1151
|
Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med 2003; 9:702-12. [PMID: 12778169 DOI: 10.1038/nm0603-702] [Citation(s) in RCA: 1182] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Emerging evidence suggests that bone marrow-derived endothelial, hematopoietic stem and progenitor cells contribute to tissue vascularization during both embryonic and postnatal physiological processes. Recent preclinical and pioneering clinical studies have shown that introduction of bone marrow-derived endothelial and hematopoietic progenitors can restore tissue vascularization after ischemic events in limbs, retina and myocardium. Corecruitment of angiocompetent hematopoietic cells delivering specific angiogenic factors facilitates incorporation of endothelial progenitor cells (EPCs) into newly sprouting blood vessels. Identification of cellular mediators and tissue-specific chemokines, which facilitate selective recruitment of bone marrow-derived stem and progenitor cells to specific organs, will open up new avenues of research to accelerate organ vascularization and regeneration. In addition, identification of factors that promote differentiation of the progenitor cells will permit functional incorporation into neo-vessels of specific tissues while diminishing potential toxicity to other organs. In this review, we discuss the clinical potential of vascular progenitor and stem cells to restore long-lasting organ vascularization and function.
Collapse
Affiliation(s)
- Shahin Rafii
- Department of Hematology-Oncology and Genetic Medicine, Cornell University Medical College, New York, New York, 10021 USA.
| | | |
Collapse
|
1152
|
Abstract
Cell transplantation is gaining a growing interest as a potential new means of improving the prognosis of patients with cardiac failure. The basic assumption is that left ventricular dysfunction is largely due to the loss of a critical number of cardiomyocytes and that it can be partly reversed by implantation of new contractile cells into the postinfarction scars. Primarily for practical reasons, autologous skeletal myoblasts have been the first to undergo clinical trials but other cell types are also considered, particularly bone marrow stem cells, which are attractive because of their autologous origin and their purported cardiomyocyte/endothelial transdifferentiation potential in response to cues provided by the target organ. However several key issues still need to be addressed including (1) the optimal type of donor cells, (2) the mechanism by which cell engraftment improves cardiac function, (3) the optimization of cell survival, and (4) the potential benefits of cell transplantation in non-ischemic heart failure. In parallel to the experimental studies designed to address these issues clinical trials are under way and should hopefully allow assessing to what extent cell transplantation may improve the outcome of patients with heart failure.
Collapse
Affiliation(s)
- Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Paris, France.
| |
Collapse
|
1153
|
Rubart M, Wang E, Dunn KW, Field LJ. Two-photon molecular excitation imaging of Ca2+ transients in Langendorff-perfused mouse hearts. Am J Physiol Cell Physiol 2003; 284:C1654-68. [PMID: 12584115 DOI: 10.1152/ajpcell.00469.2002] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The ability to image calcium signals at subcellular levels within the intact depolarizing heart could provide valuable information toward a more integrated understanding of cardiac function. Accordingly, a system combining two-photon excitation with laser-scanning microscopy was developed to monitor electrically evoked [Ca(2+)](i) transients in individual cardiomyocytes within noncontracting Langendorff-perfused mouse hearts. [Ca(2+)](i) transients were recorded at depths </=100 microm from the epicardial surface with the fluorescent indicators rhod-2 or fura-2 in the presence of the excitation-contraction uncoupler cytochalasin D. Evoked [Ca(2+)](i) transients were highly synchronized among neighboring cardiomyocytes. At 1 Hz, the times from 90 to 50% (t(90-50%)) and from 50 to 10% (t(50-10%)) of the peak [Ca(2+)](i) were (means +/- SE) 73 +/- 4 and 126 +/- 10 ms, respectively, and at 2 Hz, 62 +/- 3 and 94 +/- 6 ms (n = 19, P < 0.05 vs. 1 Hz) in rhod-2-loaded cardiomyocytes. [Ca(2+)](i) decay was markedly slower in fura-2-loaded hearts (t(90-50%) at 1 Hz, 128 +/- 9 ms and at 2 Hz, 88 +/- 5 ms; t(50-10%) at 1 Hz, 214 +/- 18 ms and at 2 Hz, 163 +/- 7 ms; n = 19, P < 0.05 vs. rhod-2). Fura-2-induced deceleration of [Ca(2+)](i) decline resulted from increased cytosolic Ca(2+) buffering, because the kinetics of rhod-2 decay resembled those obtained with fura-2 after incorporation of the Ca(2+) chelator BAPTA. Propagating calcium waves and [Ca(2+)](i) amplitude alternans were readily detected in paced hearts. This approach should be of general utility to monitor the consequences of genetic and/or functional heterogeneity in cellular calcium signaling within whole mouse hearts at tissue depths that have been inaccessible to single-photon imaging.
Collapse
Affiliation(s)
- Michael Rubart
- Wells Center for Pediatric Research, Indianapolis, Indiana 46202, USA.
| | | | | | | |
Collapse
|
1154
|
Abstract
Until recently, adults stem cells, defined by their self-renewal and differentiation abilities, were thought to be tissue-specific. This concept has been challenged by bone marrow transplantation experiments in mice, demonstrating generation of cells of different phenotype after transplantation of marrow or muscle cells. The term "plasticity" has been coined to explain this phenomenon which could be due to the persistence in adult tissues, of stem cells with multidifferentiation ability or to the "transdifferentiation" ability of some adult cells committed to differentiation, under the influence of unknown environmental cues. The relationship of the cells at the origin of the stem cells plasticity with a new type of mesodermal cell designed under the term of "multipotent adult progenitor cell" (MAPC) remains to be determined. The discovery of this latter is a major advance in this field as the MAPC have isolated from the adult bone marrow and presents certain characteristics of embryonic stem cells with the demonstration of their totipotency towards many tissues, including hematopoiesis. The discovery of the adult stem cell plasticity phenomenon in general, represents a major change in our concepts of stem and developmental biology and possibly the basis for the development of future cell therapy protocols.
Collapse
|
1155
|
Haznedaroglu IC, Oztürk MA. Towards the understanding of the local hematopoietic bone marrow renin-angiotensin system. Int J Biochem Cell Biol 2003; 35:867-80. [PMID: 12676173 DOI: 10.1016/s1357-2725(02)00278-9] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The classical view of the renin-angiotensin system (RAS) as a circulating endocrine system has evolved to organ- and tissue-based systems that perform paracrine/autocrine functions. Angiotensin II (Ang II), the dominant effector peptide of the RAS, regulates cellular growth in a wide variety of tissues in (patho)biological states. In 1996, we hypothesized that there exists a locally active RAS in the bone marrow affecting the growth, production, proliferation and differentiation of hematopoietic cells. Evidences supporting this hypothesis are growing. Ang II, through interacting with Ang II type 1 (AT1) receptor stimulates erythroid differentiation. This stimulatory effect of Ang II on erythropoiesis was completely abolished by a specific AT1 receptor antagonist, losartan. AT1a receptors are present on human CD34(+) hematopoietic stem cells. Ang II increases hematopoietic progenitor cell proliferation and this effect was also blocked by losartan. Angiotensin-converting enzyme (ACE) is involved in enhancing the recruitment of primitive stem cells into S-phase in hematopoietic bone marrow by degrading tetrapeptide AcSDKP. ACE inhibitors modified the circulating hematopoietic progenitors in healthy subjects. RAS may also affect pathological/neoplastic hematopoiesis. Renin has been isolated from leukemic blast cells. Higher bone marrow ACE levels in acute leukemic patients suggested that ACE is produced at higher quantities in the leukemic bone marrow. In this review, the 'State of the Art' of the local bone marrow RAS is summarized. A local RAS in the bone marrow can mediate, in an autocrine/paracrine fashion, some of the principal steps of hematopoietic cell production. To show a causal link between the components of RAS and the other regulatory hematopoietic growth factors is not only an academic curiosity. Elucidation of such a local bone marrow system may offer novel therapeutic approaches in pathologic/neoplastic conditions.
