Cardiac stem cells and methods for isolation of same

Information

  • Patent Grant
  • 8772030
  • Patent Number
    8,772,030
  • Date Filed
    Monday, September 26, 2011
    13 years ago
  • Date Issued
    Tuesday, July 8, 2014
    10 years ago
Abstract
Method for the isolation, expansion and preservation of cardiac stem cells from human or animal tissue biopsy samples to be employed in cell transplantation and functional repair of the myocardium or other organs. Cells may also be used in gene therapy for treating cardiomyopathies, for treating ischemic heart diseases and for setting in vitro models to study drugs.
Description
BACKGROUND OF THE INVENTION

1. Field of the Invention


The invention concerns a method for the isolation and expansion of cardiac stem cells derived from postnatal cardiac tissue biopsy.


The invention deals with a method for the isolation, expansion and preservation of cardiac stem cells from human or animal tissue biopsy samples to be employed in cell transplantation and functional repair of the myocardium or other organs.


The cells may also be used in gene therapy, for treating genetic cardiomyopathies by expressing the healthy gene in cells from biopsies of subjects with genetic defects, propagating the cells in vitro and then transplanting them in the patient; for treating ischemic heart diseases by inducing the release of angiogenic growth factors by the transplanted cells; and for the setting of an in vitro models to study drugs.


2. Prior Art


Stem cells (SC) are able to replicate and to differentiate in response to appropriate signals, thus enabling the formation or regeneration of specialized tissues.


It was thought that cardiomyocytes were terminally differentiated cells; however, emerging evidence has shown the modest potential of these cells to proliferate in animal models and in heart transplant patients (1-4).


The limited ability of adult cardiomyocytes to undergo mitosis and to regenerate the myocardium after injury leads to a permanent deficiency in the number of functioning cells, with the development and progression of cardiac insufficiency. In the end stage of the disease, the alternative treatment to transplantation is the implantation of SC in the injured myocardium (cardiomyoplasty). This method has produced promising results in animal models and has been experimented also in humans. However, the problem of having a source and an availability of SC remains (5-7).


While embryonic SC (undifferentiated cells from the embryo that can produce a wide range of specialized cells and that can be derived from the cell mass inside blastocytes which, in humans, form 4-5 days after fertilization of the ovum) have a marked capability to proliferate and differentiate, their potential immunogenicity, arrhythmogenicity, and ethical issues in particular, have limited their use. Moreover, embryonic SC are pluripotent, consequently their use carries a potential risk of generating teratomas (as occurs in animal models). Hence, before these cells can be used, they need to be differentiated in vitro in cardiomyocytes.


There exist various types of cardiomyocytes (ventricular, atrial, sinus node, Purkinje, with pacemaker functions, etc.). Embryonic SC have the potential capability to generate these cardiomyocyte phenotypes in vitro but the yield is insufficient. Furthermore, the in vivo proliferative capability of cardiomyocytes derived from embryonic SC appears to be limited by the growth of multinucleate cells.


An alternative is to use adult SC (undifferentiated cells found in differentiated tissue that are able to proliferate, reproduce and differentiate into the specialized cell types of the tissues whence they were isolated) preferably obtained from the same patient, which would afford the advantage of allowing autologous transplantation without the need for immunosuppressive therapy. For this purpose, skeletal myoblasts (satellite cells) have been employed; however, they differentiate into skeletal myocytes with morphologic and functional properties differing from those of the cardiac muscle. The inability of skeletal myoblasts to transdifferentiate into cardiomyocytes and to couple with them could give rise to arrhythmias or other anomalies.


SC derived from bone marrow offer an attractive alternative, Mesenchymal SC (MSC) of the bone marrow can differentiate into cardiomyocytes in vitro (treated with DNA-demethylating agents) and in vivo where, however, in the presence of fibrosis, they mostly generate fibroblast-like cells. Hematopoietic SC (HSC) of the bone marrow (so-called side population cells [SPcells]) are pluripotent in that they can generate vascular epithelium, smooth muscle cells and cardiomyocytes. But the functional and electrophysiologic properties of HSC- and MSC-derived cardiomyocytes are not well characterized, and the use of undifferentiated cells instead of cardiomyocytes could give rise to in vivo differentiation into fibroblasts rather than muscle cells or to the development of tumors.


Although human cardiomyocytes have been conventionally considered terminally differentiated cells (i.e. unable to re-enter the cell cycle and to divide), indirect evidence accumulating over the past two years has suggested the existence of adult SC in the heart. These cells are ideal candidates for cardioplasty in that they need no reprogramming, give rise only to cells present in the heart, i.e. cardiomyocytes and vessels (endothelial cells and smooth muscles) and may, because this is their physiologic function, survive in transplant patients, integrate into the surrounding tissues and carry out their functions for longer periods without causing any damage. Patent applications WO 03/008535 and WO 03/006950 concern methods to derive cardiomyocytes from embryonic SC. Patent applications WO 02/13760 and WO 02/09650 deal with the use of adult SC (particularly hematopoietic and/or cardiac cells, without indicating a method to isolate them, also in combination) to repair cardiac injury or in treating cardiovascular diseases in general.


Patent application WO 99/49015 deals with the isolation of pluripotent cardiac SC of the adult p53−/− mouse. In particular, the description concerns the heart-derived pluripotent SC that differentiate and proliferate to produce a variety of cell types, including cardiocytes, fibroblasts, smooth muscle cells, skeletal muscle cells, keratinocytes, osteoblasts and chondrocytes. The cells may be employed in methods to treat patients with cardiac tissue necrosis. The SC proliferate and differentiate to produce cardiocytes that replace the necrotic tissue.


However, the method differs from that of the present invention, which was based on the assumption that the cardiac muscle cells, the striate muscles and the smooth muscle cells derived from a common precursor, the myoblast. Furthermore, there is no in vivo evidence from cardiomyopathic animals that supports the applicability of the method. Lastly, the methods differ substantially. In the method described in patent WO 99/49015, adult p53−/− mouse hearts are fragmented, dissociated with DNAse and collagenase. After centrifugation, the sediment myocytes are isolated on a discontinuous gradient (Percoll) and plated on a medium containing 5% FBS and then on a medium containing 15% FBS 20 days later. Between days 20 and 26, small (<5 μm) round, nonadherent, slow-growth, phase-bright cells with a high nucleus-to-cytoplasm ratio form in the suspension. These cells continue to live in the suspension for about 1.5 months in the presence of 10% horse serum. Then the cells remain suspended also without the addition of horse serum. The nonadherent SC do not form colonies in methylcellulose and proliferate in the presence of serum, SCF, aFGF, bFGF, and cFGF. In the absence of horse serum, the nonadherent cells differentiate into differently appearing adherent cells the authors have identified by mainly morphologic criteria as cardiocytes, chondrocytes, fibroblasts, smooth muscle cells, skeletal muscle myoblasts, pericytes, and other cells the authors have called adherent SC. About one fourth to one fifth of these cells is positive to alkaline phosphatase (osteoblasts and endothelial cells); all cells are negative to acetylated LDL (absence of endothelial cells) and to myosin heavy chain (MF20). The cells undergo mitosis when stimulated by bFGF, aFGF and cFGF. In the absence of serum, they differentiate into cells resembling a fried egg (myocytes), After treatment with ascorbic acid/α-GP, they differentiate into chondrocyte-like cells.


Adherent cells cloned by limiting dilution give rise to mesenchymal cells, including osteoblasts, chondrocytes, adipocytes and myocytes, although they cannot be clearly identified due to often inappropriate morphologic criteria and markers. All the cells tested negative to acetylated LDL (absence of endothelial cells). None of the 11 isolated clones could be induced to differentiate toward a single mesenchymal lineage.


The isolation of the cardiac-derived SC of neonate mice (1-4 days) is also described, wherein the passage of myocytes on human fibronectin is added to eliminate the fibroblasts. However, no data are given about the characteristics of the isolated SC. Furthermore, the cells isolated with the previous method do not give rise to the formation of an essential component of the heart tissues, i.e. vessels and endothelium.


DESCRIPTION OF THE INVENTION

The method of the present invention employs heart biopsy tissue as starting material, hence an elective material that cannot be used in the method described in patent application WO 99/49015, since the material was insufficient. After fragmenting the biopsy specimen and possibly using dissociating agents (e.g. trypsin, EDTA and collagenase), the fragments are plated and added to a medium containing 10% FBS; 10-30 days later, fibroblast-like adherent cells grow from the explants over which small round, phase-bright cells migrate that tend to cluster but are either not or only weakly adherent. The cells are isolated by washing and mild dissociation (e.g. EDTA, trypsin-EDTA for 2-3 min). The cells are then plated on polylysine-treated cellular substrates in an appropriate medium unlike that used in the previous technique, in that it is horse-serum-free and contains other growth factors; after 2-3 days cell aggregates (cardiospheres) arise that tend to grow as floating formations. The authors have found that the cardiac-forming cells are postnatal SC that can be advantageously employed for reimplantation in the myocardium.


These cells are able to multiply, while maintaining their origin characteristics for a period (at least 60 days) that is long enough to markedly enrich the cell population. Mechanical disaggregation of the cardiospheres (CS) by repeated pipetting and changing the medium every 3 days increases the number of CS (about 100-fold every 10 days) for at least the first 20 days. Given the number of SC that can be derived from a biopsy and their ability to multiply in vitro, it is thought that they can be used to replace a greater amount of tissue than that removed.


Certain cells in the CS present stem-cell markers (ckit, sca-1, CD34) that are able to differentiate toward the main components of the myocardium (cardiomyocytes and vessels). As evaluated by immunohistochemistry and/or RT-PCR, certain cells spontaneously express, particularly at the border of the CS, markers for cardiomyocyte (troponin I, ANP, myosin heavy chain) and for endothelial cells (von Willebrand factor and Ve-cadherin). The human CS, in a co-culture with rat myocytes, beat spontaneously. When inoculated subcutaneously in SCID mice, the murine CS give rise to growths containing cardiac muscle tissue and vessels within several days.


The authors have thus demonstrated that the SC can be derived in a reproducible manner from biopsy tissue of the atrium, ventricle and auricle of human subjects aged from 1 month to 70 years. The CS pertaining to the invention can be cryopreserved, and they maintain their functional characteristics after thawing.


Adult cardiac SC with similar characteristics can also be isolated from the mouse. In particular, to better understand cell differentiation in CS, several breeds of transgenic mice were studied; the findings confirmed the results obtained with human cells.


Lastly, the authors have shown in an animal model that human CS can be used for cardioplasty. When inoculated in the infarcted area (transthoracic cauterization or LAD ligation) of a SCID mouse, the cells give rise to cardiac tissue that presents good integration with the host tissue, as observed by morphology and immunohistochemistry studies.


Hence, the isolation and expansion of CS by the method of the invention is novel and advantageous compared with that described in the previous technique in terms of the origin of the sample, the methods of isolation and expansion and the morphologic and functional characteristics of the derived cells.


DETAILED DESCRIPTION OF THE INVENTION

The method comprises the following steps: biopsy sample obtained under sterile conditions and transported to the laboratory; preparation of fragments sized large enough to allow diffusion of nutrients present in the culture medium; distribution of fragments on culture plates and incubation under conditions appropriate for cell survival and growth; sampling of culture medium and cells and transfer to other culture plates under conditions adequate for cell expansion.


An object of the invention is a method to obtain stem cells able to repair damaged myocardiac tissue, comprising the following steps:

    • a) take a biopsy specimen of cardiac tissue and keep it in an appropriate culture medium;
    • b) treat the specimen under appropriate conditions with mild mechanical and/or chemical and/or enzymatic techniques to obtain tissue fragments sized large enough to allow the diffusion of nutrients present in the medium;
    • c) leave the tissue fragments to adhere to appropriate solid supports and maintain them in a medium containing convenient serum and/or growth factors;
    • d) allow the cells to grow, changing the medium partially or completely, until multicellular structures form that are either weakly adherent or do not adhere to the support;
    • e) separate said multicellular structures from the rest of the culture;
    • f) treat said multicellular structures by mild dissociation until most of the small phase-bright spherical cells detach but maintain their morphologic and functional characteristics;
    • g) plate the cells on culture substrates treated with polylysine or other agents promoting the adhesion of the culture to the support in a medium containing at least the minimal essential constituents for the growth of mammalian cells;
    • h) possibly repeat steps d) to g) at least once;
    • i) select the cells that aggregate in phase-bright spheroid formations (cardiospheres);
    • l) electively promote the formation of new cardiospheres by mild dissociation thereof and new formation;
    • m) eventually cryopreserve the cardiospheres for use after thawing.


Preferably stem cells are derived from non-embryonic cardiac tissue biopsies.


In one embodiment of the invention at least one of the steps follows treatment with oxygen concentrations different from that normally present in the atmosphere in order to modify the biologic characteristics of the cultures.


Experts in the field will understand that the CS derived with the procedure of the invention may be able to generate continuous cell lines following spontaneous transformation or transformation induced by chemical, physical or biologic agents.


In another embodiment the cells giving rise to and/or constituting cardiospheres are fused with other cells.


In another embodiment the cells giving rise to and/or constituting cardiospheres are used for nuclear transfer to and from other cells.


In another embodiment the cells giving rise to and/or constituting cardiospheres are grown in at least one stage on biodegradable and/or biocompatible supports.


In another embodiment the cells giving rise to and/or constituting cardiospheres are cultured in bioreactors and/or fermenters.


It is another object of the invention cells giving rise to and/or constituting cardiospheres able to repair myocardiac tissue obtainable according to the method of previous claims. Preferably said cells are to be used in gene therapy. Preferably said cells are to be used for nuclear transfers to and from other cells. The CS derived with the method of the invention can be variously used in the repair of myocardiac tissue, for nuclear transfer from and to other cells, in gene therapy for cardiopathies of genetic origin.





BRIEF DESCRIPTION OF FIGURES

FIG. 1—CS proliferation. A1-1A4) Phase micrographs of floating CSs (cultured from <24 h to >48 h) derived from a primary culture of a human atrial bioptical sample. 1B1-1B3) Proliferation curves of human and mouse CSs (human CSs were derived from 8 different subjects (1B1) and from pre- and post-natal mouse hearts (1B2 and 1B3) respectively), in the presence (1B1 and 1B2) and in absence (1B3) of 3.5% serum. Number of spheres refers to the mean number per well from which 90% of the spheres where withdrawn at each time-point for further analysis. Note the different pattern of proliferation between the human and mouse CSs and the rapid rise of the curves, followed by an irreversible decline in the serum-free conditions. 1C1-1C4) Fluorescence analysis of a single cell (1C2) (obtained from a dissociated GFP-expressing CS), when plated by limiting dilution on mitomycin-treated STO-fibroblast-coated 96-wells plates in CGM, over the course of the generation of the GFP-labeled clone. This clone could be passaged and expanded on poly-D-Lysine coat (1C3). 1D1-1D2) x-Gal staining of a eGFP/MLC3F clone (obtained as those human) after 48 hours exposure of growth factors-free medium: in these conditions cells in the clone become more flattened with many nuclei showing a blue color, demonstrating that a differentiation process occurred.


