PROVISION OF NEW CARDIOMYOCYTE PROGENITOR CELLS AND CARDIOMYOCYTES DERIVED THEREFROM

Abstract
The present invention relates to cardiomyocyte progenitor cells (CMPCs), to methods for their isolation and to the use of this CMPCs for the provision of cardiomyocytes, by way of differentiating the obtained CMPCs with a demethylating agent. The present invention also relates to pharmaceuticals compositions comprising cardiomyocytes for use in a cardiomyocyte replacement therapy and/or for the treatment in myocardial infarctation or for ameliorating the effects of myocardial infarctation.
Description

The present invention relates to cardiomyocyte progenitor cells (CMPCs), preferably human CMPCs (hCMPCs). Methods for the isolation of such cells are also provided. The present invention also relates to the use of the CMPCs for the provision of cardiomyocytes, by way of differentiating the obtained CMPCs with a demethylating agent. Pharmaceutical compositions comprising these cardiomyocytes for use in a cardiomyocyte replacement therapy and/or for the treatment of myocardial infarction or for ameliorating the effects of myocardial infarction are also provided. These compositions can be used in cardiomyocyte replacement therapies and/or for the treatment of myocardial infarction or for ameliorating the effects of myocardial infarction. In a further aspect, the present invention relates to the use of the cardiomyocytes obtainable by the methods of the invention in screening methods, e.g. drug screening methods.


A variety of documents is cited throughout this specification. The disclosure content of said documents (including any manufacturer's specifications, instructions etc.) is herewith incorporated by reference; however, there is no admission that any document cited is indeed prior art as to the present invention.


If not defined otherwise, the terms used herein have the meaning and scope that they normally have in the relevant art of medicine or biochemistry, particularly in the field of cardiology, immunology, cell biology, electrophysiology and molecular biology.


Myocardial infarction is a common, severe cardiovascular disorder and a major cause of heart failure and cardiac death. Poor tissue perfusion and defective cardiomyocyte function are underlying mechanisms for the development of myocardial dysfunction, and the inability of remaining cardiomyocytes to regenerate and compensate the loss of ventricular cell mass. Stem cells have been studied intensively as a source of new cardiomyocytes to ameliorate injured myocardium and improve cardiac function1-4. The potential therapeutic benefit of stem cell transplantation has been investigated in animal models using bone marrow cells5-7 cardiac stem cells8, embryonic stem (ES) cells9,10 and foetal cardiomyocytes11,12 injected at the site of cardiac injury. The results reported in these animal studies has already let to the initiation of several clinical trials although at present only using bone marrow derived cells and skeletal myoblasts have been assessed in clinical trials13-16. The developmental plasticity of bone marrow cells to differentiate into cardiomyocytes has been questioned17,18 and the predominant in vivo effect of bone marrow or endothelial progenitor cells may be neoangiogenesis but not muscle regeneration. Autologous transplantation of skeletal myoblasts is confounded by the induction of life-threatening arrhythmias despite partial integration, survival and contribution to cardiac contractility. Another source of transplantable cardiomyocytes are cardiomyocytes derived from human embryonic stem cells (hES) cells. Although hES cells can be directed into the cardiomyocyte lineage, with a fetal phenotype19, differentiation is not homogenous despite recent improvements in protocols20,21. Furthermore, immunogenic, arrhythmogenic and ethical problems will limit their clinical use.


Accordingly, the technical problem underlying the present invention was the provision of means and methods for cardiomyocyte replacement therapies.


This technical problem is solved by the provision of the embodiments as characterized in the claims.


Recently, different cardiac stem cell populations have been identified in the heart2, namely, cells expressing stem cell factor receptor (c-Kit8), stem cell antigen-1 (Sca-122), or homeodomain transcription factor (islet-123) on their cell surface, side population cells (SP24) and cells able to grow in cardiospheres25.


Although rodent cardiac cell populations have been shown to be capable of differentiation into cardiomyocytes, either in vitro or in vivo, there has been no undisputed evidence to date, that any of the human cardiac stem cell populations are able to differentiate into functional mature cardiomyocytes. The different human cardiac stem cells populations isolated so far, only differentiated into cardiomyocytes in vitro when co-cultured with neonatal cardiomyocytes.


We isolated human cardiomyocyte progenitor cells (hCMPCs) from human heart tissue using an anti-Sca-1 antibody, although a bonafide Sca-1 determinant in human cells had been disputed. The cells we selected using a Sca-1 antibody from both fetal and adult human heart proved amenable to expansion in culture. The progenitor cell population isolated from the human heart were defined as immunophenotypically distinct from previously described cardiac stem and progenitor cells (reviewed in1,3). Most remarkable in our study was the isolation and culture of hCMPCs from atrial biopsies of adult patients undergoing cardiac surgery, in particular coronary artery bypass graft (CABG). The unexpectedly high frequency with which this was possible opens perspectives, e.g. for autologous transplantation at a later date then the initial surgery if cultures were carried out under clinically compatible conditions.


Several groups have isolated cell populations from the rodent and human heart8,22-25 and all of these cardiac-derived stem cell populations are distinct from the CMPCs disclosed herein, based on their origin (i.e. the part of the heart they were isolated from), their gene expression profile, their expression profile of cell surface markers and their growth characteristics. Significant differences are partly summarized in the following Table 2 and are further explained herein below.














TABLE 2







CMPC of the invention
c-kit
Isl-1
Sca-1 (mouse)




















Marker expression
Sca-1+/c-kit+/−/CD31+
Sca-1+/c-kit+/CD31−
Sca-1−/c-kit−/CD31−
Sca-1+/c-kit−/CD31−



Nkx2.5+/Gata4+/Mef2c+/−
Nkx2.5+/Gata4+/Mef2c+
Nkx2.5+/Gata4+
Nkx2.5−



(prior to differentiation)
(only when grown in diff medium)
Isl1+



Isl1+
Isl1−
ND



CD34−/CD45−
CD34−/CD45−


Isolation
Sca-1 expression
C-kit expression
co-culture with cardiac
Sca-1 expression





fibroblasts results in the





formation of Isl1+





clusters


Growth
Adherent cells when cultured
Non adherent fraction replated
co-culture with cardiac



on gelatin coated dish

fibroblasts


Location
Auricle of the heart;
Human ventricle
Human post-natal heart, atria
Mouse heart



Adult human heart in atria

ventricle and OFT



Foetal heart in atria, AV



boundary and within the



ventricle









An apparent difference which can be observed between the cells described in the art and the isolated cells (CMPCs) underlying the present invention is the expression of Sca-1 or a Sca-1 like epitope, (a term which will be explained herein below) and CD31 on the cell surface.


The present invention, thus, relates in general to isolated cardiomyocyte progenitor cells (CMPCs) which are characterized by Sca-1 or a Sca-1 like epitope and CD31 on their cell surface. “Isolated” refers to material removed from its original natural environment, and thus is altered “by the hand of man” from its natural state.


Furthermore, we provide the first evidence for functional differentiation of human myocardial derived progenitor cells into cardiomyocytes. Importantly, none of the previously described human cardiac stem cell populations have been shown to be capable of differentiation into cardiomyocytes in vitro, except when co-cultured with neonatal cardiomyocytes. Messina et al25 stimulated explants grown from cardiac biopsies with cardiotropin and generated cardiospheres. While mouse cardiospheres were spontaneously started to beat, with primitive cells at the inside and beating myocytes at the edges, human cardiospheres only differentiated when co-cultured with rat cardiomyocytes Isolated mouse Isl-1+ progenitor cells differentiated into cardiomyocytes when co-cultured with neonatal cardiomyocytes23, as was also shown for mouse cardiac SP cells32.


The present invention, thus, relates in a further embodiment to the cardiomyocyte progenitor cells (CMPCs) as described herein, wherein these cells are capable of differentiation into cardiomyocytes in the absence of co-cultured neonatal cardiomyocytes, for example in the absence of neonatal rat cardiomyocytes.


The term “in the absence of co-cultured neonatal cardiomyocytes” means that the isolated human cardiomyocyte progenitor cells (hCMPCs) of the invention have not to be cultivated in the presence of exogenously added cardiomyocytes as described in the art, to differentiate in cardiomyocytes. The “absence” however does not exclude that small portions of endogenous cardiomyocytes are still present. Small portions in this regard includes that up to about 5, 4, 3, 2, 1 or 0.5% of the total cell population as isolated by the methods of the invention as described herein, consist of endogenous cardiomyocytes, i.e. those which were isolated together with the envisaged CMPCs of the invention. These co-isolated endogenous cardiomyocytes, however, are regularly absent after about 2 passages. In order to exclude a contamination with co-isolated cardiomyocytes, it is normally sufficient to use a CPMC passage >8. To the more, it is preferred that the CMPCs which were isolated for example by the methods of the invention normally contain <5, 4, 3, 2, 1, or 0.5% contamination with other cell types, for example with fibroblasts. The quality of the CMPCs may be observed by standard staining methods in order to exclude contamination with, for example fibroblasts, if desired.


The CMPCs of the invention are preferably of human origin but it is also envisaged that these cells are derived from other mammals like for example, horse, camel, swine, goat, cattle, rabbit, cat, dog, mouse, or rat. Human CMPCs (hCMPCs) are particularly preferred.


“Human cardiomyocyte progenitor cells (hCMPCs)” of the invention are cells which can be characterised as follows:

  • (A) Morphologically, hCMPCs are less than (about) 40 μm in diameter and exhibit a high nucleus-to-cytoplasm ratio. “High” nucleus to cytoplasm ration means in this regard that at least about 50, 60, 70 or 80% of the volume of the cell is filled by the nucleus. Early isolated cells (i.e. cells at early passages, for example passage 5) are round/cuboidal cells that will elongate to a small fibroblast shape.
  • (B) Growth characteristics: hCMPCs can be clonally expanded and, after an initial lag phase of (about) 3-10 days, divide (about) once every 18-26 hours. When cultured on gelatine coated dishes, hCMPCs grow as adherent cultures.
  • (C) Biochemistry; Immunology:
    • (a) they lack CD11b, CD13, CD14, CD29, CD34, CD45, CD68, CD71, CD133, SSEA-3 and SSEA-4 when determined in FACS analysis; “lack” means that no significant levels are detectable in a standard FACS analysis
    • (b) they express Sca-1, CD105, CD31, CD59 and low levels of c-kit when determined in FACS analysis;
    • (c) they express Islet-1 (Isl-1), Nkx2.5, Gata-4 and low levels of mef2c in RT-PCR (for example when using the primers as shown in Table 1 in the appended examples and when using the PCR-protocol exemplified in Example 3)
    • (d) they lack the expression of endothelial-, smooth muscle cell-, or cardiomyocyte specific genes (VE-cadherin, Von Willebrand factor for endothelial cells; Desmin or smooth muscle cell heavy chain myosin for smooth muscle cells and cardiac actin, MLC2v for cardiomyocytes). “Lack the expression” means in this regard that the cells do not express significant levels of the genes when evaluated with Blotting or micro array techniques.
    • (e) they express the potassium inward rectifiers Kir2.1 and 2.2 as determined by RT-PCR. Kir2.x modulates the Ik1 current, involved in late repolarization and important for stabilizing the resting membrane potential of cardiomyocytes. Immature cardiomyocytes often lack, or have low Ik1 currents, resulting in higher resting membrane potentials, shorter action potentials, and increased excitability.
    • (f) they express latent TGFβ binding protein 4 (mRNA e.g. in RT-PCR and protein by western blotting), Fibroblast growth factor receptor 4 (mRNA e.g. in RT-PCR and protein by western blotting), and Vascular endothelial growth factor receptor 2 (mRNA e.g. in RT-PCR and protein by FACS).


“Cardiomyocytes” are cells which are, inter alia, characterised by the formation of cross striations, the expression of sarcomeric proteins (α-actinin, Troponin I, β-MHC, titin, desmin, MLC-2V), and atrial natriuretic factor (ANF). Cardiomyocytes are furthermore characterised by the expression of connexins on the cell membrane, by gap junctional communication, by the presence of a ventricular action potential and by excitation-contraction coupling. Cardiomyocytes normally have a diameter of over 120 μm.


The cardiomyocytes disclosed herein (e.g. those derived from the CMPCs of the invention and/or obtainable by the methods of the invention) are furthermore characterized by the following technical features which makes them unique in comparison to cardiomyocytes disclosed in the art. They are more mature then the stem cell derived cardiomcytes described so far, based on their low maximal diastolic membrane potential which is much lower then the ones from human embryonic stem cells or mouse Isl-1 derived cardiomyocytes. The maximale diastolic or resting membrane potential is caused by the difference in ionic charge across the membrane of the cell during phase 4 of the action potential. The normal resting membrane potential in the adult ventricular myocardium is about −85 to −95 mV. This potential is determined by the selective permeability of the cell membrane to various ions. Up to date, the only other reported resting membrane potential from human stem cell derived cardiomyocyte is those from embryonic stem cells. Embryonic stem cell derived cardiomyocytes have a maximal diastolic membrane potential of about −48 mV, while CMPC derived cardiomyocytes have a maximal resting membrane potential of about −67 mV or lower.


In a further embodiment, the present invention relates to the cardiomyocytes as described herein. These cardiomyocytes are obtainable by differentiating the CMPCs of the invention by way of the methods described herein. These cardiomyocytes can be comprised in a pharmaceutical and/or diagnostic composition which will be exemplified in more detail below.


To isolate CMPCs from heart tissue (fetal or adult), preferably from humans, atrial biopsies, e.g. derived from the auricle of the heart were cut into small pieces which were subsequently incubated in collagenase/protease solution in PBS with 0.5% serum either 2 hours at 37 degrees centigrade or overnight at 4 degrees centigrade. This was followed by differential centrifugation (50 g for 5 min) to separate the large cardiomyocytes from the small non-myocyte fractions in which the CMPCs are resident. The collagenase that can be used in this regard is for example type I from SIGMA (1 mg/ml) or Worthington Type II (0.5 mg/ml). This procedure resulted in a nucleated cell suspension virtually depleted of cardiomyocytes. Using an anti Sca-1 antibody coupled to magnetic beads (obtainable e.g. from Miltenyi Biotec, Sunnyvale, Calif.) a cell fraction with a diameter of about <50 μm and high growth potential was isolated by magnetic cell sorting (MACS, Miltenyi Biotec, Sunnyvale, Calif.), following the manufactures protocol. (FIG. 1b). Sca-1 positive cells were eluted from the column by washing with PBS supplemented with 2% fetal calf serum (FCS) and cultured on 0.1% gelatin coated dishes in M199 (Gibco)/EGM (3:1) supplemented with 10% FCS (Gibco), 10 ng/ml basic Fibroblast growth factor (bFGF), 5 ng/ml epithelial growth factor (EGF), 5 ng/ml insulin like growth factor (IGF-1) and 5 ng/ml hepatocyte growth factor (HGF). After a lag phase of 3 days, the cells started to proliferate and colonies formed from small spindle-shaped cells with a high nucleus-to-cytoplasm ratio (FIG. 1a). hCMPCs could be clonally expanded. After limited dilution, single cells grown in a 96-well plate generated colonies with a 5-25% efficiency.


We have also used the following methods to isolate CMPCs:

  • (a) the nucleated cell suspension virtually depleted of cardiomyocytes were incubated with Sca-1 antibody from R&D (according to the manufacturers instructions) and we came back with rat secondary antibody coupled on magnetic beads to isolate Sca-1 positive cells; or
  • (b) we have also used a Sca-1 antibody labelled with FITC and either FACS-sorted the cells, or used an anti-FITC antibody coupled to magnetic beads to isolate Sca-1 positive cells.


“Sca-1 antibodies” that can be used in accordance with the present invention are exemplified in the following but it has to be understood that the methods of the invention are not limited to these specific antibodies:


Rat monoclonal Scat antibody clone D7 (Abcam), Rat-anti-Mouse Ly6a/e clone D7 (cederlane, eBioscience), Rat monoclonal Sca-1 clone 177228 (R&D system), Anti-Mouse Sca-1 (Ly-6A/E) Monoclonal Antibody, Clone 38/42 Leinco Technologies (Rat Anti-Ly-6A/E Monoclonal Antibody, Clone E13-161.7 (Pharmingen, Stemcell technologies). These antibodies can either be not conjugated, or conjugated to biotin, FITC, TRITC or other well-known labels known in this field (Immunology) and labels to that a secondary isolation step can be performed to. Such labels and secondary isolation steps are mentioned herein and are also well-known to the skilled person. It is, for example possible to use an FITC-labelled Sca-1 antibody. The Sca-1 positive cells are thus sortable, e.g., by FACS. The explanations given herein equally apply to other antibodies which might be used alternatively or in combination with Sca-1, e.g., CD31 antibodies or functional fragments thereof. It is thus also possible to sort the CMPCs of the invention with CD31 antibodies or a combination of CD31 and Sca-1 antibodies.


The present invention relates in a further aspect to a method for the isolation of cardiomyocyte progenitor cells (e.g. human CMPCs), said method comprising or essentially consisting of the steps of:

    • (a) dissociate heart tissue e.g. the auricle of the heart; and
    • (b) enrichment of CMPCs.


“Heart tissue” includes all parts of the heart which comprise the CMPCs of the invention. Said heart tissue preferably is derived from the atrium or parts of the atrium of the heart, e.g. the auricle. The term “auricle” relates to a small conical pouch that projects from each atrium of the heart.


The dissociation step of the heart tissue as described herein elsewhere, which is preferably enzymatically but can also be mechanically, is optionally followed by a differential centrifugation step or comparable methods like for example a centrifugation in a Percoll gradient. Such methods are well known to the skilled person and are explained in detail in standard laboratory handbooks like Sambrook et al. A differential centrifugation or a comparable method causes the separation of mixtures such as cellular particles in a medium at various centrifugal forces to separate particles of different density, size, and shape from each other. Such a step will result in a nucleated cell suspension virtually depleted of cardiomyocytes. The differential centrifugation which may be used in the methods of the invention could be exemplarily performed with a force of about 50 g for about 5 min. “Virtually depleted” means in this regard that small portions of other cells, like cardiomyocytes, can be present in the non-myocyte fraction, where the CMPCs are also present. Small portions in this regard includes that up to about <5, 4, 3, 2, 1, or 0.5% of the total cell population as isolated by the methods of the invention consists of other cells, i.e. not the envisaged CMPCs of the invention. Following the separation, CMPCs will be sorted from the non-myocyte fraction. As mentioned earlier, it is preferred that the CMPCs which were isolated for example by the methods of the invention normally contain <5, 4, 3, 2, 1, or 0.5% contamination with other cell types, for example with fibroblasts. The quality of the CMPCs may be observed by standard staining methods in order to exclude contamination with, for example fibroblasts, if desired.


Methods to dissociate heart tissue, for example the auricle of the heart, either enzymatically or mechanically, are well-known to the skilled reader and to the more exemplified in the appended examples.


In a preferred embodiment of the methods of the invention, enzymatically dissociation is used, and the enzyme is preferably a collagenase although other proteases might also be employed. The tissue can for example also be dissociated with a combination of Collagenase (Worthington) and Pancreatine (Alkemi A0585,0100 Applichem). The Sca-1 epitope is, however, sensitive for trypsin and thus trypsin should be avoided if it is intended to isolate the cells with anti-Sca-1 binding agents as described herein elsewhere. It will be understood, however, that other enzymatically dissociation methods are also known which may be used alternatively for example based on the manufacturers instructions and/or based on standard laboratory books. Protocols for tissue dissociations are known in the art and frequently published for example in laboratory handbooks. These dissociation-methods are also included in the scope of this invention as long as the methods result in dissociated cell suspensions, which comprise the cardiomyocyte progenitor cells (CMPCs) of the invention.


“Sca-1” is a mouse-antigen, particularly an 18 kDa phosphatidylinositol-anchored protein which is a member of the Ly-6 family of GPI-linked surface proteins. The Ly-6 family is involved in regulation and function of T cell activation. Sca-1 is a major phenotypic marker for mouse hematopoietic progenitor/stem cell (HSC) and has been used as a marker of HSC in mice of both Ly-6A/E haplotypes. The corresponding “human” counterpart of Sca-1 is not yet identified. A “Sca-1-like epitope” thus relates to orthologs or analogs of the mouse Sca-1. “Orthologs” have evolved from a common ancestral gene by specification and thus relate to the “same” protein in a different species, while “analogs” refers to two proteins that have the same or similar function, but that have evolved separately. Normally, orthologs or analogs of mouse Sca-1 are polypeptides which have the same or similar functions but differ from the mouse Sca-1 e.g. by post-translational modifications, by amino acid sequence differences, or by both. Thus, cardiomyocyte progenitor cells (hCMPCs) are within the scope of this invention which have an epitope, i.e. a “Sca-1 like epitope” on their surface which is characterized by its ability to specifically react with Sca-1 antibodies, preferably those exemplified above, in such a way that it is at least possible to sort these “Sca-1” positive cells by routine sorting methods also described herein (for example MACS). It has to be understood that “Sca-1” on cells, (besides mouse cells), is used herein interchangeably with “Sca-1-like epitope” on these cells (besides mouse cells which express the Sca-1 epitope).