Collapse
Affiliation(s)
- Ibrahim C Haznedaroglu
- Department of Hematology and Internal Medicine, Hacettepe University Medical School, Sihhiye, Ankara TR-06100, Turkey.
| | | |
Collapse
|
1156
|
Beck H, Voswinckel R, Wagner S, Ziegelhoeffer T, Heil M, Helisch A, Schaper W, Acker T, Hatzopoulos AK, Plate KH. Participation of bone marrow-derived cells in long-term repair processes after experimental stroke. J Cereb Blood Flow Metab 2003; 23:709-17. [PMID: 12796719 DOI: 10.1097/01.wcb.0000065940.18332.8d] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Bone marrow-derived cells participate in remodeling processes of many ischemia-associated diseases, which has raised hopes for the use of bone marrow as a source for cell-based therapeutic approaches. To study the participation of bone marrow-derived cells in a stroke model, bone marrow from C57BL/6-TgN(ACTbEGFP)1Osb mice that express green fluorescent protein (GFP) in all cells was transplanted into C57BL/6J mice. The recipient mice underwent permanent occlusion of the middle cerebral artery, and bone marrow-derived cells were tracked by fluorescence. The authors investigated the involvement of bone marrow-derived cells in repair processes 6 weeks and 6 months after infarction. Six weeks after occlusion of the artery, more than 90% of the GFP-positive cells in the infarct border zone were microglial cells. Very few GFP-positive cells expressed endothelial markers in the infarct/infarct border zone, and no bone marrow-derived cells transdifferentiated into astrocytes, neurons, or oligodendroglial cells at all time points investigated. The results indicate the need for additional experimental studies to determine whether therapeutic application of nonselected bone marrow will replenish brain cells beyond an increase in microglial engraftment.
Collapse
Affiliation(s)
- Heike Beck
- GSF-Research Center for Environment & Health, Institute for Clinical Molecular Biology and Tumor Genetics, Munich, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1157
|
Nakamura T, Sano M, Songyang Z, Schneider MD. A Wnt- and beta -catenin-dependent pathway for mammalian cardiac myogenesis. Proc Natl Acad Sci U S A 2003; 100:5834-9. [PMID: 12719544 PMCID: PMC156287 DOI: 10.1073/pnas.0935626100] [Citation(s) in RCA: 203] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Acquisition of a cardiac fate by embryonic mesodermal cells is a fundamental step in heart formation. Heart development in frogs and avians requires positive signals from adjacent endoderm, including bone morphogenic proteins, and is antagonized by a second secreted signal, Wnt proteins, from neural tube. By contrast, mechanisms of mesodermal commitment to create heart muscle in mammals are largely unknown. In addition, Wnt-dependent signals can involve either a canonical beta-catenin pathway or other, alternative mediators. Here, we tested the involvement of Wnts and beta-catenin in mammalian cardiac myogenesis by using a pluripotent mouse cell line (P19CL6) that recapitulates early steps for cardiac specification. In this system, early and late cardiac genes are up-regulated by 1% DMSO, and spontaneous beating occurs. Notably, Wnt3A and Wnt8A were induced days before even the earliest cardiogenic transcription factors. DMSO induced biochemical mediators of Wnt signaling (decreased phosphorylation and increased levels of beta-catenin), which were suppressed by Frizzled-8Fc, a soluble Wnt antagonist. DMSO provoked T cell factor-dependent transcriptional activity; thus, induction of Wnt proteins by DMSO was functionally coupled. Frizzled-8Fc inhibited the induction of cardiogenic transcription factors, cardiogenic growth factors, and sarcomeric myosin heavy chains. Likewise, differentiation was blocked by constitutively active glycogen synthase kinase 3beta, an intracellular inhibitor of the Wntbeta-catenin pathway. Conversely, lithium chloride, which inhibits glycogen synthase kinase 3beta, and Wnt3A-conditioned medium up-regulated early cardiac markers and the proportion of differentiated cells. Thus, Wntbeta-catenin signaling is activated at the inception of mammalian cardiac myogenesis and is indispensable for cardiac differentiation, at least in this pluripotent model system.
Collapse
Affiliation(s)
- Teruya Nakamura
- Center for Cardiovascular Development and Department of Biochemistry, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
1158
|
Kraitchman DL, Heldman AW, Atalar E, Amado LC, Martin BJ, Pittenger MF, Hare JM, Bulte JWM. In vivo magnetic resonance imaging of mesenchymal stem cells in myocardial infarction. Circulation 2003; 107:2290-3. [PMID: 12732608 DOI: 10.1161/01.cir.0000070931.62772.4e] [Citation(s) in RCA: 507] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND We investigated the potential of magnetic resonance imaging (MRI) to track magnetically labeled mesenchymal stem cells (MR-MSCs) in a swine myocardial infarction (MI) model. METHODS AND RESULTS Adult farm pigs (n=5) were subjected to closed-chest experimental MI. MR-MSCs (2.8 to 16x107 cells) were injected intramyocardially under x-ray fluoroscopy. MRIs were obtained on a 1.5T MR scanner to demonstrate the location of the MR-MSCs and were correlated with histology. Contrast-enhanced MRI demonstrated successful injection in the infarct and serial MSC tracking was demonstrated in two animals. CONCLUSIONS MRI tracking of MSCs is feasible and represents a preferred method for studying the engraftment of MSCs in MI.
Collapse
Affiliation(s)
- Dara L Kraitchman
- Johns Hopkins University, School of Medicine, Department of Radiology, 601 N Caroline St, Suite 4231, Baltimore, Md 21287-0845, USA.
| | | | | | | | | | | | | | | |
Collapse
|
1159
|
Itescu S, Schuster MD, Kocher AA. New directions in strategies using cell therapy for heart disease. J Mol Med (Berl) 2003; 81:288-96. [PMID: 12698252 DOI: 10.1007/s00109-003-0432-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2002] [Accepted: 11/12/2002] [Indexed: 01/22/2023]
Abstract
Congestive heart failure remains a major public health problem and is frequently the end result of cardiomyocyte apoptosis and fibrous replacement after myocardial infarction, a process referred to as left ventricular remodeling. Cardiomyocytes undergo terminal differentiation soon after birth and are generally considered to irreversibly withdraw from the cell cycle. In response to ischemic insult adult cardiomyocytes undergo cellular hypertrophy, nuclear ploidy, and a high degree of apoptosis. A small number of human cardiomyocytes retain the capacity to proliferate and regenerate in response to ischemic injury. However, whether these cells are derived from a resident pool of cardiomyocyte stem cells or from a renewable source of circulating bone marrow-derived stem cells that home to the damaged myocardium is at present not known. Replacement and regeneration of functional cardiac muscle after an ischemic insult to the heart could be achieved by either stimulating proliferation of endogenous mature cardiomyocytes or resident cardiac stem cells or by implanting exogenous donor-derived or allogeneic cells such as fetal or embryonic cardiomyocyte precursors, bone marrow derived mesenchymal stem cells, or skeletal myoblasts. The newly formed cardiomyocytes must integrate precisely into the existing myocardial wall in order to augment synchronized contractility and avoid potentially life-threatening alterations in the electrical conduction of the heart. A major impediment to survival of the implanted cells is altered immunogenicity by prolonged ex vivo culture conditions. In addition, concurrent myocardial revascularization is required to ensure viability of the repaired region and prevent further scar tissue formation. Human adult bone marrow contains endothelial precursors which resemble embryonic angioblasts and can be used to induce infarct bed neovascularization after experimental myocardial infarction. This results in protection of cardiomyocytes against apoptosis, induction of cardiomyocyte proliferation and regeneration, long-term salvage and survival of viable myocardium, prevention of left ventricular remodeling, and sustained improvement in cardiac function. It is reasonable to anticipate that cell therapy strategies for ischemic heart disease will need to incorporate (a) a renewable source of proliferating, functional cardiomyocytes, and (b) angioblasts to generate a network of capillaries and larger size blood vessels for supply of oxygen and nutrients to both the chronically ischemic endogenous myocardium and to the newly implanted cardiomyocytes
Collapse
Affiliation(s)
- Silviu Itescu
- Transplantation Immunology, Columbia-Presbyterian Medical Center, 630 West 168th Street, PH 14 Central, New York, NY 10032, USA.
| | | | | |
Collapse
|
1160
|
Abstract
Since the establishment of cell lines derived from human embryonic stem (ES) cells, it has been speculated that out of such "raw material," we could some day produce all sorts of replacement parts for the human body. Human pluripotent stem cells can be isolated from embryonic, fetal, or adult tissues. Enormous self-renewal capacity and developmental potential are the characteristics of ES cells. Somatic stem cells, especially those derived from hematopoietic tissues, have also been reported to exhibit developmental potential heretofore not considered possible. The initial evidences for the plasticity potential of somatic stem cells were so encouraging that the opponents of ES cell research used them as arguments for restricting ES cell research. In the past months, however, critical issues have been raised challenging the validity and the interpretation of the initial data. Whereas hematopoietic stem-cell therapy has been a clinical reality for almost 40 years, there is still a long way to go in basic research before novel therapy strategies with stem cells as replacement for other organ systems can be established. Given the present status, we should keep all options open for research in ES cells and adult stem cells to appreciate the complexity of their differentiation pathways and the relative merits of various types of stem cells for regenerative medicine.
