Human Stem Cells for Tissue Repair
Louisa Wirthlin,David Hess,Ping Zhou,Jan Nolta
DOI: https://doi.org/10.1016/j.bbmt.2007.11.011
2008-01-01
Biology of Blood and Marrow Transplantation
Abstract:The primary application for hematopoietic stem cell transplantation has traditionally been to reconstitute blood cell lineages that had formed abnormally because of genetic mutations, or that had been eradicated to treat a disease such as leukemia. However, in recent years, compelling data has suggested that stem cells from the bone marrow might have far greater potential, and might be able to repair damaged or diseased tissues outside of the blood-forming compartment. Much attention has been paid to the concept of “stem cell plasticity,” and the hope that stem cells could be used to repair damaged tissues has generated immense excitement in many fields.Several types of adult human stem cells have been identified and studied in tissue repair applications. Most popular are hematopoietic stem cells, which can reconstitute the entire blood system, and also mesenchymal stem cells, which have the potential to form bone, cartilage, muscle, and fat. Our group studies tissue repair by both hematopoietic and mesenchymal stem cells, isolated from bone marrow, cord blood, and adipose tissue, in immune-deficient mouse models of injury and disease.Immune-deficient mouse models of tissue damage provide a system in which human stem cell migration to sites of damage and subsequent contribution to repair can be carefully evaluated. Popular strains have been discussed in a review paper [1Meyerrose T.E. Herrbrich P. Hess D.A. Nolta J.A. IImmune-deficient mouse models for analysis of human stem cells.Biotechniques. 2003; 35: 1262-1272PubMed Google Scholar], and newer strains such as NOD/SCID/IL2RG−/− (deficient in the common gamma chain of IL-2 and other lymphoid cytokines) are available. The beta-glucuronidase (GUSB)-deficient NOD/SCID/MPSVII mouse [2Hofling A.A. Vogler C. Creer M.H. Sands M.S. Engraftment of human CD34+ cells leads to widespread distribution of donor-derived cells and correction of tissue pathology in a novel murine xenotransplantation model of lysosomal storage disease.Blood. 2003; 101: 2054-2063Crossref PubMed Scopus (35) Google Scholar] allows the sensitive detection of individual unmarked donor cells (mouse, human, canine, etc.) in engrafted tissues by normal levels of the enzyme, without reliance on the continued expression of human cell surface markers or in situ hybridization. A simple substrate reaction is used on tissue slides. This mouse is highly useful for tissue repair studies, because the transplanted cells can be found in the tissues, by flow cytometry or on slides, without the use of antibody technology. This reduces background and errors in interpretation. Another benefit is that the transplanted cells do not need to be first manipulated to tag them with a vector, membrane dye, or other particle that would identify them later; they are detected based only on their innate expression of the enzyme beta-glucuronidase, absent in the mice.Immune-deficient mice provide easily accessible in vivo models for studying human stem cell migration, engraftment, and contribution to the repair of tissue damage. They can be used to test tissue repair strategies, because most of the cytokines, chemokines, and inflammatory modulators involved with stem cell recruitment and activation are conserved from mouse to humans. Hematopoietic stem cells from adult sources such as bone marrow (BM), mobilized peripheral blood (MPB), adipose tissue, and umbilical cord blood (UCB) have been shown, in our laboratory and others, to promote tissue repair in immune-deficient mouse models. Different populations of stem cells have been described to contribute to the regeneration of muscle [3Galmiche M.C. Koteliansky V.E. Briere J. Herve P. Charbord P. Stromal cells from human long-term marrow cultures are mesenchymal cells that differentiate following a vascular smooth muscle differentiation pathway.Blood. 1993; 82: 66-76Crossref PubMed Google Scholar, 4Williams J.T. Southerland S.S. Souza J. Calcutt A.F. Cartledge R.G. Cells isolated from adult human skeletal muscle capable of differentiating into multiple mesodermal phenotypes.Am Surg. 1999; 65: 22-26PubMed Google Scholar], liver [5Petersen B.E. Bowen W.C. Patrene K.D. et al.Bone marrow as a potential source of hepatic oval cells.Science. 1999; 284: 1168-1170Crossref PubMed Scopus (2177) Google Scholar, 6Lagasse E. Connors H. Al-Dhalimy M. et al.Purified hematopoietic stem cells can differentiate into hepatocytes in vivo.Nat Med. 2000; 6: 1229-1234Crossref PubMed Scopus (2127) Google Scholar, 7Theise N.D. Badve S. Saxena R. et al.Derivation of hepatocytes from bone marrow cells in mice after radiation-induced myeloablation.Hepatology. 2000; 31: 235-240Crossref PubMed Scopus (895) Google Scholar, 8Theise N.D. Nimmakayalu M. Gardner R. et al.Liver from bone marrow in humans.Hepatology. 2000; 32: 11-16Crossref PubMed Scopus (1153) Google Scholar], heart [9Jackson K.A. Majka S.M. Wang H. et al.Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells.J Clin Invest. 2001; 107: 1395-1402Crossref PubMed Scopus (1764) Google Scholar, 10Orlic D. Kajstura J. Chimenti S. et al.Bone marrow cells regenerate infarcted myocardium.Nature. 2001; 410: 701-705Crossref PubMed Scopus (4670) Google Scholar, 11Wu G.D. Nolta J.A. Jin Y.S. et al.Migration of mesenchymal stem cells to heart allografts during chronic rejection.Transplantation. 2003; 75: 679-685Crossref PubMed Scopus (163) Google Scholar, 12Zimmet J.M. Hare J.M. Emerging role for bone marrow derived mesenchymal stem cells in myocardial regenerative therapy.Basic Res Cardiol. 2005; 100: 471-481Crossref PubMed Scopus (126) Google Scholar], and to regenerate vasculature [13Crisa L. Cirulli V. Smith K.A. Ellisman M.H. Torbett B.E. Salomon D.R. Human cord blood progenitors sustain thymic T-cell development and a novel form of angiogenesis.Blood. 1999; 94: 3928-3940PubMed Google Scholar, 14Peichev M. Naiyer A.J. Pereira D. et al.Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors.Blood. 2000; 95: 952-958Crossref PubMed Google Scholar, 15Davidoff A.M. Ng C.Y. Brown P. et al.Bone marrow-derived cells contribute to tumor neovasculature and, when modified to express an angiogenesis inhibitor, can restrict tumor growth in mice.Clin Cancer Res. 2001; 7: 2870-2879PubMed Google Scholar], although the mechanisms by which they accomplish this are still not well understood. Stem cells are known, however, to secrete a variety of cytokines and growth factors that have both paracrine and autocrine activities. One theory of tissue repair and regeneration by adult stem cells is that the mechanism of action is based upon the innate functions of the stem cells: the injected stem cells home to the injured area, in particular to hypoxic and/or inflamed areas, and release trophic factors that hasten endogenous repair. These secreted bioactive factors suppress the local immune system, enhance angiogenesis, inhibit fibrosis and apoptosis, and stimulate recruitment, retention, mitosis, and differentiation of tissue-residing stem cells. These effects, which are referred to as trophic effects, are distinct from the direct differentiation of stem cells into the tissue to be regenerated.Data from our lab and others show that injected adipose and BM-derived MSC lodge in multiple tissues following various routes of administration into sublethally irradiated immune-deficient mice [16Meyerrose T.E. De Ugarte D.A. Hofling A.A. et al.In vivo distribution of human adipose-derived mesenchymal stem cells in novel xenotransplantation models.Stem Cells. 2007; 25: 220-227Crossref PubMed Scopus (154) Google Scholar]. Yet in models of acute local injury, MSC appear to preferentially home to, or accumulate in, the damaged tissue [11Wu G.D. Nolta J.A. Jin Y.S. et al.Migration of mesenchymal stem cells to heart allografts during chronic rejection.Transplantation. 