US20140274765A1 - Cardiac stem cell and myocyte secreted paracrine factors and uses thereof - Google Patents
Cardiac stem cell and myocyte secreted paracrine factors and uses thereof Download PDFInfo
- Publication number
- US20140274765A1 US20140274765A1 US14/134,301 US201314134301A US2014274765A1 US 20140274765 A1 US20140274765 A1 US 20140274765A1 US 201314134301 A US201314134301 A US 201314134301A US 2014274765 A1 US2014274765 A1 US 2014274765A1
- Authority
- US
- United States
- Prior art keywords
- protein
- proteins
- cells
- precursor
- cardiac
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Abandoned
Links
- 210000000130 stem cell Anatomy 0.000 title claims abstract description 64
- 230000000747 cardiac effect Effects 0.000 title claims abstract description 41
- 210000000107 myocyte Anatomy 0.000 title abstract description 37
- 230000003076 paracrine Effects 0.000 title description 20
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 138
- 102000004169 proteins and genes Human genes 0.000 claims abstract description 136
- 238000000034 method Methods 0.000 claims description 30
- 210000001519 tissue Anatomy 0.000 claims description 21
- 238000009168 stem cell therapy Methods 0.000 claims description 18
- 238000009580 stem-cell therapy Methods 0.000 claims description 18
- 102100028252 Brain acid soluble protein 1 Human genes 0.000 claims description 17
- 210000004165 myocardium Anatomy 0.000 claims description 17
- 108010039419 Connective Tissue Growth Factor Proteins 0.000 claims description 13
- 108090000964 Insulin-like growth factor binding protein 2 Proteins 0.000 claims description 13
- 102100026802 72 kDa type IV collagenase Human genes 0.000 claims description 12
- 101000627872 Homo sapiens 72 kDa type IV collagenase Proteins 0.000 claims description 12
- 102000004379 Adrenomedullin Human genes 0.000 claims description 11
- 101800004616 Adrenomedullin Proteins 0.000 claims description 11
- 101710087660 Brain acid soluble protein 1 Proteins 0.000 claims description 11
- 102100039364 Metalloproteinase inhibitor 1 Human genes 0.000 claims description 11
- ULCUCJFASIJEOE-NPECTJMMSA-N adrenomedullin Chemical compound C([C@@H](C(=O)N[C@@H](CCC(N)=O)C(=O)NCC(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)NCC(=O)N[C@@H]1C(N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=2C=CC=CC=2)C(=O)NCC(=O)N[C@H](C(=O)N[C@@H](CSSC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CO)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCC(N)=O)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(N)=O)[C@@H](C)O)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCSC)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=CC=C1 ULCUCJFASIJEOE-NPECTJMMSA-N 0.000 claims description 11
- 102100038381 Cystatin-M Human genes 0.000 claims description 10
- 206010019280 Heart failures Diseases 0.000 claims description 10
- 101000884770 Homo sapiens Cystatin-M Proteins 0.000 claims description 10
- 239000000523 sample Substances 0.000 claims description 10
- 108050006599 Metalloproteinase inhibitor 1 Proteins 0.000 claims description 9
- 102100029470 Apolipoprotein E Human genes 0.000 claims description 8
- 101710095339 Apolipoprotein E Proteins 0.000 claims description 8
- 239000002243 precursor Substances 0.000 claims description 8
- 210000002966 serum Anatomy 0.000 claims description 8
- 101000935689 Homo sapiens Brain acid soluble protein 1 Proteins 0.000 claims description 6
- 108010033276 Peptide Fragments Proteins 0.000 claims description 6
- 102000007079 Peptide Fragments Human genes 0.000 claims description 6
- 102100026262 Metalloproteinase inhibitor 2 Human genes 0.000 claims description 4
- 238000001574 biopsy Methods 0.000 claims description 4
- 210000004369 blood Anatomy 0.000 claims description 4
- 239000008280 blood Substances 0.000 claims description 4
- 102100026632 Mimecan Human genes 0.000 claims description 3
- 108010031374 Tissue Inhibitor of Metalloproteinase-1 Proteins 0.000 claims description 3
- 230000008901 benefit Effects 0.000 claims description 3
- 239000012472 biological sample Substances 0.000 claims description 3
- 230000017074 necrotic cell death Effects 0.000 claims description 3
- 206010002383 Angina Pectoris Diseases 0.000 claims description 2
- 102100037985 Dickkopf-related protein 3 Human genes 0.000 claims description 2
- 101710099550 Dickkopf-related protein 3 Proteins 0.000 claims description 2
- 206010020880 Hypertrophy Diseases 0.000 claims description 2
- 102100022710 Insulin-like growth factor-binding protein 2 Human genes 0.000 claims description 2
- 108091013859 Mimecan Proteins 0.000 claims description 2
- 238000012216 screening Methods 0.000 claims description 2
- 102000002723 Atrial Natriuretic Factor Human genes 0.000 claims 2
- 102000002734 Collagen Type VI Human genes 0.000 claims 2
- 108010043741 Collagen Type VI Proteins 0.000 claims 2
- 102100036706 Interleukin-1 receptor-like 1 Human genes 0.000 claims 2
- FPIPGXGPPPQFEQ-OVSJKPMPSA-N all-trans-retinol Chemical compound OC\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C FPIPGXGPPPQFEQ-OVSJKPMPSA-N 0.000 claims 2
- 102000001187 Collagen Type III Human genes 0.000 claims 1
- 108010069502 Collagen Type III Proteins 0.000 claims 1
- 102000012432 Collagen Type V Human genes 0.000 claims 1
- 108010022514 Collagen Type V Proteins 0.000 claims 1
- 102000015225 Connective Tissue Growth Factor Human genes 0.000 claims 1
- 108010014258 Elastin Proteins 0.000 claims 1
- 102100033167 Elastin Human genes 0.000 claims 1
- 108700003107 Interleukin-1 Receptor-Like 1 Proteins 0.000 claims 1
- 108050006602 Metalloproteinase inhibitor 2 Proteins 0.000 claims 1
- 101710187800 Natriuretic peptides A Proteins 0.000 claims 1
- 108010050808 Procollagen Proteins 0.000 claims 1
- 108010031372 Tissue Inhibitor of Metalloproteinase-2 Proteins 0.000 claims 1
- 208000037892 acute myocardial injury Diseases 0.000 claims 1
- 235000019169 all-trans-retinol Nutrition 0.000 claims 1
- 239000011717 all-trans-retinol Substances 0.000 claims 1
- 210000004748 cultured cell Anatomy 0.000 claims 1
- 108010028069 procathepsin B Proteins 0.000 claims 1
- 210000004027 cell Anatomy 0.000 abstract description 73
- 230000001225 therapeutic effect Effects 0.000 abstract description 7
- 210000002216 heart Anatomy 0.000 description 21
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 18
- 101800001288 Atrial natriuretic factor Proteins 0.000 description 17
- 102400001282 Atrial natriuretic peptide Human genes 0.000 description 17
- 101800001890 Atrial natriuretic peptide Proteins 0.000 description 17
- NSQLIUXCMFBZME-MPVJKSABSA-N carperitide Chemical compound C([C@H]1C(=O)NCC(=O)NCC(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@H](C(NCC(=O)N[C@@H](C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)NCC(=O)N[C@@H](CSSC[C@@H](C(=O)N1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CO)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(O)=O)=O)[C@@H](C)CC)C1=CC=CC=C1 NSQLIUXCMFBZME-MPVJKSABSA-N 0.000 description 17
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 14
- 102100031168 CCN family member 2 Human genes 0.000 description 13
- 241000700159 Rattus Species 0.000 description 13
- 230000027455 binding Effects 0.000 description 13
- 230000008929 regeneration Effects 0.000 description 13
- 238000011069 regeneration method Methods 0.000 description 13
- 108010067003 Interleukin-33 Proteins 0.000 description 12
- 102000017761 Interleukin-33 Human genes 0.000 description 12
- 102000004372 Insulin-like growth factor binding protein 2 Human genes 0.000 description 11
- 108020004999 messenger RNA Proteins 0.000 description 11
- 238000002560 therapeutic procedure Methods 0.000 description 11
- 241001465754 Metazoa Species 0.000 description 10
- 102000001708 Protein Isoforms Human genes 0.000 description 10
- 108010029485 Protein Isoforms Proteins 0.000 description 10
- 238000011282 treatment Methods 0.000 description 10
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 8
- 108090000712 Cathepsin B Proteins 0.000 description 8
- 102000004225 Cathepsin B Human genes 0.000 description 8
- 102000019197 Superoxide Dismutase Human genes 0.000 description 8
- 108010012715 Superoxide dismutase Proteins 0.000 description 8
- 238000004458 analytical method Methods 0.000 description 8
- 239000012091 fetal bovine serum Substances 0.000 description 8
- 239000012528 membrane Substances 0.000 description 8
- 230000035755 proliferation Effects 0.000 description 8
- 230000004083 survival effect Effects 0.000 description 8
- 230000035899 viability Effects 0.000 description 8
- 230000006378 damage Effects 0.000 description 7
- 230000004069 differentiation Effects 0.000 description 7
- 238000002347 injection Methods 0.000 description 7
- 239000007924 injection Substances 0.000 description 7
- 239000003446 ligand Substances 0.000 description 7
- 239000002609 medium Substances 0.000 description 7
- 210000004379 membrane Anatomy 0.000 description 7
- 108090000765 processed proteins & peptides Proteins 0.000 description 7
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 6
- 108090000631 Trypsin Proteins 0.000 description 6
- 102000004142 Trypsin Human genes 0.000 description 6
- 208000027418 Wounds and injury Diseases 0.000 description 6
- 238000001514 detection method Methods 0.000 description 6
- 208000019622 heart disease Diseases 0.000 description 6
- 230000001965 increasing effect Effects 0.000 description 6
- 208000014674 injury Diseases 0.000 description 6
- 238000004949 mass spectrometry Methods 0.000 description 6
- 208000010125 myocardial infarction Diseases 0.000 description 6
- 239000012588 trypsin Substances 0.000 description 6
- 108010035532 Collagen Proteins 0.000 description 5
- 102000008186 Collagen Human genes 0.000 description 5
- 150000001413 amino acids Chemical group 0.000 description 5
- 210000004413 cardiac myocyte Anatomy 0.000 description 5
- 238000004113 cell culture Methods 0.000 description 5
- 230000030833 cell death Effects 0.000 description 5
- 229920001436 collagen Polymers 0.000 description 5
- 210000002950 fibroblast Anatomy 0.000 description 5
- 238000012544 monitoring process Methods 0.000 description 5
- 238000004366 reverse phase liquid chromatography Methods 0.000 description 5
- 239000000243 solution Substances 0.000 description 5
- UZOVYGYOLBIAJR-UHFFFAOYSA-N 4-isocyanato-4'-methyldiphenylmethane Chemical compound C1=CC(C)=CC=C1CC1=CC=C(N=C=O)C=C1 UZOVYGYOLBIAJR-UHFFFAOYSA-N 0.000 description 4
- 108010088751 Albumins Proteins 0.000 description 4
- 102000009027 Albumins Human genes 0.000 description 4
- 108010014064 CCCTC-Binding Factor Proteins 0.000 description 4
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical group O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 4
- 102000019223 Interleukin-1 receptor Human genes 0.000 description 4
- 108050006617 Interleukin-1 receptor Proteins 0.000 description 4
- 241000699670 Mus sp. Species 0.000 description 4
- 102100027671 Transcriptional repressor CTCF Human genes 0.000 description 4
- 210000004556 brain Anatomy 0.000 description 4
- 230000001413 cellular effect Effects 0.000 description 4
- 239000012634 fragment Substances 0.000 description 4
- 238000005259 measurement Methods 0.000 description 4
- 238000002360 preparation method Methods 0.000 description 4
- 239000003656 tris buffered saline Substances 0.000 description 4
- 230000002861 ventricular Effects 0.000 description 4
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 4
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 108090000672 Annexin A5 Proteins 0.000 description 3
- 102000004121 Annexin A5 Human genes 0.000 description 3
- 102000014914 Carrier Proteins Human genes 0.000 description 3
- 108010061617 Cystatin M Proteins 0.000 description 3
- 102000012177 Cystatin M Human genes 0.000 description 3
- 108010052285 Membrane Proteins Proteins 0.000 description 3
- 241000699666 Mus <mouse, genus> Species 0.000 description 3
- 102400001018 Proadrenomedullin N-20 terminal peptide Human genes 0.000 description 3
- 101800000795 Proadrenomedullin N-20 terminal peptide Proteins 0.000 description 3
- 108010026552 Proteome Proteins 0.000 description 3
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 3
- 239000006180 TBST buffer Substances 0.000 description 3
- 238000002835 absorbance Methods 0.000 description 3
- 230000004913 activation Effects 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 108091008324 binding proteins Proteins 0.000 description 3
- 230000033228 biological regulation Effects 0.000 description 3
- 210000000038 chest Anatomy 0.000 description 3
- 230000003750 conditioning effect Effects 0.000 description 3
- 238000011161 development Methods 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- 230000029087 digestion Effects 0.000 description 3
- 238000002474 experimental method Methods 0.000 description 3
- 239000000834 fixative Substances 0.000 description 3
- 210000002064 heart cell Anatomy 0.000 description 3
- 210000005003 heart tissue Anatomy 0.000 description 3
- 238000001727 in vivo Methods 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- 238000002372 labelling Methods 0.000 description 3
- 230000007774 longterm Effects 0.000 description 3
- 210000004072 lung Anatomy 0.000 description 3
- 238000002595 magnetic resonance imaging Methods 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 230000010412 perfusion Effects 0.000 description 3
- 238000011002 quantification Methods 0.000 description 3
- 230000001172 regenerating effect Effects 0.000 description 3
- 210000001567 regular cardiac muscle cell of ventricle Anatomy 0.000 description 3
- 230000008439 repair process Effects 0.000 description 3
- 230000004044 response Effects 0.000 description 3
- 230000004043 responsiveness Effects 0.000 description 3
- 238000004007 reversed phase HPLC Methods 0.000 description 3
- 238000004885 tandem mass spectrometry Methods 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 238000002054 transplantation Methods 0.000 description 3
- 230000002792 vascular Effects 0.000 description 3
- 102000002659 Amyloid Precursor Protein Secretases Human genes 0.000 description 2
- 108010043324 Amyloid Precursor Protein Secretases Proteins 0.000 description 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- 102000018233 Fibroblast Growth Factor Human genes 0.000 description 2
- 108050007372 Fibroblast Growth Factor Proteins 0.000 description 2
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 2
- 102100037362 Fibronectin Human genes 0.000 description 2
- 108010067306 Fibronectins Proteins 0.000 description 2
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 2
- 101000645296 Homo sapiens Metalloproteinase inhibitor 2 Proteins 0.000 description 2
- 208000026350 Inborn Genetic disease Diseases 0.000 description 2
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 2
- 239000007760 Iscove's Modified Dulbecco's Medium Substances 0.000 description 2
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 2
- 229930182816 L-glutamine Natural products 0.000 description 2
- 102000018697 Membrane Proteins Human genes 0.000 description 2
- 108010006035 Metalloproteases Proteins 0.000 description 2
- 102000005741 Metalloproteases Human genes 0.000 description 2
- 108020004711 Nucleic Acid Probes Proteins 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 102000010780 Platelet-Derived Growth Factor Human genes 0.000 description 2
- 108010038512 Platelet-Derived Growth Factor Proteins 0.000 description 2
- 102000005353 Tissue Inhibitor of Metalloproteinase-1 Human genes 0.000 description 2
- 206010000891 acute myocardial infarction Diseases 0.000 description 2
- 210000004504 adult stem cell Anatomy 0.000 description 2
- 125000000539 amino acid group Chemical group 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 239000000427 antigen Substances 0.000 description 2