Collapse
Affiliation(s)
- Anthony D Ho
- Department of Medicine V, University of Heidelberg, Germany.
| | | |
Collapse
|
1161
|
Schwartz K, Vilquin JT. Building the translational highway: toward new partnerships between academia and the private sector. Nat Med 2003; 9:493-5. [PMID: 12724751 DOI: 10.1038/nm0503-493] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Ketty Schwartz
- Unité Inserm 582, Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France.
| | | |
Collapse
|
1162
|
Abstract
In the past 4 years, multiple reports have suggested that stem cells derived from adult tissues can differentiate outside their tissue of origin, challenging long-accepted tenets of developmental biology. This concept of stem-cell "plasticity" has helped to galvanize research on stem cells due to the myriad therapeutic possibilities. However, there are wide discrepancies in the reported frequencies of so-called transdifferentiation events, from recent reports of negative data to reports of the contribution in some tissues and systems reaching as much as 20%. The evidence for and against stem-cell plasticity is reviewed here as well as some of the possible sources of the experimental variation.
Collapse
Affiliation(s)
- Margaret A Goodell
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030, USA.
| |
Collapse
|
1163
|
Abstract
Recent studies have demonstrated the existence of several populations of primitive cells in mouse and human bone marrow that have the capacity, both in vitro and in vivo, to give rise to cells of all three germ layers. Mesenchymal/stromal stem cells and hematopoietic stem cells are the leading candidates for this activity that some believe may recapitulate the potential of embryonic stem cells. Very little is known about the molecular controls for this adult stem cell activity commonly referred to as transdifferentiation or plasticity. Regeneration of a large number of cell types and tissues has been investigated with one of the most extensively studied being the myocardium. These studies involved ligation of the left coronary artery in adult mice and the direct injection or mobilization of bone marrow stem cells. Using this protocol we, and others, have observed the generation of new cardiomyocytes and endothelial cells in the zone of ischemic myocardium. This approach has progressed to clinical trials at several academic institutions. Although the preliminary findings from these trials do not permit unequivocal conclusions, they do suggest that safety and feasibility are not significant problems that would argue against extending these trials in a large, randomized, double-blinded study. When considerations such as these are addressed, cell therapy may become a new modality in the treatment of heart patients.
Collapse
Affiliation(s)
- Donald Orlic
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
1164
|
Oh H, Wang SC, Prahash A, Sano M, Moravec CS, Taffet GE, Michael LH, Youker KA, Entman ML, Schneider MD. Telomere attrition and Chk2 activation in human heart failure. Proc Natl Acad Sci U S A 2003; 100:5378-83. [PMID: 12702777 PMCID: PMC154353 DOI: 10.1073/pnas.0836098100] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The "postmitotic" phenotype in adult cardiac muscle exhibits similarities to replicative senescence more generally and constitutes a barrier to effective restorative growth in heart disease. Telomere dysfunction is implicated in senescence and apoptotic signaling but its potential role in heart disorders is unknown. Here, we report that cardiac apoptosis in human heart failure is associated specifically with defective expression of the telomere repeat- binding factor TRF2, telomere shortening, and activation of the DNA damage checkpoint kinase, Chk2. In cultured cardiomyocytes, interference with either TRF2 function or expression triggered telomere erosion and apoptosis, indicating that cell death can occur via this pathway even in postmitotic, noncycling cells; conversely, exogenous TRF2 conferred protection from oxidative stress. In vivo, mechanical stress was sufficient to down-regulate TRF2, shorten telomeres, and activate Chk2 in mouse myocardium, and transgenic expression of telomerase reverse transcriptase conferred protection from all three responses. Together, these data suggest that apoptosis in chronic heart failure is mediated in part by telomere dysfunction and suggest an essential role for TRF2 even in postmitotic cells.
Collapse
Affiliation(s)
- Hidemasa Oh
- Center for Cardiovascular Development, and Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1165
|
Caplice NM, Bunch TJ, Stalboerger PG, Wang S, Simper D, Miller DV, Russell SJ, Litzow MR, Edwards WD. Smooth muscle cells in human coronary atherosclerosis can originate from cells administered at marrow transplantation. Proc Natl Acad Sci U S A 2003; 100:4754-9. [PMID: 12665618 PMCID: PMC153628 DOI: 10.1073/pnas.0730743100] [Citation(s) in RCA: 264] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis is the major cause of adult mortality in the developed world, and a significant contributor to atherosclerotic plaque progression involves smooth muscle cell recruitment to the intima of the vessel wall. Controversy currently exists on the exact origin of these recruited cells. Here we use sex-mismatched bone marrow transplant subjects to show that smooth muscle cells throughout the atherosclerotic vessel wall can derive from donor bone marrow. We demonstrate extensive recruitment of these cells in diseased compared with undiseased segments and exclude cell-cell fusion events as a cause for this enrichment. These data have broad implications for our understanding of the cellular components of human atherosclerotic plaque and provide a potentially novel target for future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Noel M Caplice
- Division of Cardiovascular Diseases, Molecular Medicine Program, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
1166
|
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. [PMID: 12639990 DOI: 10.1172/jci200316502] [Citation(s) in RCA: 452] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bone marrow harbors cells that have the capacity to differentiate into cells of nonhematopoietic tissues of neuronal, endothelial, epithelial, and muscular phenotype. Here we demonstrate that bone marrow-derived cells populate pancreatic islets of Langerhans. Bone marrow cells from male mice that express, using a CRE-LoxP system, an enhanced green fluorescent protein (EGFP) if the insulin gene is actively transcribed were transplanted into lethally irradiated recipient female mice. Four to six weeks after transplantation, recipient mice revealed Y chromosome and EGFP double-positive cells in their pancreatic islets. Neither bone marrow cells nor circulating peripheral blood nucleated cells of donor or recipient mice had any detectable EGFP. EGFP-positive cells purified from islets express insulin, glucose transporter 2 (GLUT2), and transcription factors typically found in pancreatic beta cells. Furthermore, in vitro these bone marrow-derived cells exhibit - as do pancreatic beta cells - glucose-dependent and incretin-enhanced insulin secretion. These results indicate that bone marrow harbors cells that have the capacity to differentiate into functionally competent pancreatic endocrine beta cells and that represent a source for cell-based treatment of diabetes mellitus. The results generated with the CRE-LoxP system also suggest that in vivo cell fusion is an unlikely explanation for the "transdifferentiation" of bone marrow-derived cells into differentiated cell phenotypes.
Collapse
Affiliation(s)
- Andreea Ianus
- Department of Pathology, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | |
Collapse
|
1167
|
Szilvassy SJ, Ragland PL, Miller CL, Eaves CJ. The marrow homing efficiency of murine hematopoietic stem cells remains constant during ontogeny. Exp Hematol 2003; 31:331-8. [PMID: 12691921 DOI: 10.1016/s0301-472x(03)00005-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Several recent studies have established the potential clinical utility of hematopoietic stem cells (HSCs) not only for marrow rescue but also for regenerating diseased or damaged nonhematopoietic tissues. These findings have focused renewed interest in understanding the in vivo trafficking patterns of HSCs from different sources. Previous experiments have suggested that the half-life of HSCs in the circulation is short, although the actual proportion that return to the bone marrow (BM) following transplantation has not been previously quantitated. The present study was undertaken to measure this fraction and compare the values obtained for functionally defined HSCs from adult murine BM and day-14 fetal liver (FL). METHODS The number of HSCs that could be recovered from the BM of lethally irradiated mice 24 hours after intravenous injection of Ly-5 congenic BM or FL cells was determined by limiting-dilution competitive repopulating unit (CRU) assays in secondary mice. RESULTS The marrow seeding efficiency of both adult BM- and FL-CRU able to produce lymphoid and myeloid progeny for 5-26 weeks posttransplant was approximately 10%. FL-CRU generated clones that were approximately threefold larger than those produced by BM-CRU. Interestingly, clones produced by "homed" HSCs were approximately twofold smaller than those produced by freshly isolated HSCs. Differences were also seen in the proportions of lymphoid vs myeloid progeny generated by fresh and homed HSCs. CONCLUSIONS These data suggest common mechanisms regulating the BM homing of long-term repopulating HSCs throughout ontogeny despite subtle differences in the size and composition of the clones they generate.