A “Sca-1 binding agent” is preferably an antibody or a functional fragment thereof, which is capable of binding to the Sca-1 epitope or the Sca-1 like epitope described herein. The “binding” in this regard means that the Sca-1 positive cells (e.g. the cells of the invention) are bound by the binding agent in such a way that the cells can be subsequently sorted by means and methods well known in the art and also described herein. The minimum requirement of a “binding agent” as used herein is, thus, that it is able to bind specifically to Sca-1 positive cells and/or cells expressing a Sca-1 like epitope and allows for the sorting or separation of these cells. The term “specifically binding” in connection with the binding agent used in accordance with the present invention means that the binding agent, e.g. the antibody etc. does not or essentially does not cross-react with other polypeptides. Antibodies that bind to the Sca-1 or a Sca-1 like epitope but do not or do not essentially bind to other polypeptides on the cell surface are considered specific and are, therefore, preferably selected for the methods of the invention.


In a preferred embodiment of the methods of the invention, said CMPCs are enriched with a Sca-1 binding agent. In a particularly preferred embodiment of the method of the invention, said binding agent is an anti-Sca-1 antibody or a functional fragment thereof.


The term “enriched” means that the relative number of the Sca-1 positive cells is increased in the cell suspension after the cell sorting when compared to the other cells (e.g. cardiomyocytes) which were also contained in the initial cell suspension. Preferably, the CMPCs are enriched, i.e. their relative cell number is increased to at least 50, 60, 70, 80, 90, 95% or even more. Protocols for such an “enrichment” are exemplified herein, for example the methods to isolate CMPCs as described herein.


As described herein above, the cardiomyocyte progenitor cells (CMPCs) are characterized by Sca-1 or a Sca-1 like epitope and CD31 on their cell surface. Thus, it is also envisaged that the CMPCs of the invention are enriched with a CD31 binding agent, e.g. an CD31 specific antibody or functional fragments thereof. The explanations given in the section above for example those which relate to the scope and meaning of “binding agent” and “functional fragments of antibodies” and “specific binding” equally apply to the CD31 marker which is also detectable on the CMPCs of the invention. To the more, each marker which is present on the surface of the CMPCs of the invention (e.g. CD105) might be used alternatively or in combination to the afore mentioned ones (CD31 and Sca-1) to enrich CMPCs, provided that this enrichment results in a cell fraction wherein the relative cell number of the CMPCs as described herein is increased to at least 50, 60, 70, 80, 90, 95% or even more.


In a further embodiment, it is also envisaged to enrich the cells with a Sca-1 and an CD31 binding agent, e.g. antibodies directed to these epitopes. Means and methods to detect such bound antibodies or fragments thereof are well-known and also described herein.


Anti-Sca-1 antibodies are well known in the art and also exemplified above. The list of these specific Sca-1 antibodies is however exemplarily and in no way limits the group of Sca-1 antibodies which might be employed when carrying out the methods of the invention.


CD31 antibodies are also well-known and exemplified by mouse α-CD31 monoclonal antibody clone WM59 (BD biosciences), α-human CD31 clone 1F11 (Beckman coulter), mouse α-CD31 clone 158-2B3 (Cell signaling), α-CD31 clone HC1/6 (Chemicon), mouse α-human CD31 clone 9G11 (R&D systems).


CD105 antibodies are also well-known and exemplified by mouse α-CD105 clone 35 or clone 266 (BD biosciences), Goat α-human CD105 polyclonal antibody (R&D systems), mouse α-human CD105 clone 166707 of clone 166713 (R&D systems), α-CD105 clone 8E11 or clone P3D1 (Chemicon).


Other antibodies which are specific for Sca-1 or a Sca-1 like epitope or CD31 or CD105 or other epitopes useful in the methods of the invention can be easily produced by methods well-known in the art. For example it is possible to use cell lines secreting antibodies to essentially any desired substance that produces an immune response. RNA encoding the light and heavy chains of the immunoglobulin can then be obtained from the cytoplasm of the hybridoma. The 5′ end portion of the mRNA can be used to prepare cDNA to be inserted into an expression vector. The DNA encoding the antibody or its immunoglobulin chains can subsequently be expressed in cells, preferably mammalian cells. Depending on the host cell, renaturation techniques may be required to attain proper conformation of the antibody. If necessary, point substitutions seeking to optimize binding or stability of the antibody may be made in the DNA using conventional cassette mutagenesis or other protein engineering methodology such as is disclosed herein. Furthermore, antibodies or fragments thereof to the aforementioned epitopes (Sca-1) can be obtained by using methods which are described, e.g., in Harlow and Lane “Antibodies, A Laboratory Manual”, CSH Press, Cold Spring Harbor, 1988. Other suitable antibodies as well as methods for testing the effectiveness of such antibodies are detailed in WO02071061.


For the production of antibodies in experimental animals, various hosts including goats, rabbits, rats, mice, and others, may be immunized by injection with polypeptides of the present invention or any fragment or oligopeptide or derivative thereof which has immunogenic properties. Techniques for producing and processing polyclonal antibodies are known in the art and are described in, among others, Mayer and Walker, eds., “Immunochemical Methods in Cell and Molecular Biology”, Academic Press, London (1987). Polyclonal antibodies also may be obtained from an animal, preferably a mammal. Methods for purifying antibodies are known in the art and comprise, for example, immunoaffinity chromatography. Depending on the host species, various adjuvants or immunological carriers may be used to increase immunological responses. When derivatives of said antibodies are obtained by the phage display technique, surface plasmon resonance as employed in the BIAcore system can be used to increase the efficiency of phage antibodies which bind to an epitope of the peptide or polypeptide of the invention (Schier, Human Antibodies Hybridomas 7 (1996), 97-105; Malmborg, J. Immunol. Methods 183 (1995), 7-13). In many cases, the binding phenomena of antibodies to antigens is equivalent to other ligand/anti-ligand binding.


The production of chimeric antibodies is described, for example, in WO89/09622. A further source of antibodies to be utilized in accordance with the present invention are so-called xenogenic antibodies. The general principle for the production of xenogenic antibodies such as human antibodies in mice is described in, e.g., WO 91/10741, WO 94/02602, WO 96/34096 and WO 96/33735.


The production of recombinant antibodies is described, for example, in R. Kontermann, S. Dübel: Antibody Engineering, Springer Lab Manual 2001. The antibody which is used in accordance with the uses or methods of the invention may be a monoclonal or a polyclonal antibody (see Harlow and Lane, “Antibodies, A Laboratory Manual”, CSH Press, Cold Spring Harbor, USA, 1988) or a derivative of said antibody which retains or essentially retains its binding specificity. Preferred derivatives of such antibodies are chimeric antibodies comprising, for example, a mouse or rat variable region and a human constant region.


The term “functional fragment” as used herein refers to fragments of the antibodies as specified herein which retain or essentially retain the binding specificity of the antibodies like, separated light and heavy chains, Fab, Fab/c, Fv, Fab′, F(ab′)2. The term “antibody” also comprises bifunctional antibodies and antibody constructs, like single chain Fvs (scFv) or antibody-fusion proteins. The term “scFv fragment” (single-chain Fv fragment) is well understood in the art and preferred due to its small size and the possibility to recombinantly produce such fragments.


The binding agent or antibody etc. may be affixed to a carrier, for example to magnetic beads, column material or solid carriers like polystyrene plates or the like, which makes it possible to separate cells which specifically bind to the so affixed binding agents. Means and methods in this regard are well-known to the skilled person and furthermore explained in the appended examples.


In a preferred embodiment of the methods of the invention, the CMPCs are enriched by MACS.


“MACS” means magnetic activated cell sorting and is a well-known and reliable method for the separation of cells according to specific cell surface markers. Cells stained sequentially e.g. with biotinylated antibodies, fluorochrome-conjugated avidin, and superparamagnetic biotinylated-microparticles (about 100 nm diameter) are separated on high gradient magnetic (HGM) columns. Unlabelled cells pass through the column, while labelled cells are retained. The retained cells can be easily eluted. For example the Anti-Sca-1 MicroBead Kit provided by Miltenyi Biotec includes all reagents for fluorescent and indirect magnetic labelling of Sca-1+ cells, using Anti-Sca-1-FITC and Anti-FITC MicroBeads. This Kit was used in the appended examples, but it will be understood that also other MACS-systems are at hand which might also be used.


In a further preferred embodiment of the methods of the invention, the CMPCs are enriched by FACS. “FACS” means fluorescent-activated cell sorting which is a well-known method. As cells or particles pass through the FACS-instrument they can be selectively charged, based on user defined parameters, and can be deflected into separate paths of flow directed to different collection tubes. It is therefore possible to separate defined populations of cells from an original mix with a high degree of accuracy and speed by way of antibody-labeling of the cell suspension.


It will be understood that the other enrichment-methods are also contemplated, e.g. Enzyme-Linked ImmunoSorbent (ELISA)-techniques or columns with antibodies affixed thereto etc.


In a further embodiment, the present invention relates to the CMPCs as defined herein, which are obtainable or obtained by the method(s) as described herein.


To phenotypically characterize the isolated CMPCs of the invention which were enriched by the methods disclosed herein, freshly isolated cells were examined for cell surface expression of haematopoietic markers (CD45 and CD34), stem cell marker (c-Kit), CD105 and CD31 (Pecam-1) by FACS analysis. To this end several antibodies were used which are detailed herein in the examples. Such antibodies are well-known to the skilled reader (see for example the catalogues and WebPages of the respective manufacturers of such antibodies for example Beckmann Coulter) but might be replaced by other antibodies which are also able to detect the mentioned epitopes. The isolated CMPCs were negative for CD34, CD45, but exhibited expression of CD31, CD105 and moderate levels of c-kit (FIG. 1c) when tested under the experimental conditions exemplified in the appended examples.


RT-PCR analysis of the cells revealed that they do not express Oct4, a marker for pluripotent ES cells. These results indicated that the cardiac derived progenitor cells disclosed herein do not belong to the haematopoietic stem cell or previously described cardiac stem cell population.


To further characterize CMPCs of the invention, the expression of genes specific for early cardiac development was examined by RT-PCR using cultured cells that had been passaged 8-10 times (FIG. 1d) and using the primers as listed in Table 1. As can be seen in FIG. 1d, cultured hCMPCs express c-Kit and Islet-1 as well as GATA-4, Nkx2.5 and Mef2C, suggesting that they are more restricted to the cardiac lineage. Besides mRNA, CMPCs also expressed GATA-4, Nkx2.5 and Islet-1 protein (FIG. 1e), although they did not express cardiac genes, like bMHC, α-actinin, ANF, MLC2A or MLC2V (FIG. 1d). Interestingly, the subcloned hCMPCs showed the same characteristics as the nonclone-derived CMPCs.


Also adult hCMPCs were isolated from enzymatically dissociated atrial biopsies, using an anti Sca-1 antibody and the methods described hereinbefore. Seeding these cells on gelatin-coated dishes in culture medium resulted in growing progenitor colonies in 24 out of 33 cases. Adult hCMPCs had the same stem-cell-like morphology with a large nucleus and little cytoplasm; no loss of proliferation potential was noted after 23 passages. Three to 6 weeks after isolation adequate numbers of cells were obtained for further analysis. FACS-analysis revealed that cultured adult hCMPCs were CD34 negative/CD45 negative and 5% of the cells expressed c-kit. The gene-expression profile was analyzed by RT-PCR and revealed that adult hCMPCs had the same expression profile as fetal derived hCMPCs (FIG. 1). Immunofluorescent staining revealed that almost all adult cells were Islet-1+, although predominantly cytoplasmic.


We have also established differentiation of CMPCs of the invention into tubular vascular structures (angiogenesis) on Matrigel, said “tubular vascular structures” which are sometimes also denoted herein as “tubular structures” containing aligned endothelial cells encircled by smooth muscle cells. The formation of these newly formed capillary tubes (i.e. the “tubular structures” containing aligned endothelial cells encircled by smooth muscle cells) can be induced by the addition of VEGF (see Example 8). The formation of these newly formed capillary tubes can be visualised for example by staining α-SMA (a smooth muscle actin) on smooth muscle cells and/or PECAM-1 (platelet endothelial cell adhesion molecule-1, CD31) on endothelial cells (see also appended Example 8). Antibodies which can be used in this regard are exemplified in the following: SMA antibody: AB5694 from abcam, clone 1A4, Sigma, cat. Number: A2547, or Dako, cat. Number M08510. Endothelial cells: PECAM-1 clone 9G11 or CD34 clone Qbend/10 form Biogenex or CD31 from Sigma and ve-cadherin from Santa Cruz.


Angiogenesis is the generation and formation of new capillary blood vessels, a process fundamental for processes like wound healing and reproduction. It's also involved in pathological processes (rheumatoid arthritis, tumor growth and metastasis).


“Matrigel” is the trade name for a gelatinous protein mixture secreted by mouse tumor cells and marketed by BD Biosciences. This mixture resembles the complex extracellular environment found in many tissues and is used as a substrate for cell culture.


The present invention thus relates in a further aspect to an in vitro method for the differentiation of CMPCs into tubular structures or vascular sprouts comprising the steps of:


(a) providing the CMPCs of the invention, preferably on Matrigel and (or any other gelatinous protein mixture which resembles the complex extracellular environment; and


(b) optionally treating said CMPCs with an biologically active agent having the capabilities to induce angiogenesis or sprouting angiogenesis (like e.g. VEGF).


It is preferred that the CMPCs to be differentiated in the above in vitro method for the differentiation of CMPCs into tubular structures or vascular sprouts are seeded on Matrigel and/or any other gelatinous protein mixture which resembles the complex extracellular environment.


In a further embodiment, the present invention relates to the “tubular structures” or vascular sprouts as described herein. These “tubular structures” or “vascular sprouts” are obtainable by differentiating the CMPCs of the invention by way of the methods described hereinabove. These “tubular structures” or “vascular sprouts” can be comprised in the pharmaceutical and/or diagnostic compositions of the present invention and may be furthermore used in the screening assays/methods of the present invention.


The present invention relates in a further aspect to the use of the CMPCs defined herein for the provision of cardiomyocytes and/or tubular structures and/or vascular sprouts.


In preferred embodiments of the uses, methods etc. described herein the CMPCs of the invention are propagated/expanded in vitro (exemplified in Example 1/culture conditions of the Sca-1 isolated cells) in order to further expand their cell number, before the CMPCs are actually differentiated into the cardiomyocytes or into tubular structures or vascular sprouts which were mentioned before.


We have also established differentiation of CMPCs of the invention into adipocytes. To this end, CMPCs of the invention are seeded and cultured until they have reached a confluence of 90-100% in SP++ (SP++: EBM-2+EGM-2 single qouts Cat. No. CC-4176+2% FBS). Subsequently, the medium is replaced by DMEM 4.5 g/l glucose+Na pyruvaat containing 10% FBS, 1 uM dexamethasone, 0.5 mM 3-Isobutyl-1-methylxanthine (IBMX), 10 ug/ml Insulin, 0.2 mM indomethacin and Pen/strep. After some days, droplets of fat are deposited around the nucleus. Every 3 days the medium is refreshed.


The present invention thus relates in a further aspect to an in vitro method for the differentiation of CMPCs into adipocytes comprising the steps of:


(a) providing the CMPCs of the invention, and


(b) treating said CMPCs with IBMX, indomethacin and insulin.


For a cell type to be suitable for cardiomyocyte replacement therapies, it should not only be able to proliferate but also differentiate efficiently into cardiomyocytes. To study the capacity of the hCMPCs of the invention to differentiate into cardiomyocytes, cells were plated at 105 per cm2 in differentiation medium containing ascorbic acid and treated with 5-azacytidine (which is a demethylating agent). Three weeks after treatment, hCMPCs formed elongated aggregates and spontaneously beating areas were observed. We thus demonstrated that hCMPCs are able to differentiate into mature cardiomyocytes in vitro after 5-azacytidine treatment in the presence of an antioxidant namely ascorbic acid, even after prolonged passage.


The present invention thus relates to an in vitro method for the differentiation of CMPCs into cardiomyocytes, comprising the steps of:

    • (a) providing the CMPCs of the invention; and
    • (b) treating said CMPCs with a demethylating agent.


As mentioned before, the CMPCs of the invention are surprisingly able to differentiate into cardiomyocytes even in the absence of co-cultured neonatal cardiomyocytes. It is thus preferred that these differentiation methods of the invention are carried out in the absence of co-cultured neonatal cardiomyocytes. It is also envisaged that all the differentiation methods described herein (cardiomyocytes and/or tubular structures) can be carried out in the absence of co-cultured neonatal cardiomyocytes.


DNA methylation is an epigenetical mechanism that plays crucial roles in cellular differentiation and tissue development in embryogenesis. A “demethylating agent” as used herein is an agent which is able to demethylate DNA, for example by way of inhibiting the intracellular DNA methyltransferase(s). Such agents are well-known to the skilled person and include for example 5-aza-cytidine or 5-aza-2′-deoxycytidine, which are preferred, but the invention is not limited thereto.


In a preferred embodiment, said hCMPCs are treated with the demethylating agent mentioned above in the presence of an antioxidant agent, preferably ascorbic acid. An “antioxidant agent” in this regard refers to an anorganic or organic chemical that reduces the rate of oxidation reactions. Further examples for antioxidants which might be used in the context of the present invention are 2-mercapthoethanol, tocopherol or BHA but it will be understood that further antioxidant agents might be used as long as they exert the desired effect of cell differentiation described herein.


In a preferred embodiment, the present invention thus relates to the CMPCs defined hereinabove, said cells being capable of differentiation into cardiomyocytes in vitro after 5-aza-cytidine or 5-aza-2′-deoxycytidine treatment in the presence of ascorbic acid.


Apart from antioxidant agents like ascorbic acid27, TGFβ family members, including TGFβ and BMP, have also been found to promote cardiomyogenic differentiation in ES cells, and be critical for the expression of cardiac specific markers.9,10,26,28. Furthermore, TGFβ stimulated myogenic differentiation of c-kit+ bone marrow stem cells29. Therefore, we explored the capacity of TGFβ to improve cardiomyogenic differentiation of hCMPCs.


To determine whether CMPCs of the invention were capable of responding to TGFβ ligand we examined their ability to phosphorylate smad proteins. TGFβ exerts its effect by binding to a complex of type I and type II receptor and phosphorylating Smads30. TGFβ phosphorylates Smad2 while BMP phosphorylates Smad1. As shown in FIG. 3a CMPCs show clear BMP induced Smad1 phosphorylation as well as TGFβ induced Smad2 phosphorylation indicating that they express the appropriate TGFβ and BMP receptor combinations to respond to ligand stimulation. To examine the effect of TGFβ on cardiomyocyte differentiation, hCMPCs were plated at 2×104 cells per cm2 in differentiation medium, treated with 5-aza for 24 hours and cultured for up to 3 weeks in differentiation medium supplemented with TGFβ (1 ng/ml). RT-PCR analysis showed that expression of several cardiac genes like Troponin-I, ANP, cardiac-actin and desmin was already observed 7 days after the initiation of differentiation in the presence of TGFβ, but these genes were undetectable in its absence (FIG. 3B). MLC2v was normally detected in cultures without TGFβ only after 3 weeks but was already present after two weeks in its presence. Western blot analysis showed that the expression of Nkx2.5 and α-actinin in CMPCs increased dramatically after 7 days of culture in the presence of TGFβ (FIG. 3c). Immunofluorescent staining demonstrated that the percentage of α-actinin positive cells increased to nearly 100% (FIG. 3d). Addition of an ALK5 kinase inhibitor to the cell cultures known to block TGFβ signaling31, inhibited the differentiation of hCMPCs into cardiomyocytes (data not shown). These results indicated that TGFβ stimulation increases the differentiation of hCMPCs into cardiomyocytes to nearly 100%.


Accordingly, it is preferred that the in vitro methods for the differentiation of CMPCs into cardiomyocytes as defined herein further or alternatively comprise the step of treating the CMPCs with a TGF-β family member. TGFβ exists in at least three known subtypes in humans, TGF-β1, TGF-β2 and TGF-β3. The TGFβ superfamily includes several proteins, for example TGF-β, activin and bone morphogenetic proteins (BMPs) BMP2 through BMP7. A member of the TGFβ-family thus includes any of the aforementioned substances either alone or in any desired combination. It is also envisaged to provide recombinant cells which express and secrete one or more members of the TGFβ-family instead of providing/adding the TGFβ-family member per se. Examples of recombinant cells include the CMPCs and/or cardiomyocytes or any other cell which is (a) able to express and secrete a member of the TGF-β family and (b) is biocompatible either with the CMPCs/cardiomyocytes and/or with the subject to be treated (see for example the description of the pharmaceutical compositions below).