- 102000036639 antigens Human genes 0.000 description 2
- 108091007433 antigens Proteins 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 210000001185 bone marrow Anatomy 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- YKYOUMDCQGMQQO-UHFFFAOYSA-L cadmium dichloride Chemical compound Cl[Cd]Cl YKYOUMDCQGMQQO-UHFFFAOYSA-L 0.000 description 2
- 230000006037 cell lysis Effects 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- -1 chromosome Proteins 0.000 description 2
- 239000003636 conditioned culture medium Substances 0.000 description 2
- 230000001143 conditioned effect Effects 0.000 description 2
- 238000011109 contamination Methods 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 238000002592 echocardiography Methods 0.000 description 2
- 230000000694 effects Effects 0.000 description 2
- 230000003511 endothelial effect Effects 0.000 description 2
- 229940088598 enzyme Drugs 0.000 description 2
- 210000002919 epithelial cell Anatomy 0.000 description 2
- 229940126864 fibroblast growth factor Drugs 0.000 description 2
- 238000000684 flow cytometry Methods 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 208000016361 genetic disease Diseases 0.000 description 2
- 239000005556 hormone Substances 0.000 description 2
- 229940088597 hormone Drugs 0.000 description 2
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 2
- 230000002452 interceptive effect Effects 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- 239000006166 lysate Substances 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 238000013508 migration Methods 0.000 description 2
- 230000005012 migration Effects 0.000 description 2
- 238000010232 migration assay Methods 0.000 description 2
- 210000001700 mitochondrial membrane Anatomy 0.000 description 2
- 239000000203 mixture Substances 0.000 description 2
- 230000002107 myocardial effect Effects 0.000 description 2
- 230000002644 neurohormonal effect Effects 0.000 description 2
- 239000002853 nucleic acid probe Substances 0.000 description 2
- 239000013610 patient sample Substances 0.000 description 2
- 102000004196 processed proteins & peptides Human genes 0.000 description 2
- 238000012545 processing Methods 0.000 description 2
- 238000004393 prognosis Methods 0.000 description 2
- 230000002062 proliferating effect Effects 0.000 description 2
- 238000000575 proteomic method Methods 0.000 description 2
- 208000002815 pulmonary hypertension Diseases 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 229960005322 streptomycin Drugs 0.000 description 2
- 230000035882 stress Effects 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 238000010200 validation analysis Methods 0.000 description 2
- 210000003556 vascular endothelial cell Anatomy 0.000 description 2
- 210000005166 vasculature Anatomy 0.000 description 2
- 239000011800 void material Substances 0.000 description 2
- ZKRFOXLVOKTUTA-KQYNXXCUSA-N 9-(5-phosphoribofuranosyl)-6-mercaptopurine Chemical compound O[C@@H]1[C@H](O)[C@@H](COP(O)(O)=O)O[C@H]1N1C(NC=NC2=S)=C2N=C1 ZKRFOXLVOKTUTA-KQYNXXCUSA-N 0.000 description 1
- ATRRKUHOCOJYRX-UHFFFAOYSA-N Ammonium bicarbonate Chemical compound [NH4+].OC([O-])=O ATRRKUHOCOJYRX-UHFFFAOYSA-N 0.000 description 1
- 229910000013 Ammonium bicarbonate Inorganic materials 0.000 description 1
- 206010002388 Angina unstable Diseases 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 206010007556 Cardiac failure acute Diseases 0.000 description 1
- 102100028892 Cardiotrophin-1 Human genes 0.000 description 1
- 208000024172 Cardiovascular disease Diseases 0.000 description 1
- 102000005600 Cathepsins Human genes 0.000 description 1
- 108010084457 Cathepsins Proteins 0.000 description 1
- 241000700199 Cavia porcellus Species 0.000 description 1
- KDXKERNSBIXSRK-RXMQYKEDSA-N D-lysine Chemical compound NCCCC[C@@H](N)C(O)=O KDXKERNSBIXSRK-RXMQYKEDSA-N 0.000 description 1
- 230000006820 DNA synthesis Effects 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 102000009024 Epidermal Growth Factor Human genes 0.000 description 1
- 102400001368 Epidermal growth factor Human genes 0.000 description 1
- 101800003838 Epidermal growth factor Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 description 1
- 102100024785 Fibroblast growth factor 2 Human genes 0.000 description 1
- 206010016654 Fibrosis Diseases 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 101150007870 HEMA3 gene Proteins 0.000 description 1
- 208000013875 Heart injury Diseases 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000777550 Homo sapiens CCN family member 2 Proteins 0.000 description 1
- 101000852968 Homo sapiens Interleukin-1 receptor-like 1 Proteins 0.000 description 1
- 101001013150 Homo sapiens Interstitial collagenase Proteins 0.000 description 1
- 101000669513 Homo sapiens Metalloproteinase inhibitor 1 Proteins 0.000 description 1
- 101000585365 Homo sapiens Sulfotransferase 2A1 Proteins 0.000 description 1
- 206010062767 Hypophysitis Diseases 0.000 description 1
- 208000001953 Hypotension Diseases 0.000 description 1
- 206010021143 Hypoxia Diseases 0.000 description 1
- 206010061216 Infarction Diseases 0.000 description 1
- 102000004877 Insulin Human genes 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 102000004374 Insulin-like growth factor binding protein 3 Human genes 0.000 description 1
- 108090000965 Insulin-like growth factor binding protein 3 Proteins 0.000 description 1
- 102000004883 Insulin-like growth factor-binding protein 6 Human genes 0.000 description 1
- 108090001014 Insulin-like growth factor-binding protein 6 Proteins 0.000 description 1
- 102000000380 Matrix Metalloproteinase 1 Human genes 0.000 description 1
- 208000009525 Myocarditis Diseases 0.000 description 1
- 206010028851 Necrosis Diseases 0.000 description 1
- 239000000020 Nitrocellulose Substances 0.000 description 1
- CTQNGGLPUBDAKN-UHFFFAOYSA-N O-Xylene Chemical compound CC1=CC=CC=C1C CTQNGGLPUBDAKN-UHFFFAOYSA-N 0.000 description 1
- 101800002327 Osteoinductive factor Proteins 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 241000276498 Pollachius virens Species 0.000 description 1
- 239000004698 Polyethylene Substances 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 102100026651 Pro-adrenomedullin Human genes 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 208000007718 Stable Angina Diseases 0.000 description 1
- 102100029867 Sulfotransferase 2A1 Human genes 0.000 description 1
- 101710097834 Thiol protease Proteins 0.000 description 1
- 108090000190 Thrombin Proteins 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 208000007814 Unstable Angina Diseases 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 101150109862 WNT-5A gene Proteins 0.000 description 1
- 102000043366 Wnt-5a Human genes 0.000 description 1
- 108700020483 Wnt-5a Proteins 0.000 description 1
- 230000001594 aberrant effect Effects 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 150000001298 alcohols Chemical class 0.000 description 1
- 235000012538 ammonium bicarbonate Nutrition 0.000 description 1
- 239000001099 ammonium carbonate Substances 0.000 description 1
- 239000012491 analyte Substances 0.000 description 1
- 210000003484 anatomy Anatomy 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 239000002246 antineoplastic agent Substances 0.000 description 1
- 210000000702 aorta abdominal Anatomy 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 230000004872 arterial blood pressure Effects 0.000 description 1
- 230000003416 augmentation Effects 0.000 description 1
- 210000002469 basement membrane Anatomy 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- OWMVSZAMULFTJU-UHFFFAOYSA-N bis-tris Chemical compound OCCN(CCO)C(CO)(CO)CO OWMVSZAMULFTJU-UHFFFAOYSA-N 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 210000001124 body fluid Anatomy 0.000 description 1
- 239000010839 body fluid Substances 0.000 description 1
- 108010041776 cardiotrophin 1 Proteins 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000021164 cell adhesion Effects 0.000 description 1
- 238000010609 cell counting kit-8 assay Methods 0.000 description 1
- 230000012292 cell migration Effects 0.000 description 1
- 238000001516 cell proliferation assay Methods 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 229940044683 chemotherapy drug Drugs 0.000 description 1
- 210000001612 chondrocyte Anatomy 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 235000008504 concentrate Nutrition 0.000 description 1
- 239000012141 concentrate Substances 0.000 description 1
- 238000012790 confirmation Methods 0.000 description 1
- 210000002808 connective tissue Anatomy 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 210000004351 coronary vessel Anatomy 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 239000012153 distilled water Substances 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 239000002934 diuretic Substances 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 238000000132 electrospray ionisation Methods 0.000 description 1
- 238000002101 electrospray ionisation tandem mass spectrometry Methods 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- 210000002889 endothelial cell Anatomy 0.000 description 1
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 description 1
- 229940116977 epidermal growth factor Drugs 0.000 description 1
- 235000013861 fat-free Nutrition 0.000 description 1
- 230000004761 fibrosis Effects 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 238000002695 general anesthesia Methods 0.000 description 1
- 230000005484 gravity Effects 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 210000000020 growth cone Anatomy 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 208000021822 hypotensive Diseases 0.000 description 1
- 230000001077 hypotensive effect Effects 0.000 description 1
- 230000001146 hypoxic effect Effects 0.000 description 1
- 230000000984 immunochemical effect Effects 0.000 description 1
- 238000003365 immunocytochemistry Methods 0.000 description 1
- 238000011532 immunohistochemical staining Methods 0.000 description 1
- 238000003364 immunohistochemistry Methods 0.000 description 1
- 238000012744 immunostaining Methods 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 230000007574 infarction Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 230000003601 intercostal effect Effects 0.000 description 1
- 201000004332 intermediate coronary syndrome Diseases 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 238000005040 ion trap Methods 0.000 description 1
- 229910052742 iron Inorganic materials 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 210000005240 left ventricle Anatomy 0.000 description 1
- 235000014666 liquid concentrate Nutrition 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 210000005244 lower chamber Anatomy 0.000 description 1
- 210000003712 lysosome Anatomy 0.000 description 1
- 230000001868 lysosomic effect Effects 0.000 description 1
- 108010082117 matrigel Proteins 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 210000000663 muscle cell Anatomy 0.000 description 1
- 208000031225 myocardial ischemia Diseases 0.000 description 1
- 208000002089 myocardial stunning Diseases 0.000 description 1
- 210000000651 myofibroblast Anatomy 0.000 description 1
- 230000001452 natriuretic effect Effects 0.000 description 1
- 210000004498 neuroglial cell Anatomy 0.000 description 1
- 229920001220 nitrocellulos Polymers 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 102000039446 nucleic acids Human genes 0.000 description 1
- 150000007523 nucleic acids Chemical class 0.000 description 1
- 230000011164 ossification Effects 0.000 description 1
- 230000036542 oxidative stress Effects 0.000 description 1
- 238000002751 oxidative stress assay Methods 0.000 description 1
- 210000003540 papillary muscle Anatomy 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 239000008363 phosphate buffer Substances 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 210000003635 pituitary gland Anatomy 0.000 description 1
- 238000007747 plating Methods 0.000 description 1
- 239000004417 polycarbonate Substances 0.000 description 1
- 229920000515 polycarbonate Polymers 0.000 description 1
- 229920000573 polyethylene Polymers 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 239000012460 protein solution Substances 0.000 description 1
- 229940024999 proteolytic enzymes for treatment of wounds and ulcers Drugs 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 239000012146 running buffer Substances 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 210000000329 smooth muscle myocyte Anatomy 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- VSIVTUIKYVGDCX-UHFFFAOYSA-M sodium;4-[2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)tetrazol-2-ium-5-yl]benzene-1,3-disulfonate Chemical compound [Na+].COC1=CC([N+]([O-])=O)=CC=C1[N+]1=NC(C=2C(=CC(=CC=2)S([O-])(=O)=O)S([O-])(=O)=O)=NN1C1=CC=C([N+]([O-])=O)C=C1 VSIVTUIKYVGDCX-UHFFFAOYSA-M 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 230000009870 specific binding Effects 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 238000012453 sprague-dawley rat model Methods 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 238000000528 statistical test Methods 0.000 description 1
- 238000011476 stem cell transplantation Methods 0.000 description 1
- 238000002660 stem cell treatment Methods 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- QTTDXDAWQMDLOF-UHFFFAOYSA-J tetrasodium 3-[[4-[[4-[(6-amino-1-hydroxy-3-sulfonatonaphthalen-2-yl)diazenyl]-6-sulfonatonaphthalen-1-yl]diazenyl]naphthalen-1-yl]diazenyl]naphthalene-1,5-disulfonate Chemical compound [Na+].[Na+].[Na+].[Na+].Nc1ccc2c(O)c(N=Nc3ccc(N=Nc4ccc(N=Nc5cc(c6cccc(c6c5)S([O-])(=O)=O)S([O-])(=O)=O)c5ccccc45)c4ccc(cc34)S([O-])(=O)=O)c(cc2c1)S([O-])(=O)=O QTTDXDAWQMDLOF-UHFFFAOYSA-J 0.000 description 1
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 1
- 230000004797 therapeutic response Effects 0.000 description 1
- 229960004072 thrombin Drugs 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 230000007306 turnover Effects 0.000 description 1
- VBEQCZHXXJYVRD-GACYYNSASA-N uroanthelone Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CS)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(C)C)[C@@H](C)O)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CS)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC(N)=O)C(C)C)[C@@H](C)CC)C1=CC=C(O)C=C1 VBEQCZHXXJYVRD-GACYYNSASA-N 0.000 description 1
- 210000004509 vascular smooth muscle cell Anatomy 0.000 description 1
- 229940124549 vasodilator Drugs 0.000 description 1
- 239000003071 vasodilator agent Substances 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 238000012800 visualization Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- 238000011706 wistar kyoto rat Methods 0.000 description 1
Images
Classifications
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
- G01N33/6893—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
- G01N33/6863—Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
- G01N33/6869—Interleukin
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
- G01N33/6887—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids from muscle, cartilage or connective tissue
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/74—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/92—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving lipids, e.g. cholesterol, lipoproteins, or their receptors
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/32—Cardiovascular disorders
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/52—Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis
Definitions
- the invention relates to secreted proteins from cardiac stem cells (cardiospheres and cardiosphere-derived cells) or myocytes for diagnostic and/or therapeutic use.