Collapse
Affiliation(s)
- Stephen J Szilvassy
- Blood and Marrow Transplant Program, University of Kentucky, Lexington, Ky., USA.
| | | | | | | |
Collapse
|
1168
|
Lögdberg L, Sgan SL, Larsen CP, Hillyer CD. Islet transplantation, stem cells, and transfusion medicine. Transfus Med Rev 2003; 17:95-109. [PMID: 12733103 DOI: 10.1053/tmrv.2003.50006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Despite the widespread use of exogenous insulin, morbidity and mortality caused by type 1 diabetes mellitus (DM) continue to place a significant burden on society, both in terms of human suffering and cost. The transplantation of vascularized pancreas, usually performed concurrently with renal transplantation, can cure type 1 DM, as shown by results in more than 15000 such transplants over about 30 years. Transplantation of isolated pancreatic islets, instead of the whole organ, however, offers an attractive alternative that minimizes surgery and its complications. Although islet transplantation initially met with only modest success (only about 9% insulin independence at 1 year posttransplant), recent changes in patient selection criteria, number and treatment of islets transplanted, and better immunosuppressive regimens dramatically improved the results; spawning widespread enthusiasm for islet transplantation. Despite this promise, organ/islet availability remains an important limitation to this technology. A solution to the problem of limited materials for transplantation may be in the use of stem/progenitor cells. This article reviews the background of the current enthusiasm for pancreatic islet cell transplantation, highlights future research trends in the field, and suggests that the new islet-related cellular therapies belong within the domain of transfusion medicine.
Collapse
Affiliation(s)
- Lennart Lögdberg
- Department of Pathology, The Carlos and Marguerite Mason Transplantation Biology Research Center, Emory University School of Medicine, Atlanta 30322, GA, USA.
| | | | | | | |
Collapse
|
1169
|
Hanazono Y, Asano T, Ueda Y, Ozawa K. Genetic manipulation of primate embryonic and hematopoietic stem cells with simian lentivirus vectors. Trends Cardiovasc Med 2003; 13:106-10. [PMID: 12691674 DOI: 10.1016/s1050-1738(02)00253-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
During the past several years, many articles have described how human embryonic stem (ES) cells and adult hematopoietic stem cells (HSCs) can differentiate into cardiac muscle, blood vessels, and various other types of cells. The articles raised the expectation that these stem cells may become useful for the treatment of a variety of diseases, including cardiovascular diseases. Genetic manipulation of ES cells and HSCs would be important for such future applications of the cells. Until now, retroviral vectors have been used primarily for stable expression of transgenes in murine ES cells and HSCs. Because murine models may not predict reliably the biology of ES cells and HSCs in humans, we have utilized primate ES cells and HSCs as targets of gene transfer. We have shown that primate ES cells and HSCs can be transduced efficiently with lentiviral vectors derived from the simian immunodeficiency virus, and that the high transgene expression persists without transcriptional silencing. This highly efficient gene transfer method allows for safe and faithful gene delivery to primate ES cells and HSCs to test potential research and therapeutic applications.
Collapse
Affiliation(s)
- Yutaka Hanazono
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical School, Kawachi, Tochigi, Japan.
| | | | | | | |
Collapse
|
1170
|
Watanabe Y, Haruyama T, Akaike T. Liver-derived matrix metalloproteinase-9 (gelatinase B) recruits progenitor cells from bone marrow into the blood circulation. Biol Pharm Bull 2003; 26:564-8. [PMID: 12673046 DOI: 10.1248/bpb.26.564] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Matrix metalloproteinases (MMPs) are involved in invasive cell behavior, embryonic development and organ remodeling. In this report, we investigated the role of liver-derived MMP-9 in the in vivo system at liver injury. Liver injury induced MMP-9 expression in the liver 3 to 12 h after intravenous administration of anti-Fas antibody, followed by the expression of the activity and the protein detected by zymography and Western blotting, respectively, in the blood circulation. Interestingly, the MMP-9 expression was accompanied by the recruitment of hematopoietic progenitor cells from bone marrow into the circulation. The recruitment was blocked by a specific MMP-9 inhibitor, R94138, which did not affect the Fas-mediated liver injury or induced expression of MMP-9. Compulsive expression of mutant active MMP-9 in the liver also recruited the progenitor cells into the circulation. In contrast, partial hepatectomy, which treatment does not directly injure hepatocytes, did not recruit progenitor cells despite the increased expression of MMP-9 in the circulation. These results suggest that liver-derived MMP-9 induced by liver injury plays an essential role in the recruitment of hematopoietic progenitor cells from bone marrow into the blood circulation.
Collapse
Affiliation(s)
- Yoshifumi Watanabe
- Department of Biomolecular Engineering, Tokyo Institute of Technology, Yokohama, Japan.
| | | | | |
Collapse
|
1171
|
Troyer DL, Weiss ML, Mitchell KE, Martin PL, Davis DL. Incorporation of bovine bone marrow stromal cells into porcine foetal tissues after xenotransplantation. Anat Histol Embryol 2003; 32:98-101. [PMID: 12797531 DOI: 10.1046/j.1439-0264.2003.00428.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Bovine bone marrow stromal cells (BMSCs) were injected into the liver of foetal pigs at about 40 days of gestation to test whether these cells could populate developing tissue, and if so, which ones. Approximately 40 days after injection, the foetuses were harvested and tissue sections from many areas of the body were analysed for the presence of bovine cells using two different methods. First, using PCR, bovine repetitive DNA was found to be present in DNA extracted from foetal pig tissues. Secondly, using oligonucleotide primed in situ synthesis (PRINS), the in situ presence of bovine cells was found within porcine tissue sections. PRINS-labelled cells were found within cartilage, perichondrium, connective tissue and smooth muscle. These data suggest that bovine BMSCs integrate throughout the foetal pig.
Collapse
Affiliation(s)
- D L Troyer
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KA 66506-5802, USA.
| | | | | | | | | |
Collapse
|
1172
|
Preston SL, Alison MR, Forbes SJ, Direkze NC, Poulsom R, Wright NA. The new stem cell biology: something for everyone. Mol Pathol 2003; 56:86-96. [PMID: 12665626 PMCID: PMC1187299 DOI: 10.1136/mp.56.2.86] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The ability of multipotential adult stem cells to cross lineage boundaries (transdifferentiate) is currently causing heated debate in the scientific press. The proponents see adult stem cells as an attractive alternative to the use of embryonic stem cells in regenerative medicine (the treatment of diabetes, Parkinson's disease, etc). However, opponents have questioned the very existence of the process, claiming that cell fusion is responsible for the phenomenon. This review sets out to provide a critical evaluation of the current literature in the adult stem cell field.
Collapse
Affiliation(s)
- S L Preston
- Department of Histopathology, Barts and the London School of Medicine and Dentistry, Turner Street, London E1 2AD, UK.
| | | | | | | | | | | |
Collapse
|
1173
|
Arsic N, Zentilin L, Zacchigna S, Santoro D, Stanta G, Salvi A, Sinagra G, Giacca M. Induction of functional neovascularization by combined VEGF and angiopoietin-1 gene transfer using AAV vectors. Mol Ther 2003; 7:450-9. [PMID: 12727107 DOI: 10.1016/s1525-0016(03)00034-0] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vectors based on the adeno-associated virus (AAV) deliver therapeutic genes to muscle and heart at high efficiency and maintain transgene expression for long periods of time. Here we report about the synergistic effect on blood vessel formation of AAV vectors expressing the 165 aa isoform of vascular endothelial growth factor (VEGF165), a powerful activator of endothelial cells, and of angiopoietin-1 (Ang-1), which is required for vessel maturation. High titer AAV-VEGF165 and AAV-Ang-1 vector preparations were injected either alone or in combination in the normoperfused tibialis anterior muscle of rats. Long term expression of VEGF165 determined massive cellular infiltration of the muscle tissues over time, with the formation of a large set of new vessels. Strikingly, some of the cells infiltrating the treated muscles were found positive for markers of activated endothelial precursors (VEGFR-2/KDR and Tie-2) and for c-kit, an antigen expressed by pluripotent bone marrow stem cells. Expression of VEGF165 eventually resulted in the formation of structured vessels surrounded by a layer of smooth muscle cells. Presence of these arteriolae correlated with significantly increased blood perfusion in the injected areas. Co-expression of VEGF165 with angiopoietin-1-which did not display angiogenic effect per se-remarkably reduced leakage of vessels produced by VEGF165 alone.
Collapse
Affiliation(s)
- Nikola Arsic
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | | | | | | | | | | | | | | |
Collapse
|
1174
|
Wu GD, Nolta JA, Jin YS, Barr ML, Yu H, Starnes VA, Cramer DV. Migration of mesenchymal stem cells to heart allografts during chronic rejection. Transplantation 2003; 75:679-85. [PMID: 12640309 DOI: 10.1097/01.tp.0000048488.35010.95] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSC) are pluripotent progenitors for a variety of cell types, including fibroblasts and muscle cells. Their involvement in the tissue repair of allografts during the development of chronic rejection has been hypothesized, but not yet substantiated, by experimental evidence. METHODS Rat MSC were isolated from circulation using an aortic pouch allograft as a trapping device. The plasticity of these cells was examined in differentiation cultures. One of the resulting MSC lines was immortalized and transduced to express a marker gene. The -labeled cells were then transferred to F344 rats bearing Lewis (LEW) cardiac allografts to measure their localization and contribution to graft tissue repair. RESULTS The MSC isolated from circulation exhibited multipotential for differentiation in culture, developing into various lineages including osteoblasts, lipocytes, chondrocytes, myotubes, and fibroblasts. Intravenous engraftment of the -labeled cells into recipients of heart transplant resulted in migration of the beta-gal+ cells into the lesions of chronic rejection in the cardiac grafts and homing of the cells to the bone marrow. The majority of beta-gal+ cells present in the allografts exhibited fibroblast phenotypes, and a small number of the cells expressed desmin, indicative of myocyte differentiation. CONCLUSION MSC vigorously migrated into the site of allograft rejection. This data suggests that they may be attracted to this site to actively participate in tissue repair during chronic rejection. In addition, given the robust migration, the inhibition of MSC differentiation toward fibroblast progeny and induction toward the myocyte lineage may serve as a new strategy for treatment of chronic rejection and allograft tissue repair.