2003; 75: 679-685Crossref PubMed Scopus (163) Google Scholar]. Our group has studied the trafficking of human BM and UCB-derived aldehyde dehydrogenase (ALDH)+, CD34+, and more highly purified subpopulations, and of BM and adipose tissue-derived mesenchymal stem cells to damaged heart, liver, muscle, pancreas, and to the ischemic limbs of immune-deficient mice. Cell trafficking and functional outcomes from intravenous injection of the stem cell populations versus nonstem cell controls (ie, ALDH hi versus ALDH lo sorted populations) were assessed. Trafficking was determined using fluorescent nanoparticle labeling and in vivo dynamic imaging. Endpoints were determined by FACS analysis and by tissue staining using the NOD/SCID/MPSVII mouse, where unmarked donor-derived cells can be easily identified using a simple enzymatic substrate reaction. Within each study, mice were assessed in groups large enough to achieve statistical significance. The groups that had received the stem cell populations demonstrated improved regional blood flow and tissue function, compared with the nonstem cell-injected control groups, at various time points after transplantation. However, very few donor marker-positive cells were found incorporated into vascular structures or in the repaired tissue. Tissue staining in the damaged regions revealed that the transplanted human cells had undergone very few cell divisions after homing to the area of hypoxia or inflammation.The paucity of human cells remaining in the tissue after repair suggest that the tissue improvements that were observed were not the result of generation of transplanted cell-derived endothelial cells, muscle, liver, or beta cells, but suggest that cytokines secreted from transplanted cells potentiated angiogenic activity and tissue repair from endogenous murine cells. Although the combinations of factors involved are still being established, our data indicate that adult human stem cells do not become a significant part of the damaged tissue, but rapidly home to and persist only temporarily at a site of hypoxia or inflammation to exert significant trophic effects on tissue repair.It is important to note that, with over 400 mice now analyzed in these tissue repair experiments, no adult human stem cell-derived tumors have ever been observed. The mouse strains used have no capacity to reject the human cells, and thus were a valid model for testing the tumorigenesis abilities of adult stem cells (Dao, Bauer et al, submitted). Both MSC and HSC have been tested extensively without a single occurrence of a stem cell-derived tumor. However, the immune-compromised mice were susceptible to human leukemias, caused by preexisting leukemic or preleukemic cells in the transplanted cell populations. The spontaneous emergence of the leukemias would likely not have occurred in the presence of functioning NK cells in the mice.In contrast to the hematopoietic and mesenchymal cells that have been isolated using standard techniques, novel populations of cells have been identified by investigators who “think outside the box,” as exemplified by exciting work done in the laboratory headed by Mariusz Ratajczak. In addition to cytokines, Ratajczak et al. [17Ratajczak J. Wysoczynski M. Hayek T. Janowska-Wieczorek A. Rataczak H.Z. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication.Leukemia. 2006; 20: 1487-1495Crossref PubMed Scopus (1039) Google Scholar] have shown that microvesicles (MV) can play a role in cell-cell interactions. MV are tiny, between 100 nm to 1 μm, are shed by cells, and can be vehicles for transferring mRNA and can activate various signaling pathways. Ratajczak's group found that microparticles (MP), MV shed from platelets, can be used to heighten the rapidity of HSC engraftment as well as enhance the level of engraftment in lethally irradiated mice [18Janowska-Wieczorek A. Majka M. Kijowski J. et al.Platelet-derived microparticles bind to hematopoietic stem/progenitor cells and enhance their engraftment.Blood. 2001; 98: 3143-3149Crossref PubMed Scopus (280) Google Scholar]. Furthermore, when MV from human embryonic stem cells were added to HSCs, they increased the pluripotency of the cells as well as upregulating expression of Oct-4, Rex-1, and Nanog, cellular markers of pluripotency [19Ratajczak J. Miekus K. Kucia M. et al.Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery.Leukemia. 2006; 20: 847-856Crossref PubMed Scopus (1140) Google Scholar].Mitochondria have also been shown to play a role in cellular repair. It has been shown that MSC, as well as other cell types, are able to transfer their mitochondria to other cells to rescue the cells from death [20Spees J.L. Olson S.D. Whitney M.J. Prockop D.J. Mitochondrial transfer between cells can rescue aerobic respiration.Proc Natl Acad Sci USA. 2006; 103: 1283-1288Crossref PubMed Scopus (660) Google Scholar, 21Redmond Jr., D.E. Bjugstad K.B. et al.Behavioral improvement in a primate Parkinson's model is associated with multiple homeostatic effects of human neural stem cells.Proc Natl Acad Sci USA. 2007; 104: 12175-12180Crossref PubMed Scopus (283) Google Scholar]. In this study, MSC were added to cells treated with ethidium bromide and were able to restore the cell's aerobic respiration as well as their ability to expand. The restoration of cellular functions was attributed to active mitochondrial transfer. Coculture with mitochondria alone did not result in restored function, nor was there nuclear DNA present, indicating that the effects were not because of cell fusion.Adult stem cells cannot create an entire tissue, as can embryonic cells. However, under the right conditions, adult stem cells can act as the “paramedics of the body,” rushing to areas of hypoxia and rapidly causing revascularization and releasing trophic factors to aide in tissue repair, early after tissue injury. In a seminal study by Evan Snyder's group, using a primate Parkinsonian model, human Neural Stem Cells (hNSC) were injected unilaterally and were found to traverse to the other, damaged side of the brain. The stem cells were found to both elicit endogenous repair in the damaged host neurons as well as to differentiate into mature neurons [20Spees J.L. Olson S.D. Whitney M.J. Prockop D.J. Mitochondrial transfer between cells can rescue aerobic respiration.Proc Natl Acad Sci USA. 2006; 103: 1283-1288Crossref PubMed Scopus (660) Google Scholar]. The active travel of the hNSC is further support for the active rescue nature of adult stem and progenitor cells.In addition to the cell types described above, human embryonic stem cells are being explored, with the hope that they can repair all tissues and will more robustly “become” the tissue, as has been proven for murine embryonic stem cells. Because these cells were first reported in 1998, the field is new, but extremely promising. Better animal transplantation models are needed, and more models of disease and tissue injury in animals that are immune deficient, so can accept the human cell graft, will need to be developed to further strengthen these efforts. The combination of embryonic and adult stem cell treatments could also prove beneficial for tissue repair, perhaps providing long- and short-term engraftment and support systems for the injured tissues. More experiments need to be done to further investigate the many prospects of this burgeoning field, to bring the most effective stem cell-based therapies to our many community members who urgently need them. The primary application for hematopoietic stem cell transplantation has traditionally been to reconstitute blood cell lineages that had formed abnormally because of genetic mutations, or that had been eradicated to treat a disease such as leukemia. However, in recent years, compelling data has suggested that stem cells from the bone marrow might have far greater potential, and might be able to repair damaged or diseased tissues outside of the blood-forming compartment. Much attention has been paid to the concept of “stem cell plasticity,” and the hope that stem cells could be used to repair damaged tissues has generated immense excitement in many fields. Several types of adult human stem cells have been identified and studied in tissue repair applications. Most popular are hematopoietic stem cells, which can reconstitute the entire blood system, and also mesenchymal stem cells, which have the potential to form bone, cartilage, muscle, and fat. Our group studies tissue repair by both hematopoietic and mesenchymal stem cells, isolated from bone marrow, cord blood, and adipose tissue, in immune-deficient mouse models of injury and disease. Immune-deficient mouse models of tissue damage provide a system in which human stem cell migration to sites of damage and subsequent contribution to repair can be carefully evaluated. Popular strains have been discussed in a review paper [1Meyerrose T.E. Herrbrich P. Hess D.A. Nolta J.A. IImmune-deficient mouse models for analysis of human stem cells.Biotechniques. 2003; 35: 1262-1272PubMed Google Scholar], and newer strains such as NOD/SCID/IL2RG−/− (deficient in the common gamma chain of IL-2 and other lymphoid cytokines) are available. The beta-glucuronidase (GUSB)-deficient NOD/SCID/MPSVII mouse [2Hofling A.A. Vogler C. Creer M.H. Sands M.S. Engraftment of human CD34+ cells leads to widespread distribution of donor-derived cells and correction of tissue pathology in a novel murine xenotransplantation model of lysosomal storage disease.Blood. 2003; 101: 2054-2063Crossref PubMed Scopus (35) Google Scholar] allows the sensitive detection of individual unmarked donor cells (mouse, human, canine, etc.) in engrafted tissues by normal levels of the enzyme, without reliance on the continued expression of human cell surface markers or in situ hybridization. A simple substrate reaction is used on tissue slides. This mouse is highly useful for tissue repair studies, because the transplanted cells can be found in the tissues, by flow cytometry or on slides, without the use of antibody technology. This reduces background and errors in interpretation. Another benefit is that the transplanted cells do not need to be first manipulated to tag them with a vector, membrane dye, or other particle that would identify them later; they are detected based only on their innate expression of the enzyme beta-glucuronidase, absent in the mice. Immune-deficient mice provide easily accessible in vivo models for studying human stem cell migration, engraftment, and contribution to the repair of tissue damage. They can be used to test tissue repair strategies, because most of the cytokines, chemokines, and inflammatory modulators involved with stem cell recruitment and activation are conserved from mouse to humans. Hematopoietic stem cells from adult sources such as bone marrow (BM), mobilized peripheral blood (MPB), adipose tissue, and umbilical cord blood (UCB) have been shown, in our laboratory and others, to promote tissue repair in immune-deficient mouse models. Different populations of stem cells have been described to contribute to the regeneration of muscle [3Galmiche M.C. Koteliansky V.E. Briere J. Herve P. Charbord P. Stromal cells from human long-term marrow cultures are mesenchymal cells that differentiate following a vascular smooth muscle differentiation pathway.Blood. 