FIG. 2—CS characterization. 2A1-2A10) Fluorescence-confocal analysis of BrdU-labeled human CSs for cardiac differentiation markers: 6 μm scans (from the periphery to the center of the sphere) and final pictures (small and large images respectively). BrdU (green; 2A1-2A10), cTnI (red; 2A1-2A5) and ANP (red; 2A6-2A10). 2B1-2B4) Confocal analysis of human CSs after 12 h of culture: CD-34 (2B4), CD-31 (2B3), KDR (2B2) and c-Kit (2B1) labeling of CS-generating cells at the beginning of sphere formation. 2C1-2C3) Fluorescence phenotype analysis of human CSs (cryosections): cTnI (red; 2C1), sarcomeric myosin (2C2) and vWf (green; 2C3). 2D1-2D2), Fluorescence phenotype analysis of human partially dissociated-CSs, after four days of culture on collagen coat in CEM: cTnI (red; 2D2) expression appears in the cytoplasm of the human cells (migrated from the sphere) showing a triangular shape with a row arrangement). 2E1-2E3) Fluorescence analysis of partially dissociated eGFP-labeled human CSs at 96 h of co-culture with rat cardiomyocytes: the same green cells that showed a synchronous contraction with cardiocytes, express cTnI. 2F1-2F3) Fluorescent analysis of connexin-43 expression (red) in eGFP-labeled human CSs co-cultured with rat cardiomyocytes (as in panel e): a punctuate red fluorescence is present in the cell membrane of human cells. 2G1-2G6) Phase micrograph of CSs from MLC3F-nlacZ (2G1-2G2) and cTnI-nlacZ mice (2G3-2G6): nuclear lacZ expression mainly localized in the external layers of both embryo (2G1 and 2G3/4) and adult CSs (2G2 and 2G5/6), after a short time from their formation (2G4 and 2G6) and after a few days of culture. Nuclei of cells (derived from partially dissociated CSs, cultured for 5 days on collagen-coated surfaces) are also blue stained. 2H) Florescence analysis of a spontaneously differentiated mouse CS: as suggested from the synchronous contraction showed in culture, cTnI (red) is expressed in the sphere and the cells migrated; in the last, sarcomeres are also evident. 2I1-2I6) Fluorescence and phase analysis of CSs from GFP-cKit (2I1, 2I2, 2I4, and 2I5), GFP-cKit/MLC3F-nLacZ (2I3) and GFP-cKit/cTnI-nlacZ (2I6) mice. GFP-labeled cells were present a few minutes after their seeding in culture with CGM, at the beginning of the generation of the CSs, later in their inner mass and after their migration out from the oldest adherent spheres (arrows). GFP-labeled cells did not co-localize with the blue-stained ones (arrows) in CSs from GFP-cKit/MLC3F-nLacZ and GFP-cKit/cTnI-nlacZ mice; fluorescent cells were present also in the CSs' growth area (arrows) (right upper and lower panels; 2I3 and 2I6, respectively). Fluorescence, phase (small) and merged (large) images. 2J1-2J3, FACS analysis of post-natal mouse CSs-derived cells. A time course at 0 and 6 days was performed and the phenotype profile for CD34, cKit, Cd31 and sea-1 expression was analyzed and showed as percentage of positive events (2J1). Data are presented as mean±SD (n=3). *Indicates a statistically significant difference from TO.


FIG. 3—In vivo analysis. 3A1-3A5) Ectopic transplantation in SCID mouse of CSs from MLC3F-nlacZ/BS-eGFP mouse (3A1-3A5). Fluorescence analysis of unfixed cryosections (3A1-3A2; 3A4) from the subcutaneous dorsal inoculum (day 17): GFP-cells seemed to have migrated from the spheres while clusters of vessel-like structures could be observed mainly in the external area (insert). Staining for SMA of one of these cryosections showed positive immunoreaction of the sphere and some cells within the inoculum (3A5). 3B1-3B6) Fluorescence ([;] 3B1, 3B2, and [3B4] 3B5) and phase analysis ([;] 3B4 and [3B5] 3B6) of fixed and immuno-stained cryosections from dorsal inoculum of CSs from MLC3F-nlacZ/CD-1 and cTnI-lacZ/CD-1 mice: tubular structures were stained for sarcomeric myosin and cTnI (middle and lower panels respectively). X-Gal staining labeled the cells within and those migrating from a CS (3B2). Endothelial markers (SMA and Ve-cadherin), stained the vasculature (“black-holes”) (3B1; see also 3A3). 3C1-3C8) Orthotopic transplantation on a SCID-bg mouse, of cryopreserved human CSs into the viable myocardium bordering a freshly produced infarct. Confocal analysis of cryosectioned left ventricular heart after 18 days from the coronary ligature, shows that (3C1-3C4) cardiomyocytes expressing MHC (red) in the regenerating myocardium (particularly those indicated by the two central arrows), stain positive also for lamin A/C (green) (a specific human nuclear marker). In these cells MHC expression is evident mainly in the perinuclear area. Lamin A/C-labeled cells (red) are present in newly generated capillaries staining for smooth a-actin (see 3C9-3C12), and PECAM (3C5-3C8); connexin-43 (red) [0], as in the co-culture experiments, lines cytoplasmic membrane of some human cell (green) in the regenerating myocardium. 3D) Table 1. Effect of human CSs orthotopic transplantation on echocardiographic index of myocardial performance. Data are presented as mean±SD. Abbreviations: LVIDd, left ventricular internal dimension at end diastole; AWThd, anterior wall thickness; FS, fractional shortening; EF, ejection fraction. *: vs CAL+CSs p<0.05, §: vs CALp<0.05



FIGS. 4A-4B. 4a) (left) RT-PCR analysis of human CS from pediatric (PCS), adult (aCS) subjects and cardiac fragments (H) (ANF, NKx2.S, Ve-cadherin, GAPDH), and 4b) (right) RT-PCR analysis of murine CS (mCS) and of mouse heart fragments (H) (α-MHC, TnC, cardiac a-actin, GAPDH).





METHODS AND MATERIALS

Tissue Samples


The human tissue came from myocardiac biopsies of adult or other patients who underwent open heart surgery (aortocoronary bypass, cardiac valve replacement, tetralogy of Fallot, ventricular septum defect) or heart transplantation for advanced dilated cardiomyopathy or post-infarction chronic congestive cardiomyopathy. The murine tissue came from the hearts of previously characterized homozygous MLC3F-nLacZ mice (8) homozygous troponin-I-nLacZ (9) and EGFP/ckit (10) CD1-crossed mice. The mice show localized nuclear expression (cardiac and skeletal) of the trans gene for, β-galactosidase of the myosin light chain promoter, a tissue-specific nuclear expression (exclusively cardiac) of the trans gene for troponin-I and a cytoplasmic expression of the EGFP trans gene of the ckit promoter (the gene in these cell experiments), respectively. BS-EGFP mice (11), which show generalized expression of cytoplasmic GFP, were used as base strains. The crossed MLC3F-nLacZ/EGFP, MLC3F-nLac-Z/EGFP-ckit, Tn-I-nLac-Z/EGFP-ckit mice were bred according to experimental protocol. The human cardiac tissue biopsies were preserved in serum-free IMDM (Euroclone) at 00 C and maintained at this condition until arrival in the laboratory (within 2 h).


Processing, Isolation and Cryopreservation of Sphere-Forming Cell


After careful dissection of the macroscopically visible connective tissue, the samples were cut into 1-2 mm3 pieces, washed 3 times with Ca++/Mg++-free phosphate buffered solution (PBS, Invitrogen) and sequentially digested 3 times for S min each at 370 C with 0.2% trypsin (Gibco) and 0.1% collagenase IV (Sigma). The obtained cells, the bulk of which are elements of contaminating blood, were discarded and the remaining tissue fragments were washed with complete explant medium (CEM) [IMDM supplemented with 10% fetal calf serum (FCS) (Hyclone), 100 mg/ml penicillin, 100 U/ml streptomycin (Gibco), 2 mM L-glutamine (Gibco), 0.1 mM 2-mercaptoethanol (Sigma). The tissue pieces were then fixed to Petri dishes (Falcon) by light scraping with a scalpel on a plastic surface. The explants with cultured at 37° C. in 5% CO2 in complete IMDM. The murine cardiac tissues were treated similarly, except for the embryonic hearts, where enzyme digestion prior to explant digestion was omitted and the organs were partially dissociated with a 25 gauge needle. After a period of 1 to 3 weeks (depending on the origin of the sample, i.e. a shorter period for the embryonic tissue and a longer one for the adult tissue), a layer of fibroblast-like cells forms that derive from or surround the explants. The explants are then periodically treated (every 6-10 days, 4 times maximum) to isolate the sphere-forming cells. To remove only the phase-bright cells, which migrate from the explants to the outer cell layer, the medium is removed, and the material is collected by washing it twice with Ca++—Mg++-free PBS and once with 0.53 mM EDTA (Versene, Gibco) for 1-2 min, followed by mild trypsinization with 0.5 g/L-0, 53 mM Trypsin-EDTA (Gibco) at room temperature for another 2-3 min under visual microscopy control. After the cells are collected, complete medium is added to the explants, whereas the cells obtained by washing and enzymatic treatment are collected by centrifugation (1200 rpm for 7 min) and resuspended in cardiosphere-growing medium (CGM) (35% complete IMDM/65% DMEM-Ham's F-12 mix with 2% B27 [Gibco], 0.1 mM 2-mercaptoethanol, 10 ng/ml EGF (Prepotek EC, Ltd.), 40 ng/ml bFGF (prepotek EC, Ltd.), 4 nM cardiotrophin-1 (RD), 40 nM thrombin (Sigma) (final concentrations), antibiotics and L-Glu as in the complete medium. Depending on the number of cells obtained (from 104 to 4×105 cells/explant), the cells were resuspended by repipetting them and then plating about 2×105 cells/ml on poly-D-lysine (BD) coated multi-well plates. After 12-24 h, several cells begin to divide and after 48 h, cell groups form that are often surrounded by a thin membrane and that can grow as floating spheres and adherent spheres. The growth medium is partially changed every 2-3 days, and the spheres are mechanically triturated using a pipette or 1 ml needles. For cryopreservation, the spheres (washed in Ca++-Mg++-free PBS and Versene) are resuspended in the freezing medium (complete IMDM/DMEM-Ham-F-12 50: 50, 5% B27, 10% DMSO). To calculate the growth curves, all the spheres are counted during the first week of growth, and then 90% of the spheres are removed at defined times (and used for RT-PCR or immunohistochemical analysis); after adding CGM and mechanically triturating the residual spheres, they are left to proliferate until the next sampling, when they are recounted. BrdU labeling is performed for 12 h on the newly generated spheres and at defined times in the other spheres, as indicated (Roche). For clonal analysis, the human CSs are infected with a third-generation lentiviral vector, pRRLsin.PPT-PGK.GFP expressing green fluorescent protein (GFP), as described elsewhere (12). After being washed twice, the GFP-labeled CSs are dissociated into single cells by trituration in Ca++/Mg++-free PBS, Versene, and 1× trypsin-EDTA solutions in sequence, resuspended in CGM, and then seeded at a presumed concentration of 1 cell/well in 96-well plates coated with a feeder layer of mitomycin-C-treated STO fibroblasts (2 μg/ml), For differentiation on a substrate-coated surface, Ca++/Mg++-free PBS-washed, centrifuged and partially dissociated CSs are repeatedly pipetted and then seeded in a small volume of CEM (200-300 μl) on type I collagen-(Sigma) or Matrigel-(Falcon) coated dishes and cultured for 3-6 days.


In Vivo Analysis


For heterotopic transplantation, about 60 pooled CS obtained from pre- and postnatal EGFP/MLC3F-nLacZ or EGFP/TnI-nLacZ or MLC3F/nLacZ, TnI-nLacZ mice were washed twice in PBS and suspended in 100 μl of Matrigel (BD) and subcutaneously injected into the dorsal region of anesthetized (ketamine, 35 mg/kg i.m.) adult NOD-SCID mice. Transplanted-cardiosphere survival and function were monitored by direct palpation of beating through the skin. After about 3 weeks, the mice were sacrificed and the isolated inoculum was embedded in OCT for immunocytochemical analysis. After thawing, 10-day cultures of cryopreserved human CS derived from ventricular and atrial cardiac explants from adult subjects were used for orthotopic transplantation. About 20 washed and partially dissociated CS were suspended in 3 μl PBS and injected in the infarcted myocardiac area using a 27 gauge needle and a Hamilton syringe. Myocardiac infarction was induced as described elsewhere (13) with slight modifications. Briefly, the recipient NOD-SCID mice (anesthetized with ketamine [35 mg/kg]+xylazine [5 mg/kg] i.p.) underwent transthoracic cauterization (Surgitron 140 v) with a modified electrocautery probe inserted through the internal intercostal muscle in the fourth intercostal space on the anterior surface of the heart. Electrocauterization (ca. 40 W) was applied twice for 1 sec in the cutting mode before the CS were injected (the same volume of PBS was injected in the control mice). In some mice myocardial infarction has been also induced by LAD ligation. After about 3 weeks, the mice were sacrificed and the isolated heart was embedded in OCT after extensive washing in PBS and fixing with paraformaldehyde (4%) in PBS pH 7.4.


Immunocytochemistry


Immunocytochemistry on tissue sections and on cell cultures was performed as described elsewhere (14) using the following antibodies: monoclonal anti-human-cTnI, anti-human-cardiac-MHC, anti-human nucleus and polyclonal (PAb) anti-human ANP (Chemicon); mAb anti-CD-31, CD-34 (BD Biosciences), mAb anti-human Cripto-1 (RD), monoclonal anti-Ve-cadherin, anti-sea-I, mAb anti-mouse-cKit (Pharmigen), mAb anti-human-c-Kit (DAKO); pAb anti-human-von-Willebrand-factor and mAb anti-human-KDR (Sigma); mAb MF20 and pAb anti-mouse/human MHC (14), anti-desmine and anti-Smooth-Muscle-Actin (Sigma), mAb anti-humanimouse-cTnI (15), donated by S. Schiaffino (Dept. of Pathology, Univ. of Padua), pAb anti-mouse-flk-1 (Santa Cruz, USA). β-galactosidase activity was detected by light microscopy, as described elsewhere (14).