Collapse
Affiliation(s)
- Gordon D Wu
- Department of Cardiothoracic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA. gdw@.usc.edu
| | | | | | | | | | | | | |
Collapse
|
1175
|
Preffer FI, Dombkowski D, Sykes M, Scadden D, Yang YG. Lineage-negative side-population (SP) cells with restricted hematopoietic capacity circulate in normal human adult blood: immunophenotypic and functional characterization. Stem Cells 2003; 20:417-27. [PMID: 12351812 DOI: 10.1634/stemcells.20-5-417] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Side-population (SP) cells are a recently described rare cell population detected in selected tissues of various mammalian species, but not yet described in the peripheral circulation. In the present study, we have identified for the first time SP cells in lineage-negative adult human blood and have provided an extensive functional and immunophenotypic characterization of these cells. Adult peripheral blood was depleted of mature leukocyte cell types by density gradient and immunomagnetic separation. The SP cell population was identified by its characteristic Hoechst 33342 profile. Immunophenotypic analysis revealed that blood SP cells expressed high levels of CD45, CD59, CD43, CD49d, CD31, and integrin markers and lacked CD34. Highly purified SP cells were put into cobblestone area-forming cell (CAFC), long-term culture-initiating cell (LTC-IC), and liquid cell culture assays; repopulating assays were performed utilizing nonobese diabetic/severe combined immunodeficient mice. Circulating SP cells were shown to exhibit verapamil sensitivity and a low growth rate. LTC-IC, CAFC, and engraftment assays indicated that circulating SP cells had lost the multipotentiality described in murine bone marrow SP cells. However, outgrowth of mature cell types from liquid cell culture suggests the presence of common lymphoid (T and natural killer) and dendritic cell precursor(s) within circulating SP cell populations. The absence of SP cell growth in the LTC-IC, CAFC, and repopulating assays might be intrinsic to the tissue source (marrow versus blood) or species (mouse versus human) tested. Thus, human blood SP cells, although rare, may serve as a source of selected leukocyte progenitor cells. The immunophenotype of circulating SP cells may provide clues to their seeding and homing capacity.
Collapse
Affiliation(s)
- Frederic I Preffer
- Department of Pathology, Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA.
| | | | | | | | | |
Collapse
|
1176
|
Deb A, Wang S, Skelding KA, Miller D, Simper D, Caplice NM. Bone marrow-derived cardiomyocytes are present in adult human heart: A study of gender-mismatched bone marrow transplantation patients. Circulation 2003; 107:1247-9. [PMID: 12628942 DOI: 10.1161/01.cir.0000061910.39145.f0] [Citation(s) in RCA: 245] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Recent studies have identified cardiomyocytes of extracardiac origin in transplanted human hearts, but the exact origin of these myocyte progenitors is currently unknown. METHODS AND RESULTS Hearts of female subjects (n=4) who had undergone sex-mismatched bone marrow transplantation (BMT) were recovered at autopsy and analyzed for the presence of Y chromosome-positive cardiomyocytes. Four female gender-matched BMT subjects served as controls. Fluorescence in situ hybridization (FISH) for the Y chromosome was performed on paraffin-embedded sections to identify cells of bone marrow origin with concomitant immunofluorescent labeling for alpha-sarcomeric actin to identify cardiomyocytes. A total of 160 000 cardiomyocyte nuclei were analyzed approximating 20 000 nuclei per patient. The mean percentage of Y chromosome-positive cardiomyocytes in patients with sex-mismatched BMT was 0.23+/-0.06%. Not a single Y chromosome-positive cardiomyocyte was identified in any of the control patients. Immunofluorescent costaining for laminin and chromosomal ploidy analysis with FISH showed no evidence of either pseudonuclei or cell fusion in any of the chimeric cardiac myocytes identified. CONCLUSIONS These data establish for the first time human bone marrow as a source of extracardiac progenitor cells capable of de novo cardiomyocyte formation.
Collapse
Affiliation(s)
- Arjun Deb
- Division of Cardiovascular Diseases and Molecular Medicine Program, Mayo Clinic, Rochester, Minn 55905, USA
| | | | | | | | | | | |
Collapse
|
1177
|
Abstract
The inability of adult cardiomyocytes to divide to a significant extent and regenerate the myocardium after injury leads to permanent deficits in the number of functional cells, which can contribute to the development and progression of heart failure. The transplantation of skeletal myoblasts or stem cells or cardiomyocytes derived from them into the injured myocardium is a novel and promising approach in the treatment of cardiac disease and the restoration of myocardial function. In this article, skeletal myoblasts and embryonic and bone marrow stem cells are discussed in the context of their potential therapeutic use in cardiac failure. The state of the art in both laboratory and clinic is presented. We discuss current and intrinsic limitations of cardiac cellular transplantation and suggest directions for future research.
Collapse
Affiliation(s)
- Rutger J Hassink
- University Medical Center, Heart Lung Center, Department of Cardio-Thoracic Surgery, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
1178
|
El Oakley RM, Seow KK, Tang TPL, Kok CW, Teh M, Lim YT, Lim SK. Whole bone marrow transplantation induces angiogenesis following acute ischemia. Redox Rep 2003; 7:215-8. [PMID: 12396666 DOI: 10.1179/135100002125000532] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Several recent studies have shown that purified subsets of bone marrow (BM) cells can differentiate into endothelial, cardiac, and other cell types. During coronary artery bypass graft (CABG) surgery, sternal BM is routinely discarded. To determine if this BM can be used to induce angiogenesis and augment perfusion of the cardiac tissues after CABG, a simplified and more practical approach of using whole BM extract was tried to determine whether it would be adequate for the induction of BM-derived angiogenesis in experimental acute limb ischemia. BM was prepared from FVB/N-TgN(TIE2 lacZ)182 Sato (Tie2-lacZ) or B6.129S7-Gtrosa 26 (Rosa 26) mice that express beta-galactosidase (beta-gal) in endothelial cells and most adult tissues, respectively. Acute limb ischemia was induced in either C57BL6/J or FVB/N mice by double ligation of the left femoral artery just distal to the profunda femoral artery branch. Occlusion of the ligated artery was verified by angiography. The study group (n = 31) received an intramuscular injection of 50 micro l containing 1 x 10(6) BM cells, 5 mm proximal to the site of ligation. Experimental controls (n = 21) had an intramuscular injection of 50 micro l of saline. Angiogenesis in the mice was assessed by histological analysis. BM-derived beta-gal(+) cells were observed to aggregate in the vicinity of the ligated artery and not in the injected musculature BM-derived endothelial cells were incorporated within capillaries and small size blood vessels near the site of ligation. Generation of BM-derived blood vessels in experimental acute limb ischemia does not require purification of specific subset of cells. The elimination of cell purification will enhance the ease of using BM transplantation in generating blood vessels.
Collapse
|
1179
|
Pagani FD, DerSimonian H, Zawadzka A, Wetzel K, Edge ASB, Jacoby DB, Dinsmore JH, Wright S, Aretz TH, Eisen HJ, Aaronson KD. Autologous skeletal myoblasts transplanted to ischemia-damaged myocardium in humans. Histological analysis of cell survival and differentiation. J Am Coll Cardiol 2003; 41:879-88. [PMID: 12628737 DOI: 10.1016/s0735-1097(03)00081-0] [Citation(s) in RCA: 323] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES We report histological analysis of hearts from patients with end-stage heart disease who were transplanted with autologous skeletal myoblasts concurrent with left ventricular assist device (LVAD) implantation. BACKGROUND Autologous skeletal myoblast transplantation is under investigation as a means to repair infarcted myocardium. To date, there is only indirect evidence to suggest survival of skeletal muscle in humans. METHODS Five patients (all male; median age 60 years) with ischemic cardiomyopathy, refractory heart failure, and listed for heart transplantation underwent muscle biopsy from the quadriceps muscle. The muscle specimen was shipped to a cell isolation facility where myoblasts were isolated and grown. Patients received a transplant of 300 million cells concomitant with LVAD implantation. Four patients underwent LVAD explant after 68, 91, 141, and 191 days of LVAD support (three transplant, one LVAD death), respectively. One patient remains alive on LVAD support awaiting heart transplantation. RESULTS Skeletal muscle cell survival and differentiation into mature myofibers were directly demonstrated in scarred myocardium from three of the four explanted hearts using an antibody against skeletal muscle-specific myosin heavy chain. An increase in small vessel formation was observed in one of three patients at the site of surviving myotubes, but not in adjacent tissue devoid of engrafted cells. CONCLUSIONS These findings represent demonstration of autologous myoblast cell survival in human heart. The implanted skeletal myoblasts formed viable grafts in heavily scarred human myocardial tissue. These results establish the feasibility of myoblast transplants for myocardial repair in humans.