1993; 82: 66-76Crossref PubMed Google Scholar, 4Williams J.T. Southerland S.S. Souza J. Calcutt A.F. Cartledge R.G. Cells isolated from adult human skeletal muscle capable of differentiating into multiple mesodermal phenotypes.Am Surg. 1999; 65: 22-26PubMed Google Scholar], liver [5Petersen B.E. Bowen W.C. Patrene K.D. et al.Bone marrow as a potential source of hepatic oval cells.Science. 1999; 284: 1168-1170Crossref PubMed Scopus (2177) Google Scholar, 6Lagasse E. Connors H. Al-Dhalimy M. et al.Purified hematopoietic stem cells can differentiate into hepatocytes in vivo.Nat Med. 2000; 6: 1229-1234Crossref PubMed Scopus (2127) Google Scholar, 7Theise N.D. Badve S. Saxena R. et al.Derivation of hepatocytes from bone marrow cells in mice after radiation-induced myeloablation.Hepatology. 2000; 31: 235-240Crossref PubMed Scopus (895) Google Scholar, 8Theise N.D. Nimmakayalu M. Gardner R. et al.Liver from bone marrow in humans.Hepatology. 2000; 32: 11-16Crossref PubMed Scopus (1153) Google Scholar], heart [9Jackson K.A. Majka S.M. Wang H. et al.Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells.J Clin Invest. 2001; 107: 1395-1402Crossref PubMed Scopus (1764) Google Scholar, 10Orlic D. Kajstura J. Chimenti S. et al.Bone marrow cells regenerate infarcted myocardium.Nature. 2001; 410: 701-705Crossref PubMed Scopus (4670) Google Scholar, 11Wu G.D. Nolta J.A. Jin Y.S. et al.Migration of mesenchymal stem cells to heart allografts during chronic rejection.Transplantation. 2003; 75: 679-685Crossref PubMed Scopus (163) Google Scholar, 12Zimmet J.M. Hare J.M. Emerging role for bone marrow derived mesenchymal stem cells in myocardial regenerative therapy.Basic Res Cardiol. 2005; 100: 471-481Crossref PubMed Scopus (126) Google Scholar], and to regenerate vasculature [13Crisa L. Cirulli V. Smith K.A. Ellisman M.H. Torbett B.E. Salomon D.R. Human cord blood progenitors sustain thymic T-cell development and a novel form of angiogenesis.Blood. 1999; 94: 3928-3940PubMed Google Scholar, 14Peichev M. Naiyer A.J. Pereira D. et al.Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors.Blood. 2000; 95: 952-958Crossref PubMed Google Scholar, 15Davidoff A.M. Ng C.Y. Brown P. et al.Bone marrow-derived cells contribute to tumor neovasculature and, when modified to express an angiogenesis inhibitor, can restrict tumor growth in mice.Clin Cancer Res. 2001; 7: 2870-2879PubMed Google Scholar], although the mechanisms by which they accomplish this are still not well understood. Stem cells are known, however, to secrete a variety of cytokines and growth factors that have both paracrine and autocrine activities. One theory of tissue repair and regeneration by adult stem cells is that the mechanism of action is based upon the innate functions of the stem cells: the injected stem cells home to the injured area, in particular to hypoxic and/or inflamed areas, and release trophic factors that hasten endogenous repair. These secreted bioactive factors suppress the local immune system, enhance angiogenesis, inhibit fibrosis and apoptosis, and stimulate recruitment, retention, mitosis, and differentiation of tissue-residing stem cells. These effects, which are referred to as trophic effects, are distinct from the direct differentiation of stem cells into the tissue to be regenerated. Data from our lab and others show that injected adipose and BM-derived MSC lodge in multiple tissues following various routes of administration into sublethally irradiated immune-deficient mice [16Meyerrose T.E. De Ugarte D.A. Hofling A.A. et al.In vivo distribution of human adipose-derived mesenchymal stem cells in novel xenotransplantation models.Stem Cells. 2007; 25: 220-227Crossref PubMed Scopus (154) Google Scholar]. Yet in models of acute local injury, MSC appear to preferentially home to, or accumulate in, the damaged tissue [11Wu G.D. Nolta J.A. Jin Y.S. et al.Migration of mesenchymal stem cells to heart allografts during chronic rejection.Transplantation. 2003; 75: 679-685Crossref PubMed Scopus (163) Google Scholar]. Our group has studied the trafficking of human BM and UCB-derived aldehyde dehydrogenase (ALDH)+, CD34+, and more highly purified subpopulations, and of BM and adipose tissue-derived mesenchymal stem cells to damaged heart, liver, muscle, pancreas, and to the ischemic limbs of immune-deficient mice. Cell trafficking and functional outcomes from intravenous injection of the stem cell populations versus nonstem cell controls (ie, ALDH hi versus ALDH lo sorted populations) were assessed. Trafficking was determined using fluorescent nanoparticle labeling and in vivo dynamic imaging. Endpoints were determined by FACS analysis and by tissue staining using the NOD/SCID/MPSVII mouse, where unmarked donor-derived cells can be easily identified using a simple enzymatic substrate reaction. Within each study, mice were assessed in groups large enough to achieve statistical significance. The groups that had received the stem cell populations demonstrated improved regional blood flow and tissue function, compared with the nonstem cell-injected control groups, at various time points after transplantation. However, very few donor marker-positive cells were found incorporated into vascular structures or in the repaired tissue. Tissue staining in the damaged regions revealed that the transplanted human cells had undergone very few cell divisions after homing to the area of hypoxia or inflammation. The paucity of human cells remaining in the tissue after repair suggest that the tissue improvements that were observed were not the result of generation of transplanted cell-derived endothelial cells, muscle, liver, or beta cells, but suggest that cytokines secreted from transplanted cells potentiated angiogenic activity and tissue repair from endogenous murine cells. Although the combinations of factors involved are still being established, our data indicate that adult human stem cells do not become a significant part of the damaged tissue, but rapidly home to and persist only temporarily at a site of hypoxia or inflammation to exert significant trophic effects on tissue repair. It is important to note that, with over 400 mice now analyzed in these tissue repair experiments, no adult human stem cell-derived tumors have ever been observed. The mouse strains used have no capacity to reject the human cells, and thus were a valid model for testing the tumorigenesis abilities of adult stem cells (Dao, Bauer et al, submitted). Both MSC and HSC have been tested extensively without a single occurrence of a stem cell-derived tumor. However, the immune-compromised mice were susceptible to human leukemias, caused by preexisting leukemic or preleukemic cells in the transplanted cell populations. The spontaneous emergence of the leukemias would likely not have occurred in the presence of functioning NK cells in the mice. In contrast to the hematopoietic and mesenchymal cells that have been isolated using standard techniques, novel populations of cells have been identified by investigators who “think outside the box,” as exemplified by exciting work done in the laboratory headed by Mariusz Ratajczak. In addition to cytokines, Ratajczak et al. [17Ratajczak J. Wysoczynski M. Hayek T. Janowska-Wieczorek A. Rataczak H.Z. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication.Leukemia. 2006; 20: 1487-1495Crossref PubMed Scopus (1039) Google Scholar] have shown that microvesicles (MV) can play a role in cell-cell interactions. MV are tiny, between 100 nm to 1 μm, are shed by cells, and can be vehicles for transferring mRNA and can activate various signaling pathways. Ratajczak's group found that microparticles (MP), MV shed from platelets, can be used to heighten the rapidity of HSC engraftment as well as enhance the level of engraftment in lethally irradiated mice [18Janowska-Wieczorek A. Majka M. Kijowski J. et al.Platelet-derived microparticles bind to hematopoietic stem/progenitor cells and enhance their engraftment.Blood. 2001; 98: 3143-3149Crossref PubMed Scopus (280) Google Scholar]. Furthermore, when MV from human embryonic stem cells were added to HSCs, they increased the pluripotency of the cells as well as upregulating expression of Oct-4, Rex-1, and Nanog, cellular markers of pluripotency [19Ratajczak J. Miekus K. Kucia M. et al.Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery.Leukemia. 