- Stem cells hold the promise to revolutionize future reparative medicine through the development of stem cell-based therapies.
- Transplanting stem cells (either embryonic or adult derived) into damaged myocardium is emerging as a novel means for acute repair and as an alternative to organ transplantation or ventricular assist devices in the treatment of end-stage heart failure.
- the crux for the success of this therapy will lie in being able to manipulate proliferating ES cells to differentiate specifically into cardiac muscle upon demand, and to predict which patients will benefit from such intervention.
- An alternative approach is to harness the endogenous stem cells and remaining viable myocytes to regenerate the needed myocardium. This could be accomplished if a contusive environment at the site of injury could be therapeutically produced through the use (or augmentation) of soluble proteins and or paracine factors.
- adult stem cells in mature tissues and organs such as bone marrow, brain, skin or liver has been demonstrated. Although adult stem cells are mostly considered to differentiate into cell types of tissue of their origin, they have also been found to form specialized cell types of other tissues. This transdifferentiation was reported for bone marrow stem cells, which can differentiate into e.g. cardiac cells (myocytes) and can induce cardiac regeneration. Although the heart has for considerable time been considered to be a terminally differentiated organ with cells not able to self-regenerate after injury or damage, the discovery of stem cells residing in heart has opened the possibility of their use for autologous heart cell repair.
- cardiac cells myocytes
- CSCs adult cardiac stem cells
- CSps cardiospheres
- CDCs cardiosphere-derived cells
- the present inventors have found that secreted proteins from cardiac stem cells or myocytes can be useful therapeutically in cardiac regenerative treatment by addition or by inducing enhanced cellular expression of one or more of proteins in isolated or cultured stem cells or myocytes prior to, at the time or following administration of therapy or they can be directly administered to the patient for enhancement of endogenous innate cardiac regeneration. Furthermore, detection of one or more of these proteins (fragment, isoform or modified form) and their binding partner(s) for diagnostic/prognostic assessment of patient viability and responsiveness to treatment and/or assessment of the regenerative potential of stem cells, directly. Thus, detection of one or more of these proteins and/or their binding partners allows better assessment and clinical intervention including regenerative therapy.
- Interleukin-1 receptor ST2
- its ligand IL33 as well as other paracrine factors including brain acid soluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M, insulin-like growth factor binding protein 2, and minecan, (for additional proteins, see Table 1) may increase the ability of stem cells (including cardiac or cardiac derived stem cells) to hone, survive or differentiation in the myocardial infracted zone as well as increase survival of the injured myocardium, fibroblasts or vascular tissue.
- stem cells including cardiac or cardiac derived stem cells
- cardiac myocytes healthy or injured
- ANP atrial natriuretic peptide
- apolipoprotein E matrix metallopeptidase 2
- metalloproteinase inhibitor 1 adrenomedullin
- connective tissue growth factor a similar manner.
- these proteins could affect the viability of the myocytes or their ability to transgenerate or alter other cell types (eg. vascular smooth muscle cells or endothelial cells).
- paracrine factors from either or both stem cells and/or myocytes maybe used as a diagnostic monitor or prognostic indicator of i) their ability to be involved in regeneration or ii) viability of the cells.
- paracrine factor could enhance therapeutically the viability of the stem cells and surrounding cardiac or vascular tissue and or ability to differentiate.
- measurement of the paracrine factor in setting of heart failure, AMI or angina may predict outcome as a reflection of their ability for endogenous or therapeutic application of stem cell therapy.
- this is a method to screen for paracrine factors.
- Diagnostic and prognostic markers can be used to measure cardiac stem cell and myocyte secreted factors and their binding partners for the in vivo assessment of the regeneration potential of individual patients.
- ST2 its ligand IL33 and/or including brain acid soluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M, insulin-like growth factor binding protein 2 and minecan, atrial natriuretic peptide (ANP), apolipoprotein E, matrix metallopeptidase 2, metalloproteinase inhibitor 1, adrenomedullin and/or connective tissue growth factor (for additional proteins, see Table 1) can be used alone or in combination to provide a measurement of viability or efficacy of stem cell treatment or the long term regeneration potential of the injured myocardium (including but not exclusive to myocyte, fibroblast, endothelial, smooth muscle cells) in patients.
- Diagnostic/prognostic markers can be used to measure cardiac stem cell and myocyte secreted paracrine factors and/or their binding partners for the assessment of the potency, viability and/or efficacy of the exogenous stem cells prior to therapeutic application.
- ST2 its ligand IL33 and/or including brain acid soluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M insulin-like growth factor binding protein 2 and minecan, atrial natriuretic peptide (ANP), apolipoprotein E, matrix metallopeptidase 2, metalloproteinase inhibitor 1, adrenomedullin and/or connective tissue growth factor (for additional proteins, see Table 1), can be used alone or in combination to assess test viability, efficacy and suitability of stem cells or other cell types for transplantation and regeneration therapy.
- ADP atrial natriuretic peptide
- apolipoprotein E matrix metallopeptidase 2
- Diagnostic/prognostic markers can be used alone or in combination to assess clinical outcome following acute myocardial infarction or heart failure as an indicator of the patients inherent ability to repair/regenerate injured myocardium by measuring one or more of the factors including ST2, IL33, brain acid soluble protein 1, cathepsin
- Therapeutic target or protein application of paracrine factor(s) and/or their binding partners can be used through endogenous administration or the use of a method to enhancement in vivo cellular release in individual patents in order to improve cardiac heart regeneration.
- ST2 its ligand IL33 and/or including brain acid soluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M, insulin-like growth factor binding protein 2 and minecan, atrial natriuretic peptide (ANP), apolipoprotein E, matrix metallopeptidase 2, metalloproteinase inhibitor 1, adrenomedullin and/or connective tissue growth factor (or their binding partners) could be increased alone or in combination, in the myocardium (at or near site of injury) to enhance the in vivo cardiac and vascular regeneration and stem cell action.
- Protein(s) could be administered at the time of heart injury (e.g. at time of myocardial infarction or during development of heart failure),
- Paracrine factor(s) and/or their binding partners can be used in stem cell culture to be used in regeneration therapy.
- ST2 it ligand IL33 and/or including (but not exclusive) brain acid soluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M, insulin-like growth factor binding protein 2 and minecan, atrial natriuretic peptide (ANP), apolipoprotein E, matrix metallopeptidase 2, metalloproteinase inhibitor 1, adrenomedullin and connective tissue growth factor (see secreted factors Table 1), alone or in combination, can be added to the media of the cell culture to enhance preparation of stem cells prior to, at the time or following administration to patients.
- ABP atrial natriuretic peptide
- apolipoprotein E matrix metallopeptidase 2
- metalloproteinase inhibitor 1 adrenomedullin and connective tissue growth factor
- Personalized medicine applications can be used in stem cell therapy, through the monitoring of paracrine proteins and their binding protein will allow clinician intervene, choose effective dose and time course of stem cells and proteins therapy during the course of stem cell therapy.
- All proteins listed in the Tables herein maybe therapeutically important for cell survival, proliferation or deformation in the injured area of the heart. As well, they may be biomarkers for assessment of cell viability and effectiveness, responsiveness of the patent to stem cell therapy and long term prognosis of development of heart failure or survival. Equivalent proteins from humans or other species including isoforms, splice varients and polymorphorisms/SNPs are also expected to be effective. These are known to and can be tested by those of skill in the art without undue experimentation. Proteins considered to be particularly useful are listed in Table 2.
- Another embodiment of the invention is a method to screen for paracrine factors from cells including stem cells.
- the method uses reversed phase HPLC to separate the intact proteins prior to MS.
- FIG. 1 Cardiac cell analysis strategy
- FIG. 2 Representative chromatograms and reproducibility obtained by RPLC analysis for CSps, CDCs and NRVms with 9 final fractions used for ESI-MS/MS protein identification after trypsin digestion. Eight chromatograms are overlaid (100 ⁇ g/run) to demonstrate the reproducibility. Fraction containing albumin was not analyzed.
- FIGS. 3A-3C Influence of proteins IL33, BASP1 and CTCF at different concentrations on proliferation of CDCs.
- BASP1 and CTCF increases proliferation of CDCs.
- Soluble proteins secreted from stem cells of any cell type including the myocytes near or at the site of injury can serve as interactive signals to the local environment, influencing survival, differentiation and stem cell engraftment as well as affects on the injured myocardium.
- any combination of the proteins, protein isoforms, protein polymorphorism, peptide fragment(s) protein with post-translational modifications thereof examined herein, or others can be assayed with a method of the invention. For example, one can first measure the amount of expression of apolipoprotein E precursor compared to a baseline value, to determine if a significantly elevated or decreased amount is present in a patient sample. One can then further measure the amount of expression of adrenomedullin and/or Dkk3 protein compared to the baseline value. A significant increase (e.g. at least a statistically significant increase) in the amount of expression of one or more proteins, e.g. as listed in Table 2, compared to the baseline value indicates a greater likelihood that the patient is in need of, or would benefit from stem cell therapy.
- a significant increase e.g. at least a statistically significant increase
- peptide fragment any fragment of a protein of interest of at least 10 amino acid residues, preferably at least 15, 20, 15, or 30 amino acid residues up to fragments that may be only a few (e.g. 1, 2, 3, 4, 5, 10, 15, etc.) residues shorter than the full length protein.
- protein, polypeptide and peptide are used interchangeably herein.
- amino acid sequences of the proteins of the invention are well-known and can be determined routinely, as well as downloaded from various known databases using the provided GenBank Accession numbers. See, e.g., the world wide web site, ncbi.nlm.nih.gov.
- the amount of expression of a protein of the invention can be determined by measuring the amount of the protein, or by measuring the amount of mRNA encoding the protein.
- the amount of a protein can be determined using any routine method known in the art, e.g. MS, an antibody, etc.
- the method can encompass binding the protein to an antibody which is specific for it, under conditions that are effective for specifically binding the protein to the antibody.
- an antibody or any sensor may be contacted with a histological preparation, and the amount of protein is determined by immunohistochemical staining.
- the amount of an mRNA can be determined using a nucleic acid probe for the mRNA.
- the method can encompass hybridizing the mRNA to a nucleic acid probe which is specific for it, under conditions that are effective for specifically hybridizing the mRNA and the probe.
- Some methods involve the use of antibodies, any binding ligand or mass spectrometry tagged peptide specific for a protein of interest.
- Antibodies suitable for use in such assays are commercially available, or can be prepared routinely. Methods for preparing and using antibodies in assays for proteins of interest are conventional, and are described, e.g., in Green et al., Production of Polyclonal Antisera, in Immunochemical Protocols (Manson, ed.), (Humana Press 1992); Coligan et al., in Current Protocols in Immunology , Sec.
- antibodies can be used, including, e.g., polyclonal, monoclonal (mAbs), recombinant, humanized or partially humanized, single chain, Fab, and fragments thereof.
- the antibodies can be of any isotype, e.g., IgM, various IgG isotypes such as IgG 1 ′ IgG 2a , etc., and they can be from any animal species that produces antibodies, including goat, rabbit, mouse, chicken or the like.
- the term, an antibody “specific for” a protein means that the antibody recognizes a defined sequence of amino acids, or epitope, in the protein, and binds selectively to the protein and not generally to proteins unintended for binding to the antibody. The parameters required to achieve specific binding can be determined routinely, using conventional methods in the art.
- the baseline value for such measurements can be an average or mean from a population of normal subjects, i.e. those not suffering from heart disease, or, e.g. may be earlier measurements taken from the same patient, for example when progress of treatment is being monitored. Suitable baseline values can be determined by those of skill in the art without undue experimentation.
- a “baseline value” refers to the expression, as determined by the levels (amounts) of mRNA and/or protein, in normal tissue (e.g., the same type of tissue as the tested tissue, such as normal cardiac tissue or normal serum), from normal subjects that do not have heart disease. If desired, a pool of the same tissues from normal subjects can be used.
- Such baseline values may be available in a database compiled from the values and/or may be determined based on published data or on retrospective studies of patients' tissues, and other information as would be apparent to a person of ordinary skill implementing a method of the invention. Suitable baseline values may be selected using statistical tools that provide an appropriate confidence interval so that measured levels that fall outside the standard value can be accepted as being aberrant from a diagnostic perspective, and predictive of success in treatment, diagnosis or prognosis.
- a “significant” increase in the amount of a protein or mRNA can refer to a difference which is reproducible or statistically significant, as determined using statistical methods that are appropriate and well-known in the art, generally with a probability value of less than five percent chance of the change being due to random variation. Some such statistical tests are described in the Examples herein.
- a significant increase in the amount of mRNA or protein compared to a baseline value can be at least about 2.5-fold (e.g., at least about 5-fold, 10-fold, 20-fold, 25-fold, or more) higher.
- Methods for obtaining samples and preparing them for analysis are conventional and well-known in the art.
- a “subject” or “patient”, as used herein, includes any animal that has, or may have, heart disease, including experimentally induced heart disease, for example in laboratory animals (such as mouse, rat, rabbit, or guinea pig), farm animals, and domestic animals or pets (such as a cat or dog).
- laboratory animals such as mouse, rat, rabbit, or guinea pig
- domestic animals or pets such as a cat or dog
- Non-human primates and human patients are included.
- Heart failure can be induced following heart attack or other injuries to the heart (pulmonary hypertension), viral infections, genetic disorders and many other affectors that weaken the heart muscle or vasculature.
- AMI after AMI when heart muscle has been injured or destroyed, regeneration is required to replace the lost muscle cells and other cells required for functioning heart.
- sample includes any biological sample, for example whole blood, serum, cardiac tissue, etc. obtained from a subject or patient.
- a sample also includes stem cell cultures, e.g. for therapeutic use in such patients.
- stem cell therapy is meant the administration of cardiac myocytes or equivalent cells as described herein to a cardiac patient or to an animal with experimentally induced heart disease.
- the paracrine factor(s) could alter the honing, survival, proliferation and differentiation of all stem cells and progenitor cells found in the body including EPC, mesochymal, and hemopoetic stem cell.