Collapse
Affiliation(s)
- Francis D Pagani
- Section of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1180
|
Abstract
Cell plasticity is a central issue in stem cell biology. In many recent discussions, observation of cell fusion has been seen as a confounding factor which calls into question published results concerning cell plasticity of, particularly, adult stem cells. An examination of the voluminous literature of "somatic cell fusion" suggests the relatively frequent occurrence of "spontaneous" cell fusion and shows that the complicated cellular phenotypes which it can give rise to have long been recognized. Here, a brief overview of this field is presented, with emphasis on studies of special relevance to current work on cell plasticity.
Collapse
Affiliation(s)
- Joseph J. Lucas
- Department of Pediatrics, National Jewish Medical and Research Center, Denver, CO U.S.A
| | - Naohiro Terada
- Department of Pathology, University of Florida College of Medicine, Gainesville, FL, U.S.A. (Author for correspondence; E-mail
| |
Collapse
|
1181
|
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; 111:843-50. [PMID: 12639990 PMCID: PMC153767 DOI: 10.1172/jci16502] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2002] [Accepted: 02/11/2003] [Indexed: 12/20/2022] Open
Abstract
Bone marrow harbors cells that have the capacity to differentiate into cells of nonhematopoietic tissues of neuronal, endothelial, epithelial, and muscular phenotype. Here we demonstrate that bone marrow-derived cells populate pancreatic islets of Langerhans. Bone marrow cells from male mice that express, using a CRE-LoxP system, an enhanced green fluorescent protein (EGFP) if the insulin gene is actively transcribed were transplanted into lethally irradiated recipient female mice. Four to six weeks after transplantation, recipient mice revealed Y chromosome and EGFP double-positive cells in their pancreatic islets. Neither bone marrow cells nor circulating peripheral blood nucleated cells of donor or recipient mice had any detectable EGFP. EGFP-positive cells purified from islets express insulin, glucose transporter 2 (GLUT2), and transcription factors typically found in pancreatic beta cells. Furthermore, in vitro these bone marrow-derived cells exhibit - as do pancreatic beta cells - glucose-dependent and incretin-enhanced insulin secretion. These results indicate that bone marrow harbors cells that have the capacity to differentiate into functionally competent pancreatic endocrine beta cells and that represent a source for cell-based treatment of diabetes mellitus. The results generated with the CRE-LoxP system also suggest that in vivo cell fusion is an unlikely explanation for the "transdifferentiation" of bone marrow-derived cells into differentiated cell phenotypes.
Collapse
Affiliation(s)
- Andreea Ianus
- Department of Pathology, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | |
Collapse
|
1182
|
Parmar K, Sauk-Schubert C, Burdick D, Handley M, Mauch P. Sca+CD34- murine side population cells are highly enriched for primitive stem cells. Exp Hematol 2003; 31:244-50. [PMID: 12644022 DOI: 10.1016/s0301-472x(02)01074-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE The aim of this study was to characterize murine side population (SP) stem cells and SP cell subpopulations for primitive stem cell capacity. MATERIALS AND METHODS SP cells, characterized by a specific Hoechst dye efflux pattern, were isolated by flow cytometric analysis and sorting from murine adult whole bone marrow (WBM). Different subpopulations of SP cells were isolated by staining with anti-Sca and anti-CD34 antibodies. Primitive stem cell content of SP cells and SP subsets were determined by cobblestone area-forming cell (CAFC) frequencies. RESULTS Measurement of CAFC frequencies revealed that SP cells are greatly enriched for both primitive stem cells (day-28-35 CAFC) and somewhat more mature hematopoietic cells (day-14-21 CAFC) compared to WBM. The day-28 and day-35 CAFC enrichments in SP cells vs WBM cells were 1065 and 471, respectively. Analysis of the subpopulations of SP cells revealed that SP(+)Sca(-)CD34(+) cells contained almost exclusively day-7 CAFC and had little day-28-35 CAFC activity. SP(+)Sca(+)CD34(+) cells had high day-7-14 CAFC frequencies, but lower day-35 CAFC frequencies compared to SP(+)Sca(+)CD34(-) cells. SP(+)Sca(+)CD34(-) cells contained very low day-7 CAFC activity, but nearly 2200 times the day-28-35 CAFC activity as normal bone marrow. To evaluate the influence of Hoechst dye efflux capacity, we divided the SP tail into four groups of cells. The SP cells with lowest efflux of Hoechst dye contained the highest progenitor activity (day-7-14 CAFC). The highest day-35 CAFC frequencies, nearly 6000 times those of normal marrow, were seen in the SP cells with the greatest efflux of the Hoechst dye. CONCLUSIONS Murine SP cells contain both progenitor and primitive populations of hematopoietic stem cells. The most primitive stem cells measured in the in vitro CAFC assay mark for Sca(+) and CD34(-) and have a high ability to efflux Hoechst dye. Isolation of these cells may provide the means to directly study mechanisms of primitive stem cell damage.
Collapse
Affiliation(s)
- Kalindi Parmar
- Department of Radiation Oncology, Brigham and Women's Hospital, and the Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass 02115, USA
| | | | | | | | | |
Collapse
|
1183
|
Abstract
UNLABELLED Although stem cells have held the fascination of scientists for years, the attention of the general public has recently been captured by the derivation of human embryonic stem cells. In this review we describe the historical experiments leading up to the isolation of human embryonic stem cells and discuss recent advances in our understanding of both embryonic and somatic stem cells. Select examples are used to illustrate the potential of stem cells, both in the sense of their ability to differentiate into specific cell types and in the sense of their power to treat various diseases and conditions. Also discussed are recent studies describing current progress toward the treatment of Parkinson disease, spinal cord injuries, diabetes, and cardiac disease. TARGET AUDIENCE Obstetricians & Gynecologists, Family Physicians LEARNING OBJECTIVES After completion of this article, the reader will be able to describe the various types of stem cells, outline potential clinical uses of stem cells, and summarize the somatic cell transdifferentiation debate.
Collapse
Affiliation(s)
- Kristina C Pfendler
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, California, USA.
| | | |
Collapse
|
1184
|
Abstract
The phenomenon of regeneration is of growing interest to medical researchers. Until recently this was an area in which research in flatworms and newts predominated, but there is now a proliferation of research concerning regeneration in virtually all of the organs, not only the heart. One of the object is restoration of function to a failing heart through cell transplantation, and there have been many reports seeking donor sources of somatic stem cells, i.e.: stem cells in marrow or skeletal muscle and ES cells, beginning with those in embryonic myocardial cell transplant experiments. In particular, reports of mesenchymal stem cell differentiation into nerve cell, myocardial cell, skeletal muscle cell, and vascular endothelial cell series have drawn attention to cell plasticity, and clinical applications are awaited.
Collapse
Affiliation(s)
- Satoshi Gojo
- Department of Cardiovascular Surgery, Saitama Medical Center, Kawagoe, Japan.
| | | |
Collapse
|
1185
|
Nishida M, Li TS, Hirata K, Yano M, Matsuzaki M, Hamano K. Improvement of cardiac function by bone marrow cell implantation in a rat hypoperfusion heart model. Ann Thorac Surg 2003; 75:768-73; discussion 773-4. [PMID: 12645691 DOI: 10.1016/s0003-4975(02)04388-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Local bone marrow cell implantation can induce angiogenesis. In the present study we investigated whether angiogenesis induced by bone marrow cell implantation improves deteriorated cardiac function in a rat heart model of hypoperfusion. METHODS A hypoperfusion heart model was created in Dark Agouti rats by ligating the left anterior descending artery placed against a copper wire (phi275 microm), then pulling out the wire immediately. The left ventricular (LV) anterior wall was injected directly at six points, each with 1 x 10(7) bone marrow cells in 10 microL of phosphate-buffered saline or with phosphate-buffered saline only, respectively. Echocardiography was performed to evaluate the cardiac function 7, 30, 60, and 90 days after treatment. Microvessel density and blood flow in the LV anterior wall were estimated 60 days after treatment. RESULTS Both the increase of LV end-systolic diameter and the decrease of percent of fractional shortening caused by myocardial ischemia were attenuated effectively by bone marrow cell implantation treatment. Bone marrow cell implantation treatment also increased the levels of angiopoietin-1 and vascular endothelial growth factor in the LV anterior wall. The microvessel density, blood flow, and thickness of the LV anterior wall significantly also increased after bone marrow cell implantation treatment compared with those after phosphate-buffered saline injection. CONCLUSIONS The local implantation of autologous bone marrow cells induced angiogenesis and improved the perfusion of ischemic myocardium, thereby preventing LV remodeling and improving deteriorated cardiac function caused by myocardial hypoperfusion.