The present invention thus relates to an in vitro method for the differentiation of CMPCs into cardiomyocytes, comprising the steps of:

    • (a) providing the CMPCs of the invention; and
    • (b) treating said CMPCs with a demethylating agent and/or an antioxidant agent and/or a TGF-β family member.


It will be understood that the so-treated CMPCs are treated for a time which is sufficient to allow the CMPCs to differentiate into cardiomyocytes. The differentiation can be determined for example by immunofluorescence analysis of contractile protein expression, e.g. of α-actinin. Furthermore, it will be understood that if the cells start to beat, they are differentiated. The cross-striations can also be determined using light microscopy. It is also envisaged to modify the CMPCs using a cardiomyocyte specific promoter coupled to a detectable marker gene for example to GFP so that one can follow the differentiation of the cells more easily. Suitable cardiac promoters are for example human cardiac actin promoter and the Mlc-2V promoter.


Four weeks after stimulation, mRNA expression of cardiac differentiation markers was analyzed by RT-PCR. Before differentiation, only GATA-4 and Nkx2.5 were expressed, while after 4 weeks of stimulation, hCMPCs-derived cardiomyocytes expressed the CM-specific genes bMHC, ANP, Troponin I and cardiac α-actin (FIG. 2a). Moreover, no Myo-D mRNA could be detected, indicating that there was no differentiation towards skeletal myocytes. To characterize the cardiomyocytes further, immunofluorescent staining for sarcomeric proteins was carried out. hCMPC-derived cardiomyocytes exhibited sarcomeric striations when double stained with α-actinin and troponin-I, organized in separated bundles (FIG. 2b). Labeling of the cells with antibodies raised against other components of the contractile apparatus revealed a pattern of cross striations positive for titin, desmin, MLC2v, and β-MHC (FIG. 2c). Double labeling of α-actinin with the intercalated disk related proteins like N-cadherin or ZO-1 resulted in strong positive signals at the cellular boundaries of nearly all cardiomyocytes indicative of stable mechanical coupling (FIG. 2c).


The cardiomyocytes of the present invention i.e. those obtainable by the in vitro methods for the differentiation of CMPCs into cardiomyocytes as defined herein, are thus characterized by the expression of β-MHC, ANP, α-actinin, cardiac α-actin, Troponin I and/or MLC2v cardiac mRNA detected by RT-PCR and/or by the absence of Myo-D and/or Isl-1 mRNA detected by RT-PCR.


The cardiomyocytes disclosed herein furthermore are more mature then the ones described so far, based on their low maximal diastolic membrane potential which is much lower then the ones from human embryonic stem cells or mouse Isl-1 derived cardiomyocytes.


To further characterize the cardiomyocytes of the invention, we performed patch-clamp electrophysiology. Action potentials recorded from single cardiomyocytes and cardiomyocytes in small clusters (n=11) had a ventricle-like shape (FIG. 4A), as confirmed by quantification of action potential parameters (FIG. 4B). Average maximal diastolic potential was −67±8 mV and average maximal upstroke velocity was 39±4 V/s. Average APD50 and APD90 were 258±38 and 483±51 ms, respectively. Furthermore, diastolic membrane potential was rather stable, unlike cardiac pacemaker cells. Excitation-contraction coupling was demonstrated using combined calcium-imaging and current clamp experiments (FIG. 4C). Average time to peak of the transients (n=7) was 447±54 ms, time of half-inactivation was 568±42 ms.


The cardiomyocytes of the present invention i.e. those obtainable by the in vitro methods for the differentiation of CMPCs into cardiomyocytes as defined herein, are thus further characterized by their ability to show excitation-contraction coupling. Excitation-contraction coupling (ECC) is the process by which an action potential triggers a myocyte to contract.


Cardiomyocytes, suitable for transplantation, should be able to couple correctly and propagate action potentials. Therefore, we analyzed the presence of gap junctional communication in hCMPCs. Semi-quantitative RT-PCR showed that undifferentiated hCMPCs express Cx40, Cx43 and Cx45. Connexin (Cx) are structural proteins needed to form gap junctions in the cell membrane. Upon differentiation into cardiomyocytes expression of Cx40 and 43 but not Cx45 is down regulated (FIG. 5a). Immunohistochemical analysis showed that while undifferentiated hCMPCs express Cx 40, 43 and 45 in a diffuse and predominantly intracellular pattern (FIG. 5b), hCMPCs-derived cardiomyocytes express Cx40 and 43 at the cell borders in a typical gap junctional pattern. Cx45 was found both intracellular and at the cell membrane. Metabolic coupling was measured by Lucifer Yellow injection in monolayers of undifferentiated and differentiated hCMPCs, resulting in spreading of dye to 6±1 (n=14) and 17±3 (n=8) cells, respectively (FIGS. 6A and B). The difference in dye spreading was statistically significant (p<0.007).


Macroscopic gap junctional conductance in cell pairs of undifferentiated and differentiated CMPCs was 48±21 nS (n=5) and 31±4 nS (n=5), respectively (FIG. 6C). Differences in average conductance were not statistically significant. A representative recording of differentiated hCMPCs (FIG. 6D) shows a mild voltage dependent inactivation of gap junctional conductance starting at a Vj of +30 mV indicative for Cx43 channels the predominantly expressed isoform. These results indicate gap junctional communication between hCMPC derived cardiomyocytes.


Analysis of hCMPC cardiomyocyte action potential shape showed a low maximal diastolic potential, as compared to values we measured in human embryonic stem cell-derived cardiomyocytes, human fetal ventricular cardiomyocytes and mouse P19 embryonic carcinoma cell-derived cardiomyocytes (−67±8 mV compared to −48±2, −38.5±1.6 and −54±2 mV, respectively)19,34. Furthermore, and possibly as a consequence, the maximal upstroke velocity is higher (39±4 V/s compared to 7.0±0.8, 8.9±4.3 and 10.0±1.0 V/s). APD90 of hCMPCs is comparable with values measured in human embryonic stem cell-derived cardiomyocytes and human fetal ventricular cardiomyocytes (483±51 ms compared to 436.4±55.3 and 370.0±45.8 ms). hCMPCs express high levels of gap junction proteins both in the differentiated and the undifferentiated state. In hCMPC cardiomyocytes, expression is almost exclusively found on the plasma membrane. The expression pattern reflected functional gap junction channels as confirmed by a robust metabolic and electrical coupling. Interestingly, hCMPC cardiomyocytes have a phenotype that is even more mature than cultured ventricular cardiomyocytes isolated from 16 week human foetuses19, showing that our differentiation strategy enhances maturation of the cardiomyocytes.


Without being hereby bound to any theory, we consistently and repeatedly find. APD-90 values in the CMPCs as isolated with the methods described herein that are higher then week 16 myocytes as explained above. It thus appears that the cardiomyocytes derived from our CMPCs are more mature then the ones which can be found in a 16 week foetus. The differentiation potential of the CMPCs described herein is thus towards more mature cells.


In terms of future therapy, transplantation of poorly coupled skeletal myoblasts in human hearts in a clinical trial resulted in ventricular tachyarrhythmias in some patients14. Proper intercellular coupling with host heart cells will be necessary in order to preserve conduction characteristics and will be among the most important criteria for determining whether hCMPCs can be taken forward to clinical trials. The combination of high upstroke velocity, low maximal diastolic potential and efficient gap junctional communication suggests that hCMPC cardiomyocytes will sufficiently support impulse propagation when transplanted. The characterization of the hCMPCs in this study identified a distinct population of cardiac progenitor cells that can be used to study human cardiomyocyte differentiation, and serve as a suitable source for transplantable human cardiomyocytes.


Accordingly, the present invention relates to a pharmaceutical composition comprising the cardiomyocytes of the invention which are obtainable by the methods of the invention by use of the CMPCs as defined herein. The pharmaceutical composition of the invention optionally comprises a pharmaceutically active carrier and/or diluent. Examples of suitable pharmaceutical carriers are well known in the art and include phosphate buffered saline solutions, water, sterile solutions etc. Compositions comprising such carriers can be formulated by well known conventional methods.


It is also envisaged to provide a pharmaceutical composition comprising the “tubular vascular structures” and/or “vascular sprouts” of the invention which are obtainable by the methods of the invention by use of the CMPCs in the respective differentiation methods as defined herein.


In a further aspect, the present invention relates to a pharmaceutical composition comprising the CMPCs as described herein and/or to a Kit comprising this pharmaceutical composition, wherein said kit and/or pharmaceutical composition further comprises instruction manuals which guide the skilled person to differentiate these CMPCs into cardiomyocytes and/or the tubular structures and/or the vascular sprouts of the invention, for example by use of the differentiation methods of the invention. To this end, this kit and/or pharmaceutical composition may further comprise the substances/chemicals and/or cell culture equipment (culture dishes; antibodies for quality staining, differentiation agents etc.) which are useful for the differentiation protocol and, optionally, a solvent, diluent, buffer for stabilizing and/or storing the inventions compounds. To this end, it is envisaged that the pharmaceutical composition comprising the CMPCs of the invention and/or the kit comprising this pharmaceutical composition further comprises the demethylating agents, antioxidant agents members of the TGF-β family; VEGF etc. as described herein in detail in the context of the differentiation methods of the invention.


It is also envisaged to keep/maintain/encapsulate the CMPCs comprised in the pharmaceutical composition and/or the kit described in the paragraph above in, for example, biodegradable grafts. Examples of the biocompatible/biodegradable material have been exemplified herein elsewhere. To this end, it is also envisaged to keep/maintain/encapsulate the CMPCs of the invention together with members of, for example the TGF-β family (when aiming to differentiate into cardiomyocytes) and/or VEGF (when aiming to differentiating into the “tubular structures” or “vascular sprouts”) in the biocompatible/biodegradable material/graft in order to achieve the desired differentiation of the CMPCs at their envisaged destination (see the paragraph below). It is envisaged that the members of the TGF-β family and/or VEGF are contained per se within the biocompatible/biodegradable material/graft together with the CMPCs of the invention and/or to add recombinant cells which are able to express and secrete these TGF-β family members and/or VEGF, preferably in the vicinity of the CMPCs of the invention. The mentioned recombinant cells are preferably also CMPCS and/or cardiomyocytes comprising the desired member of the TGRβ-family and/or VEGF as an expressible nucleic acid, preferably under the control of a suitable promoter.


It will be understood that it is also possible to further encapsulate the TGFβ-member and/or VEGF for example into microcapsule structures. Microencapsulation can be used to slow the release of the TGFβ family member and/or VEGF into the body. This may decrease toxic side effects by preventing high initial concentrations in the subject. The release pattern is typically first-order in which the rate decreases exponentially with time until the drug source is exhausted. In this situation, a fixed amount of drug is inside the microcapsule. The concentration difference between the inside and the outside of the capsule decreases continually as the contained agent diffuses.


Consequently, it is envisaged that the last step of the differentiation of the CMPCs may occur by way of providing a pharmaceutical composition comprising the CMPCs of the invention, preferably pre-differentiated with a demethylating agent and more preferred also in the presence of an antioxidant agent (these differentiation steps have been described herein before and the so-treated cells are denoted “pre-differentiated” in the following) and a member of the TGFβ-family (either directly or produced by a recombinant cell in the vicinity of the CMPC). The present invention thus provides a prodrug comprising pre-differentiated CMPCs of the invention. It is also envisaged to keep/maintain/encapsulate the CMPCs of the invention (preferably in the pre-differentiated form) in a first biodegradable/biocompatible graft and to add the TGFβ-member (either the compounds pre se or cells producing them) in form of a second graft which is transplanted in the vicinity of the first graft and thereby to achieve the differentiation of the cells at their desired destination. Said second graft may be replaced if necessary or removed once the CMPCs have been differentiated into cardiomyocytes. The same applies equally to the tubular structures/vascular sprouts as mentioned herein before, by using the respective differentiation method described herein (including the treatment of the cells with an biologically active agent having the capabilities to induce angiogenesis or sprouting angiogenesis (like e.g. VEGF).


The present invention thus also relates to a pharmaceutical composition comprising the CMPCs of the invention together with members of the TGF-β family member in a biocompatible/biodegradable material/graft. Said pharmaceutical composition may alternatively comprise two distinct grafts, namely a first biodegradable/biocompatible graft comprising the CMPCs of the invention (preferably in the pre-differentiated form) and a second graft comprising the mentioned TGFβ-member (either the compounds pre se or cells producing these TGFβ members). It is also envisaged that the second graft is made of a non-biodegradable material, for example in case that a recombinant cell expressing and secreting one or more of the mentioned TGFβ-members is included instead of TGFβ member(s) per se. In such a case it is envisaged that the graft containing the TGFβ family member producing cells is removed once the CMPCs and/or the pre-differentiated CMPCs are differentiated into cardiomyocytes. It follows that such a graft may also be comprised of a non-biodegradable material as it is removed anyhow. Such a non-biodegradable graft, however, is preferably biocompatible, i.e. it has the quality of not having toxic or injurious effects on biological systems.


The above equally applies to pharmaceutical compositions comprising the CMPCs of the invention together with compounds like VEGF which have the capability to differentiate the CPMPs of the invention into the tubular structures of the invention or the vascular sprouts of the invention.


The pharmaceutical compositions can be administered to the subject at a suitable dose. The dosage regimen will be determined by the attending physician and clinical factors. The pharmaceutical compositions comprising the cardiomyocytes of the invention should be brought into the border zone surrounding the infarcted area via e.g. intramyocardial injection, or by engrafting the 3D structure onto the damaged myocardium.


The pharmaceutical compositions can thus be used in cardiomyocyte replacement therapy and/or for the treatment of myocardial infarction or for ameliorating the effects of myocardial infarction, i.e. the present invention relates to the use of the cardiomyocytes obtainable by the method of the invention for the preparation of a pharmaceutical composition to be used in a cardiomyocyte replacement therapy and/or for the treatment of myocardial infarction or for ameliorating the effects of myocardial infarction. “Cardiomyocyte replacement therapy” in this regard refers to the improvement of cardiac contractile force. The goal of cardiomyocyte replacement therapy is to replace the lost cardiomyocytes in the ventricular wall by viable contractile cells. Due to the dead myocytes and the development of a scar, the heart has less power to pump. Improvement of its contractile function will help to increase the ejection fraction.


As mentioned before, it is also envisaged to keep/maintain/encapsulate the cardiomyocytes and/or CMPCs as explained above and/or the tubular structures or vascular sprouts of the invention in, for example, biodegradable grafts. Examples of the biocompatible/biodegradable material include collagen, gelatin, fibrin, albumin, hyaluronic acid, heparin, chondroitin sulfate, chitin, chitosan, alginic acid, pectin, agarose, hydroxyapatite, polypropylene, polyethylene, polydimethylsiloxane, polymer or copolymer of glycolic acid, lactic acid or amino acid, a mixture of two or more kinds of the biocompatible materials above, and the like. A particularly preferred biocompatible material is collagen. It is also preferred to mix collagen with other biocompatible material described in the above. Collagen may be any collagen and includes, for example, collagen soluble in acid, collagen solubilized by enzyme (for example, atelocollagen, etc.), collagen solubilized by alkali, collagen having modified amino acid side chains, bridged collagen, collagen produced by genetic engineering, or the like. Collagen having modified amino acid side chains includes, for example, succinyl or methyl collagen, or the like. Bridged collagen includes, for example, collagen treated with glutaraldehyde, hexamethylenediisocyanate or polyepoxy compound, or the like (Fragrance J., 1989-12, 104-109, Japanese Patent KOKOKU (examined) No. 7 (1995)-59522)). The pharmaceutical compositions comprising the cardiomyocytes of the invention in such a biodegradable graft can then easily be brought into the border zone surrounding the infarcted area or onto the damaged myocardium.


It is further envisaged that the mentioned biocompatible/biodegradable grafts containing the cardiomyocytes and/or the CMPCs (preferably the pre-differentiated CMPCs) and/or the tubular structures or vascular sprouts of the invention of the invention and/or the member of the TGFβ-family or an biologically active agent having the capabilities to induce angiogenesis or sprouting angiogenesis (like e.g. VEGF) (depending on which differentiation result is to be achieved) are superimposed on a stent or stent graft. In the medical field, a stent is an expandable wire form or perforated tube (for example perforated by means of laser cutting) that is normally inserted into a natural conduit of the body to prevent or counteract a disease-induced localized flow constriction and/or to support weak points in arteries. An endovascular stent graft is a tubular device which is composed of fabric supported by another rigid structure, usually metal. This rigid structure is also called a stent.


It is also envisaged to superimpose the biocompatible/biodegradable grafts containing the cardiomyocytes and/or the CMPCs (preferably the pre-differentiated CMPCs) and/or the tubular structures or vascular sprouts of the invention and/or the member of the TGFβ-family or an biologically active agent having the capabilities to induce angiogenesis or sprouting angiogenesis (like e.g. VEGF) (depending on which differentiation result is to be achieved) on an artificial heart valve or a biological heart valve. An artificial heart valve is a device which is implanted in the heart of a subject, for example a human. These mechanical heart valves are prosthetics designed to replicate the function of the natural valves of the heart. Biological heart valves which are heart valves of animals (i.e. xenografts), like pigs, which undergo several chemical procedures in order to make them suitable for implantation in the human heart are also within the scope of the present invention. Alternatively, transplanted heart valves of humans are envisaged (allograft or homograft).


In alternative embodiments the biodegradable graft may be replaced by a biocompatible but non-degradable graft. Biocompatible means that it has the quality of not having toxic or injurious effects on biological systems, particular on the subject to be treated


It is further envisaged that the cardiomyocytes and/or the CMPCs (preferably the pre-differentiated CMPCs) and/or the tubular structures or vascular sprouts of the invention and/or the member of the TGFβ-family or an biologically active agent having the capabilities to induce angiogenesis or sprouting angiogenesis (like e.g. VEGF) (depending on which differentiation result is to be achieved) is(are) directly superimposed (which means without a biocompatible/biodegradable graft) on the above-mentioned devices, i.e. onto the stents, heart valves etc. Alternatively, the surface of the above mentioned devices can be coated with adhesive polymeric layers including but not limited to for example poly-L-Lysine (PLL) or fibronectin (FN).


The terms “treatment”, “treating” and the like are used herein to generally mean obtaining a desired pharmacological and/or physiological effect. The effect may be therapeutic in terms of partially or completely curing a disease and/or adverse effect attributed to the disease. The term “treatment” as used herein covers any treatment of a disease in a mammal, particularly a human, and includes: (a) inhibiting the disease, i.e. arresting its development; or (b) relieving the disease, i.e. causing regression of the disease. The present invention is directed towards treating patients with myocardial infarction. Accordingly, a treatment of the invention would involve inhibiting or relieving any medical condition related to myocardial infarction.


“Myocardial infarction” literally means that there is destruction of heart muscle cells due to a lack of oxygen. The resulting oxygen shortage causes damage and potential death of heart tissue.


The hCMPC-derived cardiomyocytes of the invention had a rather mature electrical phenotype in culture, and therefore will serve as a source of autologous transplantable cardiomyocytes.


The present invention thus relates in a further embodiment to the use of the cardiomyocytes obtainable by the methods of the invention for the preparation of a pharmaceutical composition, wherein said composition comprises or consists of cardiomyocytes which were differentiated from CMPCs as defined herein, and wherein said CMPCs were derived from the patient to be treated (i.e. autologous treatment). It will be understood that “autologous” refers to the cells of the invention that are reimplanted in the same individual as they come from (autografts).


In contrast, cells transplanted from a different individual are referred to as allogeneic or as an allograft. Such allogeneic cells, their uses, and pharmaceutical compositions comprising them, are also within the scope of the present invention.


In an further aspect, the present invention relates to the use of the CMPCs of the invention for the preparation of a pharmaceutical composition for the treatment of myocardial infarction or for ameliorating the effects of myocardial infarction, wherein said “preparation” explicitly includes the differentiating methods of the present invention (i.e. the methods for the differentiation of the CMPCs of the invention to the cardiomyocytes of the invention as explained herein).


In a further aspect, the present invention relates to a method for the treatment of myocardial infarction or for ameliorating the effects of myocardial infarction in a subject, comprising the methods of the invention as mentioned above (i.e. the methods for providing the cardiomyocytes of the invention) and further administering the so obtained cardiomyocytes to a subject in need thereof (e.g. the same subject from which the CMPCs were derived from (autograft) or to a different individual (allograft)).