- Administration of cells can be by any means known in the art, for example, by local injection, infusion,. Cells would be pretreated at optimal concentration most likely between pg/ml-1 ⁇ g/ml for a 10,000 cells (in 1 ml).
- heart disease is meant by stable and unstable angina, myocardial ischemia, myocardial stunning, acute myocardial infarction, (minor necrotic cell death, heart failure induced by myocardial infraction, genetic disease, pulmonary hypertension and other injury to the myocardium which makes the heart work harder.
- treated is meant that an effective amount of a chemotherapeutic drug or other anti-cancer procedure is administered to the subject.
- An “effective” treatment refers to a treatment that elicits a detectable response (e.g. a therapeutic response) in the subject.
- an effective amount is meant any amount that will elicit a detectable clinical response.
- a significant increase in the amount of expression of a secreted protein of the invention compared to the baseline value indicates that a subject is likely to be responsive to stem cell therapy.
- a subject that is “likely” to be responsive has greater than, e.g., at least about: a 25%, more likely 50%, 75%, chance to show an improvement in clinical symptoms.
- secreted proteome (“secretome”)
- CSps and CDC) proteins secreted into media conditioning adult cardiac stem cells
- FIG. 1 proteins secreted from neonatal rat ventricular myocytes after analysis by reversed phase liquid chromatography and identification by mass spectrometry.
- the cells were obtained under optimized conditions that minimize cell lysis, allowing us to distinguish between secreted proteins and those proteins that would be liberated upon cell death due to necrosis and/or apoptosis.
- CSps cardiac tissue specimens from septum or left ventricule of Wistar Kyoto rats, 12 to 16 weeks old, were cut into small pieces, washed by PBS, enzymatically digested and grown in primary cultures as explants on fibronectin (from human plasma, BD Biosciences) coated Petri dishes in IMDM medium (Iscove's Modified Dulbecco's Medium, Invitrogen) supplemented with 20% fetal bovine serum (FBS), 1% penicillin-streptomycin, 1% L-glutamine and 0.1 mmol/L 2-mercaptoethanol at 37° C.
- FBS fetal bovine serum
- penicillin-streptomycin 1% L-glutamine
- 2-mercaptoethanol 0.1 mmol/L 2-mercaptoethanol
- CSps cardiac multicellular clusters
- CDCs cardiosphere-derived cells
- NRVMs Neonatal rat ventricular myocytes
- NRVMs rat ventricular myocytes
- CSps, CDCs and NRVMs were further conditioned in media containing 1%
- the liquid concentrates with secreted proteins and concentrates of media alone were mixed with solvent to final concentration of 20% (v/v) acetonitrile (ACN), 1% (v/v) trifluoroacetic acid (TFA), pH 2.3, vortexed and spun down at 18 ⁇ 1000 g for 30 min at 4° C. Samples of intact proteins were separated by RPLC in order to reduce the complexity of protein mixtures prior to mass spectrometry protein identification.
- ACN acetonitrile
- TFA trifluoroacetic acid
- FIG. 2 shows representative chromatographs of multiple runs as well as the fractions pooled for MS analysis.
- the secreted proteins are often at low concentration within the culture media, ii) the media is a large source of contamination (including albumin and iii) the need to distinguish between secreted proteins and those artificially liberated with cell death. This will be assessed this by both Annexin V labeling and increased detection of intercellular proteins (as outlined elsewhereherein).
- Table 1 there are several protein highlighted which may arise from lysed cells due to their known intracellular location and the low level quantity present in the media. The remaining proteins are known to be secreted from cells or are membrane or membrane associated proteins.
- Proteins that may play a role (alone or in combination) found exclusively in the myocyte media are apolipoprotien E, matrix metallopeptidase 2 (and although its inhibitor (metalloprotease 2 inhibitor) is not altered the ratio of these two proteins maybe critical), and metalloproteinase inhibitor 1 (and although the corresponding matrix metallopeptidase 1 is not observed, the ratio of the two proteins maybe important).
- all metalloproteinase and their inhibitors may play a role in both stem cell and myocyte function and survival in the infracted or injured area of the heart.
- Additional proteins found exclusively in the cardiac stem cell media are brain acid soluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M, and the insulin-like growth factor binding protein 2 (although not observed insulin growth factor (IGF) and other analytes that bind this protein or to the IGF receptor maybe important).
- IGF insulin growth factor
- Brain soluble protein 1 is also named BASPI protein, neuronal axonal membrane protein NAP22 and the 22 kDa neuronal tissue-enriched acidic protein. This membrane protein is involved in cell projection and growth cones and was originally thought to be present exclusively in the brain.
- Cathepsin B also named APP secretase (APPS) contains a heavy and light chain within its amino acid sequence which after processing forms a dimmer with the heavy and light chains crosslinked by a disulfide bond. This protein in its active form is a thiol protease involved in degradation and turnover of proteins and is normally associated with the lysosome.
- Cystatin E/M is also called Cystatin M/E or cystatin 6 is a protease inhibitor.
- Insulin like growth factor binding protein 2 is also called IGFB-2, IBP-2 and IGF-binding protein 2 is secrteted and prolongs the half life of the IGF and has been shown to alter growth promoting affects on IGF on cell cultures.
- IGF and the insulin like growth factor binding protein 2 have been implicated in changes to may cells types including fibroblasts, lung epithelial cells, and glial cells but to our knowledge not cardiac derived stem cells or the cardiac myocyte.
- adrenomedullin and connective tissue growth factor which are present in the media of NRMV exclusively and minecan which is found exclusively in the media of cardiac stem cells.
- Minecan also called osteoglycin or osteoinductive factor is known to induce bone formation in conjunction with TFG beta 1 or TGF beta 2, and have been implicated in regulation of collagen fibrillogeneis of the cardiac stem cells or act also on cells in the surrounding regions of the injured heart.
- Adrenomedullin or AM is processed active domain of larger protein called ADML or ADM which also contains the proadrenomedullin N-20 terminal peptide (PAMP).
- AM and PAMP are potent hypotensive and vasodilator factors that have affects throughout the body including the kidney, brain and pituitary gland. It has been found in the ventricle. This protein has been implicated in differentiation of several stem cells but not cardiac derived cells or injured myocytes. Connective tissue growth factor is also termed hypertrophic chondrocyte-specific protein 24. This protein is a connective tissue mitoattractant that has been previously reported to be secreted by vascular endothelial cells but, to our knowledge not cardiac myocytes under normal physiological conditions but has been shown to increase in hearts with ongoing myocarditis.
- AM has been shown to induce proliferation and differentiation of chondrocytes, improve cell adhesion for fibroblasts, myofibroblasts, endothelial and epithelial cells and stimulate fibroblast growth factor-induced DNA synthesis. Therefore, fibroblast growth factor alone or its ratio with respect to connective tissue growth factor may be also important.
- Soluble proteins secreted from stem cells serve as interactive signals to the local environment, influencing survival, differentiation and engraftment.
- secreted proteome proteins found in the media conditioned by adult cardiac stem cells (CSCs) or neonatal rat ventricular myocytes (NRVMs) were obtained under optimized conditions that minimize cell lysis, allowing distinction between secreted proteins and those artificially liberated with cell death.
- Conditioned media 1% serum
- 90-110 proteins were identified exclusively in media from CSCs or NRVMs, with the majority (>85%) comprising known membrane, extracellular or secreted proteins. Of these, >2 fold more proteins were detected in CSCs than NRVMs (60 vs. 27, respectively). Functional protein classes were conserved between the cell types, although proteins and/or their isoforms could differ. Of interest, 10 different collagen isoforms were observed with 5 being cell-specific. Cell-specificity was also reflected with collagen regulation as TIMP2 and MMP2 were observed in NRVMs while TIMP 1 was uniquely present in CSCs. The signaling molecules, insulin-like growth factor binding protein 6 and 3, were present in CSCs, but only isoform 7 in NRVMs.
- the cardiac specific natriuretic hormone ANP was only detected in NRVMs and not CSCs, while the soluble interleukin-1 receptor family member ST2 was exclusive to CSCs.
- ST2 is known to increase in myocytes with mechanical stress and heart failure where it has been linked to neurohormonal activation.
- CSC and NRVM-specific secretomes display unique functionality including differential secretion of two cardiovascular hormones.
- IMDM medium Iscove's Modified Dulbecco's Medium, Invitrogen
- FBS fetal bovine serum
- Day 5 to 10 the procedure of day 4 was repeated on each following plate row. Influence of proteins interleukin-33 (IL 33, ALX-522-098-0010, Apotech Corporation, USA), brain abundant membrane attached signal protein 1 (BASP1,H00010409-P01, Novus Biological, USA) and connective tissue growth factor (CTGF, CRC604B, Cell Sciences, USA) at different concentration on CDCs proliferation is shown in FIG. 3 .
- IL 33 interleukin-33
- ALX-522-098-0010 brain abundant membrane attached signal protein 1
- BASP1,H00010409-P01 Novus Biological, USA
- CTGF connective tissue growth factor
- Modified Boyden chambers were equipped with 8 ⁇ m pore-size polycarbonate filters (Neuroprobe, Gaithersburg, Md.) coated with Matrigel (BD Bioscience, Palo Alto, Calif.). Cells (stem cells or myocytes) were either untreated or pretreated with appropriate protein for 24 hours. 220 ⁇ l of migration medium (DMEM—Dulbecco's Modified Eagle Medium with 0.1% BSA) was added to lower chamber with or without platelet-derived growth factor (PDGF) for chemattract and random migration assay, respectively. Cells (10 6 /ml) were placed in upper chamber in 200 ⁇ l of migration medium, performed in triplicate. The assay was stopped after 4 hours at ° C. Cells that crossed the basement membrane and migrated to lower side of the filter were fixed and stained using HEMA3 system (Curtin, Matheson Scientific Inc., Houston, Tex.). Four random fields were counted at 400 ⁇ magnification for each filter.
- DMEM Dynamic Eagle Medium
- PDGF
- NRVMs tetramethylrhodamine ethyl ester
- TMRE tetramethylrhodamine ethyl ester
- IHC is method for identification of specific tissue components by means of specific antigen/antibody (or other sensors) reaction tagged with a visible label (or other types of chemical labels). This method makes possible to visualize the distribution and localization of specific cellular components within a cell or tissue.
- the tissue for staining is fixed. Fixative procedure is optimized based on tissue and antigen/sensor used and various fixatives can be used. Usually, the tissue is formalin fixed and paraffin embedded. Prior to staining, tissue slides are deparaffinized (e.g. in xylol) and rehydrated in graded alcohol series and can be further pretreated with proteolytic enzymes, washed in distilled water and heated in microwave oven for epitope retrieval. Then nonspecific sites are blocked with serum or blocker protein, incubated with primary antibody (1:100-1:1,000), washed and incubated with secondary antibody-enzyme conjugate (1:2,000-1:5,000), washed and incubated with substrate and finally, the stained tissue is visualized.
- tissue slides are deparaffinized (e.g. in xylol) and rehydrated in graded alcohol series and can be further pretreated with proteolytic enzymes, washed in distilled water and heated in microwave oven for epitope retrieval. Then nonspecific sites are blocked with
- the CSps-, CDC-, NRVM-media with secreted proteins/lysates or adult myocyte lysates were subjected to 1D gel electrophoresis (NuPage BisTris gels of various concentrations, 200 V, 35 to 55 minutes based on running buffer and gel concentration used), separated proteins were transferred into nitrocellulose membrane (NuPage transfer buffer, 100 V, 1 hour). Proteins on the membrane were visualized by dye Direct Blue 71, briefly destained by 40% ethanol/10% acetic acid, washed by lx Tris buffered saline solution (TBS; 20 mM Tris, 500 mM NaCl, pH 7.5) and transferred into blocking solution (5% of non fat dry milk in 1 ⁇ TBS).
- TBS Tris buffered saline solution
- the membrane was blocked overnight at 4° C., then washed by TTBS (0.1% Tween-20 in TBS) and incubated with appropriate primary antibody (1:100-1:1,000) for 4.5 hours. After that, the membrane was washed in TTBS and incubated with secondary antibody-enzyme conjugate (1:2,000-1:5,000), washed in TTBS and incubated with ImmunostarAP Substrate Pack (BioRad laboratories, Calif., USA) for 5 minutes. The membrane was placed in cassette with film and developed.
- Stem cells treated and non-treated are labeled with tracking agent such as iron as described by Terrovitis et al [4] or unique protein, chromosome, gene or chemical compound(s).
- tracking agent such as iron as described by Terrovitis et al [4] or unique protein, chromosome, gene or chemical compound(s).
- the ferumoxide-labeled CDCs (with and without treatment either exogenous protein or transgenically manipulated to produce proteins, see Smith et al. [5] for examples) is injected intramyocardially into normal (immunocompetent) rats or mice prior to or after induction of experimental of myocardial infarction. Rats may also undergo left thoracotomy in the fourth or fifth intercostals space under general anesthesia. The heart will be exposed and the cells injected directly into the myocardium at a single or multiple sites.
- the myocardial infarction can be produced by a number of methods including a permanent ligation of the left anterior descending coronary artery using a suture immediately before or after cell injection. Subsequently, the chest is closed and the animals are allowed to recover. Magnetic resonance imaging (MRI) images are obtained on a number of days after surgery such as day 2 and 21 or longer. After completion of this follow-up period, the rats will be sacrificed and the hearts subjected to histology.
- MRI Magnetic resonance imaging
- MRI For MRI, animals were anesthetized and then placed prone, head first in the magnet. ECG-gated cine images of the heart are obtained. At least 3 consecutive short-axis slices will be acquired to completely cover the area of cell injection. Signal intensity will be measured in the myocardium (remote areas and areas of cell injection); noise is measured by creating regions of interest in the lungs. Contrast-to-noise ratios (signal intensity in the remote myocardium minus signal intensity in the areas of the cell injection divided by the SD of noise) will be calculated for each slice in which the signal void is visualized.
- percent signal area will be calculated as the area of visually determined signal void (manually defined region of interest containing area obviously darker than the surrounding myocardium) divided by the total left ventricular area in the same slice. Histological analysis of cell engraftment will be performed for number of proteins and for general pathological stains.
- Echocardiography will be performed in conscious animals before and after myocardial infarction.
- the anterior chest area is shaved and 2D images and M-mode tracings are recorded from the parasternal short-axis view at the level of papillary muscles.
- abdominal aorta could be cannulated with a polyethylene catheter and the heart arrested in diastole (eg. injection of CdCl 2 ), the thorax was opened, perfusion with phosphate buffer started.
- An aortic catheter connected to a pressure reservoir is used to adjust perfusion pressure to mean arterial blood pressure while simultaneously, the LV chamber is filled with formalin.
- the coronary vasculature is perfused with fixative.