Collapse
Affiliation(s)
- Masahiko Nishida
- Division of Cardiovascular Surgery, Department of Bioregulation, Yamaguchi University School of Medicine, Ube, Yamaguchi, Japan
| | | | | | | | | | | |
Collapse
|
1186
|
Hillebrands JL, Klatter FA, Rozing J. Origin of vascular smooth muscle cells and the role of circulating stem cells in transplant arteriosclerosis. Arterioscler Thromb Vasc Biol 2003; 23:380-7. [PMID: 12615698 DOI: 10.1161/01.atv.0000059337.60393.64] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To date, clinical solid-organ transplantation has not achieved its goals as a long-term treatment for patients with end-stage organ failure. Development of so-called chronic transplant dysfunction (CTD) is now recognized as the predominant cause of allograft loss long term (after the first postoperative year) after transplantation. CTD has the remarkable histological feature that the luminal areas of intragraft arteries become obliterated, predominantly with vascular smooth muscle cells (VSMCs) intermingled with some inflammatory cells (transplant arteriosclerosis, or TA). The development of TA is a multifactorial process, and many risk factors have been identified. However, the precise pathogenetic mechanisms leading to TA are largely unknown and, as a result, adequate prevention and treatment protocols are still lacking. This review discusses the origin (donor versus recipient, bone marrow versus nonbone marrow) of the VSMCs in TA lesions. Poorly controlled influx and subsequent proliferative behavior of these VSMCs are considered to be critical elements in the development of TA. Available data show heterogeneity when analyzing the origin of neointimal VSMCs in various transplant models and species, indicating the existence of multiple sites of origin. Based on these findings, a model considering plasticity of VSMC origin in TA in relation to severity and extent of graft damage is proposed.
Collapse
Affiliation(s)
- Jan-Luuk Hillebrands
- Department Cell Biology/Section Immunology, Faculty of Medical Sciences, University of Groningen, A. Deusinglaan 1, NL-9713 AV Groningen, The Netherlands.
| | | | | |
Collapse
|
1187
|
Cousin B, André M, Arnaud E, Pénicaud L, Casteilla L. Reconstitution of lethally irradiated mice by cells isolated from adipose tissue. Biochem Biophys Res Commun 2003; 301:1016-22. [PMID: 12589814 DOI: 10.1016/s0006-291x(03)00061-5] [Citation(s) in RCA: 376] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It is suggested that hematopoietic stem cells (HSC) could be found in several tissues of mesodermic origin. Among these, adipose tissue can expand throughout adult life and its expansion is not only due to mature adipocyte hypertrophy but also to the presence of precursor cells in stroma-vascular fraction (SVF). Here we report that transplantation of cells isolated from mice adipose tissue can efficiently rescue lethally irradiated mice and results in a reconstitution of major hematopoietic lineages. Donor cells can be detected in blood and in hematopoietic tissues of recipient mice. Adipose tissue contains a significant percentage of CD34, CD45 positive cells, and SVF cells were able to give rise to hematopoietic colonies in methylcellulose. We demonstrate the presence of hematopoietic progenitors in adipose tissue by phenotypic and functional characteristics. Thus adipose tissue could be considered as an important and convenient source of cells able to support hematopoiesis.
Collapse
Affiliation(s)
- Béatrice Cousin
- UMR 5018-CNRS, CHU Rangueil, Bat L1, 1 Avenue J. Poulhès, 31403, Toulouse Cedex, France.
| | | | | | | | | |
Collapse
|
1188
|
Abstract
Stem cells are characterized by the ability to remain undifferentiated and to self-renew. Embryonic stem cells derived from blastocysts are pluripotent (able to differentiate into many cell types). Adult stem cells, which were traditionally thought to be monopotent multipotent, or tissue restricted, have recently also been shown to have pluripotent properties. Adult bone marrow stem cells have been shown to be capable of differentiating into skeletal muscle, brain microglia and astroglia, and hepatocytes. Stem cell lines derived from both embryonic stem and embryonic germ cells (from the embryonic gonadal ridge) are pluripotent and capable of self-renewal for long periods. Therefore embryonic stem and germ cells have been widely investigated for their potential to cure diseases by repairing or replacing damaged cells and tissues. Studies in animal models have shown that transplantation of fetal, embryonic stem, or embryonic germ cells may be able to treat some chronic diseases. In this review, we highlight recent developments in the use of stem cells as therapeutic agents for three such diseases: Diabetes, Parkinson disease, and congestive heart failure. We also discuss the potential use of stem cells as gene therapy delivery cells and the scientific and ethical issues that arise with the use of human stem cells.
Collapse
Affiliation(s)
- Carl T Henningson
- University of Pennsylvania School of Medicine, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
1189
|
Guo Y, Follo M, Geiger K, Lübbert M, Engelhardt M. Side-population cells from different precursor compartments. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2003; 12:71-82. [PMID: 12662438 DOI: 10.1089/152581603321210154] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The rapid efflux of the fluorescent DNA-binding dye Hoechst 33342 identifies a rare, so-called side population (SP), which rapidly expels the dye, can reconstitute the bone marrow (BM) of lethally irradiated mice, and has proven negative for most lineage markers including CD34. Because SP cells from human cell sources, such as mobilized peripheral blood [apheresis products (AP)], cord blood (CB), or BM have not been extensively characterized to date, we sought to analyze SP cells from various cell sources. We detected murine SP cells with a median frequency of 0.04% (n = 23) and a 52-fold colony-forming units (CFU) increase compared to unsorted cells (p = 0.028). The median frequency of human SP cells was 0.02% (n = 90), with highest numbers in donor AP, and lower in CB and BM. Human SP cells were mostly CD34(-) and lineage marker-negative. These showed no enrichment in CFU before expansion; however, they displayed a CFU increase after 5-7 days of cytokine-supported suspension culture (10.7-fold at day 5, 7.2-fold at day 7; n = 17) that was significant compared to both input (day 0) SP and to non-SP cells before and after expansion (p < 0.05). SP cells demonstrated a significant long-term culture-initiating cell (LTC-IC) increase of 167-fold (n = 17) as compared to non-SP cells (p = 0.002), with the highest numbers from AP specimens. We conclude that human primitive hematopoietic cells can be isolated via Hoechst staining and that SP cells of various human sources show substantial differences and represent a rare CD34(-) population with stem cell potential.
Collapse
Affiliation(s)
- Yalin Guo
- University of Freiburg Medical Center, Department of Hematology/Oncology, D-79106 Freiburg, Germany
| | | | | | | | | |
Collapse
|
1190
|
Lechner A, Habener JF. Stem/progenitor cells derived from adult tissues: potential for the treatment of diabetes mellitus. Am J Physiol Endocrinol Metab 2003; 284:E259-66. [PMID: 12531740 DOI: 10.1152/ajpendo.00393.2002] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In view of the recent success in pancreatic islet transplantation, interest in treating diabetes by the delivery of insulin-producing beta-cells has been renewed. Because differentiated pancreatic beta-cells cannot be expanded significantly in vitro, beta-cell stem or progenitor cells are seen as a potential source for the preparation of transplantable insulin-producing tissue. In addition to embryonic stem (ES) cells, several potential adult islet/beta-cell progenitors, derived from pancreas, liver, and bone marrow, are being studied. To date, none of the candidate cells has been fully characterized or is clinically applicable, but pancreatic physiology makes the existence of one or more types of adult islet stem cells very likely. It also seems possible that pluripotential stem cells, derived from the bone marrow, contribute to adult islet neogenesis. In future studies, more stringent criteria should be met to clonally define adult islet/beta-cell progenitor cells. If this can be achieved, the utilization of these cells for the generation of insulin-producing beta-cells in vitro seems to be feasible in the near future.