2006; 20: 847-856Crossref PubMed Scopus (1140) Google Scholar]. Mitochondria have also been shown to play a role in cellular repair. It has been shown that MSC, as well as other cell types, are able to transfer their mitochondria to other cells to rescue the cells from death [20Spees J.L. Olson S.D. Whitney M.J. Prockop D.J. Mitochondrial transfer between cells can rescue aerobic respiration.Proc Natl Acad Sci USA. 2006; 103: 1283-1288Crossref PubMed Scopus (660) Google Scholar, 21Redmond Jr., D.E. Bjugstad K.B. et al.Behavioral improvement in a primate Parkinson's model is associated with multiple homeostatic effects of human neural stem cells.Proc Natl Acad Sci USA. 2007; 104: 12175-12180Crossref PubMed Scopus (283) Google Scholar]. In this study, MSC were added to cells treated with ethidium bromide and were able to restore the cell's aerobic respiration as well as their ability to expand. The restoration of cellular functions was attributed to active mitochondrial transfer. Coculture with mitochondria alone did not result in restored function, nor was there nuclear DNA present, indicating that the effects were not because of cell fusion. Adult stem cells cannot create an entire tissue, as can embryonic cells. However, under the right conditions, adult stem cells can act as the “paramedics of the body,” rushing to areas of hypoxia and rapidly causing revascularization and releasing trophic factors to aide in tissue repair, early after tissue injury. In a seminal study by Evan Snyder's group, using a primate Parkinsonian model, human Neural Stem Cells (hNSC) were injected unilaterally and were found to traverse to the other, damaged side of the brain. The stem cells were found to both elicit endogenous repair in the damaged host neurons as well as to differentiate into mature neurons [20Spees J.L. Olson S.D. Whitney M.J. Prockop D.J. Mitochondrial transfer between cells can rescue aerobic respiration.Proc Natl Acad Sci USA. 2006; 103: 1283-1288Crossref PubMed Scopus (660) Google Scholar]. The active travel of the hNSC is further support for the active rescue nature of adult stem and progenitor cells. In addition to the cell types described above, human embryonic stem cells are being explored, with the hope that they can repair all tissues and will more robustly “become” the tissue, as has been proven for murine embryonic stem cells. Because these cells were first reported in 1998, the field is new, but extremely promising. Better animal transplantation models are needed, and more models of disease and tissue injury in animals that are immune deficient, so can accept the human cell graft, will need to be developed to further strengthen these efforts. The combination of embryonic and adult stem cell treatments could also prove beneficial for tissue repair, perhaps providing long- and short-term engraftment and support systems for the injured tissues. More experiments need to be done to further investigate the many prospects of this burgeoning field, to bring the most effective stem cell-based therapies to our many community members who urgently need them.
What problem does this paper attempt to address?
-
Reply to Letter from Drs. Haider and Ashraf
Boon Chin Heng,Eugene K.W. Sim,Tong Cao
DOI: https://doi.org/10.1097/01.mat.0000160799.12761.7c
2005-01-01
ASAIO Journal
Abstract:Dear Editor, It was proposed by Haider and Ashraf1 that cytokine and growth factor mediated mobilization of endogenous mesenchymal stem cells from their bone marrow niches for homing into the infarcted myocardium could be a better therapeutic strategy than the explantation and subsequent injection of these cells (either autologous or from a donated allogenic source) directly into the damaged heart.2 Besides being less invasive to the patient, it was argued that this approach would obviate the need for ex vivo processing of the putative stem cells, which could in turn reduce the risk of nonspecific differentiation into undesired lineages at the transplantation site.3–5 Nevertheless, such an approach could be severely hampered by the age related decline in the regenerative capacity of the putative mesenchymal stem cell population within the bone marrow, as has been demonstrated by a number of studies.6,7 It must be remembered that the overwhelming majority of myocardial infarct cases occur in the older segment of the population.8 Quarto and colleagues6 demonstrated in a rat model that there was an age associated decline in the ability of marrow stromal cells to repair bone, which could be the underlying cause of age related osteodegenerative diseases. Subsequently, D’Ippolito and colleagues7 reported a similar finding with the human model. This would be expected because, with increasing age, the putative mesenchymal stem cell population within the bone marrow would have gone through more rounds of cell division, in addition to having been exposed to free radicals and metabolic toxins over a longer lifespan. Hence it is no surprise that there is a gradual decline in their regenerative capacity with increasing age. Although there have been no studies to date that have conclusively demonstrated an age associated decline in the capacity of bone marrow cells to repair the infarcted heart, we could make such a conjecture based upon extrapolation from the bone repair model.6,7 The obvious solution to overcome this age related decline in regenerative capacity would be to look at donated allogenic sources of stem cells for myocardial repair. In this regard, human embryonic stem (hES) cells or their differentiated progenies would hold much promise because these potentially represent an abundant and readily available source of transplantable cells. Recently, Zandstra and colleagues9 successfully developed a scalable, robust, and reproducible process for obtaining purified cardiomyocytes from hES cells. Nevertheless, the immunologic barrier10 probably represents the most formidable challenge to the allogenic transplantation of hES cells or their differentiated progenies into the infarcted myocardium. It is hoped that this could somehow be overcome in the near future. Boon Chin Heng* Eugene Kwang-Wei Sim† Tong Cao* *Stem Cell Laboratory, Faculty of Dentistry, †Department of Surgery, Faculty of Medicine National University of Singapore.
-
Adult Human Stem Cells Exert Therapeutic Effects to Repair Damaged Tissues in Xenograft Systems Through Secretion of Trophic Factors Rather Than Direct Incorporation and Expansion.
Claus Sondergaard,David Hess,Ivana Rosova,Sarah Hohm,Louisa Wirthlin,Ryan Lahey,Matt Lindsey,Jon Walker,Gerhard Bauer,Ping Zhou,Jan Nolta
DOI: https://doi.org/10.1182/blood.v110.11.3693.3693
IF: 20.3
2007-01-01
Blood
Abstract:Human stem cells from adult sources have been shown, in our laboratory and others, to promote tissue repair. Different populations of stem cells have been shown to contribute to the regeneration of muscle, liver, heart, and vasculature, although the mechanisms by which they accomplish this are still not well understood. Stem cells are known, however, to secrete a variety of factors that have both paracrine and autocrine activities. One theory of tissue repair and regeneration by adult stem cells is that they home to hypoxic and/or inflamed areas, and release trophic factors that hasten endogenous repair. These secreted bioactive factors suppress the local immune system, enhance angiogenesis, inhibit fibrosis and apoptosis, and stimulate recruitment, retention, mitosis and differentiation of tissue-residing stem cells. These effects, which are referred to as trophic effects, are distinct from the direct differentiation of stem cells into the tissue to be regenerated. We tested human umbilical cord blood (UCB) derived CD34+, ALDHhiLin− and ALDHloLin− cells following transplantation to NOD/SCID or NOD/SCID/B2M null mice with experimentally induced acute myocardial infarction (AMI). We used combined nanoparticle labeling and whole organ fluorescent imaging to detect homing of the cells to multiple organs 48 hours post transplantation. Long term engraftment and the regenerative effect of cell treatment was assessed four weeks post transplant. There was superior homing of ALDHhiLin− cells to the site of injury, as compared to CD34+ or ALDHloLin− cells at 48 hours post transplantation. At four weeks post transplantation, ALDHhiLin− cells engrafted multiple organs, including the heart, liver and kidney, at higher frequencies than ALDHloLin− cells. We found no donor derived cardiomyocytes and only few endothelial cells of donor origin. However, there was a significant increase in the density of large caliber vessels in the central infarct zone of ALDHhiLin− cell-transplanted mice, as compared to PBS and ALDHhiLin− cell treated groups. Tissue staining in the damaged regions revealed that the transplanted human cells had undergone very few cell divisions after homing to the area of hypoxia or inflammation. It is important to note that, with over 300 mice now analyzed in tissue repair experiments in our group, no adult human stem cell-derived tumors have ever been observed, even though the mouse strains used have no capacity to reject the human cells. The paucity of human cells remaining in the tissue after repair suggest that the tissue improvements that were observed were not the result of generation of transplanted cell-derived endothelial cells or cardiac tissue, but suggest that cytokines secreted from transplanted cells potentiated angiogenic activity and tissue repair by the endogenous murine cells. Our data indicate that adult human stem cells do not become a significant part of the damaged tissue, but rapidly home to and persist only temporarily at a site of hypoxia or inflammation to exert significant trophic effects on tissue repair, and to enhance recovery of the vasculature.