In a further preferred embodiment, the present invention relates to a method for the treatment of myocardial infarction or for ameliorating the effects of myocardial infarction in a subject, comprising providing the CMPCs of the invention and further comprising the differentiating methods of the present invention (i.e. the methods for the differentiation of the CMPCs of the invention into the cardiomyocytes of the invention as explained herein) and further administering the so obtained cardiomyocytes to a subject in need thereof (e.g. the same subject from which the CMPCs were derived from (autograft) or to a different individual (allograft)).


In the context of the present invention the term “subject” means an individual in need of a treatment of an affective disorder. Preferably, the subject is a mammalian, particularly preferred a human, a horse, a camel, a dog, a cat, a pig, a cow, a goat or a fowl.


The term “administered” means administration of a therapeutically effective dose of the cardiomyocytes as disclosed herein. By “therapeutically effective amount” is meant a dose that produces the effects for which it is administered. The exact dose will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques.


The hCMPC-derived cardiomyocytes had a rather mature electrical phenotype in culture, suggesting that they may serve as a homogeneous population of human cardiomyocytes for drug screens. For drug screens, one is in need of a homologues cell culture of cardiomyocytes which does not contain significant amount of other cells heterologous thereto as this might influence the behavior/response to the drug. Having a more mature phenotype of the cardiomyocytes will mimic more the patient situation, and a more mature cardiomyocyte, as the ones of the present invention, are more fragile and might respond different then more fetal cells. The cardiomyocytes of the present invention are thus particularly useful for screening methods like drug screening methods.


In a further embodiment, it is envisaged that the invention's CMPCs and thereby also the cardiomyocytes and/or “tubular structures” derived therefrom may comprise a foreign polynucleotide sequence which allows the transcription of nucleic acids and/or the expression of proteins encoded by such nucleic acids.


“Foreign” in this regard means that the polynucleotide sequence comes from the outside of the cell (i.e. is exogenous thereto) and is artificially introduced into the cells.


Preferably, the foreign polynucleotide sequences are part of a vector, e.g. a commercially available vector. Nonlimiting examples include plasmid vectors compatible with mammalian cells, such as pUC, pBluescript (Stratagene), pET (Novagen), pREP (Invitrogen), pCRTopo (Invitrogen), pcDNA3 (Invitrogen), pCEP4 (Invitrogen), pMC1 neo (Stratagene), pXT1 (Stratagene), pSG5 (Stratagene), EBO-pSV2neo, pBPV-1, pdBPVMMTneo, pRSVgpt, pRSVneo, pSV2-dhfr, pUCTag, pIZD35, pLXIN and pSIR (Clontech) and pIRES-EGFP (Clontech). For vector modification techniques, see Sambrook and Russell (2001), loc. cit. Vectors can contain one or more replication and inheritance systems for cloning or expression, one or more markers for selection in the host, e.g., antibiotic resistance, and one or more expression cassettes.


The coding sequences inserted in the vector can be synthesized by standard methods, isolated from natural sources, or prepared as hybrids. Ligation of the coding sequences to transcriptional regulatory elements (e.g., promoters, enhancers, and/or insulators) and/or to other amino acid encoding sequences can be carried out using established methods.


Furthermore, the vectors may comprise expression control elements, allowing proper expression of coding regions in suitable hosts. Such control elements are known to the skilled artisan and may include a promoter, translation initiation codon, translation and insertion site or internal ribosomal entry sites (IRES) (Owens, Proc. Natl. Acad. Sci. USA 98 (2001), 1471-1476) for introducing an insert into the vector. Preferably, the foreign nucleic acid molecule is operatively linked to said expression control sequences allowing expression in eukaryotic cells. Particularly preferred are in this context control sequences which allow for correct expression in cardiomyocytes and/or cells derived from the heart. Such control sequences have been exemplified herein elsewhere.


Control elements ensuring expression in eukaryotic cells are well known to those skilled in the art. As mentioned above, they usually comprise regulatory sequences ensuring initiation of transcription and optionally poly-A signals ensuring termination of transcription and stabilization of the transcript. Additional regulatory elements may include transcriptional as well as translational enhancers, and/or naturally-associated or heterologous promoter regions. Possible regulatory elements permitting expression in for example mammalian host cells comprise the CMV-HSV thymidine kinase promoter, SV40, RSV-promoter (Rous sarcome virus), human elongation factor 1α-promoter, CMV enhancer, CaM-kinase promoter or SV40-enhancer.


An expression vector according to this invention is at least capable of directing the replication, and preferably the expression, of the nucleic acids and protein. Suitable origins of replication include, for example, the Col E1, the SV40 viral and the M 13 origins of replication. Suitable promoters include, for example, the cytomegalovirus (CMV) promoter, the lacZ promoter, the gal10 promoter and the Autographa californica multiple nuclear polyhedrosis virus (AcMNPV) polyhedral promoter. Suitable termination sequences include, for example, the bovine growth hormone, SV40, iacZ and AcMNPV polyhedral polyadenylation signals. Examples of selectable markers include neomycin, ampicillin, and hygromycin resistance and the like. Specifically-designed vectors allow the shuttling of DNA between different host cells e.g. between bacteria-animal cells.


It is envisaged that the foreign polynucleotide sequence comprises a nucleic acid sequence (e.g. a gene, cDNA etc) which when expressed shows an effect on proliferation, hypertrophy, maximal diastolic potential, the ability to couple correctly and propagate action potentials; on excitation-contraction coupling and/or on cardiomyocyte and/or CMPC survival or death. It is furthermore envisaged that the foreign polynucleotide sequence comprises a gene which expresses a desired gene product which might have a beneficial effect on heart tissue and/or on angiogenesis Genes which are envisaged in this regard are exemplified by but not limited to the following list: Anti apoptotic genes like Akt, pro angiogenesis like VEGF, pro growth like bFGF (but then driven by a cardiomyocyte specific promoter so it will not be on in undifferentiated cells so it will not block there differentiation. BMP2/4 which are pro myocyte differentiation.


It is also envisaged that the foreign polynucleotide sequence additionally or solely comprises a reporter gene, i.e. nucleic acid sequences encoding easily assayed proteins like for example GFP, EGFP, CAT, GAL or GUS. Reporter genes can be attached to other nucleic acid sequences so that only the reporter protein is made or so that the reporter protein is fused to another protein (fusion protein). Alternatively, only the reporter gene is expressed for example under the control of a cardiac promoter or under the control of a cardiomyocyte a or CPMC-specific promoter. Such promoters have been exemplified herein elsewhere. Suitable cardiac promoters are for example human cardiac actin promoter and the Mlc-2V promoter.


Alternatively, it is also envisaged that the cells of the invention (i.e. the CMPCs, the cardiomyocytes and/or the “tubular structures” derived therefrom) may comprise inhibitory short nucleic acid molecules like anti-sense nucleotides, iRNA, siRNA, miRNA or ribozyme. These inhibitory molecules interfere with the transcription and/or expression of a target gene.


miRNAs are short non-coding small regulatory RNAs that regulate protein expression by translational repression. In the past few years, studies have demonstrated that microRNAs (miRNAs) are important for the transcriptional regulation of target genes, serving important regulatory functions in angiogenesis, heart development, and maintenance of stem cell populations.


An siRNA approach is, for example, disclosed in Elbashir ((2001), Nature 411, 494-498)). It is also envisaged in accordance with this invention that for example short hairpin RNAs (shRNAs) are employed. The shRNA approach for gene silencing is well known in the art and may comprise the use of st (small temporal) RNAs; see, inter alia, Paddison (2002) Genes Dev. 16, 948-958. Approaches for gene silencing are known in the art and comprise “RNA”-approaches like RNAi or siRNA. Successful use of such approaches has been shown in Paddison (2002) loc. cit., Elbashir (2002) Methods 26, 199-213; Novina (2002) Mat. Med. Jun. 3, 2002; Donze (2002) Nucl. Acids Res. 30, e46; Paul (2002) Nat. Biotech 20, 505-508; Lee (2002) Nat. Biotech. 20, 500-505; Miyagashi (2002) Nat. Biotech. 20, 497-500; Yu (2002) PNAS 99, 6047-6052 or Brummelkamp (2002), Science 296, 550-553.


“Anti-sense” and “antisense nucleotides” means DNA or RNA constructs which block the expression of the naturally occurring gene product. As used herein, the terms “antisense oligonucleotide” and “antisense oligomer” are used interchangeably and refer to a sequence of nucleotide bases that allows the antisense oligomer to hybridize to a target sequence in an RNA by Watson Crick base pairing, to form an RNA: oligomer heteroduplex within the target sequence. The term “target sequence” in this regard refers to the sequence of the gene to be silenced within the cells of the invention. The antisense may have exact sequence complementarity to the target sequence or near complementarity. Such antisense oligomers may block or inhibit translation of the mRNA containing the target sequence, or inhibit gene transcription by binding to double-stranded or single stranded sequences. Preferably, said antisense oligonucleotides as used herein are “nuclease-resistant” oligomeric molecule e.g. their backbone is not susceptible to nuclease cleavage of a phosphodiester bond. Exemplary nuclease resistant antisense oligomers are oligonucleotide analogs, such as phosphorothioate and phosphate-amine DNA (pnDNA), both of which have a charged backbone, and methylphosphonate, morpholino, and peptide nucleic acid (PNA) oligonucleotides, all of which may have uncharged backbones.


Such cells as described above are well suited for, e.g., the screening methods of the invention and/or for pharmacological studies of drugs in connection with heart infarct and/or for the medical/pharmaceutical compositions/uses and/or treatments as described herein.


The present invention relates in a further aspect to the use of the cardiomyocytes and/or “tubular structures” and/or “vascular sprouts” obtainable by the method of the invention for screening methods, preferably drug screening methods, said screening methods comprising the steps of:

    • (a) providing the cardiomyocytes and/or “tubular structures” and/or “vascular sprouts” of the invention;
    • (b) bringing said cardiomyocytes and/or “tubular structures” and/or “vascular sprouts” into contact with a test substance; and
    • (c) evaluating the effect of said test substance on the phenotype of said cardiomyocytes and/or “tubular structures” and/or “vascular sprouts”.


Evaluating the effect on the phenotype of the cardiomyocytes shall mean evaluating the effect on proliferation, hypertrophy, maximal diastolic potential, the ability to couple correctly and propagate action potentials; on excitation-contraction coupling, cardiomyocyte survival or death, and induction of genes known to be involved in these processes.


Evaluating the effect on the phenotype of the “tubular structures” and/or “vascular sprouts” shall mean evaluating the effect on the thickness of the sprouts, the length of the sprouts, the number of branching points and the presence of endothelial and smooth muscle cells.


Alternatively, also the CMPCs of the invention can be employed for the screening methods of the invention. To this end, it is envisaged that (a) the CMPCs of the invention are provided and (b) said CMPCs are brought into contact with a test substance and (c) the effect of said test substance on the differentiation capability of the CMPCs is evaluated. “Differentiation capability” means the capability of the CMPCs to differentiate into either cardiomyocytes or “tubular structures” and/or “vascular sprouts”, depending on the differentiation method (i.e. the differentiation methods of the invention). Said differentiation methods are explained in great detail herein.


The screening methods, optionally comprise the steps of a) introducing a molecular library of randomized candidate nucleic acids into a plurality of test-cells (i.e. the cardiomyoctes and/or “tubular structures” and/or “vascular sprouts” of the invention), wherein each of said nucleic acids comprises a different nucleotide sequence; b) screening the plurality of test-cells for a cell exhibiting an altered phenotype. The methods may also include the steps of c) isolating the test-cell(s) exhibiting an altered phenotype; and d) isolating a candidate nucleic acid from the cell(s). The introduction (e.g. by way of retroviral vectors) and construction of suitable molecular libraries of randomized candidate nucleic acids is detailed, e.g. in WO 97/27213.


The term “test substance” in accordance with the screening methods of the present invention shall mean any biologically active substance that has an effect on proliferation, hypertrophy, cardiomyocyte survival or death, and induction of genes known to be involved in these processes. Screening of the effect on tubular structures are growth, ES and SMC survival, migration, sprout length, and branching points. Preferred compounds are nucleic acids, preferably coding for a peptide, polypeptide, antisense RNA, iRNA, siRNA, miRNA or a ribozyme or nucleic acids that act independently of their transcription respective their translation as for example an antisense RNA or ribozyme; natural or synthetic peptides, preferably with a relative molecular mass of about 1.000, especially of about 500, peptide analogs, polypeptides or compositions of polypeptides, proteins, protein complexes, fusion proteins, preferably antibodies, especially murine, human or humanized antibodies, single chain antibodies, Fab fragments or any other antigen binding portion or derivative of an antibody, including modifications of such molecules as for example glycosylation, acetylation, phosphorylation, farnesylation, hydroxylation, methylation or esterification, hormones, organic or inorganic molecules or compositions or libraries, preferably small molecules with a relative molecular mass of about 1.000, especially of about 500. The test-compounds can be recovered from products purified from natural sources or natural product mixtures, including bodily fluids, tissues and cells. These may be derived from bacteria, fungi, plants including higher plants, insects, and mammalians. It is particularly envisaged that the test-compound is a chemical and/or natural library of compounds, the latter including but not limited to libraries of natural compounds derived from flowers, green plants, fruits, vegetables, dairy products, grain and/or fungal sources.


It is envisaged that the term “contacting a cardiomyocyte and/or “tubular structure” and/or “vascular sprouts” with test substance” includes to introduce the test-compounds by suitable methods like electro/chemical poration; lipofection; bioballistics or microinjection into the cells.


It is also envisaged that the cells to be used in the screening assays of the present invention are used in a one-hybrid, three-hybrid or a one-two-hybrid-technique, which is a molecular biology technique used to discover protein-protein interactions or protein-DNA interactions by testing for physical interactions (such as binding) between two proteins or a single protein and a DNA molecule, respectively. Said terms and the biological principle behind are well known in the art.


The present invention relates in a further embodiment to a diagnostic composition comprising the cardiomyocytes and/or “tubular structures” and/or “vascular sprouts” of the invention and to a kit comprising this diagnostic composition and instructions to use, i.e. instruction manuals which guide the skilled reader on how to keep the cells viable during the test protocol. To the more, it is envisaged that the above described kit and/or diagnostic compositions further comprise the test-substances (e.g. in the form of a library of compounds) and instructions to screen for the influence of said test substance(s) on the phenotype of the cells.


In a further aspect, the present invention relates to a diagnostic composition comprising the CMPCs as described herein and/or to a kit comprising this diagnostic composition, wherein said kit and/or diagnostic composition further comprises instruction manuals which guide the skilled person to differentiate these CMPCs into cardiomyocytes and/or “tubular structures” and/or “vascular sprouts”, for example by use of the differentiation methods of the invention. To this end, this kit and/or diagnostic composition may further comprise the substances/chemicals and/or cell culture equipment (culture dishes; antibodies for quality staining etc) which are useful for the differentiation protocol and, optionally, a solvent, diluent, buffer for stabilizing and/or storing the inventions compounds. To this end, it is envisaged that the diagnostic composition comprising the CMPCs of the invention and/or the kit comprising this diagnostic composition further comprises the demethylating agents, antioxidant agents members of the TGF-β family etc as described herein in detail in the context of the differentiation methods of the invention. To the more, it is envisaged that the above described kit and/or diagnostic compositions further comprise the test-substances (e.g. in the form of a library of compounds) and instructions to screen for the influence of said test substance(s) on the phenotype of the cells.


This disclosure may best be understood in conjunction with the accompanying drawings, incorporated herein by references. Furthermore, a better understanding of the present invention and of its many advantages will be obtained from the following examples, given by way of illustration and are not intended as limiting.





THE FIGURES SHOW


FIG. 1: (A) Bright field images of CMPC cultures. Passage number is indicated. Right panel shows a MSC culture. B) Growth curve of freshly isolated CMPCs. Total number of cells per well were counted at the indicated day after plating. C) Table showing immunoreactivity of the indicated marker proteins by FACS analysis. The CMPC surface expression was compared to the levels in whole blood. (D) Semi quantitative RT-PCR on RNA isolated from undifferentiated CMPCs probed for the expression of the indicated genes. E) Immunolabeling against transcription factors GATA-4, Nkx2.5, Islet-1 and Western-Blot analysis to demonstrate the presence of GATA-4 protein.



FIG. 2: (A) Semi-quantitative RT-PCR on RNA isolated from undifferentiated CMPCs (CMPC) and differentiated CMPCs (diff CMPC) probed for the expression of Islet-1, early cardiac transcription factors Nkx2.5 and GATA-4, the contractile proteins cardiac actin, cardiac Troponin I, MLC2v, and the skeletal myoblast transcription factor Myo-D. B) Immunolabeling of differentiated cell culture 3 weeks after 5-Azacytidine treatment against the contractile proteins α-actinin and cardiac Troponin I. C) Immunolabeling against the contractile proteins desmin, titin, MLC2v, β-MHC and α-actinin as expressed in CMPCs differentiated into cardiomyocytes. Right panels show the expression profile of the intercalated disk related proteins ZO-1 and N-cadherin. Bar=20 μm.



FIG. 3: (A) Western blot analysis on protein samples isolated from CMPCs stimulated without or with 1 ng/ml TGFβ or 25 ng/ml BMP-6 and probed with α-pSmad1 or α-pSmad2. Asterisk indicates an non-specific band showing equal loading. B) Semi-quantitative RT-PCR on RNA isolated from CMPCs stimulated without or with 1 ng/ml TGFβ for the indicated days probed for the expression of the early cardiac transcription factors Nkx2.5 and Mef2C, the contractile proteins cardiac actin, cardiac Troponin I, desmin, ANP and MLC2v. β-actin was used as a control for equal RNA input. C) Western blot analysis on protein samples from CMPCs stimulated without or with 1 ng/ml TGF for the indicated days, probed for Nkx2.5 or α-actinin. D) Immunohistochemical labelling for α-actinin shows the degree of differentiation into cardiomyocytes within the cultures without (left) or with (right) TGFβ stimulation. Counterstaining of nuclei was performed using DAPI. Bar=50 μm.



FIG. 4: (A) Current clamp recording from a CMPC cardiomyocyte showing a rapid phase 0 depolarization and mild diastolic depolarization. (B) Table summarizing CMPC action potential parameters. (C) Combined calcium-imaging and current clamp recording showing excitation-contraction coupling in a CMPC cardiomyocyte. Scale bars: horizontal 500 ms, vertical −50 mV.



FIG. 5: (A) Semi-quantitative RT-PCR on RNA isolated from undifferentiated undif and differentiated (dif) CMPCs probed for the expression of Cx40, Cx43 and Cx45. β-tubulin was used as a control for equal RNA input. −RT is the control in which reverse transcriptase was omitted during the reaction. B) Immunolabeling against the connexin isoforms Cx40, Cx43 and Cx45 in undifferentiated CMPCs (top panels) and CMPCs differentiated into cardiomyocytes (bottom panels). Bar=25 μm.



FIG. 6. (A) Transfer of Lucifer Yellow from the injected cell to surrounding cells showing metabolic coupling. Dye spreading is significantly higher in differentiated CMPCs compared to undifferentiated cells. (B) Photomicrograph of a representative Lucifer Yellow injection in differentiated CMPCs, phase-contrast (upper panel) and Lucifer Yellow fluorescence (lower panel). Asterisk marks the injected cell. (C) Quantification of electrical conductance in undifferentiated and differentiated CMPC cell pairs. (D) Representative recording or gap junctional currents in a differentiated CMPC cell pair. Scale bars: horizontal 200 ms, vertical −200 pA.



FIG. 7. (A) Semi-quantitative RT-PCR on RNA isolated from undifferentiated adult (ACMPC) probed for the expression of the indicated genes. (B) Semi-quantitative RT-PCR on RNA isolated from undifferentiated adult (ACMPC) and ACMPC 4 weeks after differentiation, probed for the expression of the indicated genes. (C) Immunolabeling against Islet-1 in ACMPCs (top panel) and against α-actinin in ACMPCs differentiated into cardiomyocytes (bottom panels).



FIG. 8: Results of an in vitro angiogenesis assay. Part 1: CMPC were plated on matrigel without (A) or with 100 nM (B) miRNA-x. CMCPs formed tubular structures with endothelial (red) and SMC-like (green) cells, as can be observed in the magnification (C). Adding miRNA-x resulted in less side-branches and increased tubular structures. Part 2: CMPC were plated on matrigel. CMCPs formed tubular structures with endothelial (CD31 staining in red) and SMC-like (alpha-SMA in green) cells, as can be observed in the magnification.



FIG. 9: Results of a myocardial Infarction model in NOD/scid mice. At day 2, LV volumes were higher and EF (ejection fraction) was lower in both MI groups as compared to Sham, with no difference between the MI groups (FIG. 9). However at day 14, EF (ejection fraction) was higher and ESV (end systolic volume) was lower in the hCMPC group as compared to the MI+Medium group (P=0.001 and P=0.048 respectively). Although there was a trend towards a reduced EDV (end diastolic volume), this did not reach significance (P=0.14).