Collapse
Affiliation(s)
- Andreas Lechner
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | |
Collapse
|
1191
|
Hao HN, Zhao J, Thomas RL, Parker GC, Lyman WD. Fetal human hematopoietic stem cells can differentiate sequentially into neural stem cells and then astrocytes in vitro. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2003; 12:23-32. [PMID: 12662433 DOI: 10.1089/152581603321210109] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In some rodent models, there is evidence that hematopoietic stem cells (HSC) can differentiate into neural cells. However, it is not known whether humans share this potential, and, if so, what conditions are sufficient for this transdifferentiation to occur. We addressed this question by assessing the ability of fetal human liver CD34(+)/CD133(+)/CD3(-) hematopoietic stem cells to generate neural cells and astrocytes in culture. We cultured fetal liver-derived hematopoietic stem cells in human astrocyte culture-conditioned medium or using a method wherein growing human astrocytes were separated from cultured, nonadherent hematopoietic stem cells by a semipermeable membrane in a double-chamber co-culture system. Hematopoietic stem cell cultures were probed for neural progenitor cell marker expression (nestin and bone morphogenic protein-2 [BMP-2]) during growth in both culture conditions. RT-PCR, western blotting, and immunocytochemistry assays showed that cells cultured in either condition expressed nestin mRNA and protein and BMP-2 mRNA. HSC similarly cultured in nonconditioned medium or in the absence of astrocytes did not express either marker. Cells expressing these neural markers were transferred and cultured on poly-D-lysine-coated dishes with nonconditioned growth medium for further study. Immunocytochemistry demonstrated that these cells differentiated into astrocytes after 8 days in culture as indicated by their morphology and expression of the astrocytic markers glial fibrillary acidic protein (GFAP) and S100, as well as by their rate of proliferation, which was identical to that of freshly isolated fetal brain astrocytes. These findings demonstrate that neural precursor gene expression can be induced when human hematopoietic stem cells are exposed to a suitable microenvironment. Furthermore, the neural stem cells generated in this environment can then differentiate into astrocytes. Therefore, human hematopoietic stem cells may be an alternative resource for generation of neural stem cells for therapy of central nervous system defects resulting from disease or trauma.
Collapse
Affiliation(s)
- Hsiao-Nan Hao
- Department of Pediatrics, Wayne State University School of Medicine and Children's Research Center of Michigan, Detroit, MI 48201, USA.
| | | | | | | | | |
Collapse
|
1192
|
Abstract
The central tenet of stem cell biology is that within tissues there reside stem cells with the capacity for both self-renewal and terminal differentiation to the multiple lineages of that tissue. Over the last few years, numerous studies have challenged this paradigm by showing that tissue stem cells can differentiate to unexpected cell lineages, suggesting an enormous plasticity of differentiation. The hematopoietic stem cell, which resides within bone marrow and gives rise to all blood cells, has been the focal point of these efforts. However, recent studies have disputed the notion of hematopoietic stem cell plasticity. In truth, stem cell plasticity, strictly defined, has yet to be rigorously proven. Both animal models to carefully address outstanding issues and pilot clinical trials to explore the therapeutic potential will be key elements to advance science for the benefit of patients.
Collapse
Affiliation(s)
- Edwin M Horwitz
- Department of Hematology-Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA.
| |
Collapse
|
1193
|
|
1194
|
Abstract
Adult cardiac muscle is unable to repair itself following severe disease or injury. Because of this fundamental property of the myocardium, it was long believed that the adult myocardium is a postmitotic tissue. Yet, recent studies have indicated that new cardiac myocytes are generated throughout the life span of an adult and that extracardiac cells can contribute to the renewal of individual cells within the myocardium. In addition, investigations of the phenotypic capacity of adult stem cells have suggested that their potential is not solely restricted to the differentiated cell phenotypes of the source tissue. These observations have great implications for cardiac biology, as stem cells obtained from the bone marrow and other readily accessible adult tissues may serve as a source of replacement cardiac myocytes. In this review, we describe the evidence for these new findings and discuss their implications in context of the continuing controversy over stem cell plasticity.
Collapse
Affiliation(s)
- Leonard M Eisenberg
- Department of Cell Biology and Anatomy, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | |
Collapse
|
1195
|
|
1196
|
Tao H, Ma DD. Evidence for transdifferentiation of human bone marrow-derived stem cells: recent progress and controversies. Pathology 2003. [DOI: 10.1080/003130202201471] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
1197
|
Daley GQ, Goodell MA, Snyder EY. Realistic prospects for stem cell therapeutics. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2003; 2003:398-418. [PMID: 14633792 DOI: 10.1182/asheducation-2003.1.398] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Studies of the regenerating hematopoietic system have led to the definition of many of the fundamental principles of stem cell biology. Therapies based on a range of tissue stem cells have been widely touted as a new treatment modality, presaging an emerging new specialty called regenerative medicine that promises to harness stem cells from embryonic and somatic sources to provide replacement cell therapies for genetic, malignant, and degenerative conditions. Insights borne from stem cell biology also portend development of protein and small molecule therapeutics that act on endogenous stem cells to promote repair and regeneration. Much of the newfound enthusiasm for regenerative medicine stems from the hope that advances in the laboratory will be followed soon thereafter by breakthrough treatments in the clinic. But how does one sort through the hype to judge the true promise? Are stem cell biologists and the media building expectations that cannot be met? Which diseases can be treated, and when can we expect success? In this review, we outline the realms of investigation that are capturing the most attention, and consider the current state of scientific understanding and controversy regarding the properties of embryonic and somatic (adult) stem cells. Our objective is to provide a framework for appreciating the promise while at the same time understanding the challenges behind translating fundamental stem cell biology into novel clinical therapies.
Collapse
Affiliation(s)
- George Q Daley
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Children's Hospital, Cambridge, MA 02142, USA
| | | | | |
Collapse
|
1198
|
Schatteman GC, Awad O. Hemangioblasts, angioblasts, and adult endothelial cell progenitors. ACTA ACUST UNITED AC 2003; 276:13-21. [PMID: 14699630 DOI: 10.1002/ar.a.10131] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
After decades of speculation, proof of embryonic hemangioblasts finally emerged a few years ago. Surprisingly, at about the same time, evidence for adult hemangioblasts began to appear, and recent single-cell bone marrow transplants have confirmed their existence. Embryonic and adult hemangioblasts appear to share antigenic determinants, including CD34, ACC133, and VEGFR2, although their phenotype may be plastic. They also respond to similar factors, prominent among them vascular endothelial growth factor (VEGF). In the adult, hemangioblasts reside principally in the bone marrow, although they may subsequently leave that niche to reside in nonhematopoietic tissues. A number of studies indicate that these cells or their progeny may be a significant source of endothelial cells in adult pathologic and nonpathologic vascularization, and may participate in vascular repair. In addition to hemangioblasts, a more differentiated source of endothelial cell progenitors may be present in the blood, namely, monocytes or monocytic-like cells. The relative importance of the two cell types in vivo is not clear, though endothelial cells derived from the two sources may not be identical, and hemangioblasts seem to provide a stimulus for differentiation of the monocytes. Treatment with exogenous bone marrow-derived cells can promote neovascularization, accelerate restoration of blood flow to ischemic tissues, and improve cardiac function after infarct. Hence, there is great hope that either alone, in combination with angiogenic factors, or as gene therapy vectors, we can harness these cells to treat ischemic and vascular diseases in the relatively near future.
Collapse
Affiliation(s)
- Gina C Schatteman
- University of Iowa, Department of Exercise Science, Iowa City, Iowa 52242, USA.
| | | |
Collapse
|
1199
|
Abstract
Vascular smooth muscle cells (SMCs) originate from multiple types of progenitor cells. In the embryo, the most well studied SMC progenitor is the cardiac neural crest stem cell. Smooth muscle differentiation in the neural crest lineage is controlled by a combination of cell intrinsic factors, including Pax3, Tbx1, FoxC1, and serum response factor, interacting with various extrinsic factors in the local environment such as bone morphogenetic proteins (BMPs), Wnts, endothelin (ET)-1, and FGF8. Additional sources of multipotential cells that give rise to vascular SMCs in the embryo include proepicardial cells and possibly endothelial progenitor cells. In the adult, vascular SMCs must continually repair arterial injuries and maintain functional mass in response to changing demands upon the vessel wall. Recent evidence suggests that this is accomplished, in part, by recruiting multipotential vascular progenitors from bone marrow-derived stem cells as well as from less well defined sources within adult tissues themselves. This article will review our current understanding of the origins of vascular SMCs from multipotential stem and progenitor cells in developing as well as adult vasculature.
Collapse
Affiliation(s)
- Karen K Hirschi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
1200
|
Affiliation(s)
- Nadia Rosenthal
- European Molecular Biology Laboratory, Mouse Biology Programme, Monterotondo, Rome, Italy
| |
Collapse
|