Reverse-PCR Transcription Analysis


Reverse-PCR transcription analysis was performed as described elsewhere (16). The oligonucleotides for amplifying the genes of the CS derived from the pediatric (PCS), adult subjects (aCS) and heart fragments (H) were the following:











hNkx2,5 (150 bp)



forw 5′-CTCCCAACATGACCCTGAGT-3′



and







rev 5′-GAGCTCAGTCCCAGTTCCAA-3′,







hANF (350 bp)



forw 5′-AATCAAGTTCAGAGGATGGG-3′



and







rev 5′-AATGCATGGGGTGGGAGAGG-3′,







hVe-Cad (330 bp)



forw 5′-TCTCTGTCCTCTGCACAA-3′



and







rev 5′-ATGCAGAGGCTCATGATG-3′,







hGAPDH



forw 5′-GAAGAGCCAAGGACAGGTAC-3′



and







rev 5′-CTGCACCACCAACTGCTTAG-3;






The oligonucleotides for amplifying the genes of the murine CS and the heart fragments (H) were the following:











mMHC (302 bp)



forw 5′-GAAGAGTGAGCGGCGCATCAAGGA-3′



and







rev 5′-TCTGCTGGAGAGGTTATTCCTCG-3′,







m cardiac actin (494 bp)



forw 5′-TGTTACGTCGCCTTGGATTTTGAG-3′



and







rev 5′-AAGAGAGAGACATATCAGAAGC-3′,







m cardiac TnC (410 bp)



forw 5′-AATGGATGACATCTACAAAG-3′



and







rev 5′-TGAGCTCTTCAATGTCATCT-3′.







mGAPDH



forw 5′-CCTCTGGAAAGCTGTGGCGT-3′



and







rev 5′-TTGGAGGCCATGTAGGCCAT-3′






Results

Isolation and Expansion of CS


Sphere-generating cells were obtained by mild enzymatic digestion of explanted human atrial or ventricular biopsies and fetal, embryo and postnatal mouse hearts. Soon after the generation of a layer of fibroblast-like cells from well adherent explants, small, round phase-bright cells began to migrate over this coat. These cells could be harvested periodically by treatment with EDTA and mild trypsinization, and allowed to grow on poly-D-lysine-coated culture surfaces, in a low-serum (3.5% FCS) medium supplemented with a serum substitute (B27), growth factors (EGF and bFGF), cardiothrophin-1 (CT-1) (17) and thrombin (18), which, in the first week of culture, led to a seven-fold increase in the number of spheres with respect to that obtained using the medium supplemented with the others factors either alone or in combination. Time course observations of cells derived from both human and murine explants showed that, early after their seeding (30 min), some of these cells began to divide while still in suspension; most cells became loosely adherent, others remained in suspension and some contaminating fibroblast-like cells attached firmly to the poly-D-lysine coat. Cellular divisions were evident also from the loosely adherent cell population and produced clusters of small, round phase-bright cells [that we termed cardiospheres (CSs)] after 10-12 hours (FIG. 1a). Within 24-36 hours from their appearance, CSs increased in size and some of them detached from the culture surface; after 48-72 hours most CSs were between 20 and 150 urn in size and, when not subjected to mechanical dissociation, the largest contained dark zones within their inner mass (FIG. 1a).


Murine CSs started a spontaneous rhythmic contractile activity soon after their generation and maintained this function during their life span, while human CSs did so only when co-cultured with rat cardiomyocytes. To be sure that contraction was a new trait acquired by the cs cells, gfp-labeled human CSs (partially or totally dissociated) were co-cultured with cardiomyocytes pre-stained or not with dil. Contracting gfp-labeled cells were observed after 48 hours of co-culture; furthermore, from this time onwards, a red color stained also the green fluorescent cells, suggesting that a connection is created between the human CSs and the rat cardiac cells. In fact, labeling of human cs/rat-cardiomyocyte co-cultures (in which only human cells were pre-labeled with gfp by lentiviral infection) with cx-43, the major ventricular gap junction protein, demonstrated the typical punctuate fluorescence pattern along the cytoplasmic membrane (FIG. 2f), indicating that a functional connection is created between the two cellular populations.


CSs were found to be composed of clonally derived cells and did not simply represent cellular aggregates. In fact, when human CSs [expressing the green fluorescent protein (GFP) after infection with lentiviral vectors expressing the reporter gene] or murine CSs (derived from eGFPIMLC3F or eGFP/cTnI-mice) were dissociated and plated as single cells on mitomycin-treated STO-fibroblast-coated 96-wells or at clonal dilution on 10 cm Ø Petri dishes, fluorescent spheres that could be sub-cloned on poly-D-lysine-coated surfaces (FIG. 1c) were generated with a 1 to 10% efficiency. These sub-clonally derived CSs show the same functional and phenotypic behavior in culture: after 3 days from their appearance, some murine clones started to, and after 48 hours of culture with CEM, the majority (6/7) of these showed expression of the lac-Z trans gene within nuclei after specific histochemical staining (FIG. 1d), Equally, human clones, derived from a single GFP-labeled cell, start a synchronous beating and express cTnI after 48 hours of co-culture with rat cardiomyocytes.


Furthermore, when BrdU was added to the culture medium, virtually all cells in the small, and those of the inner part of the largest CSs, were labeled (FIG. 2a), indicating that these cells were newly generated.


Human CS-generating cells were capable of self-renewal. With periodical dissociation, together with partial substitution of the growth medium every 2-3 days, a log-phase expansion of spheres was obtained (FIG. 1b). Growth was slower for mouse CSs (owing, probably, to the more differentiated features assumed in culture such as beating) and, serum-dependent (FIG. 1b) as for the human ones.


As shown in FIG. 2a, confocal immunofluorescence analysis of BrdU labeled human CSS with anti-BrdU (green) and cardiac-troponin i (ctni) or atrial natriuretic peptide (anp) (red), revealed BrdU-positive cells particularly in the inner part of the spheres, while ctni- or anp-positive cells were mainly localized in the external layers. Furthermore, several cs-cells expressed cardiac differentiation markers (ctni, anp) while still dividing, as indicated by BrdU incorporation (FIG. 2a), suggesting that early cardiac differentiation already occurred during the log-phase growth; generally, within 2-3 weeks, some spheres became adherent, showing a more flattened morphology. Some small cells eventually migrated out from these “sun-like” spheres in the form of adherent (differentiated) or small, round cells that could generate new spheres. After thawing from cryopreservation, CSs proliferated again, maintaining their propensity to beat.


Phenotypic analysis of newly developing human and mouse CSs revealed expression of endothelial (KDR (human)/flk-1 (mouse), CD-31) and stem-cell (CD-34, ckit, sca-1) markers. As shown in FIG. 2b, CSs at the 2-10 cell stage, strongly reacted with antibodies against these antigens. In larger spheres, the expression pattern of some of these markers (particularly cKit) was similar to the BrdU labeling (positive staining in the center and in some peripheral zones generating satellite spheres).


A time course (0 and 6 days) of the quantitative characterization of CSs cells with these stem and endothelial markers was performed by FACS analysis (FIG. 21: as shown, at the beginning of their formation (T 0) the phenotype of these cells seems to reflect the epi-fluorescent microscopy analysis with about 10% of positive staining for all four phenotypes. However, at 6 days (T6) cKit appears as the only conserved marker, suggesting that the cKit+ cells could be the main ones contributing to the maintenance of proliferation, while the initial positivity to the others may reflect an early activation state, as has been indicated for CD-34 in several system (19). Fluorescence microscopy analysis, performed on cryo-sectioned human CSs revealed expression of cardiac-differentiation markers (cTnI, MHC) and also of endothelial markers [von Willebrand factor (vWf)] (FIG. 2c). When totally or partially dissociated into single-cells and cultured on collagen-coated dishes, in the same medium as the explants, mouse and humans CS-derived cells assumed a typical cardiomyocyte morphology, phenotype (FIG. d-d1, h) and function documented (in the mouse only) by spontaneous contraction.


As aforementioned, human CSs did not beat spontaneously; however, these began to beat within 24 h when co-cultured with postnatal rat cardiomyocytes, losing, after this time, their spherical shape and assuming a “sun-like” appearance. Markers of cardiac differentiation were co-expressed within GFP in human labeled CSs-cells (FIG. 2e).


To follow the differentiation process of CSs during-the pre- and post-natal age, MLC3F-nlacZ and cTnI-nlacZ mice were utilized (8,9). These mice express a form of lacZ transgene that localizes within the nucleus under the skeletal and cardiac muscle myosin light chain or cardiac troponin I promoter, respectively. CSs obtained from embryonic day 9-12, fetal day 17-18, neonatal and adult mice, showed spontaneous expression of the reporter gene in a variable percentage (10-60%) of spheres in the different culture conditions employed (FIG. 2e); moreover, as for the human ones, CS-generating cells from mice expressed stern (CD-34, sea-L, cKit) and endothelial cell markers (flk-1 CD-31).


On this basis, we utilized transgenic mice expressing the green fluorescent protein (GFP) under the control of the c-kit promoter (10), in order to further clarify the cellular origin of these spheres and to follow the pattern of their growth process. As shown in FIG. 2i, GFP-positive cells were present from the beginning of the formation of the CSs and, albeit with reduced fluorescence intensity, also later, within the mass of cells of the CSs and in cells migrating from old “sun-like” adherent CSs. Moreover, as suggested by the growth pattern of human CSs, when satellite secondary CSs appeared to detach from the primary ones, GFP-positive cells localized on the margins of the latter and in the inner part of the former.


We studied this process in double-heterozygous mice obtained from GFP-cKitIMLC3F-nlacZ or GFP-cKit/cTnI-nLacZ crossings: as shown in FIG. 2i, beta-Gal-positivity did not co-localize with GFP in cells present within the growing areas.


In conclusion, CSs appear to be a mixture of cardiac stern cells, differentiating progenitors and even spontaneously differentiated cardiomyocytes. Vascular cells were also present, depending on the sphere's size and time in culture. It is possible that, as for neurospheres (20), differentiating/differentiated cells stop dividing and/or die while stem cells continue to proliferate in an apparent asymmetric way, giving rise to many secondary spheres and to exponential growth in vitro. Mechanical dissociation favors this process. Death, differentiation and responsiveness to growth factors of the different cells within the CS, could depend on its three-dimensional architecture and on localization within the CS (21-22). The spontaneous formation of spheres is a known prerogative of neural stem cells, some tumor cell lines (LM) (22), endothelial cells (23) and fetal chicken cardiomyocytes (24). All these models (ours included), that mimic the true three-dimensional architecture of tissues, consist of spheroids of aggregated cells which develop a two-compartment system composed of a surface layer of differentiated cells and a core of unorganized cells that first proliferate and thereafter disappear over time (perhaps through apoptotic cell death). As well documented in fetal chick cardiomyocytes and endothelial cell spheroid culture, three-dimensional structure affects the sensitivity of cells to survival and growth factors (22,23). In particular, central spheroid cells do not differentiate and are dependent on survival factors to prevent apoptosis, while the cells of the surface layer seem to differentiate beyond the degree that can be obtained in two dimensional culture and become independent of the activity of survival factors. Furthermore, cell-cell contact and membrane-associated factors are known to be important for the division of neural precursor cells (25), in accordance with the notion that stem cells will only retain their pluripotency within an appropriate environment, as suggested by the “niche” hypothesis (26).


To investigate the survival and morpho-functional potential of the CSs in vivo, two sets of experiments were performed: in the first, CS cells were injected in the dorsal subcutaneous region of SCID mice; in the second, they were injected into the hearts of SCID-beige mice, acutely after myocardial infarction. The intention of ectopic transplantation experiments was to study the pattern and the behavior of growth of CSs in a neutral milieu (i.e. one without specific cardiac induction), in order to verify their unique potential of generation of the main cardiac cell types and to exclude the possibility of neoplastic transformation. For these experiments about 60 pooled spheres/inoculum/mouse from pre- and post-natal MLC3F-nlacZ/B5-eGFP TnI-nlacZ/B5-eGFP mice or MLC3F-nlacZ/CD-1 and cTnI-nlacZ/CD-1 mice, were employed. During the first 10 days, beating was appreciable through the skin over the injection site, distant from large blood vessels. On day 17, animals were sacrificed and the inoculum recognized as a translucent formation, grain-like in size, wrapped in ramified vessel-like structures. Observation of unfixed cryosections by fluorescence microscopy revealed the presence of open spheres from which cells appeared to have migrated; clusters of “black holes”, particularly in the periphery of the structure, were evident. The tissue contained tubular formations, surrounded by nuclei (Hoechst-positive), identified as cardiac sarcomeres because they were positive for cTnI and sarcomeric myosin (FIG. 3a). Alpha-SMA-positive structures (known to be transiently expressed during cardiomyogenesis (27)), were present in the remainder of the spheres and associated with the vasculature (the clusters of “black-holes”): this exhibited well-differentiated structures with a thin endothelium expressing Ve-cadherin (FIG. 3a) and a relative large lumen containing erythrocytes, indicating the establishment of successful perfusion by the host. Light microscopic observation of the inoculum, after X-Gal staining, showed strong nuclear expression of striated muscle-specific lacZ in the remainder of the spheres and in some cells close to them. No multi-differentiated structures suggesting the presence of tumor formation were observed.


To test the acquisition of functional competence and the cardiac regenerative potential of the CSs when challenged into an infarcted myocardium, orthotopic transplantation experiments with human CSs were performed. To do this, thawed (cryo-preserved) adult human CSs, coming from three atrial (one male and two female) and one ventricular (one female) biopsies were injected into the viable myocardium bordering a freshly produced infarct. Each mouse received CSs from a single passage of an explant (derived from a single subject). Four control infarcted animals were injected with an equal volume of PBS. After eighteen days from the intervention, the animals were sacrificed and infarct size was determined. Infarct size was 34.9±7.1 (3.6) and 31.9±6.9 (3.5) in the CS-treated group and PBS-injected group, respectively (p=n.s.). However, echocardiography showed better preservation of the infarcted anterior wall thickness (0.80±0.29(0.15) versus 0.60±0.20(0.08) p=n.s.) and particularly of FS % (36.85±16.43 (8.21) versus 17.87±5.95 (2.43) p<0.05) in the CS-treated group compared to the PBS-injected group (FIG. 3-table 1).


At the time of evaluation, bands of regenerating myocardium were present (with different degrees of organization and thickness) throughout most of the infarcted areas, as evaluated with hematoxilin-eosin histochemistry and MHC immunofluorescence (FIG. 3c). In the regenerating myocardium, cells expressing lamin A/C (a specific human nuclear marker) co-localize also with cardiomyocytes stained positive for MHC, newly generated capillaries stained for smooth a-actin and PECAM (FIG. 3c), and with connexin-43 expressing cells (which, as in the co-culture experiments, defines a connection between the human cells and the regenerating myocardium).


Thus CSs can be considered as clones of adult stem cells, maintaining their functional properties in vitro and in vivo also after cryo-preservation.