-
Committed-differentiation of Transplanted Bone Derived Mesenchymal Stem Cells and Their Potential to Amend Damaged Liver Functions: in Vivo Experiment with Mice
Hai-hua Lu,Gao-jun Teng,Sheng-hong Ju,Jun-hui Sun,Ai-mei Li,Ai-feng Zhang
DOI: https://doi.org/10.3760/j:issn:0376-2491.2007.04.003
2007-01-01
Abstract:OBJECTIVE:To observe whether bone mesenchymal stem cells (MSCs) have the potential to transdifferentiate into functional hepatocyte-like cells in the special "niche" as well as the therapeutic feasibility to repair damaged liver in mice.METHODS:20 nude mice were randomly divided into four groups (n = 5 in each group): Group A: 1.0 ml/kg of carbon tetrachloride (CCl(4)) (dissolved in olive oil by ratio of 1:1) was injected into the peritoneum of mice twice a week for 5 weeks. GFP-positive MSCs (1 x 10(6) cells) were injected into the caudal tail vein 1 week after the first dose of CCl(4); Group B: treated with CCl(4) as in A, but received the same volume of saline; Group C: normal nude mice with GFP-positive MSCs Transplanted in the same way as in A. Group D: normal controls. 4 weeks after the cell transplantation, all animal subjects were killed. Liver function tests (LFT), histology of HE and Masson staining as well as double immunofluorescent staining for GFP and albumin were studied in all groups.RESULTS:The hepatic fibrosis in group A & B confirmed the success of model for liver damage and there was no marked difference in the percent of the area occupied by collagen between two groups (10.5 +/- 1.5 vs 12.7 +/- 1.6, t = -2.238, P > 0.05). GFP-positive MSCs were mainly observed around portal area or interspace of lobules in group A. Some of GFP-positive cells also express albumin (35% +/- 7%). While in group B, C or D, there is no such findings. The level of serum albumin in group A was higher than that in group B (24.4 g/L +/- 3.3 g/L vs 18.6 g/L +/- 2.9 g/L, P < 0.05) while the level of ALT was also different between two groups (121 U/L +/- 21 U/L vs 192 U/L +/- 29 U/L, P < 0.05).CONCLUSION:The stimulus of persistent liver damage might enhances the migration of MSCs to the liver, in which some of the MSCs have the potential to transdifferentiate into hepatocyte-like cells. Transplantation of MSCs might amend the damaged tissue of host liver to a certain extent.
-
Models of Stem Cell Transplantation for the Repair of Non-Hematopoietic Tissues
G. Annett,L. Wirthlin,K. Pepper,C. Sondergaard,J. McGee,M. Lindsey,P. Zhou,G. Bauer,J. Nolta
DOI: https://doi.org/10.1016/j.bbmt.2009.12.307
2010-01-01
Biology of Blood and Marrow Transplantation
Abstract:Human stem cells from adult sources have been shown, in our laboratory and others, to promote the repair of damaged tissues. Different populations of stem cells contribute to the regeneration of muscle, neural tissue, liver, heart, and vasculature, although the mechanisms by which they accomplish this are still not well understood. We and others have shown that stem cells home to hypoxic and/or inflamed areas, and release bioactive factors that can suppress the local immune system, enhance angiogenesis, inhibit fibrosis and apoptosis, and stimulate recruitment, retention, mitosis and differentiation of endogenous tissue-residing stem cells. These trophic effects are distinct from the direct differentiation of stem cells into the tissue to be regenerated. To actually rebuild a non-hematopoietic tissue, the differentiated progeny of embryonic or induced pluripotent stem cells will be required. We have focused on improving rodent models in which to examine human stem cell-mediated disease correction and tissue repair, focusing primarily on liver regeneration and hypoxic tissue models of peripheral vascular disease and cardiac ischemia. Most recently we are studying mesenchymal stem cell–mediated repair of neural damage. We are interested in the mechanisms by which stem cells of different types and origins home preferentially into areas of tissue damage, and we seek to improve the robustness. To track cells into the damaged tissues in vivo, we have labeled them with fluorophore – conjugated iron oxide nanoparticles and have used novel mouse models that facilitate human cell detection. We have also used 19F magnetic resonance imaging for stem cell tracking with multiple unique perfluorocarbon nanobeacons, human/murine centromeric FISH, immunohistochemistry and quantitative PCR. Using these technologies, we have shown that human stem cells migrate from the bloodstream randomly and in moderate numbers throughout all tissues examined in cases of chronic disease or following sublethal irradiation, but that in instances of acute damage, the homing is more vigorous and specific to the site of damage. Pre-culture in hypoxia dramatically alters the phenotype and migratory characteristics of human mesenchymal stem cells. We are applying this knowledge to tissue repair strategies, to allow enhanced numbers of stem cells to migrate to the areas of hypoxic damage, to exert trophic effects that initiate revascularization and cascades of repair.
-
Bone Marrow Mesenchymal Cells: How Do They Contribute to Tissue Repair and Are They Really Stem Cells?
Yasumasa Kuroda,Masaaki Kitada,Shohei Wakao,Mari Dezawa
DOI: https://doi.org/10.1007/s00005-011-0139-9
2011-07-26
Archivum Immunologiae et Therapiae Experimentalis
Abstract:Adult stem cells typically generate the cell types of the tissue in which they reside, and thus the range of their differentiation is considered limited. Bone marrow mesenchymal stem cells (MSCs) are different from other somatic stem cells in that they differentiate not only into the same mesodermal-lineage such as bone, cartilage, and adipocytes but also into other lineages of ectodermal and endodermal cells. Thus, MSCs are a unique type of adult stem cells. In addition, MSCs home to damaged sites, differentiate into cells specific to the tissue and contribute to tissue repair. Therefore, application of MSCs in the treatment of various diseases, including liver dysfunction, myocardial infarction, and central nervous system repair, has been initiated. Because MSCs are generally harvested as adherent cells from bone marrow aspirates, however, they comprise heterogeneous cell populations and their wide-ranging differentiation ability and repair functions are not yet clear. Recent evidence suggests that a very small subpopulation of cells that assume a repair function with the ability to differentiate into trilineage cells resides among human MSCs and effective utilization of such cells is expected to improve the repair effect of MSCs. This review summarizes recent advances in the clarification of MSC properties and discusses future perspectives.
immunology
-
Bone-Marrow-Derived Mesenchymal Stem Cells for Organ Repair
Ming Li,Susumu Ikehara
DOI: https://doi.org/10.1155/2013/132642
2013-01-01
Stem Cells International
Abstract:Mesenchymal stem cells (MSCs) are prototypical adult stem cells with the capacity for self-renewal and differentiation with a broad tissue distribution. MSCs not only differentiate into types of cells of mesodermal lineage but also into endodermal and ectodermal lineages such as bone, fat, cartilage and cardiomyocytes, endothelial cells, lung epithelial cells, hepatocytes, neurons, and pancreatic islets. MSCs have been identified as an adherent, fibroblast-like population and can be isolated from different adult tissues, including bone marrow (BM), umbilical cord, skeletal muscle, and adipose tissue. MSCs secrete factors, including IL-6, M-CSF, IL-10, HGF, and PGE2, that promote tissue repair, stimulate proliferation and differentiation of endogenous tissue progenitors, and decrease inflammatory and immune reactions. In this paper, we focus on the role of BM-derived MSCs in organ repair.
cell & tissue engineering
-
Mesenchymal Stem Cell Migration and Tissue Repair
Xiaorong Fu,Ge Liu,Alexander Halim,Yang Ju,Qing Luo,Guanbin Song,Fu,Liu,Halim,Ju,Luo,Song
DOI: https://doi.org/10.3390/cells8080784
IF: 6
2019-07-28
Cells
Abstract:Mesenchymal stem cells (MSCs) are multilineage cells with the ability to self-renew and differentiate into a variety of cell types, which play key roles in tissue healing and regenerative medicine. Bone marrow-derived mesenchymal stem cells (BMSCs) are the most frequently used stem cells in cell therapy and tissue engineering. However, it is prerequisite for BMSCs to mobilize from bone marrow and migrate into injured tissues during the healing process, through peripheral circulation. The migration of BMSCs is regulated by mechanical and chemical factors in this trafficking process. In this paper, we review the effects of several main regulatory factors on BMSC migration and its underlying mechanism; discuss two critical roles of BMSCs—namely, directed differentiation and the paracrine function—in tissue repair; and provide insight into the relationship between BMSC migration and tissue repair, which may provide a better guide for clinical applications in tissue repair through the efficient regulation of BMSC migration.