FIG. 10: Results of adipocyte differentiation of the hCMPCs of the invention. FIGS. 10a to 10f show the evaluation of adipocyte differentiation marker genes leptin, adisin, ppar, Cnn1/cyr61 and glut4.





EXAMPLES

The following examples illustrate the invention. These examples should not be construed as to limit the scope of this invention. The examples are included for purposes of illustration and the present invention is limited only by the claims.


Group comparisons were made using one-way ANOVA with the Holm-Sidak post-hoc test for multiple comparisons. Statistical significance was assumed if P<0.05. All data are presented as mean±SEM.


Example 1
Isolation and Culture of Cardiomyocyte Progenitor Cells from Human Fetal Hearts

For human fetal tissue collection and atrial biopsies, individual permission using standard informed consent procedures and prior approval of the ethics committee of the University Medical Center Utrecht were obtained. Fetal hearts were collected after elective abortion followed by Langendorff perfusion with Tyrode's solution, collagenase and protease. Atrial biopsies were minced into small pieces followed by collagenase treatment. After cardiomyocyte depletion of the cell suspension, cardiomyocyte progenitor cells were isolated by magnetic cell sorting (MACS, Miltenyl Biotec, Sunnyvale, Calif.) using Sca-1-coupled beads, following the manufactures protocol. Sca-1+ cells were eluted from the column by washing with PBS supplemented with 2% fetal calf serum (FCS) and cultured on 0.1% gelatin coated dishes in M199 (Gibco)/EGM (3:1) supplemented with 10% FCS (Gibco), 10 ng/ml basic Fibroblast growth factor (bFGF), 5 ng/ml epithelial growth factor (EGF), 5 ng/ml insuline like growth factor (IGF-1) and 5 ng/ml hepatocyte growth factor (HGF).


Clonal analysis was performed by limiting-dilution method. CMPCs were dissociated into single cells and plated into the wells of a gelatine-coated 96-well plate at a density of 0.5 cell per well in culture medium.


Example 2
Differentiation of hCMPCs

To induce differentiation, cells were treated with 5 μM 5′-azacytidine (Sigma) for 72 hours in differentiation medium (Iscove's Modified Dulbecco's Medium/HamsF12 (1:1) (Gibco)) supplemented with L-Glutamine (Gibco), 2% horse serum, non-essential amino acids, Insulin-Transferrin-Selenium supplement, and 10−4 M Ascorbic Acid (Sigma)). After induction, the medium was changed every 3 days.


For TGFβ treatment, CMPCs were cultured in differentiation medium, stimulated with 5 μM 5′-azacytidine (Sigma) and 1 ng/ml TGFβ1 (Sigma) added 24 hours later. The medium was changed every 3 days.


Example 3
RNA Isolation and Reverse Transcription PCR

RNA was isolated using of Trizol (Invitrogen, Breda, The Netherlands) and reverse transcribed using oligo-dT Superscript 3 (Invitrogen). Primer sequences are given in table 1. The PCR reactions started with 2 min at 94° C. followed by 35 cycles of: 15 s at 94° C., 30 s at 55° C. and 45 s at 72° C. Products were analyzed on ethidium bromide-stained 1% agarose gel. β-tubulin or β-actin were used as RNA input control.












TABLE 1






Gene
Primers
Size








MEF2C
5′-agatacccacaacacaccacgcgcc-3′
192




5′-atccttcagagagtcgcatgcgctt-3′







IsI-1
5′-taagccaccgtcgtgtctc-3′
107




5′-tgatgaagcaactccagcag-3′







GATA-4
5′-gacaatctggttaggggaagc-3′
105




5′-accagcagcagcgaggagat-3′







NKX2.5
5′-cgccgctccagttcatag-3′
111




5′-ggtggagctggagaagacaga-3′







MLC2V
5′-gtcaatgaagccatccctgt-3′
101




5′-gcgccaactccaacgtgttct-3′







TropT
5′-gtgggaagaggcagactgag-3′
131




5′-atagatgctctgccacagc-3′







bMyHC
5′-gaagcccagcacatcaaaag-3′
118




5′-gatcaccaacaacccctacg-3′







cActin
5′-tcctgatgcgcatttttattc-3′
123




5′-aacaccactgctctagccacg-3′







Desmin
5′-acctgctcaacgtgaagatg-3′
159




5′-tggtatggacctcagaacc-3′







MyoD
5′-cggcggcggaactgctacgaa-3′
458




5′-ggggcgggggcggaaactt-3′







c-kit
5′-aagtggatggcacctgaaag-3′
138




5′-gaacttagaatcgaccggca-3′







Cx40
5′-gagaagaagcagccagagtgtgaa-3′
651




5′-GACATGCAGGGTGGTCAGGAAGATT-3′







Cx43
5′-agcgtgaggaaagtaccaaacagc-3′
666




5′-AAGAAGGCCACCTCAAAGATAGAC-3′







Cx45
5′-ctagacccactgaaaagacc-3′
494




5′-TGATTTGCTACTGGCAGTGC-3′










Example 4
Flow Cytometric Analysis

CMPCs were either used freshly isolated or as trypsinyzed cells after culture. 200.000 cells per sample was used for FACS analysis. The cells were washed twice in wash-buffer (wb: 1% FCS/PBS/0.05M azide) and re-suspended in 100 μl wb containing 0.5% antibody. Antibodies used for FACS analysis were FITC- or PE conjugated antibodies against CD31, CD34, CD105 (Endoglin), CD117 (c-kit), Sca-1, and isotype control IgGs, all from Pharmingen BD. The cells were incubated on ice in the dark for 30 minutes, washed 4 times with cold wb, resuspended in 250 μl wb and analyzed using a Beckman Coulter Cytomics FC500 FACS.


Example 5
Immunohistochemistry on Cultured Cells

Coverslips with cultured cells were rinsed in serum free medium, fixed in methanol (−20° C.) or 4% paraformaldehyde, washed with PBS and permeabilized with 0.2% Triton X-100/PBS. Non-specific binding of antibodies was blocked with 2% bovine serum albumin (BSA). Incubation with primary antibodies was performed overnight in PBS/10% normal goat serum (NGS). Antibodies used recognized GATA-4 (Santa Cruz), Islets-1 (Hybridoma Bank), Cx40 (Chemicon), Cx43 (Zymed), Cx45 (kindly provided by Dr. T. H. Steinberg, Washington University, St. Louis, USA), α-actinin (Sigma), Troponin I (Chemicon) desmin (Sanbio), titin (Sigma), MLC2v (Alexis), β-MHC (kindly provided by Dr. A. F. M. Moorman, AMC Amsterdam, Netherlands), N-cadherin (Sigma) and ZO-1 (Zymed). Immunolabeling was performed using Texas Red (TR)- or fluorescein isothiocyanate (FITC)-conjugated secondary antibodies (Jackson Laboratories). All incubation steps were performed at room temperature and in between all incubation steps, cells were washed with PBS. Finally, coverslips were mounted in Vectashield (Vector Laboratories) and examined with a Nikon Optiphot-2 light microscope equipped for epifluorescence.


Example 6
Western Blot Analysis

Western blot analysis was performed as described previously35. Detection was by ECL (Amersham). PSmad1 and PSmad2 rabbit polyclonal antibodies that specifically recognize phosphorylated Smad1/5 or Smad2 respectively were used 1:1000 (Cell signaling), Nkx2.5 rabbit polyclonal 1:500 (Santa Cruz), and α-actinin mouse monoclonal 1:5000 (Sigma). β-actin detection (1:10000, Chemicon) was used as a loading control.


Example 7
Electrophysiology

A symmetrical setup with two HEKA EPC-7 patch clamp amplifiers was used to measure action potentials and electrical coupling between cells. Electrophysiology A symmetrical setup with two HEKA EPC-7 patch clamp amplifiers was used to measure action potentials and electrical coupling between cells. All signals were recorded using a custom data acquisition program (kindly provided by Dr. J. G. Zegers, AMC Amsterdam, The Netherlands) running on an Apple Macintosh computer equipped with a 12-bit National Instruments PCI-MIO-16E-4 acquisition card. Current signals were low-pass filtered at 2.5 kHz and acquired at 10 kHz. Calcium imaging combined with action potential recording was performed using a Cairn Research imaging system and an Axopatch 200B amplifier. All experiments were done using the whole cell patch clamp configuration. Action potential parameters measured from current clamp recordings were maximal diastolic potential, maximal upstroke velocity, action potential duration at 50 and 90 percent of repolarisation (APD50 and APD90).


Gap junctional macroscopic conductance in cell pairs was measured at a holding potential of −50 mV using a voltage clamp protocol that included 50 ms square pulses with an amplitude of +10 and −10 mV, followed by a 1 second square pulse ranging between −50 and +50 mV. Gap junctional conductance (gj) is defined as gj=lj/Vj, where lj and Vj denote junctional current and transjunctional voltage, respectively. By using the small prepulses for this calculation, gap junctional conductances were maximal and not inactivating. Offline analysis was done using MacDaq 8.0 (kindly provided by Dr. A. C. G. van Ginneken, AMC Amsterdam, The Netherlands) and R 2.0.1 (R Development Core Team, 2005).


Experiments were done at 20° C. or, for calcium imaging, at 37° C. For calcium imaging, cells were loaded with 10 μM Fluo-3 AM for 25 minutes at 37° C. Extracullar buffer used in all cases was a modified Tyrode's solution, containing (in mmol/L) NaCl 140, KCl 5.4, CaCl2 1.8, MgCl2 1, HEPES 15, NaHCO3 35, glucose 6, pH 7.20/NaOH.


Action potentials were recorded using a pipette solution containing (in mmol/L) KCl 130, NaCl 10, HEPES 10, MgATP 5, MgCl2 0.5, pH 7.20/KOH. Pipette buffer used for conductance measurements contained (in mmol/L) potassium gluconate 125, KCl 10, HEPES 5, EGTA 5, MgCl2 2, CaCl2 0.6, Na2ATP 4, pH 7.20/KOH. For dye injection, microelectrodes were filled with 4% w/v Lucifer Yellow in 150 mM LiCl2, 10 mM HEPES and cells were injected for 2 minutes. Cells stained with Lucifer Yellow were counted, excluding the injected cell. Patch pipettes were pulled on a Narishige PC-10 puller and fire-polished. When filled with pipette buffer, the pipette resistance ranged between 2-5 MΩ. Liquid junction potentials were calculated using Clampex (Axon Instruments) and used for offline correction.


Example 8
Microarray Profiling of the Genes Expressed in the CMPCs

RNA was isolated from undifferentiated and differentiated CMPCs. 500 ng of RNA and cRNA was made following standard illumina protocol (illumina RNA amplification kit, Ambion cat #11755. We used illumina Sentrix® HumanRef-8 Expression BeadChip cat #BD-25-201 chips, and hybridization occurred at 55 degrees Celsius.


The genes listed in the following Table 3 are expressed in the CMCPs when measured with microarray gene profiling as indicated above.