While this manuscript was in preparation, two papers have been published concerning the isolation of cardiac stem or progenitor cells from adult mammalian heart (28, 29). Isolation of these cells was based exclusively on the expression of a stem cell-related surface antigen: c-kit in the first paper and Sea-1 in the second one. In the first study (28) freshly isolated c-kitpos Lin cells from rat heart were found to be self-renewing, clonogenic and multipotent, exhibiting biochemical differentiation into the myogenic, smooth muscle cell, or endothelial cell lineage but, differently from cells grown under the conditions described here, failed to contract spontaneously. When injected into an ischemic heart these cells regenerated functional myocardium. In the second study (29), Sca-1+ cKitneg cells from mice heart were induced in vitro to differentiate toward the cardiac myogenic lineage in response to 5′-azacytidine. When given intravenously after ischemia/reperfusion, these cells homed to injured myocardium and differentiated into cardiomyocytes with and without fusion to host cells. Our data obtained on GFP-cKit transgenic mice also suggest that the adult cardiac stem cell is cKitpos. It is possible that CSs enclose a mixed population of cells that, as a niche, could promote the viability of cKit progenitors and contribute to their proliferation. The data obtained in the present paper confirm the existence of an adult cardiac stem cell. More importantly, they demonstrate for the first time that it is possible to isolate cells from very small fragments of human myocardium and expand these cells in vitro many fold (reaching numbers that would be appropriate for in vivo transplantation in patients) without loosing their differentiation potential, thus opening previously unforeseen opportunities for myocardial repair.


Transgenic Mice


To follow the differentiation process of CSs during the pre- and post-natal age, MLC3F-nlacZ and TnI-nLacZ mice were utilized. These mice express a form of the lacZ transgene that localizes within the nucleus under the skeletal and cardiac muscle myosin light chain or cardiac troponin-I promoter, respectively (8, 9). CSs derived from embryonic day 9-12, fetal day 17-18, neonatal and adult mice, showed spontaneous expression of the reporter gene in a variable percentage (10-60%) of spheres at the different culture conditions employed (FIG. 4a). Moreover, the mouse CS start to beat at the start of formation (particularly in the embryo) and continue to beat over the course of their life span. The human CS expressed stem (CD-34, sea-L, cKit) and endothelial cell markers (flk-1, CD-31).


In order to further clarify the cellular origin of these spheres and to follow the pattern of their growth process, we utilized transgenic mice expressing the green fluorescent protein (GFP) under the control of the c-kit promoter (10). GFP-positive cells were present from the beginning of the formation of the CSs and, albeit with reduced fluorescence intensity, also later. Moreover, as suggested by the growth pattern of human CSs, when satellite secondary CSs appeared to detach from the primary ones, GFP-positive cells localized on the margins of the latter and in the inner part of the former. We studied this process in double-heterozygous mice obtained from EGFP-cKit/MLC3F-nlacZ or TnI-nLacZ crossings. As shown in FIG. 4b, beta-Gal-positivity did not co-localize with EGFP in cells present within the growing areas.


Genetic Phenotype


The RT-PCR panel created on murine or human CS RNA extracts is shown in FIG. 5. A more typical profile of cardiac progenitors seems to be that of the human samples (in log-growth phase) compared with the murine samples, in which it is easier to have proliferation and differentiation occurring together.


REFERENCES



  • 1. Soonpaa M H, Field L J. Survey of studies examining mammalian cardiomyocyte DNA synthesis. Circ Res. 1998 Jul. 13; 83(1):15-26.

  • 2. Nadal-Ginard B, Kajstura J, Leri A, Anversa P. Myocyte death, growth, and regeneration in cardiac hypertrophy and failure. Circ Res. 2003 Feb. 7; 92(2):139-50.

  • 3. Laflamme M A, Myerson D, Saffitz J E, Murry C E. Evidence for cardiomyocyte repopulation by extracardiac progenitors in transplanted human hearts. Circ Res. 2002 Apr. 5; 90(6):634-40.

  • 4. Glaser R, Lu M M, Narula N, Epstein J A. Smooth muscle cells, but not myocytes, of host origin in transplanted human hearts. Circulation. 2002 Jul. 2; 106(1):17-9.

  • 5. Reffelmann T, Kloner R A. Cellular cardiomyoplasty-cardiomyocytes, skeletal myoblasts, or stem cells for regenerating myocardium and treatment of heart failure? Cardiovasc Res. 2003 May 1; 58(2):358-68.

  • 6. Dowell J D, Rubart M, Pasumarthi K B, Soonpaa M H, Field L J. Myocyte and myogenic stem cell transplantation in the heart. Cardiovasc Res. 2003 May 1; 58(2):336-50.

  • 7. Passier R, Mummery C. Origin and use of embryonic and adult stem cells in differentiation and tissue repair. Cardiovasc Res. 2003 May 1; 58(2):324-35.

  • 8. Kelly R, Alonso S, Tajbakhsh S, Cossu G, Buckingham M. Myosin light chain 3F regulatory sequences confer regionalized cardiac and skeletal muscle expression in transgenic mice. J Cell Biol. 1995 April; 129(2):383-96.

  • 9. Di Lisi R, Millino C, Calabria E, Altruda F, Schiaffino S, Ausoni S. Combinatorial cis-acting elements control tissue-specific activation of the cardiac troponin I gene in vitro and in vivo. J Biol Chem. 1998 Sep. 25; 273(39):25371-80.

  • 10. Cairns L A, Moroni E, Levantini E, Giorgetti A, Klinger F G, Ronzoni S, Tatangelo L, Tiveron C, De Fe lici M, Dolci S, Magli M C, Giglioni B, Ottolenghi S. c-kit regulatory elements required for expression in developing hematopoietic and germ cell lineages. Blood. 2003 Dec. 1; 102(12):3954-62.

  • 11. Hadjantonakis A K, Gertsenstein M, Ikawa M, Okabe M, Nagy A. Generating green fluorescent mice by germline transmission of green fluorescent ES cells. Mech Dev. 1998 August; 76(1-2):79-90.

  • 12. Follenzi A, Ailles L E, Bakovic S, Geuna M, Naldini L. Gene transfer by lentiviral vectors is limited by nuclear translocation and rescued by HIV-1 pol sequences. Nat Genet. 2000 June; 25(2):217-22.

  • 13. Brooks W W, Garibaldi B A, Conrad C H. Myocardial injury in the mouse induced by transthoracic cauterization. Lab Anim Sci. 1998 August; 48(4):374-8.

  • 14. Tajbakhsh S, Vivarelli E, Cusella-De Angelis G, Rocancourt D, Buckingham M, Cossu G. A population of myogenic cells derived from the mouse neural tube. Neuron. 1994 October; 13(4):813-21.

  • 15. Ausoni S, Campione M, Picard A, Moretti P, Vitadello M, De Nardi C, Schiaffino S. Structure and regulation of the mouse cardiac troponin I gene. J Biol Chern. 1994 Jan 7; 269(1):339-46.

  • 16. Ferrari S, Molinari S, Melchionna R, Cusella-De Angelis M G, Battini R, De Angelis L, Kelly R, Cossu G. Absence of MEF2 binding to the AfT-rich element in the muscle creatine kinase (MCK) enhancer correlates with lack of early expression of the MCK gene in embryonic mammalian muscle. Cell Growth Differ. 1997 January; 8(1):23-34.

  • 17. Pennica D, King K L, Shaw K J, et al. Expression cloning of cardiotrophinl, a cytokine that induces cardiac myocyte hypertrophy. Proc Natl Acad. Sci USA. 1995; 92: 1142-1146.

  • 18. Sabri A, Guo J, Elouardighi H, Darrow A L, Andrade-Gordon P, Steinberg S F. Mechanisms of protease-activated receptor-4 actions in cardiomyocytes. Role of Src tyrosine kinase. J Biol Chem. 2003 Mar. 28; 278(13):11714-20.

  • 19. Zammit P S, Beauchamp J R. The skeletal muscle satellite cell.: stem cell or son of stem cell? Differentiation 2001; 68: 193-204.

  • 20. Galli R, Gritti A, Bonfanti L, Vescovi M. Neural Stem Cells: An Overview. Circ Res. 2003; 92: 598-608.

  • 21. Layer P G, Robitzki A, Rothermel A, Willbold E. Of Layers And Spheres: The Reaggregate Approach In Tissue Engineering. Trends Neurosci. 2002; 25:131-134.

  • 22. Bates R C, Edwards N S, Yates J D. Spheroids And Cell Survival. Crit Rev Oncol Hematol. 2000; 36:61-74.

  • 23. Korff T, Augustin H G. Integration Of Endothelial Cells In Multicellular Spheroids Prevents Apoptosis And Induces differentiation. J cell biol. 1998; 143:1341-1352.

  • 24. Armstrong M T, Lee D Y, Armstrong P B. Regulation Of Proliferation Of The Fetal Myocardium. Dev Dyn. 2000; 219:226-36.

  • 25. Svendsen C N, Ter Borg M G, Armstrong R J, Rosser A E, Chandran S, Ostenfeld T, Caldwell M A. A New Method For The Rapid And Long Term Growth Of Human Neural Precursor Cells. J Neurosci Methods. 1998; 85:141-152.

  • 26. Schofield R. The Relationship Between The Spleen Colony-Forming Cell And The Haemopoietic Stem Cell.Blood Cells. 1978; 4:7-25.

  • 27. Kruithof B P, Van Den Hoff M J, Tesink-Taekema S, Moorman A F Recruitment Of Intra- And Extracardiac Cells Into The Myocardial Lineage During Mouse Development. Anat Rec. 2003; 271a: 303-314.

  • 28. Beltrami A P, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal-Ginard B, Anversa P. Adult Cardiac Stem Cells Are Multipotent And Support Myocardial Regeneration. Cell. 2003; 114:763-776.

  • 29. Oh H, Bradfute S B, Gallardo Td, Nakamura T, Gaussin V, Mishina Y, Pocius J, Michael Lh, Behringer Rr, Garry Dj, Entman M I, Schneider Md. Cardiac Progenitor Cells From Adult Myocardium: Homing, Differentiation, And Fusion After Infarction. Proc Natl Acad Sci USA. 2003; 100:12313-12318.


Claims
  • 1. A method for obtaining cardiospheres from a cardiac tissue biopsy sample, said method comprising the following steps: fragmenting a cardiac tissue biopsy sample to generate a plurality of cardiac tissue fragments; wherein the cardiac tissue biopsy sample is a non-embryonic cardiac tissue obtained from the ventricle, atrium or auricle of a heart;removing non-cardiac tissue or non-cardiac cells from said cardiac tissue fragments by partial enzymatic digestion of said cardiac tissue fragments;discarding said non-cardiac tissue or non-cardiac cells and retaining said partially digested cardiac tissue fragments;culturing said partially digested cardiac tissue fragments until phase-bright cells migrate from said cardiac tissue fragments;collecting said phase-bright cells;culturing said phase-bright cells on a treated surface in a culture media comprising 3.5% serum to generate one or more cardiospheres;wherein the one or more cardiospheres are spheroid structure of about 20 μm to about 150 μm in culture,wherein said cardiospheres are multicellular aggregates comprising stem cells, cardiac cells, and endothelial cells,wherein said stem cells express one or more stem cell markers selected from the group consisting of CD-34, ckit, and sca-1 within 12 hours of said cardiospheres being generated,wherein said endothelial cells express one or more endothelial markers selected from the group consisting of KDR, flk-1 and CD31 within 12 hours of said cardiospheres being generated; andharvesting said one or more cardiospheres, thereby obtaining cardiospheres.
  • 2. The method of claim 1, wherein said removing further comprises dissection of said cardiac tissue biopsy sample.
  • 3. The method of claim 1, wherein said partial enzymatic digestion is performed using trypsin, collagenase, or combinations thereof.
  • 4. The method of claim 1, wherein said culture media comprises one or more of epidermal growth factor (EGF), fibroblast growth factor (FGF), cardiotropin-1, and thrombin.
  • 5. The method of claim 1, further comprising disaggregating said harvested cardiospheres to generate disaggregated cells and culturing said disaggregated cells in said culture media to generate additional cardiospheres.
  • 6. The method of claim 5, wherein said disaggregated cells are cultured on said treated growth surface.
  • 7. The method of claim 1, wherein said treated growth surface comprises polylysine.
  • 8. The method of claim 1, wherein said culture media comprises a serum substitute.
  • 9. The method of claim 8, wherein said serum substitute comprises B27.
  • 10. The method of claim 1, wherein said culture media is horse serum free.
  • 11. The method of claim 10, wherein said culture media further comprises growth factors.
Priority Claims (1)
Number Date Country Kind
RM2003A0376 Jul 2003 IT national
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. application Ser. No. 10/567,008 filed Jul. 13, 2006 which is the U.S. National Phase application under 35 U.S.C. §371 of International Application PCT/IT2004/000421 filed Jul. 29, 2004, which claims priority to Italian Application RM2003 A 000376, filed Jul. 31, 2003. The entirety of each of these applications is hereby incorporated by reference.