cell biology
-
Early Homing Behavior of Stro-1− Mesenchyme-Like Cells Derived from Human Embryonic Stem Cells in an Immunocompetent Xenogeneic Animal Model
Zhongyuan Su,Rongrong Wu,Zhou Tan,Ying Li,Liangbiao Chen,Jingfeng Luo,Ming Zhang
DOI: https://doi.org/10.1016/j.bbrc.2010.03.033
IF: 3.1
2010-01-01
Biochemical and Biophysical Research Communications
Abstract:Mesenchymal stem cells (MSCs) have been induced to differentiate successfully from human embryonic stem cells (hES-MSCs), which could serve as an in vitro source of MSCs. However, the homing behaviors of such cells and their potential utility for liver regeneration in vivo have not been reported. We investigated factors that influenced early homing and the hepatic-directed differentiation potency of hES-MSCs in a mouse model of acute liver injury. The hES-MSCs could be detected 36 h after cell infusion and this was unaffected by the number of cell passages in culture. Pretreatment of hES-MSCs with TNF-alpha resulted in higher rates of homing of these cells to the injured liver. Interestingly most of the cells homing at an early stage expressed alpha-fetoprotein (AFP), indicating hepatic differentiation. Thus, hES-MSCs can home to the acutely injured liver at high efficiency and undergo hepatic differentiation, suggesting that these cells could be useful for treating acute human liver injury.
-
Human Bone Marrow-Derived Mesenchymal Stem Cells Transplanted into Damaged Rabbit Heart to Improve Heart Function.
Wang Jian-an,Fan You-qi,Li Chang-ling,He Hong,Sun Yong,LV Bin-jian
DOI: https://doi.org/10.1007/bf02842459
2005-01-01
Abstract:Objective: The present study was designed to test whether transplantation of human bone marrow-derived mesenchymal stem cells (hMSCs) in New Zealand rabbits with myocardial infarction can improve heart function; and whether engrafted donor cells can survive and transdifferentiated into cardiomyocytes. Methods: Twenty milliliters bone marrow was obtained from healthy men by bone biopsy. A gradient centrifugation method was used to separate bone marrow cells (BMCs) and red blood cells. BMCs were incubated for 48 h and then washed with phosphate-buffered saline (PBS). The culture medium was changed twice a week for 28 d. Finally, hematopoietic cells were washed away to leave only MSCs. Human MSCs (hMSCs) were premarked by BrdU 72 h before the transplantation. Thirty-four New Zealand rabbits were randomly divided into myocardial infarction (MI) control group and cell treated group, which received hMSCs (MI+MSCs) through intramyocardial injection, while the control group received the same volume of PBS. Myocardial infarction was induced by ligation of the left coronary artery. Cell treated rabbits were treated with 5×10 6 MSCs transplanted into the infarcted region after ligation of the coronary artery for 1 h, and the control group received the same volume of PBS. Cyclosporin A (oral solution; 10 mg/kg) was provided alone, 24 h before surgery and once a day after MI for 4 weeks. Echocardiography was measured in each group before the surgery and 4 weeks after the surgery to test heart function change. The hearts were harvested for HE staining and immunohistochemical studies after MI and cell transplantation for 4 weeks. Results: Our data showed that cardiac function was significantly improved by hMSC transplantation in rabbit infarcted hearts 4 weeks after MI (ejection fraction: 0.695±0.038 in the cell treated group ( n =12) versus 0.554±0.065 in the control group ( n =13) ( P <0.05). Surviving hMSCs were identified by BrdU positive spots in infarcted region and transdifferentiated into cardiomyocytes characterized with a positive cardiac phenotype: troponin. I. Conclusion: Transplantation of hMSCs could transdifferentiate into cardiomyocytes and regenerate vascular structures, contributing to functional improvement.
-
The origin of human mesenchymal stromal cells dictates their reparative properties
Nili Naftali-Shani,Ayelet Itzhaki-Alfia,Natalie Landa-Rouben,David Kain,Radka Holbova,Shimrit Adutler-Lieber,Natali Molotski,Elad Asher,Avishay Grupper,Eran Millet,Ariel Tessone,Eyal Winkler,Jens Kastrup,Micha S Feinberg,Dov Zipori,Meirav Pevsner-Fischer,Ehud Raanani,Jonathan Leor
DOI: https://doi.org/10.1161/JAHA.113.000253
2013-09-30
Abstract:Background: Human mesenchymal stromal cells (hMSCs) from adipose cardiac tissue have attracted considerable interest in regard to cell-based therapies. We aimed to test the hypothesis that hMSCs from the heart and epicardial fat would be better cells for infarct repair. Methods and results: We isolated and grew hMSCs from patients with ischemic heart disease from 4 locations: epicardial fat, pericardial fat, subcutaneous fat, and the right atrium. Significantly, hMSCs from the right atrium and epicardial fat secreted the highest amounts of trophic and inflammatory cytokines, while hMSCs from pericardial and subcutaneous fat secreted the lowest. Relative expression of inflammation- and fibrosis-related genes was considerably higher in hMSCs from the right atrium and epicardial fat than in subcutaneous fat hMSCs. To determine the functional effects of hMSCs, we allocated rats to hMSC transplantation 7 days after myocardial infarction. Atrial hMSCs induced greatest infarct vascularization as well as highest inflammation score 27 days after transplantation. Surprisingly, cardiac dysfunction was worst after transplantation of hMSCs from atrium and epicardial fat and minimal after transplantation of hMSCs from subcutaneous fat. These findings were confirmed by using hMSC transplantation in immunocompromised mice after myocardial infarction. Notably, there was a correlation between tumor necrosis factor-α secretion from hMSCs and posttransplantation left ventricular remodeling and dysfunction. Conclusions: Because of their proinflammatory properties, hMSCs from the right atrium and epicardial fat of cardiac patients could impair heart function after myocardial infarction. Our findings might be relevant to autologous mesenchymal stromal cell therapy and development and progression of ischemic heart disease.
-
Mesenchymal stem cells as trophic mediators
Arnold I. Caplan,James E. Dennis
DOI: https://doi.org/10.1002/jcb.20886
2006-01-01
Journal of Cellular Biochemistry
Abstract:Adult marrow-derived Mesenchymal Stem Cells (MSCs) are capable of dividing and their progeny are further capable of differentiating into one of several mesenchymal phenotypes such as osteoblasts, chondrocytes, myocytes, marrow stromal cells, tendon-ligament fibroblasts, and adipocytes. In addition, these MSCs secrete a variety of cytokines and growth factors that have both paracrine and autocrine activities. These secreted bioactive factors suppress the local immune system, inhibit fibrosis (scar formation) and apoptosis, enhance angiogenesis, and stimulate mitosis and differentiation of tissue-intrinsic reparative or stem cells. These effects, which are referred to as trophic effects, are distinct from the direct differentiation of MSCs into repair tissue. Several studies which tested the use of MSCs in models of infarct (injured heart), stroke (brain), or meniscus regeneration models are reviewed within the context of MSC-mediated trophic effects in tissue repair.
cell biology,biochemistry & molecular biology
-
Mesenchymal stem cells: biology, pathophysiology, translational findings, and therapeutic implications for cardiac disease
Adam R Williams,Joshua M Hare
DOI: https://doi.org/10.1161/CIRCRESAHA.111.243147
2011-09-30
Abstract:Mesenchymal stem cells (MSCs) are a prototypical adult stem cell with capacity for self-renewal and differentiation with a broad tissue distribution. Initially described in bone marrow, MSCs have the capacity to differentiate into mesoderm- and nonmesoderm-derived tissues. The endogenous role for MSCs is maintenance of stem cell niches (classically the hematopoietic), and as such, MSCs participate in organ homeostasis, wound healing, and successful aging. From a therapeutic perspective, and facilitated by the ease of preparation and immunologic privilege, MSCs are emerging as an extremely promising therapeutic agent for tissue regeneration. Studies in animal models of myocardial infarction have demonstrated the ability of transplanted MSCs to engraft and differentiate into cardiomyocytes and vasculature cells, recruit endogenous cardiac stem cells, and secrete a wide array of paracrine factors. Together, these properties can be harnessed to both prevent and reverse remodeling in the ischemically injured ventricle. In proof-of-concept and phase I clinical trials, MSC therapy improved left ventricular function, induced reverse remodeling, and decreased scar size. This article reviews the current understanding of MSC biology, mechanism of action in cardiac repair, translational findings, and early clinical trial data of MSC therapy for cardiac disease.
-
STEM CELL PLASTICITY: REGENERATION OF FUNCTIONAL MYOCARDIUM FROM BONE MARROW STEM CELLS (BMSC).