TABLE 3





Gene code
Accesion No
Gene code
Accesion No
Gene code
Accesion No







ZNF22
NM_006963.1
PTGDS
NM_000954.5
THBS2
NM_003247.2


MYLK
NM_053031.1
KIAA2028
NM_172069.1
COP
NM_052889.1


ADH1B
NM_000668.3
TNFRSF1B
NM_001066.2
BDNF
NM_170734.2


LOC255189
XM_172929.3
PODN
NM_153703.3
DAAM2
NM_015345.2


PTGS1
NM_080591.1
LOC401151
XM_379274.1
THBS1
NM_003246.2


DDX3Y
NM_004660.2
P8
NM_012385.1
AFAP
NM_021638.3


FLJ23403
NM_022068.1
GAS6
NM_000820.1
LOC56901
NM_020142.3


INHBA
NM_002192.1
ACTG2
NM_001615.2
MGP
NM_000900.1


ADH1C
NM_000669.2
CDKN2B
NM_004936.2
NME4
NM_005009.2


C14orf141
NM_032035.1
CYP1B1
NM_000104.2
LOC375763
XM_353489.1


MAOA
NM_000240.2
MGLL
NM_007283.4
NFIC
NM_005597.2


DHRS9
NM_005771.3
SRPUL
NM_014467.1
C20orf97
NM_021158.3


COL8A1
NM_020351.2
LTBP2
NM_000428.1
ANGPTL4
NM_016109.2


EFEMP1
NM_018894.1
LOC400610
XM_375478.1
LOC51063
NM_015916.3


FLJ31166
NM_153022.1
CaMKIINalpha
NM_018584.4
LOC375351
XM_351558.1


PLSCR4
NM_020353.1
PEG3
NM_006210.1
FLJ10948
NM_018281.1


ADH1A
NM_000667.2
C20orf100
NM_032883.1
GDF5
NM_000557.2


KIAA1199
NM_018689.1
FLJ32389
NM_144617.1
ABCA8
NM_007168.2


CASP4
NM_001225.2
TNFAIP6
NM_007115.2
FBLN5
NM_006329.2


C7
NM_000587.2
MGC33637
NM_152596.2
DF
NM_001928.2


CTSF
NM_003793.2
MFAP4
NM_002404.1
CHI3L1
NM_001276.1


BF
NM_001710.3
CTHRC1
NM_138455.2
C3
NM_000064.1


QSCN6
NM_002826.2
OLR1
NM_002543.2
C8orf13
NM_053279.1


PSMD5
NM_005047.2
CYBRD1
NM_024843.2
TRIM4
NM_033091.1


BTEB1
NM_001206.1
SPAG4
NM_003116.1
AMSH-LP
NM_020799.1


SERPINA3
NM_001085.2
LRRC17
NM_005824.1
TNFRSF11B
NM_002546.2


RUNX1
NM_001754.2
FBN1
NM_000138.2
LOC125476
NM_194281.1


UBE1L
NM_003335.2
ADAMTS5
NM_007038.1
TCF21
NM_198392.1


C1S
NM_001734.2
STARD5
NM_030574.2
MAP3K5
NM_005923.3




A2M
NM_000014.3
CDKN1A
NM_000389.2


FLJ20073
NM_017654.2
ICAM5
NM_003259.2
MGC17330
NM_052880.2


KIAA0779
NM_015008.1
TBX2
NM_005994.3
SPOCK
NM_004598.2


PPARG
NM_005037.3
CALD1
NM_033138.2
FLJ20254
NM_017727.2


LAMB2
NM_002292.2
IGFBP4
NM_001552.1
KLHL5
NM_199039.1


LOC375758
XM_353487.1
TM4SF1
NM_014220.1
C1orf24
NM_052966.1


SERPINF1
NM_002615.3
CEBPB
NM_005194.2
7h3
NM_033025.4


HNMT
NM_006895.1
PP3856
NM_145201.3
MT2A
NM_005953.2


LOC400718
XM_375656.1
EVC
NM_014556.2
TCIRG1
NM_006053.2


FAM20C
NM_020223.1
LOC375843
XM_293449.2
PLXND1
NM_015103.1


ALS2CR3
NM_015049.1
NR2F1
NM_005654.3
DKFZP566K1924
NM_015463.1


TRIM22
NM_006074.2
LTBR
NM_002342.1
C1RL
NM_016546.1


PSCD3
NM_004227.3
SELM
NM_080430.1
TCF21
NM_003206.2


PPIL3
NM_032472.3
PLAU
NM_002658.1
MXI1
NM_130439.1


TNIP2
NM_024309.2
MRF2
XM_084482.3
G1P3
NM_002038.2


LOC143903
NM_178834.2
TIMP1
NM_003254.1
B4GALT1
NM_001497.2


IFI16
NM_005531.1
PP1201
NM_022152.3
pp9099
NM_025201.3


ARC
NM_015193.2
LTBP3
NM_021070.2
LOC401428
XM_376715.1


CPR8
NM_020739.1
CSRP1
NM_004078.1
CAV1
NM_001753.3


TNFRSF1A
NM_001065.2
LOC285148
XM_209490.3
HLA-G
NM_002127.3


RFX4
NM_002920.3
IFITM3
NM_021034.1
TNFRSF21
NM_014452.3


CKLFSF3
NM_144601.2
VIP32
NM_021732.1
AREG
NM_001657.2


CDKN2A
NM_058196.1
ASS
NM_000050.2
VCAM1
NM_080682.1


NBL1
NM_005380.3
TORC3
NM_022769.1
PMP22
NM_153322.1


DDR2
NM_006182.1
BZRP
NM_007311.2
PML
NM_033238.1


RECK
NM_021111.1
C1orf24
NM_022083.1
TACC1
NM_006283.1


LOC400810
XM_375850.1
SLC9A3R2
NM_004785.1
LOC388815
XM_373927.1


FSTL3
NM_005860.1
G0S2
NM_015714.2
PALM
NM_002579.1


SIL1
NM_022464.3
RAB5C
NM_201434.1
SPINLW1
NM_020398.2


COL16A1
NM_001856.2
KIAA1838
NM_032448.1
Spc25
NM_020675.3


S100A6
NM_014624.2
LGALS8
NM_006499.3
RCN3
NM_020650.2


TNKS1BP1
NM_033396.1
KRTHB4
NM_033045.2
LRP1
NM_002332.1


TM4SF7
NM_003271.3
IGFBP7
NM_001553.1
KIAA2002
XM_370878.1


LOC154165
XM_087859.4
PTGIS
NM_000961.2
H2AFJ
NM_177925.1


TPRA40
NM_016372.1
ARHGEF6
NM_004840.1
ANGPTL2
NM_012098.2


HLA-C
NM_002117.3
LOC253013
XM_171892.1
FHL1
NM_001449.3


AK3L1
NM_016282.2
FLJ23235
NM_024943.1
DFNA5
NM_004403.1


SEC14L1
NM_003003.1
TBX6
NM_080758.1
GABARAPL1
NM_031412.1


COPZ2
NM_016429.1
FLOT1
NM_005803.2
ARH
NM_015627.1


C1QTNF5
NM_015645.1
SRGAP1
NM_020762.1
PDGFRL
NM_006207.1


HLA-DMA
NM_006120.2
RASD2
NM_014310.3
PCOLCE
NM_002593.2


CTEN
NM_032865.3
MAN2B1
NM_000528.1
XBP1
NM_005080.2


MGC15429
NM_032750.1
STAT1
NM_139266.1
SRPX
NM_006307.2


EMP1
NM_001423.1
LOC201895
NM_174921.1
ACTN1
NM_001102.2


LOC388121
XM_370864.1
COL6A1
NM_001848.1
GYPC
NM_016815.2


C20orf140
NM_144628.1
SCARF2
NM_182895.1
GNPTAG
NM_032520.3


COL1A1
NM_000088.2
TFPI
NM_006287.3
KIAA0233
NM_014745.1


STAT2
NM_005419.2
P4HA2
NM_004199.1
NEDL2
XM_038999.2


DPP4
NM_001935.2
FLJ21347
NM_022827.2
DKFZP586H2123
NM_015430.1


ACHE
NM_000665.2
FBXO32
NM_148177.1
SLC7A5
NM_003486.4


LOC124842
XM_064333.4
ANTXR1
NM_032208.1
ZNF226
NM_015919.2


ETFDH
NM_004453.1
MMP23B
NM_006983.1
LOC400741
XM_378841.1


LHX3
NM_178138.2
HIPK2
NM_022740.1
SARS
NM_006513.2


PGCP
NM_016134.2
FLJ36525
NM_182774.1
CAV2
NM_198212.1


RAB32
NM_006834.2
RAI14
NM_015577.1
STK17A
NM_004760.1


KIAA0562
NM_014704.1
B2M
NM_004048.2
HIST1H2AH
NM_080596.1


AXL
NM_001699.3
CYB5R1
NM_016243.1
DCN
NM_133507.1


LOC114990
NM_138440.1
MYH11
NM_002474.1
FOXF2
NM_001452.1


P5326
NM_031450.2
NEK6
NM_014397.3
PRO1914
NM_014106.1


LOC375531
XM_353406.1
SOD3
NM_003102.1
LOC375662
XM_351780.1


BACE2
NM_138992.1
MOCOS
NM_017947.1
NDRG1
NM_006096.2


FTL
NM_000146.2
POU2F2
NM_002698.1
LOC375528
XM_351672.1


IFI35
NM_005533.2
TNFSF13
NM_172089.1
RPS4Y2
NM_138963.1


FLJ14800
NM_032840.1
KIAA1944
XM_062545.5
MKNK2
NM_017572.2


BRODL
NM_153252.2
CCND1
NM_053056.1
FLJ10460
NM_018097.1


MED8
NM_201542.1
SAT
NM_002970.1
TNN
XM_040527.5


WDR10
NM_052990.1
PHTF2
NM_020432.2
LEMD2
NM_181336.2


RYR1
NM_000540.1
NOD9
NM_170722.1
FLJ22374
NM_032222.1


MTND5
NM_173713.1
SIAT7D
NM_175039.1
PRRG1
NM_000950.1


SMARCD2
NM_003077.2
ARNT
NM_178426.1
SEC22L2
NM_012430.2


SNTB2
NM_130845.1
AKR1C1
NM_001353.4
LOC376156
XM_352094.1


SAA4
NM_006512.1
TMSL1
NM_182792.1
CYGB
NM_134268.3


RAP2B
NM_002886.2
LOC138649
XM_059987.6
LOC374662
XM_353096.1


PPFIA4
XM_046751.4
STXBP1
NM_003165.1
LOC390690
XM_372617.1


LGALS12
NM_033101.2
MMP2
NM_004530.1
CABP2
NM_031204.1


KCNK2
NM_014217.1
FLJ14249
NM_106552.1
LOC400587
XM_375426.1


MGC40069
NM_182615.1
TIMP2
NM_003255.2
LOC400930
XM_376019.1


GAB2
NM_080491.1
TRIM44
NM_017583.3
LOC149345
XM_086502.8


S100A11
NM_005620.1
KIAA0545
XM_032278.8
TM4SF9
NM_005723.2


PLOD
NM_000302.2
F8
NM_019863.1
PTPNS1
NM_080792.1


NNAT
NM_181689.1
ANXA11
NM_001157.2
MGC13005
NM_032685.2


LMNA
NM_005572.2
MGC32065
NM_153271.1
MICAL3
XM_032997.4


SCRN1
NM_014766.2
DKFZP564O0823
NM_015393.1
HIF1A
NM_181054.1


SUMF1
NM_182760.2
KIAA1190
NM_145166.2
LMCD1
NM_014583.2


CD97
NM_001784.2
AEBP1
NM_001129.2
HMGCL
NM_000191.1


ARHE
NM_005168.2
KIAA0934
NM_014974.1
GLG1
NM_012201.1


LOC283111
XM_210181.4
LOC127545
XM_060535.1
FLJ90024
NM_153342.1


RNPEPL1
NM_018226.2
COL5A2
NM_000393.2
SULF2
NM_198596.1


HSPA12A
XM_048898.5
HT008
NM_018469.3
LOC51334
NM_016644.1


LTC4S
NM_145867.1
LOC286177
XM_379582.1
CREB5
NM_004904.1


SLC2A10
NM_030777.3
FLJ20920
NM_025149.2
MRVLDC1
NM_031484.1


PDGFC
NM_016205.1
RAFTLIN
NM_015150.1
PRO0149
NM_014117.2


HS3ST3A1
NM_006042.1
SIGLEC8
NM_014442.1
ADRB2
NM_000024.3


CRAT
NM_144782.1
SDC4
NM_002999.2
PTPNS1L2
NM_178460.1


ZSIG11
NM_015926.3
TAZ
NM_181314.1
APG10L
NM_031482.3


01. Nov
NM_144663.1
ARHC
NM_175744.3
LOC126167
XM_058997.5


GRINL1A
NM_015532.2
LOC285919
XM_212094.1
SLC1A5
NM_005628.1


KRTHA8
NM_006771.3
FLJ25224
NM_182495.3
SEMA4C
NM_017789.3


LAMB1
NM_002291.1
LOC391403
XM_372940.1
FLJ23221
NM_024579.1


MAP3K6
NM_145319.1
DP1
NM_005669.3
BTBD6
NM_033271.1


SLC9A1
NM_003047.2
NINJ1
NM_004148.2
DIPA
NM_006848.1


ABHD4
NM_022060.1
LOXL3
NM_032603.2
HSPB1
NM_001540.2


FLJ21977
NM_032213.3
SLB
XM_114272.2
MGC44287
NM_182607.2


SPARC
NM_003118.1
GRM4
NM_000841.1
KIFC3
NM_005550.2


EPB41
NM_004437.1
DKFZP564C103
NM_015654.3
DSCAML1
NM_020693.2


MVP
NM_005115.3
ACTL6
NM_016188.3
U2AF1L3
NM_144987.1


DHX8
NM_004941.1
CGI-79
NM_016024.1
KIAA1944
NM_133448.1


CTDSP1
NM_182642.1
CCL14
NM_032963.2
AD-020
NM_020141.2


ARHGEF2
NM_004723.2
HAGHL
NM_032304.1
CTSL
NM_145918.1


LOC374402
XM_353011.1
CDC42EP1
NM_007061.3
SERPINB6
NM_004568.4


MGC3200
NM_032305.1
DSCR1
NM_004414.5
SLC9A8
XM_030524.3


TULP3
NM_003324.3
FLJ10647
NM_018166.1
NDST2
NM_003635.2


SLC35A1
NM_006416.2
RNF123
NM_022064.2
MFGE8
NM_005928.1


SF3B3
NM_012426.2
FLJ40113
NM_198079.1
PEA15
NM_003768.2


RDH5
NM_002905.1
CPA1
NM_001868.1
LRP5
NM_002335.1


CISH
NM_145071.1
TANK
NM_004180.2
NMNAT1
NM_022787.2


FSTL1
NM_007085.3
C6orf29
NM_032794.1
ERO1L
NM_014584.1


LOC375172
XM_351416.1
NAP1L5
NM_153757.1
ABCC13
NM_172024.1


TMSL4
NM_182794.1
C7orf10
NM_024728.1
CBS
NM_000071.1


BMP1
NM_006129.2
FXYD7
NM_022006.1
ACATE2
NM_012332.1


TUBG2
NM_016437.1
NARF
NM_031968.1
FLJ32001
NM_152609.1


QTRTD1
NM_024638.2
FLJ32675
NM_173811.2
BLVRB
NM_000713.1


HECA
NM_016217.1
GRLF1
NM_004491.2
KDR
NM_002253.1


HCCA2
NM_053005.2
TAGLN
NM_003186.2
PHLDA1
NM_007350.1


HIST1H2BK
NM_080593.1
FLJ20522
NM_017861.1
CTSZ
NM_001336.2


PLEKHC1
NM_006832.1
FLJ10916
NM_018271.2
RIN2
NM_018993.2


IL4R
NM_000418.1
ZNF148
NM_021964.1
RPS3A
NM_001006.2


KIAA1205
XM_046305.3
RASSF1
NM_170712.1
FBLN1
NM_001996.2


EXT1
NM_000127.1
HIST1H2BD
NM_138720.1
KIAA1600
XM_049351.3


RHOBTB3
NM_014899.2
LOC402176
XM_377841.1
SLC35B3
NM_015948.2


MAS1
NM_002377.2
MPG
NM_002434.1
CDH11
NM_001797.2


DXS9928E
NM_004699.1
ZNF33A
NM_006974.1
ASC
NM_145183.1


PILRA
NM_178273.1
PKD1L2
NM_182740.1
RSU1
NM_152724.2


ATF5
NM_012068.2
GLS
NM_014905.2
PTD015
NM_024684.2


DKFZp779M0652
XM_374877.1
SLC39A4
NM_130849.1
IER3
NM_003897.2


PLOD2
NM_000935.1
FLJ13352
NM_024592.1
MGC29891
NM_144618.1


LOC377887
XM_352835.1
SE20-4
NM_022117.1
FREQ
NM_014286.2


SLC22A2
NM_003058.2
BCL2L2
NM_004050.2
CLK3
NM_001292.1


NFKB1
NM_003998.2
RAD51L1
NM_133510.1
MGC10812
NM_031425.1


FLJ20542
NM_017871.3
CALCRL
NM_005795.2
PJA2
NM_014819.2


MGC9850
NM_152705.1
FMOD
NM_002023.2
RAB24
NM_130781.1


UBXD2
NM_014607.2
RABGGTA
NM_182836.1
CREBL2
NM_001310.2


SH3YL1
NM_015677.1
GFOD1
NM_018988.1
LOC162427
NM_178126.2


SMOX
NM_175841.1
SIAT6
NM_174970.1
ZNF339
NM_021220.1


KIAA1033
XM_035313.6
MT1J
NM_175622.2
MGC14817
NM_032338.2


MDGA1
NM_153487.2
LOC255374
XM_171171.2
COL5A1
NM_000093.2


GADD45B
NM_015675.1
BCL2L12
NM_138639.1
LOC401082
XM_376257.1


LZTS2
NM_032429.1
FIBCD1
NM_032843.3
SH3BGRL3
NM_031286.2


TNFRSF19
NM_148957.2
CALR
NM_004343.2
ATP10D
NM_020453.2


STEAP
NM_012449.2
LZTR1
NM_006767.1
CD151
NM_139030.2


THBS3
NM_007112.3
SLC7A11
NM_014331.2
MGC33867
NM_138346.1


FLJ21673
NM_030898.2
STX1A
NM_004603.1
GNB5
NM_006578.2


NDEL1
NM_030808.2
ACTR1A
NM_005736.2
RNF24
NM_007219.2


FLJ21174
NM_024863.3
EPN2
NM_148921.2
LOC125704
XM_058931.6


MGC15482
NM_032875.1
OPRS1
NM_147158.1
LIPT1
NM_145198.1


FLJ10375
NM_018075.2
HAP1
NM_177977.1
KIAA0759
NM_015305.2


CPSF2
NM_017437.1
DIAPH2
NM_006729.2
FAD104
NM_022763.2


DKFZP566E144
NM_015523.1
MAP4K1
NM_007181.3
ARPC5
NM_005717.2


FLJ23614
NM_152695.2
CD99L2
NM_031462.1
C6orf37
NM_017633.1


LOC51315
NM_016618.1
SLC25A28
NM_031212.2
FLJ23042
NM_025157.2


SHCBP1
NM_024745.2
TBL1X
NM_005647.2
CNOT2
NM_014515.1


P24B
NM_007364.1
ZHX1
NM_007222.2
LAIR1
NM_021708.1


TRIM11
NM_145214.2
SIAT1
NM_003032.2
USF2
NM_003367.1


ARD1
NM_003491.2
FLJ11273
NM_018374.2
HSGP25L2G
NM_017510.3


LOC55831
NM_018447.1
KIAA0376
XM_037759.5
MSRB
NM_012228.2


BNIP3L
NM_004331.1
CCL21
NM_002989.2
OATL1
XM_047025.6


FLI1
NM_002017.2
CGI-128
NM_016062.1
ITCH
NM_031483.3


DKFZP434P1750
NM_015527.2
PAX7
NM_013945.1
DXS1283E
XM_047871.6


OAS1
NM_002534.1
APG7L
NM_006395.1
HGS
NM_004712.3


ADAM17
NM_021832.1
C20orf98
NM_024958.1
C20orf172
NM_024918.2


MUC3B
XM_168578.3
SDSL
NM_138432.2
CYLN2
NM_032421.1


DOCK11
NM_144658.2
PFKL
NM_002626.2
PP1665
NM_030792.4


SCARB2
NM_005506.2
NUCB2
NM_005013.1
C10orf13
NM_152429.2


CDIPT
NM_006319.2
RTN4
NM_007008.1
LOC92305
NM_138385.1


DEPDC2
NM_025170.2
IGF2R
NM_000876.1
FBLP-1
NM_017556.1


KPNA6
NM_012316.3
FIBL-6
NM_031935.1
STK3
NM_006281.1


FAT
NM_005245.1
PFAAP5
NM_014887.1
UBN1
NM_016936.2


FLJ12606
NM_024804.1
CDC10
NM_001788.2
LOC376138
XM_352084.1


SERPINE2
NM_006216.2
OGFR
NM_007346.2
AGPAT1
NM_032741.3


TRIM8
NM_030912.1
DSTN
NM_006870.2
SMAP-1
NM_017979.1


KIAA0769
NM_014824.1
TTC17
NM_018259.3
B4GALT2
NM_003780.2


UBXD1
NM_025241.1
STX5A
NM_003164.2
IKBKB
NM_001556.1


CDK5RAP2
NM_018249.3
LOC389562
XM_374236.1
GLCE
XM_290631.2


TRIM47
NM_033452.1
STK4
NM_006282.2
CD63
NM_001780.3


KLK3
NM_145864.1
PPP1R12C
NM_017607.1
FLJ34443
NM_175918.2


RAB3IL1
NM_013401.2
DHRS4
NM_021004.2
CSGlcA-T
XM_379974.1


DHX34
NM_014681.3
PHC2
NM_004427.2
TTC13
NM_024525.2


KIAA1841
XM_087056.4
C6orf72
NM_138785.1
VAMP5
NM_006634.2


IPLA2(GAMMA)
XM_291241.3
ANKRD13
NM_033121.1
RAC1
NM_006908.3


TNFRSF6B
NM_003823.2
LOC339665
XM_290973.1
FLJ11280
NM_018379.2


FLJ10415
NM_018089.1
DDX24
NM_020414.3
SMARCD3
NM_003078.2


MGC19764
XM_371039.2
MGC955
NM_024097.1
FLJ32205
NM_152561.1


FARP1
NM_005766.1
C20orf194
XM_045421.2
GMPPA
NM_013335.2


GPR1
NM_005279.2
SP140
NM_007237.2
FST
NM_013409.1


FLJ25402
NM_152567.1
SIRPB1
NM_006065.1
ZNF16
NM_006958.2


LOC134478
XM_068864.5
APOBEC3A
NM_145699.2
GCC1
NM_024523.5


SHMT2
NM_005412.3
CST3
NM_000099.2
GNA11
NM_002067.1


TNFSF15
NM_005118.2
OSBPL8
NM_020841.3
KIAA0256
NM_014701.1


TOM1
NM_005488.1
TMSL2
NM_182793.1
MAP3K3
NM_002401.3


ZCWCC2
NM_024657.2
VPS16
NM_080414.1
MKLN1
NM_013255.2


SELB
NM_021937.2
PKD2
NM_000297.2
PPP5C
NM_006247.2


MGC2749
NM_024069.2
LOC391023
XM_372773.1
GSN
NM_198252.1


SLC39A3
NM_144564.3
RGL1
NM_015149.2
RPL39
NM_001000.2


C20orf149
NM_024299.2
SART2
NM_013352.1
MGC32020
NM_152266.1


SLC17A5
NM_012434.3
SLC35A4
NM_080670.2
LOC389359
XM_374156.1


ALG3
NM_005787.3
EPS15L1
NM_021235.1
BLZF1
NM_003666.2


HNRPDL
NM_005463.2
ZNF554
NM_152303.1
SRPR
NM_003139.2


JM5
NM_007075.1
MYO1B
NM_012223.2
FLJ12221
XM_031342.1


SLC43A3
NM_017611.2
FLNB
NM_001457.1
LOC389445
XM_371857.2


KIAA0543
XM_379967.1
PRICKLE2
NM_198859.1
CNN2
NM_004368.2


DPYSL2
NM_001386.3
AKAP2
NM_147150.1
CGI-30
NM_015958.1


MGC4549
NM_032377.2
MGC19604
NM_080665.3
CTNS
NM_004937.1


GSTT1
NM_000853.1
TP53AP1
NM_007233.1
C5orf4
NM_016348.1


PHLDA2
NM_003311.2
SIAT4A
NM_173344.1
MPZL1
NM_003953.3


LOC400320
XM_375163.1
CPSF1
NM_013291.1
DKFZP434J154
NM_016003.1


EPB49
NM_001978.1
CBLN1
NM_004352.1
PTD008
NM_016145.1


FHL2
NM_001450.3
MBTD1
NM_017643.1
CALM2
NM_001743.3


CBX6
NM_014292.2
SARA1
NM_020150.3
FLJ22679
NM_032227.1


PELO
NM_015946.4
SIN3B
XM_050561.7
KRTCAP2
NM_173852.2


MGC50836
XM_171060.4
CALU
NM_001219.2
LOC399768
XM_378228.1


OCSP
NM_031945.2
LOC92912
NM_173469.1
C14orf24
NM_173607.2


KIAA0802
XM_031357.5
BIRC2
NM_001166.3
FLJ46603
NM_198530.1


MPPE1
NM_138608.1
IMPA2
NM_014214.1
DUSP6
NM_022652.2


LOC339352
XM_294910.1
MIPEP
NM_005932.1
FLJ31295
NM_152320.1


BMPR2
NM_033346.2
FBN2
NM_001999.2
FLJ00060
NM_033206.1


CYP4F3
NM_000896.1
PKD1-like
NM_024874.3
RAI
NM_006663.1


WSB2
NM_018639.3
LOC389816
XM_372161.1
ZDHHC18
NM_032283.1


CHST1
NM_003654.2
LOC374723
XM_351066.1
TMSL6
NM_181428.1


PLSCR3
NM_020360.2
KIAA0121
XM_052386.3
APG12L
NM_004707.2


TM4SF10
NM_031442.2
C15orf17
NM_020447.2
ATP6V1F
NM_004231.2


FXYD5
NM_014164.3
CTNND1
NM_001331.1
LOC402694
XM_380042.1


BBS1
NM_024649.4
FLJ22329
NM_024656.2
ZFYVE21