US Referenced Citations (261)
Number Name Date Kind
3470876 Barchilon Oct 1969 A
3964468 Schulz Jun 1976 A
4106488 Gordon Aug 1978 A
4659839 Nicolotti et al. Apr 1987 A
4921482 Hammerslag et al. May 1990 A
4960134 Webster, Jr. Oct 1990 A
5052402 Bencini et al. Oct 1991 A
5175004 Matsumura Dec 1992 A
5199950 Schmitt Apr 1993 A
5228441 Lundquist Jul 1993 A
5243167 Lundquist Sep 1993 A
5287857 Mann Feb 1994 A
5315996 Lundquist May 1994 A
5322064 Lundquist Jun 1994 A
5329923 Lundquist Jul 1994 A
5334145 Lundquist et al. Aug 1994 A
5383852 Stevens-Wright Jan 1995 A
5436128 Harpold et al. Jul 1995 A
5454787 Lundquist Oct 1995 A
5477856 Lundquist Dec 1995 A
5492825 Jan et al. Feb 1996 A
5507725 Savage et al. Apr 1996 A
5616568 Prestwich et al. Apr 1997 A
5618294 Aust et al. Apr 1997 A
5670335 Jan et al. Sep 1997 A
5685868 Lundquist Nov 1997 A
5702433 Taylor et al. Dec 1997 A
5702905 Takahashi et al. Dec 1997 A
5762069 Kelleher et al. Jun 1998 A
5782748 Palmer et al. Jul 1998 A
5824031 Cookston et al. Oct 1998 A
5840502 Van Vlasselaer Nov 1998 A
5851212 Zirps et al. Dec 1998 A
5856155 Li Jan 1999 A
5874417 Prestwich et al. Feb 1999 A
5938603 Ponzi Aug 1999 A
5955275 Kamb Sep 1999 A
5957863 Koblish et al. Sep 1999 A
5981165 Weiss et al. Nov 1999 A
6004295 Langer et al. Dec 1999 A
6074408 Freeman Jun 2000 A
6077287 Taylor et al. Jun 2000 A
6086582 Altman et al. Jul 2000 A
6099832 Mickle et al. Aug 2000 A
6102887 Altman Aug 2000 A
6132390 Cookston et al. Oct 2000 A
6165164 Hill et al. Dec 2000 A
6193763 Mackin Feb 2001 B1
6203487 Consigny Mar 2001 B1
6224587 Gibson May 2001 B1
6296630 Altman et al. Oct 2001 B1
RE37463 Altman Dec 2001 E
6326198 Emerson et al. Dec 2001 B1
6337387 Sakano et al. Jan 2002 B1
6338942 Kraus et al. Jan 2002 B2
6346099 Altman Feb 2002 B1
6358247 Altman et al. Mar 2002 B1
6361997 Huss Mar 2002 B1
6387369 Pittenger et al. May 2002 B1
6408203 Mackin Jun 2002 B2
6416510 Altman et al. Jul 2002 B1
6443949 Altman Sep 2002 B2
6478776 Rosenman et al. Nov 2002 B1
6488659 Rosenman Dec 2002 B1
6500167 Webster, Jr. Dec 2002 B1
6511471 Rosenman et al. Jan 2003 B2
6511477 Altman et al. Jan 2003 B2
6514481 Prasad et al. Feb 2003 B1
6530944 West et al. Mar 2003 B2
6540725 Ponzi Apr 2003 B1
6547787 Altman et al. Apr 2003 B1
6569144 Altman May 2003 B2
6572611 Falwell Jun 2003 B1
6577895 Altman Jun 2003 B1
6585716 Altman Jul 2003 B2
6716242 Altman Apr 2004 B1
6726654 Rosenman Apr 2004 B2
6726662 Altman Apr 2004 B2
6739342 Fredriksson et al. May 2004 B1
6783510 Gibson et al. Aug 2004 B1
6796963 Carpenter et al. Sep 2004 B2
6805860 Alt Oct 2004 B1
6818757 Lee et al. Nov 2004 B2
6866117 Moss Mar 2005 B2
6905827 Wohlgemuth et al. Jun 2005 B2
6925327 Altman Aug 2005 B2
6971998 Rosenman et al. Dec 2005 B2
6997863 Handy et al. Feb 2006 B2
7026121 Wohlgemuth et al. Apr 2006 B1
7029466 Altman Apr 2006 B2
7034008 Donahue et al. Apr 2006 B2
7037648 Marban May 2006 B1
7048711 Rosenman et al. May 2006 B2
7074175 Handy et al. Jul 2006 B2
7104988 Altman et al. Sep 2006 B2
7138275 Kremer et al. Nov 2006 B2
7156824 Rosenman Jan 2007 B2
7220582 Epstein et al. May 2007 B2
7259011 Lucas et al. Aug 2007 B2
7280863 Shachar Oct 2007 B2
7329638 Yang et al. Feb 2008 B2
7351237 Altman Apr 2008 B2
7402151 Rosenman et al. Jul 2008 B2
7452532 Alt Nov 2008 B2
7468276 Hariri Dec 2008 B2
7470425 Vacanti et al. Dec 2008 B2
7500970 Altman Mar 2009 B2
7514074 Pittenger et al. Apr 2009 B2
7517686 Kremer et al. Apr 2009 B2
7531354 Stice et al. May 2009 B2
7547301 Altman et al. Jun 2009 B2
7547674 Anversa et al. Jun 2009 B2
7553663 Kremer et al. Jun 2009 B2
7592177 Chen et al. Sep 2009 B2
7625581 Laredo et al. Dec 2009 B2
7659118 Furcht et al. Feb 2010 B2
7686799 Leonhardt et al. Mar 2010 B2
7731648 Ivkov Jun 2010 B2
7745113 Evans et al. Jun 2010 B2
7794702 Rosen et al. Sep 2010 B2
7837631 Diamond et al. Nov 2010 B2
7862810 Anversa Jan 2011 B2
7875451 Murray et al. Jan 2011 B2
7971592 Ochi Jul 2011 B2
7999025 Shumaker-Parry et al. Aug 2011 B2
8008254 Anversa Aug 2011 B2
8017389 Phillips et al. Sep 2011 B2
8119123 Anversa Feb 2012 B2
8193161 Hosoda Jun 2012 B2
8232102 Dobson et al. Jul 2012 B2
8258113 Dimmeler et al. Sep 2012 B2
8562972 Edinger et al. Oct 2013 B2
20020061587 Anversa May 2002 A1
20020098167 Anversa et al. Jul 2002 A1
20020156383 Altman et al. Oct 2002 A1
20020177772 Altman et al. Nov 2002 A1
20030135113 Altman et al. Jul 2003 A1
20030161817 Young et al. Aug 2003 A1
20030195432 Kortenbach et al. Oct 2003 A1
20030229386 Rosenman et al. Dec 2003 A1
20040014209 Lassar et al. Jan 2004 A1
20040018174 Palasis Jan 2004 A1
20040030286 Altman Feb 2004 A1
20040033214 Young et al. Feb 2004 A1
20040076619 Anversa et al. Apr 2004 A1
20040087016 Keating et al. May 2004 A1
20040102759 Altman et al. May 2004 A1
20040110287 Clarke et al. Jun 2004 A1
20040136966 Anversa et al. Jul 2004 A1
20040153139 Altman Aug 2004 A1
20040158313 Altman Aug 2004 A1
20040168341 Petersen et al. Sep 2004 A1
20050074880 Sang et al. Apr 2005 A1
20050090732 Ivkov Apr 2005 A1
20050176620 Prestwich et al. Aug 2005 A1
20050215991 Altman et al. Sep 2005 A1
20050255588 Young et al. Nov 2005 A1
20050260748 Chang et al. Nov 2005 A1
20050260750 Kerr-Conte et al. Nov 2005 A1
20050271745 Gruettner et al. Dec 2005 A1
20060018897 Lee et al. Jan 2006 A1
20060020158 Altman Jan 2006 A1
20060025713 Rosengart et al. Feb 2006 A1
20060041182 Forbes et al. Feb 2006 A1
20060078496 Altman et al. Apr 2006 A1
20060083712 Anversa Apr 2006 A1
20060084089 Fort et al. Apr 2006 A1
20060084943 Rosenman et al. Apr 2006 A1
20060142749 Ivkov Jun 2006 A1
20060165805 Steinhoff Jul 2006 A1
20060198829 Rosen et al. Sep 2006 A1
20060224111 Rosenman et al. Oct 2006 A1
20060233712 Penades et al. Oct 2006 A1
20060234375 Doronin et al. Oct 2006 A1
20060239980 Miana et al. Oct 2006 A1
20060281791 Keating et al. Dec 2006 A1
20070003528 Consigny et al. Jan 2007 A1
20070014869 Matheny Jan 2007 A1
20070020758 Giacomello et al. Jan 2007 A1
20070048383 Helmus Mar 2007 A1
20070054397 Ott et al. Mar 2007 A1
20070072291 Kremer et al. Mar 2007 A1
20070088244 Miller et al. Apr 2007 A1
20070129296 Zhou Jun 2007 A1
20070142774 Rosenman Jun 2007 A1
20070196281 Jin et al. Aug 2007 A1
20070196918 Sayre et al. Aug 2007 A1
20070197891 Shachar et al. Aug 2007 A1
20070231393 Ritter et al. Oct 2007 A1
20070248580 Garcia Castro et al. Oct 2007 A1
20070292353 Levy et al. Dec 2007 A1
20080006281 Ou et al. Jan 2008 A1
20080027313 Shachar Jan 2008 A1
20080031854 Prestwich et al. Feb 2008 A1
20080076176 Dominko et al. Mar 2008 A1
20080089874 Li et al. Apr 2008 A1
20080138416 Rauh et al. Jun 2008 A1
20080187514 Anversa Aug 2008 A1
20080267921 Marban et al. Oct 2008 A1
20080268061 Jordan et al. Oct 2008 A1
20080274998 Cohen et al. Nov 2008 A1
20080287918 Rosenman et al. Nov 2008 A1
20080297287 Shachar et al. Dec 2008 A1
20080319420 Rosenman et al. Dec 2008 A1
20090074728 Gronthos et al. Mar 2009 A1
20090081170 Riley Mar 2009 A1
20090081276 Alsberg et al. Mar 2009 A1
20090123366 Dobson et al. May 2009 A1
20090136582 Albrecht et al. May 2009 A1
20090143748 Mickley et al. Jun 2009 A1
20090148415 de la Fuente et al. Jun 2009 A1
20090148421 Anversa et al. Jun 2009 A1
20090157046 Anversa Jun 2009 A1
20090162329 Anversa et al. Jun 2009 A1
20090169525 Anversa et al. Jul 2009 A1
20090177152 Altman Jul 2009 A1
20090180998 Anversa et al. Jul 2009 A1
20090226521 Smyth et al. Sep 2009 A1
20090317369 Hosoda et al. Dec 2009 A1
20100010073 Thum et al. Jan 2010 A1
20100012880 Rampersaud et al. Jan 2010 A1
20100040587 Haag et al. Feb 2010 A1
20100068811 Marban et al. Mar 2010 A1
20100081200 Rajala et al. Apr 2010 A1
20100239538 Anversa et al. Sep 2010 A9
20100255034 Meinke et al. Oct 2010 A1
20100303716 Jin et al. Dec 2010 A1
20100303722 Jin et al. Dec 2010 A1
20100303909 Oh et al. Dec 2010 A1
20100310534 Oved et al. Dec 2010 A1
20110003003 Goldberg et al. Jan 2011 A1
20110034753 Dobson et al. Feb 2011 A1
20110064675 Hadjipanayis et al. Mar 2011 A1
20110070153 Hyde et al. Mar 2011 A1
20110070154 Hyde et al. Mar 2011 A1
20110091428 Anversa Apr 2011 A1
20110092961 Hyde et al. Apr 2011 A1
20110110897 Schwarz et al. May 2011 A1
20110111412 Tai et al. May 2011 A1
20110123500 Anversa et al. May 2011 A1
20110135577 Wu et al. Jun 2011 A1
20110152835 Anversa Jun 2011 A1
20110165068 Liu et al. Jul 2011 A1
20110256105 Marbán et al. Oct 2011 A1
20110256621 Albrecht et al. Oct 2011 A1
20120034156 Hyde et al. Feb 2012 A1
20120034157 Hyde et al. Feb 2012 A1
20120039857 Smith et al. Feb 2012 A1
20120093885 Sahoo et al. Apr 2012 A1
20120165392 Olson et al. Jun 2012 A1
20120171291 Rademacher et al. Jul 2012 A1
20120177574 Gho et al. Jul 2012 A1
20120183528 Ebert et al. Jul 2012 A1
20120201795 Ware et al. Aug 2012 A1
20120238619 Dimmeler et al. Sep 2012 A1
20120253102 Marbán et al. Oct 2012 A1
20130059006 Schmuck et al. Mar 2013 A1
20130266543 Nadal-Ginard Oct 2013 A1
20130288962 Anversa et al. Oct 2013 A1
20130295060 Yang et al. Nov 2013 A1
20130309304 Nadal-Ginard Nov 2013 A1
Foreign Referenced Citations (94)
Number Date Country
1537646 Oct 2004 CN
1772300 May 2006 CN
1785430 Jun 2006 CN
1254952 Nov 2002 EP
1970446 Sep 2008 EP
2182053 May 2010 EP
2228444 Sep 2010 EP
1631318 Nov 2010 EP
1650293 Dec 2010 EP
2371370 Oct 2011 EP
2385120 Nov 2011 EP
2446929 May 2012 EP
1945256 Jul 2012 EP
2094869 Jul 2012 EP
2486944 Aug 2012 EP
2277548 Jan 2013 EP
2005110565 Apr 2005 JP
100830889 May 2008 KR
WO 9705265 Feb 1997 WO
WO 9712912 Apr 1997 WO
WO 9804708 Feb 1998 WO
WO 9832866 Jul 1998 WO
WO 9911809 Mar 1999 WO
WO 9939624 Aug 1999 WO
WO 9949015 Sep 1999 WO
WO 9951297 Oct 1999 WO
WO 0009185 Feb 2000 WO
WO 0024452 May 2000 WO
WO 0110482 Feb 2001 WO
WO 0126585 Apr 2001 WO
WO 0126706 Apr 2001 WO
WO 0126727 Apr 2001 WO
WO 0148151 Jul 2001 WO
WO 0176679 Oct 2001 WO
WO 0176682 Oct 2001 WO
WO 0209650 Feb 2002 WO
WO 0213760 Feb 2002 WO
WO 02051489 Jul 2002 WO
WO 03006950 Jan 2003 WO
WO 03008535 Jan 2003 WO
WO 03064463 Aug 2003 WO
WO 03103611 Dec 2003 WO
WO 03103764 Dec 2003 WO
WO 2004044142 May 2004 WO
WO 2005012510 Feb 2005 WO
WO 2006052925 May 2006 WO
WO 2006065949 Jun 2006 WO
WO 2006081190 Aug 2006 WO
WO 2007019398 Feb 2007 WO
WO 2007069666 Jun 2007 WO
WO 2007100530 Sep 2007 WO
WO 2007106175 Sep 2007 WO
WO2008036776 Mar 2008 WO
WO 2008043521 Apr 2008 WO
WO 2008058273 May 2008 WO
WO 2008118820 Oct 2008 WO
WO 2008124133 Oct 2008 WO
WO 2009032456 Mar 2009 WO
WO2009058818 May 2009 WO
WO 2009062143 May 2009 WO
WO 2009062169 May 2009 WO
WO 2009073518 Jun 2009 WO
WO 2009073594 Jun 2009 WO
WO 2009073616 Jun 2009 WO
WO 2009073618 Jun 2009 WO
WO 2009056116 Jul 2009 WO
WO2009067644 Aug 2009 WO
WO 2009100137 Aug 2009 WO
WO 2009149956 Dec 2009 WO
WO 2009152111 Dec 2009 WO
WO 2010028090 Mar 2010 WO
WO 2010059806 May 2010 WO
WO 2010083466 Jul 2010 WO
WO 2010118059 Oct 2010 WO
WO 2010135570 Nov 2010 WO
WO2011029092 Mar 2011 WO
WO2011029903 Mar 2011 WO
WO 2011053901 May 2011 WO
WO 2011056685 May 2011 WO
WO 2011057249 May 2011 WO
WO 2011057251 May 2011 WO
WO2011062244 May 2011 WO
WO2011064354 Jun 2011 WO
WO2011084460 Jul 2011 WO
WO 2011121120 Oct 2011 WO
WO2011127625 Oct 2011 WO
WO 2011138328 Nov 2011 WO
WO2011143499 Nov 2011 WO
WO2012020307 Feb 2012 WO
WO2012020308 Feb 2012 WO
WO2012055971 May 2012 WO
WO2012065027 May 2012 WO
WO 2012135253 Oct 2012 WO
WO 03004626 Jan 2013 WO
Non-Patent Literature Citations (259)
Entry
Duff et al., “CD105 is important for angiogenesis: evidence and potential applications,” FASEB J, Jun. 2003, vol. 17(9), pp. 984-992.
Gu, Bispecific Antibody Targeted Stem Cell Therapy for Myocardial Repair, University of California, San Francisco and University of California Berkeley, 2008.
Kyrtatos et al., Magnetic Tagging Increases Delivery of Circulating Progenitors in Vascular Injury, J. Am. Coll. Cardiol. Intv. 2:794-802 (2009).
Lee et al., Antibody Targeting of Stem Cells to Infarcted Myocardium, Stem Cells Translational and Clinical Research 25:712-717 (2007).
Levenberg, et al. Endothelial cells derived from human embryonic stem cells. Proceedings of the National Academy of Sciences Apr. 2, 2002, vol. 99, No. 7, pp. 4391-4396.
Lum et al., The New Face of Bispecific Antibodies: Targeting Cancer and Much More, Exp. Hematol. 34: 1-6 (2006).
Payne, Using Immunomagnetic Technology and Other Means to Facilitate Stem Cell Homing, Medical Hypotheses 62: 718-720 (2004).
Zhao et al., Targeting Human CD34+ Hematopoietic Stem Cells With Anti-CD45 × Anti-Myosin Light-chain Bispecific Antibody Preserves Cardiac Function in Myocardial Infarction, J. Appl. Physiol. 104:1793-1800 (2008).