S T. Ildstad,Y Huang,B Dawn,Y Guo,A Rezazadeh,A B. Stein,S D. Prabhu,Y Gu,G Hunt,J Varma,R Bolli
DOI: https://doi.org/10.1097/00007890-200407271-00430
2004-01-01
Transplantation
Abstract:O417 Aims: Bone marrow stem cells (BMSC) have been demonstrated to have the capacity to repair damaged neural, cardiac, and skeletal tissue under selected conditions. As such, BMSC offer great promise for regenerative medicine. However, functional assessment of outcomes has been limited, and it remains controversial whether significant functional repair can be achieved. Methods: In the present studies we evaluated whether treatment of mice with hematopoietic growth factors to mobilize BMSC would enhance functional repair after myocardial infarction (MI). Three groups of mice underwent a 30 minute coronary artery occlusion followed by reperfusion. Mice were treated with G-CSF plus Flt3 ligand (FL) (★–★; n = 16); G-CSF plus SCF (SYMBOL; n = 18) and compared to vehicle-treated controls (⊠–⊠; n = 17). Left ventricular function was assessed at baseline, 48 hours after MI, and at 35 days.SYMBOLResults: Strikingly, there was a significant (P <0.05) improvement in function in the growth factor-treated groups as early as 35 days, with the most significant improvement in the group receiving FL + G-CSF. Importantly, this correlated directly with the absolute number of mobilized HSC and graft facilitating cells (FC). The most profound increase occurred with the combination of G-CSF and FL (★–★). To confirm that this functional outcome was due to BMSC, chimeras were prepared using EGFP+ BMC and enrolled in the protocol. At 35 days the hearts were retrieved and examined for EGFP+/troponin+ cardiomyocytes. There were no EGFP+ cardiomyocytes in vehicle-treated controls, while there was a significant number of EGFP+/troponin+ cardiomyocytes in the growth factor-treated chimeras, confirming that BMSC contributed directly to the newly generated cardiac tissue. Studies are underway to determine the relative contribution of each of these cellular subpopulations on functional outcome in stem cell repair. Conclusions: Optimization of function and composition of the SC product could offer significant advantage to enhance myocardial repair following MI and may hold great therapeutic potential in the clinic.Figure
-
Fundamentals of Stem Cells: Why and How Patients' Own Adult Stem Cells Are the Next Generation of Medicine
Eckhard U. Alt,Christoph Schmitz,Xiaowen Bai
DOI: https://doi.org/10.20944/preprints201904.0200.v1
2019-01-01
Abstract:Various tissue resident stem cells are receiving attention from basic scientists and clinicians as they hold certain promise for regenerative medicine. This paper is intended to clarify and facilitate the understanding, development and adoption of regenerative medicine in general and specifically of therapies based on unmodified, autologous adipose-derived regenerative cells (UA-ADRCs). To this end, results of landmark experiments on stem cells and stem cell therapy performed in the labs of the authors are summarized, the most intriguing of which are the following: (i) vascular associated mesenchymal stem cells (MSCs) can be isolated from different organs (adipose tissue, heart, skin, bone marrow and skeletal muscle) and differentiated into ectoderm, mesoderm and endoderm, providing significant support for the hypothesis of the existence of a small, ubiquitously distributed, universal vascular associated stem cell with full pluripotency; (ii) the orientation and differentiation of MSCs are driven by signals of the respective microenvironment; and (iii) these stem cells irrespective of the tissue origin exhibit full pluripotent differentiation potential without any prior genetic modification or the need for culturing. They can be obtained from a small amount of adipose tissue when using the appropriate technology for isolating the cells, and can be harvested from and re-applied to the same patient at the point of care without the need for complicated processing, manipulation, culturing, expensive equipment, or repeat interventions. These findings demonstrate the potential of UA-ADRCs for triggering the development of an entire new generation of medicine for the benefit of patients and of healthcare systems.
-
Transplanted Human Embryonic Stem Cells As Biological ‘catalysts’ for Tissue Repair and Regeneration
BC Heng,H Liu,T Cao
DOI: https://doi.org/10.1016/j.mehy.2004.11.036
IF: 4.7
2005-01-01
Medical Hypotheses
Abstract:Human embryonic stem cells have tremendous potential in the newly emerging field of regenerative medicine. Recently, it was demonstrated that the rescue of lethal cardiac defects in Id knockout mutant mouse embryos was not due to the transplanted cells giving rise to functional new tissues within the defective embryonic heart. Instead, there is indirect evidence that the observed therapeutic effect was due to various secreted factors emanating from the transplanted cells. This therefore, introduces the exciting prospect of utilizing human embryonic stem cells as biological ‘catalysts’ to promote tissue repair and regeneration in transplantation therapy. However, the immunological barrier against allogenic transplantation, as well as the teratogenic potential of human embryonic stem cells poses major technical challenges. A possible strategy to overcome the immunological barrier may be to impose a temporary regimen of immunosuppressive drugs followed by their gradual withdrawal, once adequate tissue regeneration has been achieved. Other more novel alternatives include the use of microencapsulation to block interaction with the transplant recipient’s immune system, and co-transplantation with bone marrow-derived mesenchymal stem cells, which have been demonstrated to possess immuno-suppressive properties. The teratogenic potential of human embryonic stem cells could possibly be alleviated by directing the differentiation of these cells to specific lineages prior to transplantation, or through mitotic inactivation (γ irradiation or mitomycin C exposure). Co-transplantation with autologous adult stem cells may represent a novel strategy to further enhance the ‘catalytic’ effects of human embryonic stem cells. The various factors secreted by human embryonic stem cells could then have a concentrated localized effect on relatively large numbers of co-transplanted autologous adult stem cells, which may in turn lead to enhanced repair and regeneration of the damaged tissue or organ. Moreover, there is also a possibility that synergistic interactions between the co-transplanted human embryonic stem cells and autologous adult stem cells, may somehow produce signals for the recruitment and migration of additional endogenous adult stem cells within the recipient (i.e. peripheral blood circulation, bone marrow), which could further enhance organ/tissue regeneration. Hence, the potential use of human embryonic stem cells as biological ‘catalysts’ to stimulate tissue repair and regeneration, appears to hold tremendous promise in the field of regenerative medicine. This new therapeutic strategy needs to thoroughly investigated, in view of its potentially important clinical applications.
-
Synergistic actions of hematopoietic and mesenchymal stem/progenitor cells in vascularizing bioengineered tissues.
Eduardo K Moioli,Paul A Clark,Mo Chen,James E Dennis,Helaman P Erickson,Stanton L Gerson,Jeremy J Mao
DOI: https://doi.org/10.1371/journal.pone.0003922
IF: 3.7
2008-01-01
PLoS ONE
Abstract:Poor angiogenesis is a major road block for tissue repair. The regeneration of virtually all tissues is limited by angiogenesis, given the diffusion of nutrients, oxygen, and waste products is limited to a few hundred micrometers. We postulated that co-transplantation of hematopoietic and mesenchymal stem/progenitor cells improves angiogenesis of tissue repair and hence the outcome of regeneration. In this study, we tested this hypothesis by using bone as a model whose regeneration is impaired unless it is vascularized. Hematopoietic stem/progenitor cells (HSCs) and mesenchymal stem/progenitor cells (MSCs) were isolated from each of three healthy human bone marrow samples and reconstituted in a porous scaffold. MSCs were seeded in micropores of 3D calcium phosphate (CP) scaffolds, followed by infusion of gel-suspended CD34(+) hematopoietic cells. Co-transplantation of CD34(+) HSCs and CD34(-) MSCs in microporous CP scaffolds subcutaneously in the dorsum of immunocompromised mice yielded vascularized tissue. The average vascular number of co-transplanted CD34(+) and MSC scaffolds was substantially greater than MSC transplantation alone. Human osteocalcin was expressed in the micropores of CP scaffolds and was significantly increased upon co-transplantation of MSCs and CD34(+) cells. Human nuclear staining revealed the engraftment of transplanted human cells in vascular endothelium upon co-transplantation of MSCs and CD34(+) cells. Based on additional in vitro results of endothelial differentiation of CD34(+) cells by vascular endothelial growth factor (VEGF), we adsorbed VEGF with co-transplanted CD34(+) and MSCs in the microporous CP scaffolds in vivo, and discovered that vascular number and diameter further increased, likely owing to the promotion of endothelial differentiation of CD34(+) cells by VEGF. Together, co-transplantation of hematopoietic and mesenchymal stem/progenitor cells may improve the regeneration of vascular dependent tissues such as bone, adipose, muscle and dermal grafts, and may have implications in the regeneration of internal organs.