NM_024071.2


NDUFS7
NM_024407.3
MYL5
NM_002477.1
KIAA1223
XM_048747.8


EIF4EBP1
NM_004095.2
GMPPB
NM_021971.1
AHR
NM_001621.2


MGST1
NM_145792.1
IKBKG
NM_003639.2
FLJ14360
NM_032775.2


KIAA0103
NM_014673.2
LOC375378
XM_353337.1
IL18
NM_001562.2


C20orf30
NM_014145.3
SMBP
NM_020123.2
PRIC285
NM_033405.2


GPR10
NM_004248.1
CDKN1B
NM_004064.2
SH2D3C
NM_170600.1


SCAM-1
NM_005775.2
LOC375406
XM_351591.1
ACO1
NM_002197.1


EIF3S10
NM_003750.1
LOC253982
NM_181718.3
ZNF323
NM_145909.1


C14orf123
NM_014169.2
LOC390066
XM_372359.1
RAP1A
NM_002884.1


LOC389789
XM_372140.2
PCBP3
NM_020528.1
ZNF145
NM_006006.3


AGA
NM_000027.2
COMT
NM_007310.1
ECM1
NM_022664.1


DKFZp761D221
NM_032291.1
FAP
NM_004460.2
SSX5
NM_175723.1


H2AFJ
NM_177925.1
EMILIN1
NM_007046.1
CD44
NM_000610.2


SQRDL
NM_021199.1
IFITM2
NM_006435.1
NS3TP2
NM_023927.1


MOXD1
NM_015529.1
ANGPT1
NM_001146.3
RPS4Y
NM_001008.2


FLJ30681
NM_133459.1
BRIP1
NM_032043.1
TIMP3
NM_000362.3


ZNF345
NM_003419.2
SYTL2
NM_032379.2
TRADD
NM_003789.2


CKTSF1B1
NM_013372.4
DOC1
NM_014890.1
ACAS2L
NM_032501.2


GDF15
NM_004864.1
SPON2
NM_012445.1
C1QTNF1
NM_198593.1


ADFP
NM_001122.2
TNXB
NM_032470.2
EIF1AY
NM_004681.2


COL1A2
NM_000089.2
TXNIP
NM_006472.1
LOC400693
XM_375609.1


MAP1LC3A
NM_181509.1
SLITRK6
NM_032229.2
DAB2
NM_001343.1


RARRES2
NM_002889.2
LOC374822
XM_353149.1
HCA112
NM_018487.2


LAMA4
NM_002290.2
AKR1C4
NM_001818.2
SEDL
NM_014563.2


ZNF25
NM_145011.2
HLA-B
NM_005514.4
MMP14
NM_004995.2


MGC3207
NM_032285.1
CTGF
NM_001901.1
TWIST2
NM_057179.1


C21orf6
NM_016940.1
FLJ21986
NM_024913.3
LOC400682
XM_375590.1


INMT
NM_006774.3
PRSS11
NM_002775.2
INHBE
NM_031479.3


OXTR
NM_000916.3
FLJ22625
NM_024715.2
MGC4504
NM_024111.2


LY96
NM_015364.2
STOM
NM_198194.1
ATP8B4
XM_370863.1


KRTAP10-10
NM_181688.1
EIF5A2
NM_020390.5
ZHX3
NM_015035.2


LOC255849
XM_172855.1
KCNS1
NM_002251.3
LOC387758
NM_203371.1


NNMT
NM_006169.1
SERF1A
NM_021967.1
GDF10
NM_004962.2


NFIB
NM_005596.1
KIAA1237
XM_087386.5
FLJ14525
NM_032800.1


AKR1C2
NM_001354.4
AKR1C3
NM_003739.4
CLDN11
NM_005602.4


IL1R1
NM_000877.2
LOC400738
XM_378837.1
FLJ20701
NM_017933.3


LOC399959
XM_378316.1
LOC374987
XM_351264.1
RAB13
NM_002870.2


C10orf10
NM_007021.1
MAGP2
NM_003480.1
KIAA0779
XM_098229.9


PTX3
NM_002852.2
ST6GALNAC6
NM_013443.3
LOC286343
XM_210019.1


FLJ32332
NM_144641.1
PPFIBP1
NM_003622.2
SLCO1A2
NM_005075.2


FLJ38149
XM_375563.1
LOC388019
XM_373611.1
GLMN
NM_053274.1


DDIT3
NM_004083.3
PSMB8
NM_148919.2
UGCG
NM_003358.1


PAPPA
NM_002581.3
FLJ11286
NM_018381.1
SULT2B1
NM_177973.1


LOXL4
NM_032211.5
RASL12
NM_016563.2
NPDC1
NM_015392.2


CEBPD
NM_005195.2
TREML1
NM_178174.2
ABTB1
NM_172028.1


LOC93349
NM_138402.2
PI16
NM_153370.1
LOC390046
XM_372351.2


LOC401217
XM_376443.1
DKFZp686O1689
NM_182608.2
MRPL4
NM_146387.1


BCL6
NM_001706.2
FCGRT
NM_004107.3
STXBP5
NM_139244.2


ARF4L
NM_001661.2
ANKRD25
NM_015493.3
LOC400768
XM_378883.1


TGFB1I1
NM_015927.3
K6HF
NM_004693.1
LOC400649
XM_378746.1


PCDH18
NM_019035.2
JDP2
NM_130469.2
M17S2
NM_031862.1


COL6A2
NM_001849.2
EHD2
NM_014601.2
CDC42EP5
NM_145057.2


C9orf59
NM_033387.2
SPUVE
NM_007173.3
CD59
NM_000611.4


DSIPI
NM_004089.2
B7
NM_006992.2
PROS1
NM_000313.1


PDGFRB
NM_002609.2
C9orf88
NM_022833.1
APM2
NM_006829.1


MICB
NM_005931.2
EPS8
NM_004447.3
MBNL1
NM_021038.2


HPCAL1
NM_002149.2
LTBP4
NM_003573.1
LOC378157
XM_353658.1


EPLIN
NM_016357.1
LRP10
NM_014045.2
CORO1B
NM_020441.1


PLTP
NM_182676.1
MGC15737
NM_032926.2
LOC255783
NM_178511.2


COL6A3
NM_057167.1
RGC32
NM_014059.1
WARS
NM_004184.2


RRAS
NM_006270.2
HOXA5
NM_019102.2
CXCL16
NM_022059.1


SERPING1
NM_000062.1
PRNP
NM_183079.1
SPG20
NM_015087.3


TAP1
NM_000593.4
PDLIM2
NM_198042.2
RIS1
NM_015444.1


MGC16121
NM_032762.2
RNASE4
NM_194431.1
TRAM2
NM_012288.1


NCOA7
NM_181782.2
MGC10500
NM_031477.2
LCAT
NM_000229.1


MGC3047
NM_032348.2
UACA
NM_018003.1
CPNE7
NM_153636.1


DKFZp761G0122
NM_152661.1
PPP1R3C
NM_005398.3
NEXN
NM_144573.1


PDLIM7
NM_005451.3
ARRDC4
NM_183376.1
OPTN
NM_021980.3


FLJ11196
NM_018357.2
ZYX
NM_003461.3
KCNRG
NM_199464.1


URG4
NM_017920.2
MTHFD2
NM_006636.2
FOXP1
NM_032682.3


STAT1
NM_007315.2
IQGAP1
NM_003870.2
C1QTNF6
NM_182486.1


CNTNAP1
NM_003632.1
MGC50372
NM_173566.1
MGC23284
XM_378628.1


CBX3
NM_007276.3
ACTA2
NM_001613.1
C10orf6
NM_144592.1


INPP1
NM_002194.2
SLC12A4
NM_005072.3
ACSL4
NM_004458.1


FLJ37940
NM_178534.2
BCL3
NM_005178.2
FLJ11259
NM_018370.1


ZNF498
NM_145115.1
KIAA1949
XM_166376.1
GARS
NM_002047.1


FLJ12409
NM_025105.1
LOC375460
XM_351626.1
CLIPR-59
NM_015526.1


LOC389414
XM_374176.1
LOC400653
XM_378750.1
GRK5
NM_005308.1


IL10RB
NM_000628.3
GNB3
NM_002075.2
C11orf15
NM_020644.1


EVI5
NM_005665.2
MICA
NM_000247.1
FLJ25348
NM_144569.2


LOX
NM_002317.3
VEGFB
NM_003377.3
GPR124
NM_032777.6


NXPH4
NM_007224.1
SMARCA2
NM_139045.1
CPD
NM_001304.3


MGMT
NM_002412.1
NTE
NM_006702.2
BHD
NM_144997.3


DDAH1
NM_012137.2
LOC155340
XM_055725.3
PLK3
NM_004073.2


THRA
NM_003250.4
KRT15
NM_002275.2
FBLN2
NM_001998.1


TNC
NM_002160.1
FLJ37396
NM_173671.1
CTSD
NM_001909.3


SCGF
NM_002975.2
TPSB2
NM_024164.2
RNH
NM_002939.3


MGC14276
NM_153248.2
SBBI54
NM_138334.1
ANTXR2
NM_058172.1


NRXN3
NM_138970.2
LPPR2
NM_022737.1
MED8
NM_052877.2


CYP1A2
NM_000761.2
IL1R2
NM_004633.3
PCK2
NM_004563.1


BCR
NM_021574.1
ADM
NM_001124.1
LOC343069
XM_291395.1


FTH1
NM_002032.1
HCCR1
NM_015416.2
CD1C
NM_001765.1


LOC146784
XM_378694.1
JM4
NM_007213.1
EFEMP2
NM_016938.1


KIAA1055
NM_015079.2
LAMC1
NM_002293.2
PGRMC2
NM_006320.1


PRKCDBP
NM_145040.1
FER1L3
NM_133337.1
PPGB
NM_000308.1


MACF1
NM_012090.3
OS-9
NM_006812.1
MMP17
NM_016155.2


DACH
NM_080759.1
ITGB1
NM_002211.2
MME
NM_007288.1


TM7SF1
NM_003272.1
LENG8
NM_052925.1
HTATIP2
NM_006410.3


C20orf18
NM_031227.1
FLJ35976
NM_173639.1
TAGLN2
NM_003564.1


MOBKL2A
NM_130807.2
LOC90271
XM_030445.6
PLEC1
NM_000445.1


ACSL3
NM_004457.3
USH1G
NM_173477.2
ALDH1B1
NM_000692.3


A2LP
NM_017492.2
CARS
NM_001751.3
ATP6V1E1
NM_001696.2


CSF2RB
NM_000395.1
GCLC
NM_001498.2
KIAA0746
NM_015187.1


LOC400558
XM_378631.1
VEGFC
NM_005429.2
MCM8
NM_032485.4


PSAT1
NM_021154.3
CCL2
NM_002982.2
USP4
NM_003363.2


MBNL2
NM_144778.1
F2RL3
NM_003950.1
SBP1
NM_178121.1


RAMP1
NM_005855.1
MGC15875
NM_032921.1
HOXA6
NM_024014.2


PLA2G10
NM_003561.1
PDE4C
NM_000923.1
MTND3
NM_173710.1


LOC399788
XM_374817.1
HDLBP
NM_005336.2
PPP2R3A
NM_002718.3


IGSF8
NM_052868.1
MLLT1
NM_005934.2
PHLDA3
NM_012396.1


MGC57858
NM_178508.2
MGC20446
NM_153611.2
CCNT2
NM_058241.1


MGC33894
NM_152914.1
NOX1
NM_007052.3
PLXDC2
NM_032812.7


VN1R2
NM_173856.1
C9orf25
NM_147202.1
C14orf127
NM_025152.1


SEC22L1
NM_004892.3
OLIG3
NM_175747.2
LOC401488
XM_379617.1


LXN
NM_020169.2
ADCY3
NM_004036.2
DISP2
NM_033510.1


ELL
NM_006532.1
LOXL2
NM_002318.1
SEMA3F
NM_004186.2


STAT6
NM_003153.3
FKBP5
NM_004117.2
CTBS
NM_004388.1


C14orf31
NM_152330.2
B3GAT1
NM_054025.1
MYH9
NM_002473.2


NTAN1
NM_173474.2
JWA
NM_006407.2
LOC93109
NM_138399.2


GLRX2
NM_016066.3
ATOH8
NM_032827.3
PSFL
NM_031301.1


FN1
NM_002026.1
KIAA0605
NM_014694.2
ODF2
NM_153437.1


CAST
NM_173060.1
CSAD
NM_015989.3
SCARA3
NM_016240.2


LOC169355
NM_194294.1
LAMA3
NM_198129.1
TGFB3
NM_003239.1


LOC196463
NM_173542.2
LOC401026
XM_376160.1
FTHFSDC1
NM_015440.3


HEXB
NM_000521.2
DSCR1L2
NM_013441.2
TINF2
NM_012461.1


FLJ12529
NM_024811.2
NOXA1
XM_351868.1
SETBP1
NM_015559.1


ZNF524
NM_153219.2
NOD9
NM_024618.2
HLX1
NM_021958.2


CHIC2
NM_012110.2
TNIP3
NM_024873.2
MGC22014
XM_351441.1


MRC2
NM_006039.1
LOC388504
XM_373793.1
SLC22A12
NM_153378.1


PLP2
NM_002668.1
TMSL3
NM_183049.1
IMPDH1
NM_183243.1


ANAPC2
NM_013366.3
NPIP
NM_006985.1
BACE
NM_138973.1


HAX1
NM_006118.2
LOC285043
XM_379085.1
LGALS1
NM_002305.2


KLHL8
NM_020803.3
LOC388940
XM_373980.1
PADI1
NM_013358.1


DIA1
NM_000398.3
BAIAP2
NM_006340.1
LOC388690
XM_373865.2


UBXD4
NM_181713.3
STCH
NM_006948.3
RRAGC
NM_022157.2


ANG
NM_001145.2
HRI
NM_014413.2
KIAA0924
XM_375471.1


TPM2
NM_003289.2
KCTD13
NM_178863.2
TRAM1
NM_014294.3


DPYSL5
NM_020134.2
KCNJ2
NM_000891.2
LOC338755
XM_291980.3


ZBTB4
NM_020899.2
FLJ20006
NM_017618.1
LOC389050
XM_374014.1


MGC13024
NM_152288.1
LOC388278
XM_373687.1
LOC145853
XM_096885.6


LOC387749
XM_370606.1
DBN1
NM_080881.1
FLJ90798
NM_153367.1


HDAC8
NM_018486.1
FLJ36766
NM_182623.1
FLJ14464
NM_032789.1


CRSP3
NM_004830.2
NPEPL1
NM_024663.2
GAA
NM_000152.2


LOC389739
XM_372100.1
TXNL4
NM_006701.2
LOC90410
NM_174887.2


SPRY1
NM_199327.1
ARHGEF17
NM_014786.2
LIMS2
NM_017980.2


SESN2
NM_031459.3
LASS5
NM_147190.1
TAF10
NM_006284.2


LOC401886
XM_377480.1
SPRED2
NM_181784.1
MFAP2
NM_002403.2


CLN2
NM_000391.2
PRICKLE2
XM_093799.2
ADCY6
NM_020983.2


MT1H
NM_005951.1
PHF1
NM_002636.3
STOML1
NM_004809.3


FLJ13868
NM_022744.1
NPD007
NM_020684.2
C21orf97
NM_021941.1


EIIs1
NM_152793.1
MAGEL2
NM_019066.2
FLJ14011
NM_022103.2


USP47
NM_017944.2
CTDSP2
NM_005730.2
MSCP
NM_016612.1


STK10
NM_005990.1
TGFBI
NM_000358.1
LOC283710
XM_211174.1


LOC51145
NM_016158.1
SSBP2
NM_012446.2
CROT
NM_021151.2


KIAA1036
NM_014909.2
NID67
NM_032947.3
KIAA1068
NM_015332.2


LOC400771
XM_378890.1
DKFZP434N1923
NM_030974.2
COL3A1
NM_000090.2


ATP13A
NM_020410.1
CHPF
NM_024536.4
COMMD3
NM_012071.1


NYREN18
NM_016118.3
LOC253827
NM_198080.1
C6orf145
NM_183373.2


PARVA
NM_018222.2
VKORC1
NM_024006.3
ACTN4
NM_004924.3


C14orf75
NM_153046.1
FLJ20793
NM_019022.3
CLN5
NM_006493.1


DHRS1
NM_138452.1
PCNX
NM_014982.1
KIAA0841
XM_049237.5


SCAMP2
NM_005697.3
SLC28A1
NM_004213.3
ANKFY1
NM_016376.2


N4BP2
NM_018177.2
FAM8A1
NM_016255.1
RARA
NM_000964.1


LOC91526
NM_153697.1
STUB1
NM_005861.1
NFE2L1
NM_003204.1


DKFZP434B044
NM_031476.1
FLJ40852
NM_173677.1
C6orf48
NM_016947.1


LOC115294
NM_052937.1
TK2
NM_004614.2
ACK1
NM_005781.2


APBA3
NM_004886.2
PRKAG1
NM_002733.2
KIAA1164
NM_019092.1


DNASE2
NM_001375.1
FLJ13815
XM_086186.3
ZNF440
NM_152357.1


MGC47816
NM_173642.1
TXNRD1
NM_182743.1
SCIN
NM_033128.1


TAPBP
NM_003190.3
AMFR
NM_138958.1
ARPC2
NM_005731.2


IL13RA1
NM_001560.2
WDR1
NM_005112.3
EPIM
NM_001980.2


LOC400608
XM_375475.1
TENS1
NM_022748.6
SYNE1
NM_133650.1


UGCGL2
NM_020121.2
MDS006
NM_020233.3
PIP5K2B
NM_138687.1


C10orf42
NM_138357.1
EEF1G
NM_001404.3
ANXA5
NM_001154.2


RP4-622L5
NM_019118.2
MGC9913
XM_378178.1
DNCL2A
NM_177953.1


LOC389119
NM_203370.1
ITGB1
NM_033668.1
KIRREL
NM_018240.3


RPH3AL
NM_006987.2
MGC45386
NM_198527.1
TNFRSF10B
NM_147187.1


ARK5
NM_014840.1
C16orf35
NM_012075.1
RPS4X
NM_001007.3


MI-ER1
NM_020948.1
PPP2R5A
NM_006243.2
TCEA2
NM_003195.4


LOC388952
XM_373986.1
D4ST1
NM_130468.2
DKFZp762C186
XM_170658.1


NFE2L2
NM_006164.2
CUL4B
NM_003588.2
MAP4K4
NM_004834.2


KCNH4
NM_012285.1
CGI-72
NM_032205.2
HAK
NM_052947.1


DKFZp313G1735
NM_198150.1
PRO0456
NM_014127.1
CXCL12
NM_199168.1


ISGF3G
NM_006084.3
MYST1
NM_032188.1
C19orf10
NM_019107.1


FLJ20477
NM_017837.2
PLK2
NM_006622.1
SMYD3
NM_022743.1


SGCE
NM_003919.1
LOC377678
XM_352744.1
COL4A1
NM_001845.3


FLJ11196
NM_197958.1
MAN1B1
NM_007230.1
C10orf9
NM_145012.3


C20orf108
NM_080821.1
FLJ90231
NM_173581.1
TEX261
NM_144582.2


BHLHB2
NM_003670.1
CLSTN1
NM_014944.2
GBF1
NM_004193.1


C21orf69
NM_058189.1
TRIM16
NM_006470.2
FLJ14153
NM_022736.1


SLC16A3
NM_004207.1
MAP1LC3B
NM_022818.2
DUX3
NM_012148.1


DCX
NM_178153.1
ZNF155
NM_198089.1
MAPK4
NM_002747.2


RAB22A
NM_020673.2
F8A
NM_012151.2
LOC286016
NM_203419.1


RAB5B
NM_002868.2
HSPC072
NM_014162.1
STX12
NM_177424.1


MGAT4B
NM_014275.2
LOC374596
XM_350967.1
AP1M1
NM_032493.2


RHBDF1
NM_022450.2
MGC29956
NM_144638.1
CHRAC1
NM_017444.3


C9orf80
NM_021218.1
FLJ39035
NM_182580.1
EMS1
NM_138565.1


PIM1
NM_002648.2
PRKCI
NM_002740.3
LOC57146
NM_020422.3


MTVR1
NM_152832.1
PLA2G6
NM_003560.1
WDR23
NM_181357.1


HEBP1
NM_015987.2
ECGF1
NM_001953.2
DOC-1R
NM_005851.3


AIRE
NM_000383.1
LOC91978
XM_042012.4
AES
NM_001130.5


PDLIM1
NM_020992.2
CHKL
NM_005198.3
PL6
NM_007024.4


RPL13
NM_000977.2
ATF4
NM_182810.1
C9orf89
NM_032310.2


NCOR1
NM_006311.2
KIAA1442
XM_044921.4
DCP1B
NM_152640.3


DNAJC13
NM_015268.1
IGBP1
NM_001551.1
CTSB
NM_001908.2


TSC2
NM_021056.1
NSD1
NM_022455.3
TLE2
NM_003260.3


RNF149
NM_173647.2
AIP
NM_003977.1
RPL9
NM_000661.2


LOC388536
XM_371163.2
ORC5L
NM_002553.2
DKK3
NM_013253.3


PHCA
NM_018367.3
KLF16
NM_031918.1
SAS
NM_005981.3


TEX27
NM_021943.1
AMPD2
NM_139156.1
OBRGRP
NM_017526.2


RRP4
NM_014285.4
SLC2A4RG
NM_020062.3
UPLC1
NM_017707.2


ZFP64
NM_199427.1
KIAA0882
XM_093895.6
MRPL24
NM_024540.2


APG16L
NM_030803.5
TCEA1
NM_006756.2
GNB2
NM_005273.2


ENG
NM_000118.1
LOC399972
XM_378321.1
RXRB
NM_021976.3


CD81
NM_004356.2
DHRS8
NM_016245.1
VMP1
NM_030938.2


DOK1
NM_001381.2
G1P2
NM_005101.1
CASP10
NM_032974.1


FKBP1A
NM_000801.2
TPST1
NM_003596.2
KIAA0913
NM_015037.1


LOC64744
NM_022733.1
UQCRB
NM_006294.2
CAMLG
NM_001745.2


ARAF1
NM_001654.1
LOC88523
NM_033111.2
PNAS-4
NM_016076.2


OCIA
NM_017830.1
DPAGT1
NM_001382.2
APBA1
NM_001163.2


LOC400555
XM_375379.1
FLJ12078
NM_024977.1
NFATC3
NM_173164.1


PH-4
NM_177938.1
CXXC1
NM_014593.1
PRO1843
NM_018507.1


FKBP11
NM_016594.1
FLJ21127
NM_024549.3
DDIT4
NM_019058.1


DDEF2
NM_003887.1
FLNA
NM_001456.1
DNCH1
NM_001376.2


SH3GLB1
NM_016009.2
SDFR1
NM_017455.1
SLC22A18
NM_002555.3


CREB3
NM_006368.4
COL9A2
NM_001852.3
HSPA12B
NM_052970.3


C1orf8
NM_004872.3
RPL29
NM_000992.2
C20orf177
NM_022106.1


MGC42090
NM_152774.1
HARS
NM_002109.3
GLTSCR2
NM_015710.2


D2S448
XM_056455.3
RPL32
NM_000994.2
BARHL2
NM_020063.1


RPL13A
NM_012423.2
LOC388291
XM_373692.1
RPL41
NM_021104.1


LOC127262
NM_182752.2
FBXO18
NM_178150.1
KIAA0186
NM_021067.1


EEF1D
NM_001960.2
PTK9L
NM_007284.3
RELB
NM_006509.2


NMT2
NM_004808.1
HNOEL-iso
NM_020190.1
SERPINH1
NM_001235.2


KIAA0472
XM_290898.2
MGC21874
XM_291105.1
ABC1
NM_022070.3


ICA1
NM_022308.1
KDELR3
NM_006855.2
GRN
NM_002087.1


DEGS
NM_144780.1
MDS032
NM_018467.1
USP52
NM_014871.2


TRAPPC3
NM_014408.3
FLJ10486
NM_018109.2
FLJ14281
NM_024920.3


NFKBIL2
NM_013432.3
LOC90353
NM_145232.2
SEMA3C
NM_006379.2


C17orf37
NM_032339.3
E2F6
NM_198257.1
PLCD1
NM_006225.1


PLEKHA1
NM_021622.2
ARL6IP4
NM_018694.1
ZNF578
NM_152472.1


COX15
NM_004376.3
LYK5
NM_153335.3
LOC221143
NM_174928.1


RPS14
NM_005617.2
PXN
NM_002859.1
LTBP1
NM_000627.1


LEPREL2
NM_014262.2
KIAA0992
NM_016081.2
FLJ32965
NM_182506.1


ARHGEF11
NM_014784.2
KIAA0628
NM_014789.1
MGC33338
NM_152366.2


TREX2
NM_017518.3
SCDR10
NM_198704.1
CORO1C
NM_014325.2


PPP2R2B
NM_181677.1
LOC169611
NM_182487.1
LOC401483
XM_376800.1


MGC22805
NM_144590.1
C11orf1
NM_022761.1
RAPGEFL1
NM_016339.1


RNPEP
NM_020216.3
LOC199800
XM_373810.2
MOSPD3
NM_023948.3


MGC21518
NM_145274.1
ZNF499
NM_032792.2
FVT1
NM_002035.1


PLXNB2
XM_371474.1


TP53
NM_000546.2


HIP14
NM_015336.1


FLJ11078
NM_018316.1


M6PRBP1
NM_005817.2


LOC90624
NM_181705.1


RALA
NM_005402.2


ASPSCR1
NM_024083.2


PCDHB9
NM_019119.3


LOC161247
NM_203402.1


FLJ20531
NM_017865.2


PTK7
NM_152881.2


SARDH
NM_007101.2


MGC4238
NM_032332.2


PIGS
NM_033198.2


FLJ10803
NM_018224.1


OAZ1
NM_004152.2


SPUF
NM_013349.3


TNFSF4
NM_003326.2


BANF1
NM_003860.2


FLJ22471
NM_025140.1









Example 9
Formation of Capillary Tubes

Angiogenesis is the generation and formation of new capillary blood vessels, a process fundamental for processes like wound healing and reproduction. It's also involved in pathological processes (rheumatoid arthritis, tumor growth and metastasis).