Abela et al., A New Method for Isolation of Cardiac Myocytes by Percutaneous Endomycardial Biopsy, Catheterization and Cardiovascular Diagnosis, 1996, 37:227-230
Andersen et al., “Murine ‘Cardiospheres’ Are Not a Source of Stem Cells with Cardiomyogenic Potential,” Stem Cells, 2009, vol. 27, No. 7, pp. 1571-1581.
Anversa et al., Primitive cells and tissue regeneration. Circ. Res. 92:579-92 (2003).
Ausma et al., “Dedifferentiation of atrial cardiomyocytes: from in vivo to in vitro”, Cardiovascular Research, Jul. 2002, vol. 55(1), pp. 9-12.
Balser et al., Global parameter optimization for cardiac potassium channel gating models, Biophys. J., Mar. 1990, vol. 57, pp. 433-444.
Balser et al., Local Anesthetics as Effectors of Allosteric Gating, J. Clin. Invest., Dec. 1996, vol. 98(12), pp. 2874-2886.
Barile et al., Cardiac stem cells: isolation, expansion and experimental use for myocardial regeneration. Nat. Clin. Pract. Cardiovasc. Med. 4 Suppl 1: S9-S14 (2007).
Barile et al., Endogenous Cardiac Stem Cells. Prog. Cardiovas. Dis. 50(1):31-48 (2007).
Barr et al., Gene Therapy, Jan. 1994, vol. 1(1), pp. 51-58.
Barry et al., Differential Expression of Voltage-Gated K+ Channel Subunits in Adult Rat Heart, Circulation Research, 1995, vol. 77, pp. 361-369.
Barth et al., Lentiviral vectors bearing the cardiac promoter of the Na+—Ca2+ exchanger report cardiogenic differentiation in stem cells. Mol. Ther. 16(5):957-964 (2008).
Beltrami et al., “Adult cardiac stem cells are multipotent and support myocardial regeneration,” Cell. vol. 114, No. 6, pp. 763-776 (2003).
Beltrami et al., Evidence that human cardiac myocytes divide after myocardial infarction. N. Engl. J. Med. 344: 1750-1757 (2001).
Benardeau et al., Primary culture of human atrial myocytes is associated with the appearance of structural and functional characteristics of immature myocardium. J. Mol. Cell Cardiol. 29: 1307-1320 (1997).
Bird et al., “The human adult cardiomyocyte phenotype”, Cardiovascular Research, May 1, 2003, vol. 58(2), pp. 423-434.
Bosnali et al., “Generation of transducible versions of transcription factors Oct4 and Sox2,” Biological Chemistry, Walter De Gruyter GmbH & Co., Berlin, DE, vol. 389(7), Jul. 1, 2008, pp. 851-861.
Chen et al., Vascular endothelial growth factor promotes cardiomyocyte differentiation of embryonic stem cells, Am J Physiol Heart Circ Physiol, Oct. 2006, vol. 291(4), pp. H1653-H1658.
Christmann et al., Biomaterials for the Treatment of Myocardial Infarction, J. Am. Coll. of Cardiol. (2006) vol. 48(5): 907-913.
De Pomerai et al., Influence of serum factors on the prevalence of “normal” and “foreign” differentiation pathways in cultures of chick embryo neuroretinal cells, J Embryol Exp Morphol., 1981, vol. 62, 291-308.
Deal et al., Molecular Physiology of Cardiac Potassium Channels, Physiological Reviews, Jan. 1996, vol. 76(1), pp. 49-67.
Dispersyn et al., Adult rabbit cardiomyocytes undergo hibernation-like dedifferentiation when co-cultured with cardiac fibroblasts. Cardiovasc. Res. 57: 230-240 (2001).
Dispersyn et al., Dissociation of cardiomyocyte apoptosis and dedifferentiation in infarct border zones. Eur. Heart J. 23:849-857 (2002).
Dixon et al., Quantitative Analysis of Potassium Channel mRNA Expression in Atrial and Ventricular Muscle of Rats, Circulation Research, 1994, vol. 75, pp. 252-260.
Dixon et al., Role of the Kv4.3 K+ Channel in Ventricular Muscle, Circulation Research, 1996, vol. 79, pp. 659-668.
Donahue et al., Ultrarapid, highly efficient viral gene transfer to the heart, Proc. Natl. Acad. Sci. USA 94:4664-4668 (1997).
Driesen et al., Structural adaptation in adult rabbit ventricular myocytes: influence of dynamic physical interaction with fibroblasts. Cell. Biochem. Biophys. 44: 119-128 (2006).
Driesen et al., Structural remodeling of cardiomyocytes in the border zone of infarcted rabbit heart. Mol. Cell. Biochem (2007), Mol Cell Biochem. Aug. 2007;302 (1-2) :225-32.
Engel et al., “p38 MAP kinase inhibition enables proliferation of adult mammalian cardiomyocytes”, Genes & Dev., May 2005, vol. 19, No. 10, pp. 1175-1187.
Engel et al., FGF1/p38 MAP kinase inhibitor therapy induces cardiomyocyte mitosis, reduces scarring, and rescues function after myocardial infarction, Proc Nat Acad Sci (USA), Oct. 17, 2006, vol. 103(42), pp. 15546-15551.
Eschenhagen et al., Engineering Myocardial Tissue, Circ Res (2005) vol. 97:1220-1231.
Fiset et al., Shal-type channels contribute to the Ca2+-independent transient outward K+ current in rat ventricle, J. Physiology, 1997, vol. 500(1), pp. 51-64.
Gidh-Jain et al., Differential Expression of Voltage-Gated K+ Channel Genes in Left Ventricular Remodeled Myocardium After Experimental Myocardial Infarction, Circulation Research, 1996, vol. 79, pp. 669-675.
Glover et al., Reduction of infarct size and postschemic inflammation from ATL-146e, a highly selective adenosine A2A receptor agonist in reperfused canine myocardium, Amer J Physiol Heart Circ Physiol, Apr. 2005, vol. 288(4), pp. H1851-H1858.
Good et al., β-Amyloid Peptide Blocks the Fast-Inactivating K+ Current in Rat Hippocampal Neurons, Biophysical Journal, Jan. 1996, vol. 70, pp. 296-304.
Harvey, “Molecular Determinants of Cardiac Development and Congenital Disease,” Mouse Development, Patterning, Morphogenesis, and Organogensis, Chapter 16, pp. 331-370, 2002.
Heng et al., “Incorporating protein transduction domains (PTD) within recombinant ‘fusion’ transcription factors. A novel strategy for directing stem cell differentiation?” Biomedicine and Pharmacotherapy, Elsevier, Paris, FR, vol. 59(3), Apr. 1, 2005, pp. 132-134.
Jackson et al., Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells, J Clin Invest., 2001, vol. 107(11), pp. 1395-1402.
Kaab et al., Ionic mechanism of action potential prolongation in ventricular myocytes from dogs with pacing-induced heart failure. Circulation Research, vol. 78, No. 2, 262 (1996).
Kim et al., “Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins,” Cell Stem Cell, Jun. 5, 2009, vol. 4(6), pp. 472-476.
Kuhn et al., Periostin induces proliferation of differentiated cardiomyocytes and promotes cardiac repair, Nature Medicine, Aug. 2007, vol. 13(8), pp. 962-969. Abstract only.
Kwon et al., “Cellular Manipulation of Human Embryonic Stem Cells by TAT-PDX1 Protein Transduction,” Molecular Therapy, Academic Press, San Diego, CA, US, vol. 12(1), Jul. 1, 2005, pp. 28-32.
Lee et al., Cardiac gene transfer by intracoronary infusion of adenovirus vector-mediated reporter gene in the transplanted mouse heart. J. Thorac, and Cardio. Surg., 111:246 (1996).
Li et al., Molecular, Cellular, and Functional Phenotypes of Human Cardiac Stem Cells Dependent Upon Monolayer Versus Three-Dimensional Culture Conditions, Circulation Research, Dec. 4, 2009, vol. 105(12).
Li et al., Expansion of human cardiac stem cells in physiological oxygen improves cell production efficiency and potency for myocardial repair, Cardiovascular Research, Aug. 21, 2010.
Li et al., Stem Cell Technology: Epigenetics, Genomics, Proteomics, and Metabonomics, Stem Cells 2010; vol. 28, pp: 1178-1185.
Lyngbaek et al., Cardiac regeneration by resident stem and progenitor cells in the adult heart. Basic Res. Cardiol. 102: 101-114 (2007).
Maletic-Savatic et al., Differential Spatiotemporal Expression of K+ Channel Polypeptides in Rat Hippocampal Neurons Developing in situ and in vitro, Journal of Neuroscience, May 1995, vol. 15(5), pp. 3840-3851.
Marban, Big cells, little cells, stem cells: agents of cardiac plasticity. Circ Res. 100(4):445-6 (2007).
Marshall et al., The Jellyfish Green Fluorescent Protein: A New Tool for Studying Ion Channel Express and Function, Neuron, 1995, vol. 14, pp. 211-215.
McGann et al., Mammalian myotube dedifferentiation induced by newt regeneration extract. Proc. Natl. Acad. Sci. USA 98, 13699-704 (2001).
Messina et al., Isolation and Expansion of Adult Cardiac Stem Cells from Human and Murine Heart; Oct. 29, 2004; pp. 911-921; vol. 95; Circulation Research; Cellular Biology; American Heart Association.
Montessuit et al., “Regulation of glucose transporter expression in cardiac myocytes: p38 MAPK is a strong inducer of GLUT4”, Cardiovascular Research, Oxford University Press, Oct. 1, 2004, vol. 64(1), pp. 94-104.
Montessuit et al., “Retinoic acids increase expression of GLUT4 in dedifferentiated and hypertrophied cardiac myocytes”, Basic Research in Cardiology, Steinkopff-Verlag, DA, Jan. 1, 2006, vol. 101(1), pp. 27-35.
Nadal-Ginard et al, Myocyte death, growth, and regeneration in cardiac hypertrophy and failure. Circ. Res. 92(2):139-50 (2003).
Nadal-Ginard et al., A matter of life and death: cardiac myocyte apoptosis and regeneration. J. Clin. Invest. 111: 1457-9 (2003).
Odelberg , Inducing cellular dedifferentiation: a potential method for enhancing endogenous regeneration in mammals., Semin Cell Dev. Biol., 13(5):335-43 (2002).
Odelberg et al., Dedifferentiation of mammalian myotubes induced by msx1. Cell 103(7):1099-1109 (2000).
Oh et al., “Cardiac muscle plasticity in adult and embryo by heart-derived progenitor cells,” Annals of the New York Academy of Sciences, vol. 1015, pp. 182-189 (2004).
Oh et al., Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction, Proc Natl Acad Sci USA, 2003, vol. 100(21), pp. 12313-12318.
Passier et al., Origin and use of embryonic and adult stem cells in differentiation and tissue repair. Cardiovasc. Res. 58(2):324-35 (2003).
Plotnikov et al., Biologial Pacemaker Implanted in Canine Left Bundle Branch Provides Ventricular Escape Rhythms that Have Physiologically Acceptable Rates, Circulation, Feb. 3, 2004, vol. 109, pp. 506-512.
Potapova et al., Enhanced recovery of mechanical function in the canine heart by seeding an extracellular matrix patch with mesenchymal stem cells committed to a cardiac lineage, Am. J. Phys. (2008) vol. 295:H2257-H2263.
Ribera, Homogeneous Development of Electrical Excitability via Heterogeneous Ion Channel Expression, Journal of Neuroscience, Feb. 1, 1996, vol. 16(3), pp. 1123-1130.
Risepro et al., Hand1 regulates cardiomyocyte proliferation versus differentiation in the developing heart, Development, Nov. 2006, vol. 133(22), pp. 4595-4606. Abstract only.
Rucker-Martin et al., Dedifferentiation of atrial myocytes during atrial fibrillation: role of fibroblast proliferation in vitro. Cardiovasc. Res. 55: 38-52 (2002).
Rudy, Diversity and Ubiquity of K Channels, Neuroscience, 1988, vol. 25(3), pp. 729-749.
Serodio et al., Cloning of a Novel Component of A-Type K+ Channels Operating at Subthreshold Potentials With Unique Expression in Heart and Brain, Journal of Neurophysiology, May 1996, vol. 75(5), pp. 2174-2179.
Smith et al., Regenerative potential of cardiosphere-derived cells expanded from percutanerous endomyocardial biopsy specimens, Circulation, Feb. 20, 2007, vol. 115(7), pp. 896-908.
Smith et al., Stem Cells in the heart: what's the buzz all about? Part 1: Preclinical considerations. Heart Rhythm 5(5):749-757(2008).
Smith et al., Stem Cells in the heart: what's the buzz all about? Part 2: Arrhythmic risks and clinical studies. Heart Rhythm 5(6):880-887 (2008).
Srivastava et al., Thymosin beta4 is cardioprotective after myocardial infarction, Ann NY Acad Sci, Sep. 2007, vol. 1112, pp. 161-170. Abstract only.
Sussman et al., Myocardial aging and senescence: where have the stem cells gone? Annu Rev. Physiol. 66:29-48 (2004).
Takahashi et al., “Induction of pluripotent stem cells from adult human fibroblasts by defined factors,” Cell, Cell Press, Cambridge, MA, US, vol. 131(5), Nov. 30, 2007, pp. 861-872.
Tomita et al., Cardiac Neural Crest Cells Contribute to the Dorman Multipotent Stem Cell in the Mammalian Heart, Journal of Cell Biology, Sep. 26, 2005, vol. 170, No. 7, pp. 1135-1148.
Torella et al., Cardiac stem cell and myocyte aging, heart failure, and insulin-like growth factor-I overexpression. Circ. Res 94:514-24 (2004).
Torella et al., Resident human cardiac stem cells: role in cardiac cellular homeostasis and potential for myocardial regeneration. Nat. Clin. Pract. Cardiovasc. Med. 3 Suppl 1:S8-13 (2006).
Urbanek et al., Cardiac Stem Cells Possess Growth Factor Receptor Systems That After Activation Regenerate the Infarcted Myocardium, Improving Ventricular Function and Long-term Survival. Circ. Res. 97:663-673 (2005).
Urbanek et al., Intense myocyte formation from cardiac stem cells in human cardiac hypertrophy. Proc. Natl. Acad. Sci. USA 100(18):10440-5 (2003).
Urbanek et al., Myocardial regeneration by activation of multipotent cardiac stem cells in ischemic heart failure. Proc. Natl. Acad. Sci. USA 102(24):8692-7 (2005).