- the heart is excised, and the weights and major axis from the base to the apex of the heart is measured.
- the volume of the myocardium is computed (4) by dividing the weight by the specific gravity of muscle tissue.
- paraffin-embedding of tissue could be done. In this case tissue slices would be stained with hematoxylin and eosin or used later for immunohistochemistry.
Landscapes
- Life Sciences & Earth Sciences (AREA)
- Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Biomedical Technology (AREA)
- Immunology (AREA)
- Chemical & Material Sciences (AREA)
- Urology & Nephrology (AREA)
- Hematology (AREA)
- Cell Biology (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Microbiology (AREA)
- Biotechnology (AREA)
- Food Science & Technology (AREA)
- Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Biochemistry (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Endocrinology (AREA)
- Biophysics (AREA)
- Heart & Thoracic Surgery (AREA)
- Public Health (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Veterinary Medicine (AREA)
- Cardiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Investigating Or Analysing Biological Materials (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
- Peptides Or Proteins (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
The invention relates to secreted proteins from cardiac stem cells (cardiospheres and cardiosphere-derived cells) or myocytes for diagnostic and/or therapeutic use
Description
- The research leading to this invention was supported in part by contract # NOI-HV-28180, U.S. National Heart, Lung and Blood Institute. The U.S. Government has certain rights in the invention.
- The invention relates to secreted proteins from cardiac stem cells (cardiospheres and cardiosphere-derived cells) or myocytes for diagnostic and/or therapeutic use.
- Stem cells hold the promise to revolutionize future reparative medicine through the development of stem cell-based therapies. Transplanting stem cells (either embryonic or adult derived) into damaged myocardium is emerging as a novel means for acute repair and as an alternative to organ transplantation or ventricular assist devices in the treatment of end-stage heart failure. The crux for the success of this therapy will lie in being able to manipulate proliferating ES cells to differentiate specifically into cardiac muscle upon demand, and to predict which patients will benefit from such intervention. An alternative approach is to harness the endogenous stem cells and remaining viable myocytes to regenerate the needed myocardium. This could be accomplished if a contusive environment at the site of injury could be therapeutically produced through the use (or augmentation) of soluble proteins and or paracine factors.
- Existence of adult stem cells in mature tissues and organs such as bone marrow, brain, skin or liver has been demonstrated. Although adult stem cells are mostly considered to differentiate into cell types of tissue of their origin, they have also been found to form specialized cell types of other tissues. This transdifferentiation was reported for bone marrow stem cells, which can differentiate into e.g. cardiac cells (myocytes) and can induce cardiac regeneration. Although the heart has for considerable time been considered to be a terminally differentiated organ with cells not able to self-regenerate after injury or damage, the discovery of stem cells residing in heart has opened the possibility of their use for autologous heart cell repair. First reports about adult cardiac stem cells (CSCs) appeared in 2003 and from that time, the attempts to find the procedures for their isolation and expansion into sufficient quantity for therapeutic purposes have been made. Recently, successful methods for isolation and expansion of adult cardiac stem cells from heart biopsy specimens were reported. Endomyocardial biopsy specimen grown in primary culture developed spherical multicellular clusters, cardiospheres (CSps), which can be further plated yielding in cardiosphere-derived cells (CDCs)—expansion step to obtain reasonable numbers of cells for transplantation from small specimens in a timely manner. Cardiospheres and CDCs exhibit properties of stem cells, expressed certain markers characteristic for stem cells and promoted cardiac regeneration and function in a mouse infarct model.
- The present inventors have found that secreted proteins from cardiac stem cells or myocytes can be useful therapeutically in cardiac regenerative treatment by addition or by inducing enhanced cellular expression of one or more of proteins in isolated or cultured stem cells or myocytes prior to, at the time or following administration of therapy or they can be directly administered to the patient for enhancement of endogenous innate cardiac regeneration. Furthermore, detection of one or more of these proteins (fragment, isoform or modified form) and their binding partner(s) for diagnostic/prognostic assessment of patient viability and responsiveness to treatment and/or assessment of the regenerative potential of stem cells, directly. Thus, detection of one or more of these proteins and/or their binding partners allows better assessment and clinical intervention including regenerative therapy.
- There is a need to be able to predict patients that will respond to stem cell therapy and the ability to manipulate the efficacy of stem cell therapy in heart, the monitoring and or application (exogenous delivery or endogenous enhance production) of paracrine factors from stem cells or myocytes maybe able to met these needs.
-
-
- 1. Unique stem cell-secreted paracrine factors found exclusively in the cardiac stem cell media include
interleukin 1 receptor like protein (also named ST2) and brain acidsoluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M, insulin-like growthfactor binding protein 2 and minecan (for additional proteins, see Table 1). IL-33, the binding partner (analyte) of ST2 is also a target. All proteins which interact or bind to these proteins are also potential modulating proteins. - 2. Unique myocyte secreted paracrine factors found exclusively in the myocyte media include atrial natriuretic peptide (ANP), apolipoprotien E,
matrix metallopeptidase 2,metalloproteinase inhibitor 1, adrenomedullin and connective tissue growth factor (for additional proteins, see Table 1). Other proteins which interact or bind to these proteins are also potential modulating proteins. - 3. Use of the above-mentioned factors, and related factors, to predict susceptibility to cardiovascular disease and or efficacy of treatment of cardiac (or other types) stem cell therapy or endogenous enhancement of stem cell therapy including response of either exogenously applied stem cells or the endogenous stem cells, myocyte or other cell types present in the Injured zone (fibroblasts).
- 4. The use of the above-mentioned secreted factors and/or their binding protein(s) alone or in combination to enhance honing, survival, engraftment and efficacy of stem cell therapy or cardiac regeneration therapy.
- 5. A method monitoring one or more factors as a means of protein diagnostic(s)/prognostic(s), for example to test efficacy of stem cells prior to therapy in blood of patients or cell culture or media used to produce/harvest cells for regeneration therapy.
- 6. A method monitoring one or more factors as a means of protein diagnostic(s)/prognostic(s) to assess patents inherent long term outcome of patent without treatment as well as to determine the potential responsiveness of patent to stem cell therapy.
- 7. A method for screening for cell populations isolated from patients based on the cells ablity to secreted proteins or presence of their complementary cellular receptors to determine which cell population will be more suitable or more effective following injection into the patent.
- 8. A method of monitoring paracrine proteins and their binding proteins that allows clinician intervention during the course of stem cell therapy and the application of personalized stem cell therapy.
- 1. Unique stem cell-secreted paracrine factors found exclusively in the cardiac stem cell media include
- Interleukin-1 receptor (ST2), and its ligand IL33 as well as other paracrine factors including brain acid
soluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M, insulin-like growthfactor binding protein 2, and minecan, (for additional proteins, see Table 1) may increase the ability of stem cells (including cardiac or cardiac derived stem cells) to hone, survive or differentiation in the myocardial infracted zone as well as increase survival of the injured myocardium, fibroblasts or vascular tissue. In addition, the paracrine factors secreted from cardiac myocytes (healthy or injured) such as ANP (atrial natriuretic peptide), apolipoprotein E,matrix metallopeptidase 2,metalloproteinase inhibitor 1, adrenomedullin and connective tissue growth factor maybe work in a similar manner. Furthermore these proteins could affect the viability of the myocytes or their ability to transgenerate or alter other cell types (eg. vascular smooth muscle cells or endothelial cells). Thus, paracrine factors from either or both stem cells and/or myocytes maybe used as a diagnostic monitor or prognostic indicator of i) their ability to be involved in regeneration or ii) viability of the cells. As well, the addition of one or more paracrine factor could enhance therapeutically the viability of the stem cells and surrounding cardiac or vascular tissue and or ability to differentiate. Finally, measurement of the paracrine factor in setting of heart failure, AMI or angina may predict outcome as a reflection of their ability for endogenous or therapeutic application of stem cell therapy. In addition, this is a method to screen for paracrine factors. - Diagnostic and prognostic markers can be used to measure cardiac stem cell and myocyte secreted factors and their binding partners for the in vivo assessment of the regeneration potential of individual patients. Specifically, ST2, its ligand IL33 and/or including brain acid
soluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M, insulin-like growthfactor binding protein 2 and minecan, atrial natriuretic peptide (ANP), apolipoprotein E,matrix metallopeptidase 2,metalloproteinase inhibitor 1, adrenomedullin and/or connective tissue growth factor (for additional proteins, see Table 1) can be used alone or in combination to provide a measurement of viability or efficacy of stem cell treatment or the long term regeneration potential of the injured myocardium (including but not exclusive to myocyte, fibroblast, endothelial, smooth muscle cells) in patients. - Diagnostic/prognostic markers can be used to measure cardiac stem cell and myocyte secreted paracrine factors and/or their binding partners for the assessment of the potency, viability and/or efficacy of the exogenous stem cells prior to therapeutic application. Specifically, ST2, its ligand IL33 and/or including brain acid
soluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M insulin-like growthfactor binding protein 2 and minecan, atrial natriuretic peptide (ANP), apolipoprotein E,matrix metallopeptidase 2,metalloproteinase inhibitor 1, adrenomedullin and/or connective tissue growth factor (for additional proteins, see Table 1), can be used alone or in combination to assess test viability, efficacy and suitability of stem cells or other cell types for transplantation and regeneration therapy. - Diagnostic/prognostic markers can be used alone or in combination to assess clinical outcome following acute myocardial infarction or heart failure as an indicator of the patients inherent ability to repair/regenerate injured myocardium by measuring one or more of the factors including ST2, IL33, brain acid
soluble protein 1, cathepsin - B, Cu/Zn superoxide dismutase, cystatin E/M , insulin-like growth
factor binding protein 2 and minecan, atrial natriuretic peptide (ANP), apolipoprotein E,matrix metallopeptidase 2,metalloproteinase inhibitor 1, adrenomedullin and/or connective tissue growth factor (or their binding partners) in body fluid of patents (for additional proteins, see Table 1). - Therapeutic target or protein application of paracrine factor(s) and/or their binding partners can be used through endogenous administration or the use of a method to enhancement in vivo cellular release in individual patents in order to improve cardiac heart regeneration. Specifically, ST2, its ligand IL33 and/or including brain acid
soluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M, insulin-like growthfactor binding protein 2 and minecan, atrial natriuretic peptide (ANP), apolipoprotein E,matrix metallopeptidase 2,metalloproteinase inhibitor 1, adrenomedullin and/or connective tissue growth factor (or their binding partners) could be increased alone or in combination, in the myocardium (at or near site of injury) to enhance the in vivo cardiac and vascular regeneration and stem cell action. Protein(s) could be administered at the time of heart injury (e.g. at time of myocardial infarction or during development of heart failure), either alone, or in advance of or at the time of or following administration of endogenous stem cell. - Paracrine factor(s) and/or their binding partners can be used in stem cell culture to be used in regeneration therapy. Specifically, ST2, it ligand IL33 and/or including (but not exclusive) brain acid
soluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M, insulin-like growthfactor binding protein 2 and minecan, atrial natriuretic peptide (ANP), apolipoprotein E,matrix metallopeptidase 2,metalloproteinase inhibitor 1, adrenomedullin and connective tissue growth factor (see secreted factors Table 1), alone or in combination, can be added to the media of the cell culture to enhance preparation of stem cells prior to, at the time or following administration to patients. - Personalized medicine applications can be used in stem cell therapy, through the monitoring of paracrine proteins and their binding protein will allow clinician intervene, choose effective dose and time course of stem cells and proteins therapy during the course of stem cell therapy.
- For adult autologous stem cell transplantation in subjects for whom therapy is to be performed, it is contemplated that 106 to 108 cells will be effective. These cells would be pretreated or injected at same time of the paracrine factors are added. Similar amounts of cardiac stem cells are expected to be effective. Determination of effective dosages can be done without undue experimentation by those of skill in the art. (See, e.g. [7-9]).
- All proteins listed in the Tables herein maybe therapeutically important for cell survival, proliferation or deformation in the injured area of the heart. As well, they may be biomarkers for assessment of cell viability and effectiveness, responsiveness of the patent to stem cell therapy and long term prognosis of development of heart failure or survival. Equivalent proteins from humans or other species including isoforms, splice varients and polymorphorisms/SNPs are also expected to be effective. These are known to and can be tested by those of skill in the art without undue experimentation. Proteins considered to be particularly useful are listed in Table 2.
- Another embodiment of the invention is a method to screen for paracrine factors from cells including stem cells. The method uses reversed phase HPLC to separate the intact proteins prior to MS.
- This application claims priority to U.S. provisional application no. 61/001,792, filed Nov. 2, 2007, which is hereby incorporated by reference.
-
FIG. 1 : Cardiac cell analysis strategy -
FIG. 2 : Representative chromatograms and reproducibility obtained by RPLC analysis for CSps, CDCs and NRVms with 9 final fractions used for ESI-MS/MS protein identification after trypsin digestion. Eight chromatograms are overlaid (100 μg/run) to demonstrate the reproducibility. Fraction containing albumin was not analyzed. -
FIGS. 3A-3C : Influence of proteins IL33, BASP1 and CTCF at different concentrations on proliferation of CDCs. BASP1 and CTCF increases proliferation of CDCs. Three proteins identified as potentially important secreted proteins, IL33,BASP 1 and CTCF, were tested at different concentrations on their ability to alter proliferation rate of rat CDCs. Detailed description is given in text. Note, CTCF began to alter proliferation byday 4 while BASP1 took longer and change was seen atday 7. On the other hand, IL33 had no affect during this time frame. - Soluble proteins secreted from stem cells of any cell type including the myocytes near or at the site of injury can serve as interactive signals to the local environment, influencing survival, differentiation and stem cell engraftment as well as affects on the injured myocardium. We have identified the secretome of cardiac stem cells (cardiospheres and cardiosphere-derived cells) and compared these proteins to those specifically secreted from isolated neonatal myocytes. This has allowed us to hone in on potential regulatory pathways that will allow modulation of the stem cells differentiation into cardiac myocytes.
- The expression of any combination of the proteins, protein isoforms, protein polymorphorism, peptide fragment(s) protein with post-translational modifications thereof examined herein, or others, can be assayed with a method of the invention. For example, one can first measure the amount of expression of apolipoprotein E precursor compared to a baseline value, to determine if a significantly elevated or decreased amount is present in a patient sample. One can then further measure the amount of expression of adrenomedullin and/or Dkk3 protein compared to the baseline value. A significant increase (e.g. at least a statistically significant increase) in the amount of expression of one or more proteins, e.g. as listed in Table 2, compared to the baseline value indicates a greater likelihood that the patient is in need of, or would benefit from stem cell therapy.