-
Human mesenchymal stem cells: From immunophenotyping by flow cytometry to clinical applications
Arthur A. Nery,Isis C. Nascimento,Talita Glaser,Vinicius Bassaneze,José E. Krieger,Henning Ulrich
DOI: https://doi.org/10.1002/cyto.a.22205
IF: 4.714
2012-10-01
Cytometry Part A
Abstract:Modern medicine will unequivocally include regenerative medicine as a major breakthrough in the re-establishment of damaged or lost tissues due to degenerative diseases or injury. In this scenario, millions of patients worldwide can have their quality of life improved by stem cell implantation coupled with endogenous secretion or administration of survival and differentiation promoting factors. Large efforts, relying mostly on flow cytometry and imaging techniques, have been put into cell isolation, immunophenotyping, and studies of differentiation properties of stem cells of diverse origins. Mesenchymal stem cells (MSCs) are particularly relevant for therapy due to their simplicity of isolation. A minimal phenotypic pattern for the identification of MSCs cells requires them to be immunopositive for CD73, CD90, and CD105 expression, while being negative for CD34, CD45, and HLA-DR and other surface markers. MSCs identified by their cell surface marker expression pattern can be readily purified from patient's bone marrow and adipose tissues. Following expansion and/or predifferentiation into a desired tissue type, stem cells can be reimplanted for tissue repair in the same patient, virtually eliminating rejection problems. Transplantation of MSCs is subject of almost 200 clinical trials to cure and treat a very broad range of conditions, including bone, heart, and neurodegenerative diseases. Immediate or medium term improvements of clinical symptoms have been reported as results of many clinical studies.
cell biology,biochemical research methods
-
Human Induced Pluripotent Stem Cells Differentiate Into Functional Mesenchymal Stem Cells and Repair Bone Defects
Dmitriy Sheyn,Shiran Ben-David,Galina Shapiro,Sandra De Mel,Maxim Bez,Loren Ornelas,Anais Sahabian,Dhruv Sareen,Xiaoyu Da,Gadi Pelled,Wafa Tawackoli,Zhenqiu Liu,Dan Gazit,Zulma Gazit
DOI: https://doi.org/10.5966/sctm.2015-0311
Abstract:: Mesenchymal stem cells (MSCs) are currently the most established cells for skeletal tissue engineering and regeneration; however, their availability and capability of self-renewal are limited. Recent discoveries of somatic cell reprogramming may be used to overcome these challenges. We hypothesized that induced pluripotent stem cells (iPSCs) that were differentiated into MSCs could be used for bone regeneration. Short-term exposure of embryoid bodies to transforming growth factor-β was used to direct iPSCs toward MSC differentiation. During this process, two types of iPSC-derived MSCs (iMSCs) were identified: early (aiMSCs) and late (tiMSCs) outgrowing cells. The transition of iPSCs toward MSCs was documented using MSC marker flow cytometry. Both types of iMSCs differentiated in vitro in response to osteogenic or adipogenic supplements. The results of quantitative assays showed that both cell types retained their multidifferentiation potential, although aiMSCs demonstrated higher osteogenic potential than tiMSCs and bone marrow-derived MSCs (BM-MSCs). Ectopic injections of BMP6-overexpressing tiMSCs produced no or limited bone formation, whereas similar injections of BMP6-overexpressing aiMSCs resulted in substantial bone formation. Upon orthotopic injection into radial defects, all three cell types regenerated bone and contributed to defect repair. In conclusion, MSCs can be derived from iPSCs and exhibit self-renewal without tumorigenic ability. Compared with BM-MSCs, aiMSCs acquire more of a stem cell phenotype, whereas tiMSCs acquire more of a differentiated osteoblast phenotype, which aids bone regeneration but does not allow the cells to induce ectopic bone formation (even when triggered by bone morphogenetic proteins), unless in an orthotopic site of bone fracture. Significance: Mesenchymal stem cells (MSCs) are currently the most established cells for skeletal tissue engineering and regeneration of various skeletal conditions; however, availability of autologous MSCs is very limited. This study demonstrates a new method to differentiate human fibroblast-derived induced pluripotent stem cells (iPSCs) to cells with MSC properties, which we comprehensively characterized including differentiation potential and transcriptomic analysis. We showed that these iPS-derived MSCs are able to regenerate nonunion bone defects in mice more efficiently than bone marrow-derived human MSCs when overexpressing BMP6 using a nonviral transfection method.
-
Mesenchymal Stromal Cells and Tissue-Specific Progenitor Cells: Their Role in Tissue Homeostasis
Aleksandra Klimczak,Urszula Kozlowska
DOI: https://doi.org/10.1155/2016/4285215
2016-01-01
Stem Cells International
Abstract:Multipotent mesenchymal stromal/stem cells (MSCs) reside in many human organs and comprise heterogeneous population of cells with self-renewal ability. These cells can be isolated from different tissues, and their morphology, immunophenotype, and differentiation potential are dependent on their tissue of origin. Each organ contains specific population of stromal cells which maintain regeneration process of the tissue where they reside, but some of them have much more wide plasticity and differentiate into multiple cells lineage. MSCs isolated from adult human tissues are ideal candidates for tissue regeneration and tissue engineering. However, MSCs do not only contribute to structurally tissue repair but also MSC possess strong immunomodulatory and anti-inflammatory properties and may influence in tissue repair by modulation of local environment. This paper is presenting an overview of the current knowledge of biology of tissue-resident mesenchymal stromal and progenitor cells (originated from bone marrow, liver, skeletal muscle, skin, heart, and lung) associated with tissue regeneration and tissue homeostasis.
cell & tissue engineering
-
Transplantation of Mesenchymal Stem Cells from Human Bone Marrow Improves Damaged Heart Function in Rats.
Mai Hou,Ke-ming Yang,Hao Zhang,Wei-Quan Zhu,Fu-jian Duan,Hao Wang,Yun-hu Song,Ying-jie Wie,Sheng-shou Hu
DOI: https://doi.org/10.1016/j.ijcard.2006.03.028
IF: 4.039
2007-01-01
International Journal of Cardiology
Abstract:Background: Bone marrow-derived mesenchymal stem cells (MSCs) are of great therapeutic potential after myocardial ischemic injury. However, little is known about the biological characteristics of MSCs in patients with coronary artery disease and their effects on infracted myocardium. The present study evaluated the biological characteristics of MSCs from patients with coronary artery disease and their effects after being transplanted into infarcted myocardium using a rat model.Methods: Sternal bone marrow aspirates were taken at the time of coronary artery bypass graft surgery. Mononuclear cells isolated from bone marrow were cultured based on plastic adherence. The morphology and growth characteristics of MSCs were observed in primary and successive passages. A myocardial infarction model was created in 27 adult rats. Two weeks later, animals were randomized into two groups: culture medium (group I, n = 13) or MSCs (2 x 10(6)) from early passages labeled with BrdU (group II, n = 14) were injected into the infarcted myocardium. Echocardiography, histological examination, and reverse transcription-polymerase chain reaction (RT-PCR) were performed four weeks after cell transplantation.Results: Flow cytometry analyses demonstrated that adherent spindle cells from bone marrow are mesenchymal stem cells (positive for CD29 and CD44, but negative for CD34 and CD45). Growth curves showed that MSCs have great proliferative capability especially at early passages. MSCs implantation in the infarcted border zone improved left ventricular function significantly in group II compared with group I. However, despite improved left ventricular function, we did not observe significant regeneration of cardiac myocytes. Immunohistochemistry revealed only the expression of desmin in the engrafted MSCs, a marker of premature myocyte. Moreover, the improved left ventricular function in this study seemed to be secondary to the beneficial reverse remodeling induced by the increase of collagen in infarcted zone, the decrease in the adjacent myocardium, and the increase of neovascularization (capillary density: 192 +/- 7.8/ mm(2) in group II vs. 165 5.9/mm(2) in group I, P < 0.05). Reverse transcription-polymerase chain reaction (RT-PCR) results showed the expression levels of collagen 1, collagen III, SDF-1 (stromal cell-derived factor-1), and VEGF (vascular endothelia growth factor) in the infarcted border zone were significantly higher in the MSCs treated group.Conclusions: The MSCs from patients with coronary artery disease have a typical phenotype with highly proliferative potential and the engrafted MSCs may regulate extracellular collagens and cytokines to prevent the ventricular scar from pathologic thinning and attenuate the contractile dysfunction of the infarcted heart. (c) 2006 Elsevier Ireland Ltd. All rights reserved.