.1126/science.1068228.CrossRefPubMed
87.
Zurück zum Zitat Groth CG, Tibell A, Wennberg L, Korsgren O: Xenoislet transplantation: experimental and clinical aspects. J Mol Med. 1999, 77: 153-154. 10.1007/s001090050325.CrossRefPubMed Groth CG, Tibell A, Wennberg L, Korsgren O: Xenoislet transplantation: experimental and clinical aspects. J Mol Med. 1999, 77: 153-154. 10.1007/s001090050325.CrossRefPubMed
88.
Zurück zum Zitat Tibell A, Groth CG, Moller E, Korsgren O, Andersson A, Hellerstrom C: Pig-to-human islet transplantation in eight patients. Transplant Proc. 1994, 26: 762-763.PubMed Tibell A, Groth CG, Moller E, Korsgren O, Andersson A, Hellerstrom C: Pig-to-human islet transplantation in eight patients. Transplant Proc. 1994, 26: 762-763.PubMed
89.
Zurück zum Zitat Schumacher JM, Ellias SA, Palmer EP, Kott HS, Dinsmore J, Dempsey PK, Fischman AJ, Thomas C, Feldman RG, Kassissieh S, Raineri R, Manhart C, Penney D, Fink JS, Isacson O: Transplantation of embryonic porcine mesencephalic tissue in patients with PD. Neurology. 2000, 54: 1042-1050.CrossRefPubMed Schumacher JM, Ellias SA, Palmer EP, Kott HS, Dinsmore J, Dempsey PK, Fischman AJ, Thomas C, Feldman RG, Kassissieh S, Raineri R, Manhart C, Penney D, Fink JS, Isacson O: Transplantation of embryonic porcine mesencephalic tissue in patients with PD. Neurology. 2000, 54: 1042-1050.CrossRefPubMed
90.
Zurück zum Zitat Deacon T, Schumacher J, Dinsmore J, Thomas C, Palmer P, Kott S, Edge A, Penney D, Kassissieh S, Dempsey P, Isacson O: Histological evidence of fetal pig neural cell survival after transplantation into a patient with Parkinson's disease. Nat Med. 1997, 3: 350-353.CrossRefPubMed Deacon T, Schumacher J, Dinsmore J, Thomas C, Palmer P, Kott S, Edge A, Penney D, Kassissieh S, Dempsey P, Isacson O: Histological evidence of fetal pig neural cell survival after transplantation into a patient with Parkinson's disease. Nat Med. 1997, 3: 350-353.CrossRefPubMed
91.
Zurück zum Zitat Fink JS, Schumacher JM, Ellias SL, Palmer EP, Saint-Hilaire M, Shannon K, Penn R, Starr P, VanHorne C, Kott HS, Dempsey PK, Fischman AJ, Raineri R, Manhart C, Dinsmore J, Isacson O: Porcine xenografts in Parkinson's disease and Huntington's disease patients: preliminary results. Cell Transplant. 2000, 9: 273-278.PubMed Fink JS, Schumacher JM, Ellias SL, Palmer EP, Saint-Hilaire M, Shannon K, Penn R, Starr P, VanHorne C, Kott HS, Dempsey PK, Fischman AJ, Raineri R, Manhart C, Dinsmore J, Isacson O: Porcine xenografts in Parkinson's disease and Huntington's disease patients: preliminary results. Cell Transplant. 2000, 9: 273-278.PubMed
92.
Zurück zum Zitat Levy MF, Crippin J, Sutton S, Netto G, McCormack J, Curiel T, Goldstein RM, Newman JT, Gonwa TA, Banchereau J, Diamond LE, Byrne G, Logan J, Klintmalm GB: Liver allotransplantation after extracorporeal hepatic support with transgenic (hCD55/hCD59) porcine livers: clinical results and lack of pig-to-human transmission of the porcine endogenous retrovirus. Transplantation. 2000, 69: 272-280. 10.1097/00007890-200001270-00013.CrossRefPubMed Levy MF, Crippin J, Sutton S, Netto G, McCormack J, Curiel T, Goldstein RM, Newman JT, Gonwa TA, Banchereau J, Diamond LE, Byrne G, Logan J, Klintmalm GB: Liver allotransplantation after extracorporeal hepatic support with transgenic (hCD55/hCD59) porcine livers: clinical results and lack of pig-to-human transmission of the porcine endogenous retrovirus. Transplantation. 2000, 69: 272-280. 10.1097/00007890-200001270-00013.CrossRefPubMed
93.