- By “peptide fragment” is meant any fragment of a protein of interest of at least 10 amino acid residues, preferably at least 15, 20, 15, or 30 amino acid residues up to fragments that may be only a few (e.g. 1, 2, 3, 4, 5, 10, 15, etc.) residues shorter than the full length protein. Unless otherwise indicated, the terms “protein, “polypeptide” and “peptide” are used interchangeably herein.
- The amino acid sequences of the proteins of the invention, and the nucleic acids encoding them, are well-known and can be determined routinely, as well as downloaded from various known databases using the provided GenBank Accession numbers. See, e.g., the world wide web site, ncbi.nlm.nih.gov.
- The amount of expression of a protein of the invention can be determined by measuring the amount of the protein, or by measuring the amount of mRNA encoding the protein. The amount of a protein can be determined using any routine method known in the art, e.g. MS, an antibody, etc. For example, the method can encompass binding the protein to an antibody which is specific for it, under conditions that are effective for specifically binding the protein to the antibody. For example, an antibody or any sensor may be contacted with a histological preparation, and the amount of protein is determined by immunohistochemical staining. The amount of an mRNA can be determined using a nucleic acid probe for the mRNA. For example, the method can encompass hybridizing the mRNA to a nucleic acid probe which is specific for it, under conditions that are effective for specifically hybridizing the mRNA and the probe.
- Some methods involve the use of antibodies, any binding ligand or mass spectrometry tagged peptide specific for a protein of interest. Antibodies suitable for use in such assays are commercially available, or can be prepared routinely. Methods for preparing and using antibodies in assays for proteins of interest are conventional, and are described, e.g., in Green et al., Production of Polyclonal Antisera, in Immunochemical Protocols (Manson, ed.), (Humana Press 1992); Coligan et al., in Current Protocols in Immunology, Sec. 2.4.1 (1992); Kohler & Milstein (1975), Nature 256, 495; Coligan et al., sections 2.5.1-2.6.7; and Harlow et al., Antibodies: A Laboratory Manual, page 726 (Cold Spring Harbor Laboratory Pub. 1988).
- Any of a variety of antibodies can be used, including, e.g., polyclonal, monoclonal (mAbs), recombinant, humanized or partially humanized, single chain, Fab, and fragments thereof. The antibodies can be of any isotype, e.g., IgM, various IgG isotypes such as IgG1′ IgG2a, etc., and they can be from any animal species that produces antibodies, including goat, rabbit, mouse, chicken or the like. The term, an antibody “specific for” a protein, means that the antibody recognizes a defined sequence of amino acids, or epitope, in the protein, and binds selectively to the protein and not generally to proteins unintended for binding to the antibody. The parameters required to achieve specific binding can be determined routinely, using conventional methods in the art.
- The baseline value for such measurements can be an average or mean from a population of normal subjects, i.e. those not suffering from heart disease, or, e.g. may be earlier measurements taken from the same patient, for example when progress of treatment is being monitored. Suitable baseline values can be determined by those of skill in the art without undue experimentation.
- Thus, a “baseline value” refers to the expression, as determined by the levels (amounts) of mRNA and/or protein, in normal tissue (e.g., the same type of tissue as the tested tissue, such as normal cardiac tissue or normal serum), from normal subjects that do not have heart disease. If desired, a pool of the same tissues from normal subjects can be used. Such baseline values may be available in a database compiled from the values and/or may be determined based on published data or on retrospective studies of patients' tissues, and other information as would be apparent to a person of ordinary skill implementing a method of the invention. Suitable baseline values may be selected using statistical tools that provide an appropriate confidence interval so that measured levels that fall outside the standard value can be accepted as being aberrant from a diagnostic perspective, and predictive of success in treatment, diagnosis or prognosis.
- A significantly elevated amount of an mRNA or a protein (or peptide fragment) of the invention compared to this baseline value, then, indicates that a patient or subject is likely to be responsive to stem cell therapy. If a protein whose expression is decreased in subjects that are expected to be less responsive, a significantly reduced amount of the protein or mRNA encoding it indicates that a test subject is less likely to respond.
- A “significant” increase in the amount of a protein or mRNA, as used herein, can refer to a difference which is reproducible or statistically significant, as determined using statistical methods that are appropriate and well-known in the art, generally with a probability value of less than five percent chance of the change being due to random variation. Some such statistical tests are described in the Examples herein. For example, a significant increase in the amount of mRNA or protein compared to a baseline value can be at least about 2.5-fold (e.g., at least about 5-fold, 10-fold, 20-fold, 25-fold, or more) higher.
- Methods for obtaining samples and preparing them for analysis (e.g., for detection of the amount of protein or mRNA encoding the protein) are conventional and well-known in the art.
- A “subject” or “patient”, as used herein, includes any animal that has, or may have, heart disease, including experimentally induced heart disease, for example in laboratory animals (such as mouse, rat, rabbit, or guinea pig), farm animals, and domestic animals or pets (such as a cat or dog). Non-human primates and human patients, are included. Heart failure can be induced following heart attack or other injuries to the heart (pulmonary hypertension), viral infections, genetic disorders and many other affectors that weaken the heart muscle or vasculature. Furthermore, following AMI when heart muscle has been injured or destroyed, regeneration is required to replace the lost muscle cells and other cells required for functioning heart.
- A “sample” includes any biological sample, for example whole blood, serum, cardiac tissue, etc. obtained from a subject or patient. A sample also includes stem cell cultures, e.g. for therapeutic use in such patients.
- By “stem cell therapy” is meant the administration of cardiac myocytes or equivalent cells as described herein to a cardiac patient or to an animal with experimentally induced heart disease. The paracrine factor(s) could alter the honing, survival, proliferation and differentiation of all stem cells and progenitor cells found in the body including EPC, mesochymal, and hemopoetic stem cell. Administration of cells can be by any means known in the art, for example, by local injection, infusion,. Cells would be pretreated at optimal concentration most likely between pg/ml-1 μg/ml for a 10,000 cells (in 1 ml).
- By “heart disease” is meant by stable and unstable angina, myocardial ischemia, myocardial stunning, acute myocardial infarction, (minor necrotic cell death, heart failure induced by myocardial infraction, genetic disease, pulmonary hypertension and other injury to the myocardium which makes the heart work harder.
- By “treated” is meant that an effective amount of a chemotherapeutic drug or other anti-cancer procedure is administered to the subject. An “effective” treatment refers to a treatment that elicits a detectable response (e.g. a therapeutic response) in the subject.
- By “effective amount” is meant any amount that will elicit a detectable clinical response.
- A significant increase in the amount of expression of a secreted protein of the invention compared to the baseline value indicates that a subject is likely to be responsive to stem cell therapy. A subject that is “likely” to be responsive has greater than, e.g., at least about: a 25%, more likely 50%, 75%, chance to show an improvement in clinical symptoms.
- In order to determine the specificity of the secreted proteome (“secretome”), we compared proteins secreted into media conditioning adult cardiac stem cells (CSps and CDC) to proteins secreted from neonatal rat ventricular myocytes after analysis by reversed phase liquid chromatography and identification by mass spectrometry (
FIG. 1 ). The cells were obtained under optimized conditions that minimize cell lysis, allowing us to distinguish between secreted proteins and those proteins that would be liberated upon cell death due to necrosis and/or apoptosis. - Sample Processing Specimen preparation, CSps, CDCs and NRVMs (rat neonatal myocytes) were harvested as previously described [1-3]. Briefly, cardiac tissue specimens from septum or left ventricule of Wistar Kyoto rats, 12 to 16 weeks old, were cut into small pieces, washed by PBS, enzymatically digested and grown in primary cultures as explants on fibronectin (from human plasma, BD Biosciences) coated Petri dishes in IMDM medium (Iscove's Modified Dulbecco's Medium, Invitrogen) supplemented with 20% fetal bovine serum (FBS), 1% penicillin-streptomycin, 1% L-glutamine and 0.1 mmol/L 2-mercaptoethanol at 37° C. and 5% CO2. After a few days, round, phase-bright generated cells migrating over a layer of fibroblast-like cells arose from explants were harvested and seeded at 2-3×104 cells/mL on poly-D-lysine (BD Biosciences) coated Petri dishes in cardiosphere-growing medium composed of 35% IMDM/65% DMEM-Ham F-12 Mix (Invitrogen) and supplemented with 3.5% fetal bovine serum (FBS), 1% penicillin-streptomycin, 1% L-glutamine, 0.1 mmol/L 2-mercaptoethanol, thrombin (1 unit/mL, Sigma), B-27 (diluted at ratio 1:50, Invitrogen), basic fibroblast growth factor (bFGF, 80 ng/mL, PeproTech), epidermal growth factor (EGF, 25 ng/mL, PeproTech) and cardiotrophin-1 (4 ng/mL, PeproTech). The yielded spherical multicellular clusters—cardiospheres (CSps) were collected for proteomic analysis or further processed by plating on fibronectin-coated flasks and growing as monolayers giving finally cardiosphere-derived cells (CDCs). Neonatal rat ventricular myocytes (NRVMs) were isolated by routine methods with overnight trypsin digestion from 2 days old Sprague-Dawley rats. Cardiospheres and cardiosphere-derived cells were observed. The cells were isolated under conditions with minimal cell death (18±2% for CSps and CDCs, n=3 and 10±1% for NRVMs, n=3) as assessed by Annexin V labeling. CSps, CDCs and NRVMs were further conditioned in media containing 1%
- FBS for 48 hours. After conditioning, media with cell secreted proteins were collected, filtered and concentrated in SpeedVac concentrator. Conditioning medium alone was processed the same way and served as control.
- The liquid concentrates with secreted proteins and concentrates of media alone were mixed with solvent to final concentration of 20% (v/v) acetonitrile (ACN), 1% (v/v) trifluoroacetic acid (TFA), pH 2.3, vortexed and spun down at 18×1000 g for 30 min at 4° C. Samples of intact proteins were separated by RPLC in order to reduce the complexity of protein mixtures prior to mass spectrometry protein identification. 200 or 800 μg were injected into C18 column (50 mm, nanoporous particles, HPRP model of ProteomeLab PF 2D, Beckman Coulter, Calif., USA) in consecutive runs of 100 μg/run in order to avoid the clogging of the column by the high amount of albumin, using a linear gradient from 0% to 100% ACN/0.08% TFA over 35 minutes. The chromatogram was recorded at an absorbance wavelength of 214 nm.
FIG. 2 shows representative chromatographs of multiple runs as well as the fractions pooled for MS analysis. 68 fractions were collected per run from 7 to 24 min (0.25 min/fraction) and combined into 9 final fractions, dried in SpeedVac concentrator, neutralized by ammonium bicarbonate, trypsin digested overnight at 37° C. and analyzed by mass spectrometry for protein identification and quantification. Fractions containing major amounts of albumin (between and 17.25 min of elution time, 50-53% acetonitrile) were excluded. - Dried samples with tryptic peptides after trypsin digestion of fractions were recovered in 8 μl of 0.1% (v/v) TFA and analyzed by a ThermoFinnigan LTQ ion trap with electrospray ionization or by a LTQ Orbitrap mass spectrometer (Thermo Electron Corporation, Mass., USA). Data obtained from MS spectra were submitted to NCBInr database search by using MASCOT search engine (Matrix Science Mascot Daemon, V2.1.3 -max. missed
cleavages 2, peptide tolerance ±1.5 Da and MS/MS tol. ±0.8 Da, all species). After Mascot Daemon search, the files were transferred to Scaffold software (Version Scaffold-01—06—06, 2006 Proteome Software Inc., OR, USA) for Mascot result validation, visualization and comparison of protein identifications between individual samples. All identified proteins were further examinated for peptide and protein redundancy. The protein amino acid sequence was blasted against UniProt Knowledgebase (Swiss-Prot+TrEMBL) by using SIB - BLAST network service (ExPASy). In case of protein multiple names or homology, only one protein name was used after the original peptide sequences obtained from our MS results were checked back for matching that protein by using multiple sequence alignment program ClustalW (EMBL-EBI). Also, the confirmation of a protein isoform was done based on matching a tryptic peptide fragment to a unique amino acid sequence of isoform of the intact protein.
- Three technical issues were overcome regarding analysis of the secretome, i) the secreted proteins are often at low concentration within the culture media, ii) the media is a large source of contamination (including albumin and iii) the need to distinguish between secreted proteins and those artificially liberated with cell death. This will be assessed this by both Annexin V labeling and increased detection of intercellular proteins (as outlined elsewhereherein). In Table 1 there are several protein highlighted which may arise from lysed cells due to their known intracellular location and the low level quantity present in the media. The remaining proteins are known to be secreted from cells or are membrane or membrane associated proteins.
- To maximize proteome coverage it was also important to reduce serum content of the media which would hinder detection of low abundant proteins during proteomic analysis. Initially, we analyzed a series of CSps or CDCs obtained from rat heart biopsies to neonatal myocytes grown under different conditions ranging from 6 hours to 4 days in 0%, 1% and 2% serum. We elected to use conditioned media with 1% serum and to collect the media after 48 hours, although earlier time points also worked. The collected media was filtered, concentrated, suspended in 20% (v/v) acetonitrile:1%TFA and separated by reversed phase HPLC using a linear gradient composed of 0.1% TFA and increasing concentrations of acetonitrile from 25% to 80%. Multiple fractions were collected, neutralized, digested with trypsin and analyzed by ESI/MS/MS for protein identification and quantification.
- We were able to observe 122 non-redundant proteins exclusively in media from cardiac stem cells or neonatal myocytes, with the majority (>90%) comprising known membrane, extracellular or secreted proteins. This indicates little contamination from dead cells. In the first set of experiments, greater than 2 fold more proteins were detected in cardiac stem cells than neonatal myocytes (60 vs. 27, respectively) and although the functional protein classes were conserved between the cell types, specific proteins and/or isoforms differed. Not surprisingly, we observed a large number of different collagen isoforms (10) with half (5) being cell-specific. Cell-specificity was also reflected with collagen regulation as TIMP2 and MMP2 were observed in neonatal myocytes and
TIMP 1 uniquely present in the cardiac stem cells. Interestingly, a recent study showed that proteins secreted from ESC (normal and hypoxic) could inhibit hydrogen peroxide induced apoptosis, and this inhibition was due, at least in part, to TIMP. On the other hand, MMP2 Is increased in heart failure in both animal models and patient samples. - Notable was the detection of two paracrine factors—the cardiac specific atrial natriuretic peptide or ANP was only detected in neonatal myocytes and not cardiac stem cells, while the soluble interleukin-1 receptor family member or ST2 was exclusive to cardiac stem cells (Table 1). ST2 is known to increase in myocytes with mechanical stress and heart failure where it has been linked to neurohormonal activation (e.g. (70,71) and reduces the endogenous affect of IL-33 (its ligand) of reducing hypertrophy and fibrosis. Furthermore, recently indirectly indicated at the gene level in C3H10T1/2 cells (a proliferative cell line) where ST2 is increased as is Wnt-5a, a know stem cell signaling factor.