Principle

In this case the assay is set up in order to test the influence of diverse compounds (i.e. chemical compounds or miRNA etc.) on tube formation by CMPC (cardio mycocyte progenitor cells). In order for these cells to form tubes these cells have to differentiate into smooth muscle cells and endothelial cells.


The formation of these newly formed capillary tubes is visualised by staining α-SMA (α smooth muscle actin) on smooth muscle cells and PECAM-1 (platelet endothelial cell adhesion molecule-1, CD31) on endothelial cells.


Materials and Methods

Reagents

    • a. CMPC (adult or fetal cardio mycocyte progenitor cells)
    • b. EBM-2 (endothelial cell basal medium). Clonetics, Cat. No. CC-3156
    • c. EGM-2: EBM-2+EGM-2 single qouts Cat. No. CC-4176+2% FBS
    • d. VEGF (vascular endothelial growth factor) S
    • e. 24-well cell culture cluster, flat bottom with lid. Corning Costar Cat. No. 3524
    • f. Microscope cover glasses 12 mm Ø No. 1 Cat. No. 01 115 20 Marienfeld GmbH & Co. KG.
    • g. Monoclonal anti actin, α-Smooth muscle, antibody produced in mouse. Sigma Cat. No. A2547 0.2 ml clone 1A4.
    • h. Alexa Fluor 488, goat anti mouse SFX kit. Invitrogen Cat. No. A31619
    • i. PECAM-1 (C-20) goat polyclonal IgG. Santa Cruz Biotechnology Cat. No. sc-1505 lot no. B169.
    • j. Donkey anti goat Cy3
    • k. Hoechst
    • l. Buffer Hoechst
    • m. In vitro angiogenesis assay kit. Chemicon International Cat. No. ECM625
    • n. PBS (phosphate buffered saline)
    • o. Water (demineralised)
    • p. Block buffer: 0.1% Saponin (supplier, Cat. No.) and 2% BSA (Sigma, Cat. No.) in PBS
    • q. 4% Para formaldehyde
    • r. Moviol


Apparatus

    • a. pipettes
    • b. microscopy
    • c. tweezers
    • d. Incubator 37° Celsius 5% CO2
    • e. Humidified box
    • f. Tissues
    • g. 200 μl pipette tips
    • h. scissors
    • i. refrigerator
    • j. freezer
    • k. parafilm


In Vitro Angiogenesis Assay

    • a. Make use of CMPC (adult/fetal)
    • b. Cells are cultured in SP++ (SP++: see reagent (c) in the section above, i.e. EBM-2+EGM-2 single qouts Cat. No. CC-4176+2% FBS). For cultivation use a split ratio between 1:3 and 1:10. Cells can be grown until passage number 22-24 after passage 22-24 the phenotype of the cell will change.


Day Before Start of Experiment:

    • a. Place sterilised (either by temperature or EtOH) 12 mm Ø microscope cover glasses into 24 well plate(s)
    • b. Put 24-well plate(s) together with 200 μl pipette tips at −20° C. overnight.
    • c. Thaw ECMatrix gel solution overnight on ice at 4° C.


Start of Experiment

    • a. Sterilise scissors in flow cabinet.
    • b. Place 24-well plate on ice in flow cabinet
    • c. Place ice cold 200 μl pipette tips in flow cabinet
    • d. Keep ECMatrix on ice before usage.
    • e. Cut off tip of 200 μl pipette tip and pipette 90 μl ECMatrix into one well of a 24-well plate. (work as quick and cold as possible) Repeat until all wanted wells are coated.
    • f. Make sure ECMatrix is even distributed in each well, then put plate for minimal one hour at 37° C.


Intermezzo


If test material is miRNA or DNA transfect cells before seeding cells onto ECMatrix If test material is a chemical compound just ad compound after/during seeding cells. Always take proper controls

    • g. After one hour seed 15.000 cell/well (0.5 ml/well) in a 24-well ECMatrix coated plate in EGM-2 medium with * % FBS with an extra addition of 100 ng/ml VEGF. Incubate cells for 24 h at 37° C. and 5% CO2. 0.5%-10%
    • h. If vessel formation has occurred take pictures before going further. This in order to safe and ensure results if anything goes wrong during staining.


Staining for α-SMA and PECAM-1

    • a. Remove supernatant and fixate cells with 4% Para formaldehyde during 15 minutes at RT 500 μl/well.
    • b. Wash twice with 1 ml PBS
    • c. Block 30 minutes with blocking buffer 500 μl/well
    • d. Meanwhile prepare primary anti body solution. Both primary antibodies can be used at the same time on the same sample.
    • e. Use 50 μl primary antibody solution for one sample (is one Microscope cover glasses 12 mm Ø). Dilute PECAM-1 1:200 in block buffer and α-SMA 1:40 in block buffer.
    • f. Pipette 50 μl primary antibody solution onto parafilm.
    • g. Take the Microscope cover glass(es) out of the 24-well plate with a pair of tweezers. Put upside down on the 50 μl antibody solution(s).
    • h. Place parafilm with samples in humidified box and incubate overnight at 4° C.
    • i. After incubation place microscope cover glasses back into a 24 well plate and wash 3 times with 1 ml PBS.
    • j. During last wash step prepare the first secondary antibody solution by diluting 1:400 Cy3 donkey anti-goat in PBS. Again 50 μl for each sample is used.


While Working with Secondary Antibodies Keep Samples Protected from Light as Much as Possible.

    • k. Pipette 50 μl first secondary antibody solution onto parafilm and add microscope cover glasses. (see step g)
    • l. Incubate during 1 hour at RT in the dark.
    • m. After incubation place Microscope cover glasses back into 24-well plate and wash 3 times with 1 ml PBS
    • n. During last wash step prepare the second secondary antibody solution by diluting 1:400 488 nm goat anti-mouse in PBS. Again 50 μl for each sample is used.
    • o. Pipette 50 μl second secondary antibody solution onto parafilm and add microscope cover glasses. (see step g)
    • p. Incubate at room temperature for 1 hour.


The results of an in vitro angiogenesis assay are depicted in FIG. 8.


Example 10
Human Foetal Cardiomyocyte Progenitor Cells Improve Left Ventricular Systolic Function after Myocardial Infarction in NOD/scid Mice

Recently, the existence of cardiomyocyte progenitor cells (CMPCs) that reside in the heart became appreciated. We isolated CMPCs from human foetal heart and showed that these human CMPCs (hCMPCs) can differentiate into functional cardiomyocytes in vitro. In the present study we investigated whether these cells are able to engraft in the ischemic myocardium and improve left ventricular function in an immune-compromised mouse myocardial infarction model.


Methods: hCMPCs were isolated from human fetal hearts by magnetic cell sorting based on expression of SCA-1. Myocardial infarction (MI) was induced in immune-compromised NOD/scid mice. Twenty minutes after MI, hCMPCs labelled with eGFP (hCMPC group, 2.0×10̂5 cells in 20 μl, n=11) or vehicle only (MI+Medium group, n=12) were injected into the infarcted area. Sham operated mice (Sham) were used as baseline (n=10). Two and 14 days after induction of MI, cardiac function was serially assessed using a vertical 9.4T animal MRI. Mice were then sacrificed and the engraftment and differentiation of injected cells was assessed by immunohistochemistry.


Results: At day 2, LV volumes were higher and EF (ejection fraction) was lower in both MI groups as compared to Sham, with no difference between the MI groups (FIG. 9). However at day 14, EF (ejection fraction) was higher and ESV (end systolic volume) was lower in the hCMPC group as compared to the MI+Medium group (P=0.001 and P=0.048 respectively). Although there was a trend towards a reduced EDV (end diastolic volume), this did not reach significance (P=0.14).


Conclusions: Foetal hCMPC engraft in the acutely infarcted myocardium, and improve LV systolic function. These results indicate the potential of hCMPC to be used in cell-based therapy for the treatment of IHD.


Example 11
Formation of Adipocytes

CMPCs of the invention are seeded and cultured until they have reached a confluence of 90-100% in SP++ (SP++: EBM-2+EGM-2 single qouts Cat. No. CC-4176+2% FBS). Subsequently, the medium is replaced by DMEM 4.5 g/l glucose+Na pyruvat containing 10% FBS, 1 uM dexamethasone, 0.5 mM 3-Isobutyl-1-methylxanthine (IBMX), 10 ug/ml Insulin, 0.2 mM indomethacin and Pen/strep. After some days, droplets of fat are deposited around the nucleus. Every 3 days the medium is refreshed.


To confirm adipocyte differentiation adipocytes were stained with Oil red according to the following protocol:


0.7 g Oil Red O stock solution FW 408.5, Sigma O-0625 was mixed with 200 ml Isopropanol; stirred O/N, then filtered with 0.2 im and stored at +4° C. The Oil Red O Working Solution was prepared from 6 parts Oil Red O stock; 4 parts dH2O; mixed and let sit at room temperature for 20 min and filtered with 0.2 im.


Most of the medium was removed and cells were fixated by adding 4% paraformaldehyde. The mixture was incubated for 5 min at room temperature, the formalin discarded and the same volume of fresh formalin was added. After incubation for at least 1 hour the cells can be kept in formalin for a couple of days before staining. Parafilm was wrapped around the plate to prevent drying and covered with aluminum foil. All the formalin was removed with a small transfer pipette and the wells are washed with 60% isopropanol. To the completely dry wells Oil Red O working solution was added for 10 min without touching the walls of the wells All Oil Red O was removed and immediately dH2O was added followed by washing with H2O 4 times. Then pictures were taken.


To further confirm adipocyte differentiation in vitro, RNA was isolated from differentiated and undifferentiated CMPCs. RT-PCR was performed on Leptin ((from the Greek leptos, meaning thin) is a protein hormone with important effects in regulating body weight, metabolism and reproductive function. Leptin is expressed predominantly by adipocytes, which fits with the idea that body weight is sensed as the total mass of fat in the body), Adipsin (is serine protease that is secreted by adipocytes), PPAR γ 2 (is predominantly expressed in fat tissue), GLUT 4 (glucose carriers in muscle and adipose tissues) & CYR61/CCN1 (expression decreased markedly during osteogenic differentiation, adipogenic differentiation and chondrogenic differentiation).


Which resulted in
















HFH
hMSC




















leptin
N/A
up



adipsin
up
up



pparγ2
up
up



Cnn1/cyr61
down
down



glut4












Therefore it can be concluded that CMPCs act in the same way as hMSC when differentiated in vitro. Therefore, the CMPCs of the invention are capable of differentiating into adipocytes. The results are shown in FIGS. 10a to 10f.


CITED REFERENCES



  • 1. Emanueli, C., Lako, M., Stojkovic, M. & Madeddu, P. In search of the best candidate for regeneration of ischemic tissues: are embryonic/fetal stem cells more advantageous than adult counterparts? Thromb. Haemost. 94, 738-749 (2005).

  • 2. Leri, A., Kajstura, J. & Anversa, P. Cardiac stem cells and mechanisms of myocardial regeneration. Physiol Rev. 85, 1373-1416 (2005).

  • 3. Smits, A. M., van Vliet, P., Hassink, R. J., Goumans, M. J. & Doevendans, P. A. The role of stem cells in cardiac regeneration. J. Cell Mol. Med. 9, 25-36 (2005).

  • 4. Van Laake, L. W., Van Hoof, D. & Mummery, C. L. Cardiomyocytes derived from stem cells. Ann. Med. 37, 499-512 (2005).

  • 5. Fukuda, K. & Fujita, J. Mesenchymal, but not hematopoietic, stem cells can be mobilized and differentiate into cardiomyocytes after myocardial infarction in mice. Kidney Int. 68, 1940-1943 (2005).

  • 6. Mangi, A. A. et al. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat. Med. 9, 1195-1201 (2003).

  • 7. Orlic, D. et al., Bone marrow cells regenerate infarcted myocardium. Nature 410, 701-705 (2001).

  • 8. Beltrami, A. P. et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 114, 763-776 (2003).

  • 9. Behfar, A. et al. Stem cell differentiation requires a paracrine pathway in the heart. FASEB J. 16, 1558-1566 (2002).

  • 10. Menard, C. et al. Transplantation of cardiac-committed mouse embryonic stem cells to infarcted sheep myocardium: a preclinical study. Lancet 366, 1005-1012 (2005).

  • 11. Rubart, M. et al. Physiological coupling of donor and host cardiomyocytes after cellular transplantation. Circ. Res. 92, 1217-1224 (2003).

  • 12. Muller-Ehmsen, J. et al. Rebuilding a damaged heart: long-term survival of transplanted neonatal rat cardiomyocytes after myocardial infarction and effect on cardiac function. Circulation 105, 1720-1726 (2002).

  • 13. Schachinger, V. et al. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction: final one-year results of the TOPCARE-AMI Trial. J. Am. Coll. Cardiol. 44, 1690-1699 (2004).

  • 14. Menasche, P. et al. Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction. J. Am. Coll. Cardiol. 41, 1078-1083 (2003).

  • 15. Smits, P. C. et al. Catheter-based intramyocardial injection of autologous skeletal myoblasts as a primary treatment of ischemic heart failure: clinical experience with six-month follow-up. J. Am. Coll. Cardiol. 42, 2063-2069 (2003).

  • 16. Meyer, G. P. et al. Intracoronary bone marrow cell transfer after myocardial infarction: eighteen months' follow-up data from the randomized, controlled BOOST (BOne marrOw transfer to enhance ST-elevation infarct regeneration) trial. Circulation 113, 1287-1294 (2006).

  • 17. Balsam, L. B. et al. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature 428, 668-673 (2004).

  • 18. Murry, C. E. et al. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature 428, 664-668 (2004).

  • 19. Mummery, C. et al. Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells. Circulation 107, 2733-2740 (2003).

  • 20. Passier, R. et al. Increased cardiomyocyte differentiation from human embryonic stem cells in serum-free cultures. Stem Cells 23, 772-780 (2005).

  • 21. Xu, C., Police, S., Rao, N. & Carpenter, M. K. Characterization and enrichment of cardiomyocytes derived from human embryonic stem cells. Circ. Res. 91, 501-508 (2002).

  • 22. Oh, H. et al. Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc. Natl. Acad. Sci. U.S.A. 100, 12313-12318 (2003).

  • 23. Laugwitz, K. L. et al. Postnatal isl1+ cardioblasts enter fully differentiated cardiomyocyte lineages. Nature 433, 647-653 (2005).

  • 24. Pfister, O. et al. Circ. Res. 97, 52-61 (2005).

  • 25. Messina, E. et al. Isolation and expansion of adult cardiac stem cells from human and murine heart. Circ. Res. 95, 911-921 (2004).

  • 26. Slager, H. G., Van Inzen, W., Freund, E., Van den Eijnden-Van Raaij A J & Mummery, C. L. Transforming growth factor-beta in the early mouse embryo: implications for the regulation of muscle formation and implantation. Dev. Genet. 14, 212-224 (1993).

  • 27. Takahashi, T. et al. Ascorbic acid enhances differentiation of embryonic stem cells into cardiac myocytes. Circulation 107, 1912-1916 (2003).

  • 28. Sachinidis, A. et al. Cardiac specific differentiation of mouse embryonic stem cells. Cardiovasc. Res. 58, 278-291 (2003).

  • 29. Li, T. S. et al. Regeneration of infarcted myocardium by intramyocardial implantation of ex vivo transforming growth factor-beta-preprogrammed bone marrow stem cells. Circulation 111, 2438-2445 (2005).

  • 30. ten Dijke, P. & Hill, C. S, New insights into TGF-beta-Smad signalling. Trends Biochem. Sci. 29, 265-273 (2004).

  • 31. Inman, G. J. et al. SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7. Mol. Pharmacol. 62, 65-74 (2002).

  • 32. Mouquet, F. at al. Restoration of cardiac progenitor cells after myocardial infarction by self-proliferation and selective homing of bone marrow-derived stem cells. Circ. Res. 97, 1090-1092 (2005).

  • 33. Flanders, K. C., Holder, M. G. & Winokur, T. S. Autoinduction of mRNA and protein expression for transforming growth factor-beta S in cultured cardiac cells. J. Mol. Cell Cardiol. 27, 805-812 (1995).

  • 34. van der Heyden, M. A. et al. P19 embryonal carcinoma cells: a suitable model system for cardiac electrophysiological differentiation at the molecular and functional level. Cardiovasc. Res. 58, 410-422 (2003).

  • 35. Goumans, M. J. et al. Balancing the activation state of the endothelium via two distinct TGF-beta type I receptors. EMBO J. 21, 1743-1753 (2002).


Claims
  • 1. Enriched human cardiomyocyte progenitor cells (CMPCs) which are characterized by Sca-1 or Sca-1-like epitopes and CD31 on their cell surfaces.
  • 2. A method for enriching human cardiomyocyte progenitor cells (CMPCs), comprising the steps of: (a) dissociating heart tissue; and(b) enriching CMPCs with a Sca-1 binding agent and/or a CD31 binding agent.
  • 3-5. (canceled)
  • 6. The method of claim 2, wherein said binding agent is an anti-Sca-1 antibody and/or an anti CD31 antibody.
  • 7. The method of claim 2, wherein said enriching in step b) is by MACS or FACS.
  • 8. Enriched human cardiomyocyte progenitor cells (CMPCs) which are characterized by Sca-1 or a Sca-1 like epitopes and CD31 on their cell surfaces, which are obtainable by a method of claim 2.
  • 9. CMPCs according to claim 1, which are capable of differentiation into cardiomyocytes in vitro after 5-Aza treatment in the presence of ascorbic acid.
  • 10-11. (canceled)
  • 12. An in vitro method for the differentiation of CMPCs into cardiomyocytes, comprising the steps of: (a) providing CMPCs according to claim 1;(b) treating said CMPCs with a demethylating agent; and(c) allowing the so-treated CMPCs to differentiate into cardiomyocytes.
  • 13. The method of claim 12, wherein said demethylating agent is 5-azacytidine or 5-aza-2′-deoxycytidine.
  • 14. The method of claim 12, wherein said CMPCs are treated with said demethylating agent in the presence of an antioxidation agent.
  • 15. The method of claim 12 further comprising the step of treating the CMPCs with a TGF-β family member.
  • 16. The method of claim 15, wherein said TGF-β family member is TGF-β or BMP.
  • 17. (canceled)
  • 18. A pharmaceutical composition comprising cardiomyocytes obtainable by the method of claim 12.
  • 19-25. (canceled)
  • 26. A method to treat myocardial infarction or to ameliorate the effects of myocardial infarction which method comprises administering to a subject in need of such treatment or amelioration an effective amount of the pharmaceutical composition of claim 18.
  • 27. The method of claim 26, wherein cardiac contractile force is improved.
  • 28. A screening method to identify a substance that affects the phenotype of the cardiomyocytes obtainable by the method of claim 12, which method comprises the steps of: (a) bringing said cardiomyocytes into contact with a test substance; and(b) evaluating the effect of said test substance on the phenotype of said cardiomyocytes, thereby identifying a substance that affects the phenotype of the cardiomyocytes.
  • 29. The method of claim 28, wherein said test substance is a drug.
Priority Claims (2)
Number Date Country Kind
EP07000374.4 Jan 2007 EP regional
EP08000174.6 Jan 2008 EP regional
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/EP2008/000119 1/9/2008 WO 00 4/19/2010