Zurück zum Zitat Switzer WM, Michler RE, Shanmugam V, Matthews A, Hussain AI, Wright A, Sandstrom P, Chapman LE, Weber C, Safley S, Denny RR, Navarro A, Evans V, Norin AJ, Kwiatkowski P, Heneine W: Lack of cross-species transmission of porcine endogenous retrovirus infection to nonhuman primate recipients of porcine cells, tissues, or organs. Transplantation. 2001, 71: 959-965. 10.1097/00007890-200104150-00022.CrossRefPubMed Switzer WM, Michler RE, Shanmugam V, Matthews A, Hussain AI, Wright A, Sandstrom P, Chapman LE, Weber C, Safley S, Denny RR, Navarro A, Evans V, Norin AJ, Kwiatkowski P, Heneine W: Lack of cross-species transmission of porcine endogenous retrovirus infection to nonhuman primate recipients of porcine cells, tissues, or organs. Transplantation. 2001, 71: 959-965. 10.1097/00007890-200104150-00022.CrossRefPubMed
94.
Zurück zum Zitat Dinsmore JH, Manhart C, Raineri R, Jacoby DB, Moore A: No evidence for infection of human cells with porcine endogenous retrovirus (PERV) after exposure to porcine fetal neuronal cells. Transplantation. 2000, 70: 1382-1389. 10.1097/00007890-200011150-00020.CrossRefPubMed Dinsmore JH, Manhart C, Raineri R, Jacoby DB, Moore A: No evidence for infection of human cells with porcine endogenous retrovirus (PERV) after exposure to porcine fetal neuronal cells. Transplantation. 2000, 70: 1382-1389. 10.1097/00007890-200011150-00020.CrossRefPubMed
95.
Zurück zum Zitat Martin U, Steinhoff G, Kiessig V, Chikobava M, Anssar M, Morschheuser T, Lapin B, Haverich A: Porcine endogenous retrovirus (PERV) was not transmitted from transplanted porcine endothelial cells to baboons in vivo. Transpl Int. 1998, 11: 247-251. 10.1007/s001470050136.CrossRefPubMed Martin U, Steinhoff G, Kiessig V, Chikobava M, Anssar M, Morschheuser T, Lapin B, Haverich A: Porcine endogenous retrovirus (PERV) was not transmitted from transplanted porcine endothelial cells to baboons in vivo. Transpl Int. 1998, 11: 247-251. 10.1007/s001470050136.CrossRefPubMed
96.
Zurück zum Zitat Paradis K, Langford G, Long Z, Heneine W, Sandstrom P, Switzer WM, Chapman LE, Lockey C, Onions D, Otto E: Search for cross-species transmission of porcine endogenous retrovirus in patients treated with living pig tissue. The XEN 111 Study Group. Science. 1999, 285: 1236-1241. 10.1126/science.285.5431.1236.CrossRefPubMed Paradis K, Langford G, Long Z, Heneine W, Sandstrom P, Switzer WM, Chapman LE, Lockey C, Onions D, Otto E: Search for cross-species transmission of porcine endogenous retrovirus in patients treated with living pig tissue. The XEN 111 Study Group. Science. 1999, 285: 1236-1241. 10.1126/science.285.5431.1236.CrossRefPubMed
Metadaten
Titel
Tissue engineering and cell based therapies, from the bench to the clinic: The potential to replace, repair and regenerate
Publikationsdatum
01.12.2003
Erschienen in
Reproductive Biology and Endocrinology / Ausgabe 1/2003
Elektronische ISSN: 1477-7827
DOI
https://doi.org/10.1186/1477-7827-1-102

Weitere Artikel der Ausgabe 1/2003

Reproductive Biology and Endocrinology 1/2003 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.