- Additional proteins that were found to be exclusively present in the media analyzed from cardiac stem cells or myocytes are listed in Table 1. Proteins that may play a role (alone or in combination) found exclusively in the myocyte media are apolipoprotien E, matrix metallopeptidase 2 (and although its inhibitor (
metalloprotease 2 inhibitor) is not altered the ratio of these two proteins maybe critical), and metalloproteinase inhibitor 1 (and although the correspondingmatrix metallopeptidase 1 is not observed, the ratio of the two proteins maybe important). In fact, all metalloproteinase and their inhibitors may play a role in both stem cell and myocyte function and survival in the infracted or injured area of the heart. Additional proteins found exclusively in the cardiac stem cell media are brain acidsoluble protein 1, cathepsin B, Cu/Zn superoxide dismutase, cystatin E/M, and the insulin-like growth factor binding protein 2 (although not observed insulin growth factor (IGF) and other analytes that bind this protein or to the IGF receptor maybe important). - Brain
soluble protein 1 is also named BASPI protein, neuronal axonal membrane protein NAP22 and the 22 kDa neuronal tissue-enriched acidic protein. This membrane protein is involved in cell projection and growth cones and was originally thought to be present exclusively in the brain. Cathepsin B also named APP secretase (APPS) contains a heavy and light chain within its amino acid sequence which after processing forms a dimmer with the heavy and light chains crosslinked by a disulfide bond. This protein in its active form is a thiol protease involved in degradation and turnover of proteins and is normally associated with the lysosome. Cystatin E/M is also called Cystatin M/E orcystatin 6 is a protease inhibitor. Insulin like growthfactor binding protein 2 is also called IGFB-2, IBP-2 and IGF-bindingprotein 2 is secrteted and prolongs the half life of the IGF and has been shown to alter growth promoting affects on IGF on cell cultures. IGF and the insulin like growthfactor binding protein 2 have been implicated in changes to may cells types including fibroblasts, lung epithelial cells, and glial cells but to our knowledge not cardiac derived stem cells or the cardiac myocyte. - In another set of experiments, we carried out independent duplicate analysis of the secretome from CDC, CSps and NRVMs obtained from left ventricle of rat hearts.
- We identified a total of 161 proteins. With this set of data we were able to expand our list of potential candidate paracrine factors. Of specific interest is adrenomedullin and connective tissue growth factor which are present in the media of NRMV exclusively and minecan which is found exclusively in the media of cardiac stem cells. Minecan also called osteoglycin or osteoinductive factor is known to induce bone formation in conjunction with
TFG beta 1 orTGF beta 2, and have been implicated in regulation of collagen fibrillogeneis of the cardiac stem cells or act also on cells in the surrounding regions of the injured heart. - Adrenomedullin or AM is processed active domain of larger protein called ADML or ADM which also contains the proadrenomedullin N-20 terminal peptide (PAMP). AM and PAMP are potent hypotensive and vasodilator factors that have affects throughout the body including the kidney, brain and pituitary gland. It has been found in the ventricle. This protein has been implicated in differentiation of several stem cells but not cardiac derived cells or injured myocytes. Connective tissue growth factor is also termed hypertrophic chondrocyte-
specific protein 24. This protein is a connective tissue mitoattractant that has been previously reported to be secreted by vascular endothelial cells but, to our knowledge not cardiac myocytes under normal physiological conditions but has been shown to increase in hearts with ongoing myocarditis. - Furthermore, AM has been shown to induce proliferation and differentiation of chondrocytes, improve cell adhesion for fibroblasts, myofibroblasts, endothelial and epithelial cells and stimulate fibroblast growth factor-induced DNA synthesis. Therefore, fibroblast growth factor alone or its ratio with respect to connective tissue growth factor may be also important.
- Soluble proteins secreted from stem cells serve as interactive signals to the local environment, influencing survival, differentiation and engraftment. To characterize the secreted proteome (“secretome”), proteins found in the media conditioned by adult cardiac stem cells (CSCs) or neonatal rat ventricular myocytes (NRVMs) were obtained under optimized conditions that minimize cell lysis, allowing distinction between secreted proteins and those artificially liberated with cell death.
- CSCs were grown from rat septal or left ventricular explants and NRVMs were isolated under conditions with minimal cell death (18±2% [CSCs, n=3] and 10±1% [NRVMs, n=3] by Annexin V labeling). Conditioned media (1% serum) was collected after 48 hours, filtered, concentrated, resuspended in 20% (v/v) acetontile:1%TFA and separated by reversed phase HPLC. Collected fractions were digested with trypsin and analyzed by ESI/MS/MS for protein identification and quantification.
- 90-110 proteins were identified exclusively in media from CSCs or NRVMs, with the majority (>85%) comprising known membrane, extracellular or secreted proteins. Of these, >2 fold more proteins were detected in CSCs than NRVMs (60 vs. 27, respectively). Functional protein classes were conserved between the cell types, although proteins and/or their isoforms could differ. Of interest, 10 different collagen isoforms were observed with 5 being cell-specific. Cell-specificity was also reflected with collagen regulation as TIMP2 and MMP2 were observed in NRVMs while
TIMP 1 was uniquely present in CSCs. The signaling molecules, insulin-like growthfactor binding protein - From these results, it is apparent that CSC and NRVM-specific secretomes display unique functionality including differential secretion of two cardiovascular hormones.
- Day 1: cardiosphere-derived cells from rat (CDCs) or neonatal myocytes or adult myocytes in IMDM medium (Iscove's Modified Dulbecco's Medium, Invitrogen) supplemented with 20% fetal bovine serum (FBS) were plated into 96-well plate in 8 rows except for blanks (last 2 wells in each row). Media (without the cells) were plated into last 2 wells of 1st row. Cells were incubated in 37° C. incubator for 48 hours.
- Day 3: the cells in wells were checked under microscope (the cells must be attached to the well bottoms). The media from all wells were aspired except for those of the 1st row using sterile Pasteur pipette. Then, 100 tl of appropriate protein solutions with increasing
concentrations 1 pg/ml, 1 ng/ml, 100 ng/ml, 1 μg/ml in IMDM medium supplemented with 10% FBS (treated cells; each in duplicate) and 100 μl of the same media without a protein (controls—untreated cells; in duplicate) were added to attached cells in wells of each row. Last 2 wells in each row were filled with 100 μl of only media (blanks). - 10 μl of Cell Counting Kit-8 solution (Dojindo laboratories, Japan) were added to the 1st row of the plate (avoiding a direct light exposure). The cells in plate were incubated in 37° C. incubator for 2 hours and after that the absorbances of solutions in 1st row wells were read (λ=450 nm, SPECTRAmax M2, Molecular Devices, Sunnyvale, Calif.).
- Day 4: 10 μl of Cell Counting Kit WST-8 solution were added to the 2nd row of the plate, the cells in plate were incubated in 37° C. incubator for 2 hours and after that the absorbances of solutions were read.
-
Day 5 to 10: the procedure ofday 4 was repeated on each following plate row. Influence of proteins interleukin-33 (IL 33, ALX-522-098-0010, Apotech Corporation, USA), brain abundant membrane attached signal protein 1 (BASP1,H00010409-P01, Novus Biological, USA) and connective tissue growth factor (CTGF, CRC604B, Cell Sciences, USA) at different concentration on CDCs proliferation is shown inFIG. 3 . - Modified Boyden chambers were equipped with 8 μm pore-size polycarbonate filters (Neuroprobe, Gaithersburg, Md.) coated with Matrigel (BD Bioscience, Palo Alto, Calif.). Cells (stem cells or myocytes) were either untreated or pretreated with appropriate protein for 24 hours. 220 μl of migration medium (DMEM—Dulbecco's Modified Eagle Medium with 0.1% BSA) was added to lower chamber with or without platelet-derived growth factor (PDGF) for chemattract and random migration assay, respectively. Cells (106/ml) were placed in upper chamber in 200 μl of migration medium, performed in triplicate. The assay was stopped after 4 hours at ° C. Cells that crossed the basement membrane and migrated to lower side of the filter were fixed and stained using HEMA3 system (Curtin, Matheson Scientific Inc., Houston, Tex.). Four random fields were counted at 400× magnification for each filter.
- To evaluate the effect of oxidative stress, the cells (NRVMs, myocytes or stem cells) were exposed to hydrogen peroxide for a fixed period of time (1 hour of 100 μmol/L H2O2). Mitochondrial membrane potential (determinant of myocytes viability) was measured by using flow cytometric analysis of tetramethylrhodamine ethyl ester (TMRE) loaded cells. TMRE (100 nmol/L) was loaded for 20 min in the dark at 37° C. Myocytes were subjected to flow cytometry by activation with the 488 nm wavelength. Fluorescence was monitored and influence of H2O2 on changes of mitochondrial membrane potential was determined.
- Effect of the Proteins on Cell Morphology Combined with Immunochistochemistry (Immunostaining, Immunocytochemistry, IHC).
- IHC is method for identification of specific tissue components by means of specific antigen/antibody (or other sensors) reaction tagged with a visible label (or other types of chemical labels). This method makes possible to visualize the distribution and localization of specific cellular components within a cell or tissue.
- First, the tissue for staining is fixed. Fixative procedure is optimized based on tissue and antigen/sensor used and various fixatives can be used. Usually, the tissue is formalin fixed and paraffin embedded. Prior to staining, tissue slides are deparaffinized (e.g. in xylol) and rehydrated in graded alcohol series and can be further pretreated with proteolytic enzymes, washed in distilled water and heated in microwave oven for epitope retrieval. Then nonspecific sites are blocked with serum or blocker protein, incubated with primary antibody (1:100-1:1,000), washed and incubated with secondary antibody-enzyme conjugate (1:2,000-1:5,000), washed and incubated with substrate and finally, the stained tissue is visualized.
- The CSps-, CDC-, NRVM-media with secreted proteins/lysates or adult myocyte lysates were subjected to 1D gel electrophoresis (NuPage BisTris gels of various concentrations, 200 V, 35 to 55 minutes based on running buffer and gel concentration used), separated proteins were transferred into nitrocellulose membrane (NuPage transfer buffer, 100 V, 1 hour). Proteins on the membrane were visualized by dye Direct Blue 71, briefly destained by 40% ethanol/10% acetic acid, washed by lx Tris buffered saline solution (TBS; 20 mM Tris, 500 mM NaCl, pH 7.5) and transferred into blocking solution (5% of non fat dry milk in 1×TBS). The membrane was blocked overnight at 4° C., then washed by TTBS (0.1% Tween-20 in TBS) and incubated with appropriate primary antibody (1:100-1:1,000) for 4.5 hours. After that, the membrane was washed in TTBS and incubated with secondary antibody-enzyme conjugate (1:2,000-1:5,000), washed in TTBS and incubated with ImmunostarAP Substrate Pack (BioRad laboratories, Calif., USA) for 5 minutes. The membrane was placed in cassette with film and developed.
- Stem cells treated and non-treated are labeled with tracking agent such as iron as described by Terrovitis et al [4] or unique protein, chromosome, gene or chemical compound(s). For example, the ferumoxide-labeled CDCs (with and without treatment either exogenous protein or transgenically manipulated to produce proteins, see Smith et al. [5] for examples) is injected intramyocardially into normal (immunocompetent) rats or mice prior to or after induction of experimental of myocardial infarction. Rats may also undergo left thoracotomy in the fourth or fifth intercostals space under general anesthesia. The heart will be exposed and the cells injected directly into the myocardium at a single or multiple sites.
- The myocardial infarction can be produced by a number of methods including a permanent ligation of the left anterior descending coronary artery using a suture immediately before or after cell injection. Subsequently, the chest is closed and the animals are allowed to recover. Magnetic resonance imaging (MRI) images are obtained on a number of days after surgery such as
day 2 and 21 or longer. After completion of this follow-up period, the rats will be sacrificed and the hearts subjected to histology. - MRI: For MRI, animals were anesthetized and then placed prone, head first in the magnet. ECG-gated cine images of the heart are obtained. At least 3 consecutive short-axis slices will be acquired to completely cover the area of cell injection. Signal intensity will be measured in the myocardium (remote areas and areas of cell injection); noise is measured by creating regions of interest in the lungs. Contrast-to-noise ratios (signal intensity in the remote myocardium minus signal intensity in the areas of the cell injection divided by the SD of noise) will be calculated for each slice in which the signal void is visualized. In addition, percent signal area will be calculated as the area of visually determined signal void (manually defined region of interest containing area obviously darker than the surrounding myocardium) divided by the total left ventricular area in the same slice. Histological analysis of cell engraftment will be performed for number of proteins and for general pathological stains.
- Echocardiography will be performed in conscious animals before and after myocardial infarction. The anterior chest area is shaved and 2D images and M-mode tracings are recorded from the parasternal short-axis view at the level of papillary muscles. In addition, it maybe needed to evaluate the LV pressure and +and −dP/dt in the closed-chest preparation. (for example see Rota et al. [6])
- As well the abdominal aorta could be cannulated with a polyethylene catheter and the heart arrested in diastole (eg. injection of CdCl2), the thorax was opened, perfusion with phosphate buffer started. An aortic catheter connected to a pressure reservoir is used to adjust perfusion pressure to mean arterial blood pressure while simultaneously, the LV chamber is filled with formalin. After perfusion with buffer, the coronary vasculature is perfused with fixative. Subsequently, the heart is excised, and the weights and major axis from the base to the apex of the heart is measured. The volume of the myocardium is computed (4) by dividing the weight by the specific gravity of muscle tissue. Furthermore, paraffin-embedding of tissue could be done. In this case tissue slices would be stained with hematoxylin and eosin or used later for immunohistochemistry.
- References cited herein are hereby incorporated by reference.
-
- [1] Messina E, De Angelis L, Frati G, Morrone S, Chimenti S, Fiordaliso F, Salio M, Battaglia M, Latronico M V G, Coletta M, Vivarelli E, Frati L, Cossu G, Giacomello A. Isolation and expansion of adult cardiac stem cells from human and murine heart. Circ Res. 2004;95:911-921.
- [2] Smith R R, Barile L, Cho H C, Leppo M K, Hare J M, Messina E, Giacomello A, Abraham M R, Marban E. Regenerative potential of cardiosphere-derived cells expanded from percutaneous endomyocardial biopsy specimen. Circulation. 2007;115:896-908.
- [3] Iravanian S, Nabutovsky Y, Kong C R, Saha S, Bursac N, Tung L. Functional reentry in cultured monolayers of neonatal cardiac cells. Am J Physiol Heart Circ Physiol. 2003;285:H449-H456.