Ventura et al., Hyaluronan Mixed Esters of Butyric and Retinoic Acid Drive Cardiac and Endothelial Fate in Term Placenta Human Mesenchymal Stem Cells and Enhance Cardiac Repair in Infarcted Rat Hearts, JBC (2007) vol. 282(19):14243-14252.
Von Harsdorf, Can cardiomyocytes divide? Heart 86: 481-482 (2001).
Wagner, The state of the art in antisense research, Nature Medicine, Nov. 1995, vol. 1(11), pp. 1116-1118.
Walder et al., Up-regulation of neural stem cell markers suggests the occurrence of dedifferentiation in regenerating spinal cord. Dev. Genes Evol. 213: 625-630 (2003).
Wu et al., Cellular Therapy and Myocardial tissue engineering: the role of adult stem and progenitor cells. Eur. J. of Cardio-Thoracic Surg. 30:770-781 (2006).
Yu et al., “Induced pluripotent stem cell lines derived from human somatic cells,” Science, American Association for the Advancement of Science, US, vol. 318(5858), Dec. 21, 2007, pp. 1917-1920.
Zammit et al., The skeletal muscle satellite cell: stem cell or son of stem cell? Differentiation 68: 193-204 (2001).
Zhang et al., “Do cardiac stem cells arise from cardiomyocyte dedifferentiation?”, Circulation Research, Nov. 2006, vol. 99(11), p. 1278. Abstract only.
Zhou et al., “Generation of Induced Pluripotent Stem Cells Using Recombinant Proteins,” Cell Stem Cell, Cell Press, US, vol. 4(5), May 1, 2009, pp. 381-384.
Web page titled: bioptome.com—Scholten Surgical Instructions; downloaded from <http://www.bioptome.com/pages.php?page=Products>, 2001, first date of publication unknown, printed on Nov. 1, 2005.
Abdel-Latif, A., et al., Adult bone marrow-derived cells for cardiac repair: a systematic review and meta-analysis. Arch Intern Med, 2007. 167(10): p. 989-97.
Alibini et al., A Rapid in Vitro Assay for Quantitating the Invasive Potential of Tumor Cells, Cancer Research, vol. 47:3239-3245 (1987).
Ames BN, Shigenaga MK, Hagen TM. Oxidants, antioxidants, and the degenerative diseases of aging. Proc Natl Acad Sci USA. 1993;90:7915-7922.
Barbash et al., “Systemic Delivery of Bone-Marrow-Derived Mesenchymal Stem Cells to the Infarcted Myocardium Feasibility, Cell Migration, and Body Distribution,” Circulation, Apr. 19, 2003, 108:863-868. American Heart Association, Inc.
Bearzi et al, Human Cardiac Stem Cells, PNAS, vol. 104(35): 14068-14073 (2007).
Bernanke, et al., Effects of Hyaluronic Acid on Cardioc Cushion Tissue Cells in Collagen Matrix Cultures, Texas Reports on Biology and Medicine, vol. 39:271-285 (1979).
Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabe-Heider F, Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, Jovinge S, Frisen J. Evidence for cardiomyocyte renewal in humans. Science. 2009;324:98-102.
Birks EJ, Tansley PD, Hardy J, George RS, Bowles CT, Burke M, Banner NR, Khaghani A, Yacoub MH. Left ventricular assist device and drug therapy for the reversal of heart failure. N. Engl J Med. 2006;355(18):1873-1884.
Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C. Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysis. JAMA. 2007;297:842-857.
Bredemeyer AL, Sharma GG, Huang CY, et al. ATM stabilizes DNA double-strand-break complexes during V(D)J recombination. Nature. 2006;442:466-470.
Cai et al., “Injectable glycosaminoglycan hydrogels for controlled release of human basic fibroblast growth factor,” Biomaterials (2005), 26:6054-6067, Elsevier Ltd.
Chambers et al., Functional Expression Cloning of Nanog, a Pluripotency Sustaining Facot in Embryonic Stem Cells, Cell. May 30, 2003; 113(5):643-55.
Chen CS, Squire JA, Wells PG. Reduced tumorigenesis in p53 knockout mice exposed in utero to low-dose vitamin E. Cancer. 2009;115:1563-1575.
Cheng, et al., Functional performance of human caradiosphere-derived cells delivered in an in situ polymerizable hyaluronan-gelatin hydrogel, Biomaterials (2012). Doi10.1016/j.biomaterials. 2012.04.006.
Cheng K, Li Ts, Malliaras K, Davis DR, Zhang Y, Marban E. Magnetic targeting enhances engraftment and functional benefit of iron-labeled cardiosphere-derived cells in myocardial infarction. Circ Res. 2010;106:1570-1581.
Chimenti et al., “Relative Roles of Direct Regeneration Versus Paracrine Effects of Human Cardiosphere-Dervied Cells Transplanted Into Infarcted Mice,” Circulation Research (2010) 106:971-980, American Heart Association, Inc.
Chimenti, I., et al., Abstract 3182: Paracrine Contribution versus Direct Regeneration in Cardiosphere-Derived Cell Therapy for Acute Myocardial Infarction. Circulation, 2009. 120(18—MeetingAbstracts): p. S756-a-.
ClinicalTrials.gov, Identifier NCT00893360. CADUCEUS—Cardiosphere-Derived aUtologous Stem CElls to Reverse ventricUlar dySfunction, 2009.
Conkright et al., A gene encoding an intestinal-enriched member of the Kruppel-like factor family exrpessein in intestinal epithelia cells, Nucleic Acids Res. 27 (5), 1263-1270 (1999).
Crisostomo et al., “Embryonic stem cells attenuate myocardial dysfunction and inflammation after surgical global ischemia via paracrine actions,” Am J Physiol Heart Cirl Physiol (2008) 295:H1726-H1735.
Davis DR, Kizana E, Terrovitis J, Barth AS, Zhang, Y, Smith RR, Miake J, Marban E. Isolation and expansion of functionally competent cardiac progenitor cells directly from heart bippsies. J Mol Cell Cardiol. 2010;49:312-321.
Davis DR, Zhang Y, Smith RR, et al. Validation of the cardiosphere method to culture cardiac progenitor cells from myocardial tissue. PLoS One. 2009;4:e7195.
Davis, D.R., R.R. Smith, and E. Marban, Human Cardiospheres are a Source of Stem Cells with Cardiomyogenic Potential. Stem Cells, 2010. 28(5): p. 903-4.
Del Monte. et al. (2004) Proc Natl Acad Sci USA 101, 5622-7.
Djokic M, Le Beau MM, Swinnen LJ, et al. Post-transplant lymphoproliferative disorder subtypes correlate with different recurring chromosomal abnormalities. Genes Chromosomes Cancer. 2006;45:313-318.
Dong et al. (1991) Mol. Endocrinol. 5:1633.
Drakos SG, Kfoury AG, Hammond EH, Reid BB, Revelo MP, Rasmusson BY, Whitehead KJ, Salama ME, Selzman CH, Stehlik J, Clayson SE, Bristow MR, Renlund DG, Li DY. Impact of mechanical unloading on microvasculature and associated central remodeling features of the failing human heart. J Am Coll Cardiol. 2010;56(5):382-391.
Eguchi (2004) Med. Res. Rev. 24:182.
Elliot & O'Hare, 88 Cell 223-233 (1997).
Elliot & O'Hare, Intercellular Trafficking of VP22-GFP fusion proteins., Gene Therapy 6:149 (1999).
Falck J, Coates J, Jackson SP. Conserved modes of recruitment of ATM ATR and DNAPKcs to sites of DNA damage. Nature. 2005;434:605-611.
Fehrer C, Brunauer R, Laschober G, et al. Reduced oxygen tension attenuates differentiation capacity of human mesenchymal stem cells and prolongs their lifespan. Aging Cell. 2007;6:745-757.
Foreman J, Demidchik V, Bothwell JH, et al. Reactive oxygen species produced by NADPH oxidase regulate plant cell growth. Nature. 2003;422:442-446.
Frankel & Pabo, Cell 55:1189-93 (1988).
Freyman et al., “A quantitative, randomized study evaluating three methods of mesenchymal stem cell delivery following myocardial infarction,” European Heart Journal, 2006, 27:1114-1122.
Furlani D, Li W, Pittermann E, et al. A transformed cell population derived from cultured mesenchymal stem cells has no functional effect after transplantation into the injured heart. Cell Transplant. 2009;18:319-331.
Galli, R., et al., Neural stem cells: an overview. Circ Res, 2003. 92(6): p. 598-608.
George RS, Sabharwal NK, Webb C, Yacoub MH, Bowles CT, Hedger M, Khaghani A, Birks EJ. Echocardiographic assessment of flow across continuous-flow ventricular assist devices at low speeds. J Heart Lung Transplant. 2010.
Gomez-Marquez et al. (1987) J. Immunol. 143:2740.
Gu, Bispecific Antibody Targeted Stem Cell Therapy for Myocardial Repair, University of California San Francisco and University of California Berkeley, 2008.
Gubbay et al., Nature, 6281:245-50 (1990).
Hacein-Bey-Abina et al., Science 2003; 302:415-9.
Hagege, A.A., et al., Skeletal myoblast transplantation in ischemic heart failure: long-term follow-up of the first phase I cohort of patients. Circulation, 2006. 114(1 Suppl): p. I108-13.
Hainsworth AH, Bhuiyan N, Green AR. The nitrone disodium 2,4-sulphophenyl-N-tert-butylnitrone is without cytoprotective effect on sodium nitroprusside-induced cell death in N1E-115 neuroblastoma cells in vitro. J Cereb Blood Flow Metab. 2008;28:24-28.
Haider, et al., Bone Marrow Stem Cell Transplantation for Cardiac Repair, Am. J. Phys. Heart Circ. Physiol., vol. 288:H2557-H2567 (2005).
Haj-Yahia S, Birks EJ, Dreyfus G, Khaghani A. Limited surgical approach for explanting the HeartMate II left ventricular assist device after myocardial recovery. J Thorac Cardiovasc Surg. 2008;135(2):453-454.
Heng, BC et al., “Incorporating Protein Transduction Domains (PTD) within Recombinant Fusion Transcription Factors. A Novel Strategy for Directing Stem Cell Differentiation?” Biomedicine and Pharmacotherapy, vol. 59(3):132-34 (2005).
Hochedlinger et al., Nature 441:1061-7(2006).
Ivanovic Z. Hypoxia or in situ normoxia: The stem cell paradigm. J Cell Physiol. 2009;219:271-275.
Johnston PV, Sasano T, Mills K, Evers R, Lee ST, Smith RR, Lardo AC, Lai S, Steenbergen C, Gerstenblith G, Lange R, Marban E. Engraftment, differentiation, and functional benefits of autologous cardiosphere-derived cells in porcine ischemic cardiomyopathy. Circulation. 2009;120:1075-1083.
Jutkiewicz et al. (2006) Mol. Interven. 6:162.
Karlsson et al., Nature 344 (6269), 879-882 (1990).
Karoubi et al., “Single-cell hydrogel encapsulation for enhanced survivial of human marrow stromal cells,” Biomaterials, 2009, 30:5445-5455, Elsevier Ltd.
Kutschka, et al., Collagen Matrices Enhance Survival of Transplanted Cardiomyoblasts and Contribute to Functional Improvement of Ischemic Rat Hearts, Circulation, vol. 114:I167-I173 (2006).
Kyrtatos et al., Magnetic Tagging Increases Delivery of Circulating Progenitors in Vascular Injury, J. Am. Coll. Cardiol. Intv. vol. 2:794-802 (2009).
Laflamme et al., Nat Biotechnology 25:1015-24 (2007).
Landazuri, N. and J.M. Le Doux, Complexation of retroviruses with charged polymers enhances gene transfer by increasing the rate that viruses are delivered to cells. J Gene Med, 2004. 6(12): p. 1304-19.
Lavon N, Narwani K, Golan-Lev T, et al. Derivation of euploid human embryonic stem cells from aneuploid embryos. Stem Cells. 2008;26:1874-1882.
Lee et al., Antibody Targeting of Stem Cells to Infarcted Myocardium, Stem Cells Translational and Clinical Research, vol. 25:712-717 (2007).
Leferovich et al. (2001) Proc. Natl. Acad. Sci. USA 98:9830.
Levenberg at al., Endothelial cells derived from human embryonic stem cells, PNAS, vol. 99(7): 4391-4396 (2002).
Levine M, Conry-Cantilena C, Wang Y, et al. Vitamin C pharmacokinetics in healthy volunteers: evidence for a recommended dietary allowance. Proc Natl Acad Sci USA. 1996;93:3704-3709.
Li, Z. et al., Imaging survival and function of transplanted cardiac resident stem cells. J Am Coll Cardiol, 2009. 53(14): p. 1229-40.
Liao et al., Enhanced efficiency of generating induced pluipotent stem (iPS) cells from human somatic cells by a combination of six transcription factors, Cell Research (2008), vol. 18: 600-603.
Lindsay, Curr. Op. Pharmacol. 2:587-94 (2002).
Lindsley et al. (2008) Curr. Cancer Drug Targets 8:7.
Lipinski, M.J., et al., Impact of intracoronary cell therapy on left ventricular function in the setting of acute myocardial infarction: a collaborative systematicreview and meta-analysis of controlled clinical trials. J Am Coll Cardiol, 2007. 50(18): p. 1761-7.
Lowrey et al., Proc Natl Acad Sci USA 105:2883-8 (2008).
Lum et al., The New Face of Bispecific Antibodies: Targeting Cancer and Much More, Exp. Hematol., vol. 24:1-6 (2006).
Maitra A, Arking DE, Shivapurkar N, et al. Genomic alterations in cultured human embryonic stem cells. Nat Genet. 2005;37:1099-1103.
Martens et al., “Percutaneous Cell Delivery Into the Heart Using Hydrogels Polymerizing in Situ,” Cell Transplantation (2009), 18:297-304.
Mehmel HC, Stockins B, Ruffmann K, von Olshausen K, Schuler G, Kubler W. The linearity of the end-systolic pressure-volume relationship in man and its sensitivity for assessment of left ventricular function. Circulation. 1981;63:1216-1222.
Miller ER 3rd, Pastor-Barriuso R, Dalai D, et al. Meta-analysis: high-dosage vitamin E supplementation may increase all-cause mortality. Ann Intern Med. 2005;142:37-46.
Mitsui et al., Cell. May 30, 2003; 113(5):631-42.