- [4 ] Terrovitis J, Stuber M, Youssef A, Preece S, Leppo M, Kizana E, Schär M, Gerstenblith G, Weiss R G, Marban E, Abraham M R. Magnetic resonance imaging overestimates ferumoxide-labeled stem cell survival after transplantation in the heart. Circulation. 2008 Mar 25;117(12):1555-1562.
- [5] Smith R R, Barile L, Cho H C, Leppo M K, Hare J M, Messina E, Giacomello A, Abraham M R, Marban E. Regenerative potential of cardiosphere-derived cells expanded from percutaneous endomyocardial biopsy specimens. Circulation. 2007
Feb 20;115(7):896-908. - [6] Rota M, Kajstura J, Hosoda T, Bearzi C, Vitale S, Esposito G, Iaffaldano G, Padin-Iruegas M E, Gonzalez A, Rizzi R, Small N, Muraski J, Alvarez R, Chen X, Urbanek K, Bolli R, Houser S R, Leri A, Sussman M A, Anversa P. Bone marrow cells adopt the cardiomyogenic fate in vivo. Proc Natl Acad Sci U S A. 2007
Nov 6;104(45): 17783-17788. - [7] Patel A N, Geffner L, Vina R F, Saslaysky J, Urschel H C Jr, Kormos R, Benetti F. Surgical treatment for congestive heart failure with autologous adult stem cell transplantation: a prospective randomized study. J Thorac Cardiovasc Surg. 2005;130(6):1631-8.
- [8] Assmus B, Honold J, Schachinger V, Britten M B, Fischer-Rasokat U, Lehmann R, Teupe C, Pistorius K, Martin H, Abolmaali N D, Tonn T, Dimmeler S, Zeiher A M. Transcoronary transplantation of progenitor cells after myocardial infarction. N Engl J Med. 2006 21;355(12):1222-32.)
- [9] Schuleri K H, Amado L C, Boyle A J, Centola M, Saliaris A P, Gutman M R, Hatzistergos K E, Oskouei B N, Zimmet J M, Young R G, Heldman A W, Lardo A C, Hare Early improvement in cardiac tissue perfusion due to mesenchymal stem cells. Am J Physiol Heart Circ Physiol. 2008 May;294(5):H2002-11.
Claims (5)
1-18. (canceled)
19. A method for screening a cardiac patient as a candidate for stem cell therapy, said method comprising measuring in a biological sample from the patient the amount of one or more of apolipoprotein E precursor, atriopeptigen (ANP, ANF), adrenomedullin, brain acid soluble protein 1 (BASP1 protein), cathepsin B precursor, collagen alpha-1 (VI) chain precursor, connective tissue growth factor, cystatin E/M, Dkk3 protein, insulin-like growth factor-binding protein 2 precursor, interleukin-1 receptor-like 1 precursor (ST2), matrix metallopeptidase 2, metalloproteinase inhibitor 1 precursor (TIMP-1), metalloproteinase inhibitor 2 precursor (TIMP-2), mimecan precursor, procollagen type III alpha 1, procollagen type V alpha 1, procollagen type 1 alpha 1, procollagen type VI alpha 2, tropoelastin, or peptide fragments thereof, compared to a baseline value, wherein a statistically significant amount of the one or more proteins or peptide fragments thereof compared to the baseline value indicates that the patient would be likely to benefit from stem cell therapy.
20. The method of claim 19 , wherein the statistically significant amount represents at least a 5% increase over baseline value.
21. The method of claim 19 , wherein the cardiac patient is suffering from angina, acute myocardial injury, cellular necrosis and myocardium hypertrophy or heart failure.
22. The method of claim 19 , wherein the biological sample is a whole blood sample, serum sample, plasma sample, tissue biopsy sample, a sample of cultured cells derived from tissue, or a sample of circulating stem cells such as EPC.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US14/134,301 US20140274765A1 (en) | 2007-11-02 | 2013-12-19 | Cardiac stem cell and myocyte secreted paracrine factors and uses thereof |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US179207P | 2007-11-02 | 2007-11-02 | |
PCT/US2008/012415 WO2009061382A2 (en) | 2007-11-02 | 2008-11-03 | Cardiac stem cell and myocyte secreted paracrine factors and uses thereof |
US74088710A | 2010-07-23 | 2010-07-23 | |
US14/134,301 US20140274765A1 (en) | 2007-11-02 | 2013-12-19 | Cardiac stem cell and myocyte secreted paracrine factors and uses thereof |
Related Parent Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US12/740,887 Continuation US8617877B2 (en) | 2007-11-02 | 2008-11-03 | Cardiac stem cell and myocyte secreted paracrine factors |
PCT/US2008/012415 Continuation WO2009061382A2 (en) | 2007-11-02 | 2008-11-03 | Cardiac stem cell and myocyte secreted paracrine factors and uses thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
US20140274765A1 true US20140274765A1 (en) | 2014-09-18 |
Family
ID=40626381
Family Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US12/740,887 Expired - Fee Related US8617877B2 (en) | 2007-11-02 | 2008-11-03 | Cardiac stem cell and myocyte secreted paracrine factors |
US14/134,301 Abandoned US20140274765A1 (en) | 2007-11-02 | 2013-12-19 | Cardiac stem cell and myocyte secreted paracrine factors and uses thereof |
Family Applications Before (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US12/740,887 Expired - Fee Related US8617877B2 (en) | 2007-11-02 | 2008-11-03 | Cardiac stem cell and myocyte secreted paracrine factors |
Country Status (2)
Country | Link |
---|---|
US (2) | US8617877B2 (en) |
WO (1) | WO2009061382A2 (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2016196366A1 (en) | 2015-05-29 | 2016-12-08 | The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services | Extension of replicative lifespan in diseases of premature aging using p53 isoforms |
Families Citing this family (25)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
ITRM20030376A1 (en) | 2003-07-31 | 2005-02-01 | Univ Roma | PROCEDURE FOR THE ISOLATION AND EXPANSION OF CARDIOC STAMIN CELLS FROM BIOPSIA. |
US11660317B2 (en) | 2004-11-08 | 2023-05-30 | The Johns Hopkins University | Compositions comprising cardiosphere-derived cells for use in cell therapy |
DK1937815T3 (en) | 2005-09-13 | 2015-08-03 | Nat Res Council Canada | Methods and compositions for the modulation of tumor cell activity |
ES2512018T3 (en) * | 2009-07-27 | 2014-10-23 | F. Hoffmann-La Roche Ag | Use of mimecan in the evaluation of heart failure |
US8802826B2 (en) | 2009-11-24 | 2014-08-12 | Alethia Biotherapeutics Inc. | Anti-clusterin antibodies and antigen binding fragments and their use to reduce tumor volume |
US9249392B2 (en) | 2010-04-30 | 2016-02-02 | Cedars-Sinai Medical Center | Methods and compositions for maintaining genomic stability in cultured stem cells |
US9845457B2 (en) | 2010-04-30 | 2017-12-19 | Cedars-Sinai Medical Center | Maintenance of genomic stability in cultured stem cells |
US20120083767A1 (en) * | 2010-10-01 | 2012-04-05 | The Johns Hopkins University | Implantable bioreactor for delivery of paracrine factors |
BR112014020756A2 (en) | 2012-02-22 | 2017-07-04 | Alethia Biotherapeutics Inc | co-use of a clusterin inhibitor with an egfr inhibitor to treat cancer |
JP2015521054A (en) | 2012-06-05 | 2015-07-27 | カプリコール,インコーポレイテッド | Optimized methods for generating cardiac stem cells from heart tissue and their use in cardiac therapy |
EP2882445B1 (en) | 2012-08-13 | 2019-04-24 | Cedars-Sinai Medical Center | Exosomes and micro-ribonucleic acids for tissue regeneration |
US10126308B2 (en) | 2014-02-17 | 2018-11-13 | Arizona Board Of Regents On Behalf Of Arizona State University | Methods for assessing biospecimen integrity |
US10126306B2 (en) | 2014-02-17 | 2018-11-13 | Arizona Board Of Regents On Behalf Of Arizona State University | Methods for assessing biospecimen integrity |
WO2016054591A1 (en) | 2014-10-03 | 2016-04-07 | Cedars-Sinai Medical Center | Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy |
WO2017123662A1 (en) | 2016-01-11 | 2017-07-20 | Cedars-Sinai Medical Center | Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction |
WO2017210652A1 (en) | 2016-06-03 | 2017-12-07 | Cedars-Sinai Medical Center | Cdc-derived exosomes for treatment of ventricular tachyarrythmias |
EP3515459A4 (en) | 2016-09-20 | 2020-08-05 | Cedars-Sinai Medical Center | CARDIOSPHERIC CELLS AND THEIR EXTRACELLULAR VESICLES TO DELAY OR REVERSE THE AGING PROCESS AND AGE-RELATED DISEASES |
AU2018255346B2 (en) | 2017-04-19 | 2024-05-02 | Capricor, Inc. | Methods and compositions for treating skeletal muscular dystrophy |
DE102017116204B4 (en) * | 2017-07-18 | 2024-06-27 | Universität Rostock | Methods for predicting the response to cardiovascular regeneration |
WO2019071135A1 (en) * | 2017-10-05 | 2019-04-11 | The Johns Hopkins University | Implantable bioreactor and methods for making and using same |
US11660355B2 (en) | 2017-12-20 | 2023-05-30 | Cedars-Sinai Medical Center | Engineered extracellular vesicles for enhanced tissue delivery |
US12286644B2 (en) * | 2017-12-27 | 2025-04-29 | Gen Suzuki | Cardiosphere-derived cell sheet and methods of making and using the same |
WO2019152549A1 (en) | 2018-02-05 | 2019-08-08 | Cedars-Sinai Medical Center | Methods for therapeutic use of exosomes and y-rnas |
BR112021001798A2 (en) | 2018-07-31 | 2021-04-27 | F. Hoffmann-La Roche Ag | methods to assess atrial fibrillation, to diagnose heart failure, to aid in the assessment of atrial fibrillation, to predict stroke risk, and to improve prediction accuracy, uses, and kit |
DE102018125324A1 (en) * | 2018-10-12 | 2020-04-16 | Universität Rostock | Procedure for predicting an answer to disease therapy |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6492129B1 (en) * | 1998-12-14 | 2002-12-10 | University Of Miami | Fragments of connective tissue growth factor that induce extracellular matrix synthesis, collagen synthesis and/or myofibroblast differentiation |
-
2008
- 2008-11-03 WO PCT/US2008/012415 patent/WO2009061382A2/en active Application Filing
- 2008-11-03 US US12/740,887 patent/US8617877B2/en not_active Expired - Fee Related
-
2013
- 2013-12-19 US US14/134,301 patent/US20140274765A1/en not_active Abandoned
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6492129B1 (en) * | 1998-12-14 | 2002-12-10 | University Of Miami | Fragments of connective tissue growth factor that induce extracellular matrix synthesis, collagen synthesis and/or myofibroblast differentiation |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2016196366A1 (en) | 2015-05-29 | 2016-12-08 | The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services | Extension of replicative lifespan in diseases of premature aging using p53 isoforms |
Also Published As
Publication number | Publication date |
---|---|
US20110256105A1 (en) | 2011-10-20 |
WO2009061382A2 (en) | 2009-05-14 |
US8617877B2 (en) | 2013-12-31 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US8617877B2 (en) | Cardiac stem cell and myocyte secreted paracrine factors | |
Stastna et al. | Secreted proteins as a fundamental source for biomarker discovery | |
Stastna et al. | Identification and functionality of proteomes secreted by rat cardiac stem cells and neonatal cardiomyocytes | |
EP2211182A1 (en) | Method for the assessment of severity of liver cirrhosis | |
EP2209003A1 (en) | Means and methods for differentiating between fibrosis and cirrhosis | |
US20090326053A1 (en) | Diagnostic uses of follistatin-like 1 | |
CA2876922C (en) | Bag3 as biochemical serum and tissue marker | |
US20100151457A1 (en) | Detection of Unhealthy Cell and Uses Thereof | |
Xia et al. | Label-free quantitative proteomic analysis of right ventricular remodeling in infant Tetralogy of Fallot patients | |
KR102521638B1 (en) | A marker for diagnosis of nonalcoholic fatty liver disease | |
CN110678757B (en) | Method for diagnosing or monitoring renal function or diagnosing renal dysfunction | |
CN117969822A (en) | Galectin-3, NT-ProBNP and ST2 detection kit and preparation and application thereof | |
KR100896328B1 (en) | Proglenulin useful as a diagnostic marker for metabolic disease | |
JP5466822B2 (en) | Methods for determining inflammatory processes and pharmaceutical compositions for this treatment | |
US20210396767A1 (en) | Method for Predicting Prognosis of Ischemic Disease | |
RU2728097C1 (en) | Diagnostic technique of inflammation in epicardial adipose tissue in patients with ischemic heart disease and coronary atherosclerosis | |
US9182413B2 (en) | Methods and devices for diagnosing cardiac disorders | |
US20130078653A1 (en) | Antibodies, compositions, and assays for detection of cardiac disease | |
Fu et al. | Cxcl9 modulates aging associated microvascular metabolic and angiogenic dysfunctions in subcutaneous adipose tissue | |
KUROBE et al. | Reevaluation of serum pancreatic secretory trypsin inhibitor (PSTI) as a diagnostic marker with monoclonal antibody-based two-site enzyme immunoassay | |
US20120282618A1 (en) | Methods and assays for risk prediction, diagnosis, and analysis of myocardial infarction, heart failure and reduced cardiac function | |
ES2987148A1 (en) | IN VITRO METHOD FOR THE PROGNOSIS AND/OR DIAGNOSIS OF DIABETES MELLITUS IN PATIENTS WITH AORTIC STENOSIS AND/OR FOR THE PROGNOSIS AND/OR DIAGNOSIS OF AORTIC STENOSIS IN PATIENTS WITH DIABETES MELLITUS (Machine-translation by Google Translate, not legally binding) | |
Zhang et al. | Mechanistic studies of proteomic biomarkers | |
BR112021003539A2 (en) | method for diagnosing a liver disease |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
STCB | Information on status: application discontinuation |
Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION |
|
AS | Assignment |
Owner name: NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR, MARYLAND Free format text: CONFIRMATORY LICENSE;ASSIGNOR:THE SCRIPPS RESEARCH INSTITUTE;REEL/FRAME:052166/0291 Effective date: 20200311 |
|
AS | Assignment |
Owner name: NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR, MARYLAND Free format text: CONFIRMATORY LICENSE;ASSIGNOR:THE JOHNS HOPKINS UNIVERSITY;REEL/FRAME:052229/0380 Effective date: 20200311 |