Miyazono et al. (1988) J. Biol. Chem. 263:6407.
Moss et al., Dev. Biol. 258 (2), 432-442 (2003).
Moss, A. J., et al., Prophylactic implantation of a defibrillator in patients with myocardial infarction and reduced ejection fraction. N. Engl J Med, 2002. 346(12): p. 877-83.
Murata K, Iwata T, Nakashima S, Fox-Talbot K, Qian Z, Wilkes DS, Baldwin WM. C4d deposition and cellular infiltrates as markers of acute rejection in rat models of orthotopic lung transplantation. Transplantation. 2008;86:123-129.
Nakagawa et al., Nat Biotechnol 26:101-6 (2008).
Nelson et al., Stem Cells 26:1464-73 (2008).
Nelson, T.J., et al., Repair of acute myocardial infarction by human stemness factors induced pluripotent stem cells. Circulation, 2009. 120(5): p. 408-16.
Niethammer P, Grabher C, Look AT, Mitchison TJ. A tissue-scale gradient of hydrogen peroxide mediates rapid wound detection in zebrafish. Nature. 2009;459:996-999.
Noguchi et al., Protein Transduction Technology: A Novel Therepeautic Perspective, Acta Medica Okayama (2005) vol. 60(1): 1-11.
Nussbaum, J., et al., Transplantation of undifferentiated murine embryonic stem cells in the heart: teratoma formation and immune response. Faseb J, 2007. 21(7):p. 1345-57.
Okita et al., Generation of Mouse Induced Pluripotent Stem Cells Without Viral Vectors, (2008), Science Express, 322:949-53 (Oct. 9, 2008).
Owusu-Ansah E, Banerjee U. Reactive oxygen species prime Drosophila haematopoietic progenitors for differentiation. Nature. 2009;461:537-541.
Park et al., Nature 451:141-6 (2008).
Passier et al. (2008) Nature 453:322.
Payne, Using Immunomagnetic Technologi and Other Means to Facilitate Stem Cell Homing, Medical Hypotheses, vol. 62:718-720 (2004).
Peterson, E.D., L.J. Shaw, and R.M. Califf, Risk stratification after myocardial infarction. Ann Intern Med, 1997. 126(7): p. 561-82.
Physicians ATSACoC. ATS/ACCP Statement on Cardiopulmonary Exercise Testing. American Journal of Respiratory and Critical CareMedicine. 2003;167:211-277.
Prunier et al. Am J Physiol Heart Circ Physiol (2006).
Qin K, Zhao L, Ash RD, McDonough WF, Zhao RY. ATM-mediated transcriptional elevation of prion in response to copper-induced oxidative stress. J Biol Chem. 2009;284:4582-4593.
Quevedo, H.C., et al., Allogeneic mesenchymal stem cells restore cardiac function in chronic ischemic cardiomyopathy via trilineage differentiating capacity. Proc Natl Acad Sci U S A, 2009. 106(33): p. 14022-7.
Rossi DJ, Bryder D, Seita J, et al. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature. 2007;447:725-729.
Rotwein et al. (1986) J. Biol. Chem. 261:4828).
Rubio D, Garcia-Castro J, Martin MC, et al. Spontaneous human adult stem cell transformation. Cancer Res. 2005;65:3035-3039.
Sempere et al., Genome Biol. 5 (3), R13 (2004).
Sesso HD, Buring JE, Christen WG, et al. Vitamins E and C in the prevention of cardiovascular disease in men: the Physicians' Health Study II randomized controlled trial. JAMA. 2008;300:2123-2133.
Sharkey et al. (1995) Biol. Reprod. 53:974).
Shen et al. (1988) Proc. Natl. Acad. Sci. USA 85:1947.
Shu et al., Disulfide-crosslinked hyaluronon-gelatin hydrogel films: a covalent mimic of the extracellular matrix for in vitro cell growth, Biomaterials, vol. 24:3825-3834 (2003).
Simpson et al. (2007) Stem Cells 25:2350).
Singh U, Otvos J, Dasgupta A, et al. High-dose alpha-tocopherol therapy does not affect HDL subfractions in patients with coronary artery disease on statin therapy. Clin Chem. 2007;53:525-528.
Slaughter MS, Pagani FD, Rogers JG, Miller LW, Sun B, Russell SD, Starling RC, Chen L, Boyle AJ, Chillcott S, Adamson RM, Blood MS, Camacho MT, Idrissi KA, Petty M, Sobieski M, Wright S, Myers TJ, Farrar DJ. Clinical management of continuous-flow left ventricular assist devices in advanced heart failure. J Heart Lung Transplant. 2010;29(4 Suppl):S1-39.
Smart et al., De novocardiomyocytes from within the activated adult heart after injury. Nature. (2011) pp. 1-7.
Stewart S, Winters GL, Fishbein MC, et al. Revision of the 1990 working formulation for the standardization of nomenclature in the diagnosis of heart rejection, J Heart Lung Transplant. 2005;24:1710-1720.
Takahashi et al., Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors (2007) Cell, vol. 131:1-12.
Takahashi et al., Induction of Pluripotent Stem Cells from Fibroblast Cultures, Nature Protocols, vol. 2 No. 12, 3081-9 (2007).
Takeda et al., Nucleic Acids Res. 20 (17), 4613-4620 (1992).
Takehara et al., J. Am. Coll. Cardiol. (2008) 52:1858-65.
Takeshita et al. (1993) Biochem. J. 294:271.
Ten Dijke et al. (1988) Proc. Natl. Acad. Sci. USA 85:4715).
Terrovitis J, Lautamaki R, Bonios M, Fox J, Engles JM, Yu J, Leppo MK, Pomper MG, Wahl RL, Seidel J, Tsui BM, Bengel FM, Abraham MR, Marban E. Noninvasive quantification and optimization of acute cell retention by in vivo positron emission tomography after intramyocardial cardiac-derived stem cell delivery. J Am Coll Cardiol. 2009;54:1619-1626.
Terrovitis, J.V., R.R. Smith, and E. Marban, Assessment and optimization of cell engraftment after transplantation into the heart. Circ Res. 106(3): p. 479-94, 2010.
Trevethick et al., (2008) Br J Pharmacol. 155:463.
Tsagalou EP, Anastasiou-Nana M, Agapitos E, Gika A, Drakos SG, Terrovitis JV, Ntalianis A, Nanas JN. Depressed coronary flow reserve is associated with decreased myocardial capillary density in patients with heart failure due to idiopathic dilated cardiomyopathy. J Am Coll Cardiol.2008;52(17):1391-1398.
Uemura et al., “Bone marrow Stem Cells Prevent Left Ventricular Remodeling of Ischemic Heart Through Paracrine Signaling,” Circulation Research, 2006, 98:1414-1421, American Heart Association.
Ueno S. et al., Biphasic role for WNT/beta-catenin signaling in cardiac specification in zebrafish and embyonic stem cells. PNAS 104L9685 (2007).
van der Geest, R, Quantification in Cardiac MRI, Journal of Magnetic Resonance Imaging, 10:602-608(1999).
van Gent DC, Hoeijmakers JH, Kanaar R. Chromosomal stability and the DNA doublestranded break connection. Nat Rev Genet. 2001;2:196-206.
Vela et al. (2008) Cardiovasc. Pathol. 17:1.
Wang et al. (1994) Endocrinol. 134:1416.
Wang F, Thirumangalathu S, Loeken MR. Establishment of new mouse embryonic stem cell lines is improved by physiological glucose and oxygen. Cloning Stem Cells. 2006;8:108-116.
Web Page titled; Culture Media Database—EGM-2 (Endothelial Growth Medium 2)—ID 63; downloaded from <http://bio.Ionza.com/3018.html#ext-comp-1003:tab—63:change>; printed on Jan. 14, 2013.
Wernig el al., Cell Stem Cell 2: 10-2 (2008).
Wilmut et al., Nature 385:810-3 (1997).
Wilson KD, Huang M, Wu JC. Bioluminescence reporter gene imaging of human embryonic stem cell survival, proliferation, and fate. Methods Mol Biol. 2009; 574:87-103.
Yamada Y, Sekine Y, Yoshida S, Yasufuku K, Petrache I, Benson HL, Brand DD, Yoshino I, Wilkes DS. Type v collagen-induced oral tolerance plus low-dose cyclosporine prevents rejection of mhc class i and ii incompatible lung allografts. J Immunology. 2009;1:237-246 8.
Zhao et al., Targeting Human CD34+ Hematopoietic Stem Cells With Anti-CD45 × Anti-Myosin Light-chain Bispecific Antibody Preserves Cardiac Function in Myocardial Infarction, J. Appl. Phsyiol., vol. 104:1793-1800 (2008).
Chen CS, Wells PG. Enhanced tumorigenesis in p53 knockout mice exposed in utero to high-dose vitamin E. Carcinogenesis. 2006;27:1358-1368.
Cheng et al., Transplantation of platelet gel spike with cardiosphere-derived cells boosts structural and functional benefits relative to gel transplantation alone in rats with myocardial infarction, Biomaterials, vol. 33:2872-2879 (2012).
Davis DR, Kizana E, Terrovitis J, Barth AS, Zhang Y, Smith RR, Miake J, Marban E. Isolation and expansion of functionally-competent cardiac progenitor cells directly from heart biopsies. J Mol Cell Cardiol. 2010;49:312-321.
Deregibus, et al., Endotheial progentior cell-derived microvesicles activate an angiogenic program in endothelial cells by a horizontal transfer of mRNA, Blood, 2007, 2440-2448.
Eppenberger-Eberhardt et al., Reexpression of alpha-Smooth Muscle Acting Isoform in Culture Adult Rat Cardiomyocytes, Dev Biol. Jun. 1990; 139 (2) :269-78.
Gatti et al., Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia reperfusion-induced acute and chronic kidney injury, Nephrol. Dial. Transplant., vol. 26(5):1474-1483 (2011).
Hergenreider et al., Atheroprotective communication between endothelial cells and smooth muscle cells through miRNAs, Nat. Cell Biol., vol. 14(3):249-256 (2012).
Herrera et al., Human liver stem cell-derived microvesicles accelerate hepatic regeneration in hepatectomized rats, J. Cell. Mol. Med., vol. 14(6B):1605-1618 (2010).
Hierlihy et al., The Post-natal Heart Contains a Myocardial Stem Cell Population, FEBS Letters, vol. 530(1-3):239-243 (2002).
Hullinger et al., Inhibition of miR-15 protects against cardiac ischemic injury, Circ. Res. vol. 110(1):71-81 (2012).
Jayawardena et al., MicroRNA-mediated in vitro and in vivo direct reprogramming of cardiac fibroblasts to cardiomyocytes, Circ. Res. vol. 110(11)L1465-73 (2012).
Leor, et al., Transplantation of Fetal Myocardial Tissue Into the Infarcted Myocardium of Rat, Circulation, vol. 94(9): II-332 (1996).
Mangi et al., “Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts,” Nature Medicine, Sep. 2003, 9(9):1195-1201, Nature Publishing Group.
Nakasa et al., Acceleration of muscle regeneration by local injection of muscle-specific microRNAs in rat skeletal muscle injury model, J. Cell. Mol. Med., vol. 14(10): 2495-2505 (2010).
Pike et al., “Herparin-regulated release of growth factors in vitro and angiogenic response in vivo to implanted hyaluronan hydrogels containing VEGF and bFGF,” Biomaterials, (2006) 27:5242-5241, Elsevier Ltd.
Prestwich, et al., The translational imperative: Making Cell Therapy Simple and Effective, Acta Biomaterialia, vol. 8: 4200-4207 (2012).
Shimizu et al., Fabrication of Pulsatile Cardiac Tissue Grafts Using a Novel 3-D Cell Sheet Manipulation Techniques and Temperature-Responsive Cell Culture Surfaces, Circ. Res., vol. 90(3);e40 (2002).
Singh J. Enabling Technologies for Homing and Engraftment of Cells for Therapeutic Applications. J Am Coll Cardiol Intv. 2009;2(8):803-804.
Takahashi et al., Nat Protoc 2: 3081-9 (2007).
Van Winkle et al, “Cardiogel: A Biosynthetic Extracellular Matrix for Cardiomyocyte Culture,” in Vitro Dev. Biol.—Animal, vol. 21, 1996, pp. 478-485.
Vela, et al., Quest for the cardiovascular holy grail: mammalian myocardial regeneration, Cardiovasc. Pathol. 17:1-5 (2008).
Yu et al., miR-221 and miR-222 promote Schwann cell proliferation and migration by targeting LASS2 after sciatic nerve injury, J. Cell Sci., vol. 125(11)″2675-2683 (2012).
Zhou et al., Down-Regulation of microRNA-26a Promotes Mouse Hepatocyte Proliferation during Liver Regeneration, PLoS ONE, vol. 7(4):e33577 (2012).
Assmus, et al., Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction (TOPCARE-AMI), Circulation, vol. 106: 3009-3017 (2002).
Cho et al., Secondary Sphere Formation Enhances the Functionality of Cardiac Progenitor Cells, Mol. Ther., vol. 20(9):1750-1766 (2012).
Li, T-S et al., Direct comparison of different stem cell types and subpopulations reveals superior paracrine potency and myocardial repair efficacy with cardiosphere-derived cells, J. Am. Coll. Cardiol., vol. 59(10):942-953 (2012).
Lin et al., Accelerated Growth and Prolonged Lifespan of Adipose Tissue-Derived Human Mesenchymal Stem Cells in a Medium Using Reduced Calcium and Antioxidants, Stem Cells and Development, vol. 14:92-102 (2005).
Naka et al., Regulation of Reactive Oxygen Species and Genomic Stability in Hematopoietic Stem Cells, Antiox. Redox Signaling, vol. 10)11):1883-1884 (2008).
Puceat, M., Role of Rac-GTPase and Reactive Oxygen Species in Cardiac Differentiation of Stem Cell., Antiox. Redox. Signaling, vol. 7(11-12)″1435-1439 (2005).
Vrijsen, et al., Cardiomyocyte progenitor cell-derived exosomes stimulate migration of endothelial cells, J. Cell. Mol. Med., vol. 14(5):1064-1070 (2010).
Yau et al., Beneficial Effect of Autologous Cell Transplantation on Infarcted Heart Function: Comparison Between Bone Marrow Stromal Cells and Heart Cells, Annals of Thoracic Surg, vol. 75(1):169 (2003).
Zha, et al., Complementary Function of ATM and H2AX in Development and Suppression of Genomic Instability, PNAS, vol. 105(27):9302-9306 (2008).
Related Publications (1)
Number Date Country
20120020935 A1 Jan 2012 US
Continuations (1)
Number Date Country
Parent 10567008 US
Child 13245788 US