You are on page 1of 31

Rejuvenation Research

Rejuvenation Research: http://mc.manuscriptcentral.com/rejuvenationresearch

Role of trauma cytokines and erythropoietin


Fo
and their therapeutic potential for acute and chronic
wounds
r

Journal: Rejuvenation Research


Pe

Manuscript ID: REJ-2010-1050.R1

Manuscript Type: Clinical Articles


er

Date Submitted by the


02-Jul-2010
Author:

Complete List of Authors: Bader, Augustinus; Biotechnological Biomedical Center, Department


Re

of Cell Techniques and Stem Cell Biology, University of Leipzig


Lorenz, Katrin; Biotechnological Biomedical Center, Department of
Cell Techniques and Stem Cell Biology, University of Leipzig
Richter, Anja; Biotechnological Biomedical Center, Department of
Cell Techniques and Stem Cell Biology, University of Leipzig
vi

Scheffler, Katja; Biotechnological Biomedical Center, Department of


Cell Techniques and Stem Cell Biology, University of Leipzig
Kern, Larissa; Biotechnological Biomedical Center, Department of
ew

Cell Techniques and Stem Cell Biology, University of Leipzig


Ebert, Sabine; Biotechnological Biomedical Center, Department of
Cell Techniques and Stem Cell Biology, University of Leipzig
Giri, shibashish; Biotechnological Biomedical Center, Department of
Cell Techniques and Stem Cell Biology, University of Leipzig
Behrens, Maria; Medical Writing Experts
Dornseifer, Ulf; Klinikum Bogenhausen, Department of Plastic,
Reconstructive, Hand and Burn Surgery
Macchiarini, Paolo; Hospital Clinico de Barcelona, Barcelona,
4Department of General Thoracic Surgery
Machens, Hans-Gunther; Klinikum Rechts der Isar, Technische
Universität München, 5Department of Plastic and Hand Surgery

Regeneration, Skin Aging, Stem Cells, Quality of Life, Growth


Keyword:
Factors

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 1 of 29 Rejuvenation Research

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
Fo
19
20
21
22
r
23
24
Pe

25
26
27
28
er

29
30
31
32
Re

33
34
35
36
vi

37
38
39
ew

40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 2 of 29

1
2
3 The interactive role of trauma cytokines and erythropoietin
4
5
6 and their therapeutic potential for acute and chronic wounds
7
8
9
10
Augustinus Bader1, Katrin Lorenz1, Anja Richter1, Katja Scheffler1, Larissa Kern1, Sabine
11
12
13 Ebert1, Shibashish Giri1, Maria Behrens2, Ulf Dornseifer3, Paolo Macchiarini4, Hans-Günther
14
15 Machens5
16
17 1
18
University of Leipzig, Centre for Biotechnology and Biomedicine, Department of Applied
19
20 Stem Cell Biology and Cell Techniques, Germany
Fo

21
22 2
Medical Writing Experts, Langwedel, Germany
23
24 3
Klinikum Bogenhausen, Zentrum für Schwerbrandverletzte, München
rP

25
26
4
27 Hospital Clinico de Barcelona, Dept. of General Thoracic Surgery
28
29
ee

5
Klinik für Plastische Chirurgie, Klinikum Rechts der Isar, Technische Universität München,
30
31
32 Germany
rR

33
34
35
36
37
Abstract:
ev

38
39
40 If controllable, stem cell activation following injury has the therapeutic potential for
41
iew

42
43
supporting regeneration in acute or chronic wounds. Human dermally derived stem cells
44
45 (FmSCs) were exposed to the cytokines IL-6, IL-1ß and TNF-α in the presence of
46
47 erythropoietin. Cells were cultured under ischemic conditions and phenotypically
48
49
50 characterized using flow cytometry. Topical EPO application was performed in three
51
52 independent clinical wound healing attemps. The FmSCs expressed the receptor for
53
54 erythropoietin (EPO). EPO had a strong inhibitory effect on FmSC growth in the absence of
55
56
57 IL-6 and TNF-α. With IL-6, the EPO effects were reversed to that of growth stimulation.
58
59 TNF-α had the strongest stimulatory effect. In contrast, IL-1ß had an inhibitory effect.
60
Topically applied EPO considerably enhanced wound healing and improved wound

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 3 of 29 Rejuvenation Research

1
2
3 conditions of acute and chronic wounds. Site specificity of stem cell activation is mediated by
4
5
6 IL-6 and TNF-α. In trauma, EPO ceases its inhibitory role and reverts to a clinically relevant
7
8 boosting function. EPO may be an important therapeutic tool for the topical treatment of acute
9
10
and chronic wounds.
11
12
13
14
15 INTRODUCTION
16
17
18
The human body has command of a tool box that allows it to appropriately react to injury with
19
20 an amazing site specificity to achieve a regenerative response. It has been unclear how local
Fo

21
22 stem cells are “awakened” in such instances of need. If this mechanism was better understood,
23
24
a fundamental therapeutic strategy that would allow site-specific stem cell activation in any
rP

25
26
27 area of the human body could be developed. Site specificity of tissue regeneration has been
28
29
ee

taken for granted as a “normal” process, but their underlying mechanisms, relevance for stem
30
31
32 cell-based responses and therapeutic potential have not yet been understood. Frequently, this
rR

33
34 innate capacity is not sufficient to lead to full restoration, resulting in so-called “nonhealing”
35
36
wounds.
37
ev

38
39 It is well-known that local trauma leads to the release of inflammatory cytokines, including
40
41 IL-6, IL-1β and TNF-α.1 Mesenchymal stem cells (MSCs), isolated from many human tissues,
iew

42
43
44
such as bone marrow, adipose tissue, the adult liver, peripheral blood, amniotic fluid, the
45
46 bronchial lung, the articular synovium and other fetal tissues, are a cell population that
47
48 possesses a fibroblastic-like morphology, limited but long-term viability, self-renewal
49
50
51 capacity and multilineage potential.2,3 They are characterized by similar surface antigen
52
53 expression patterns for CD14(-), CD31(-), CD34(-), CD45(-), CD71(+), CD73/SH3-SH4(+),
54
55 CD90/Thy-1(+), CD105/SH2(+), CD133(-) and CD166/ALCAM(+)4,5. MSCs can
56
57
58 differentiate into adipogenic, osteogenic, myogenic, chondrogenic and neurogenic cell types.
59
4-8
60

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 4 of 29

1
2
3 In an effort to identify new sources of mesenchymal stem cells, dermal rodent fibroblast cell
4
5
6 lines were examined for their mesenchymal potential.9-11 Publications by Toma et al. 9
and
7
10
8 Crigler et al. suggest that the adult mammalian dermis contains tissue-derived stem cells
9
10
and that these fibroblastic mesenchymal stem cells are more plastic than previously
11
12
13 appreciated. Dermis-derived fibroblastic mesenchymal stem cells have been used for
14
15 therapeutic applications such as transplantation to support bone formation.12-14 Toma et al.9
16
17
18
demonstrated the mesenchymal plasticity of primary human dermal fibroblasts in vitro with
19
20 different approaches regarding characterization and applications.15-18 Zuk et al. analyzed the
Fo

21
22 phenotypic characteristics of these fibroblasts and noted that their phenotype seems to be
23
24
similar to that of adult-derived stem cells (ADSCs).5,16,18
rP

25
26
27
28
29
ee

To continue these studies, we examined whether unselected human dermis fibroblastic


30
31
32 mesenchymal cells possess stem cell-like characteristics and are phenotypically similar to
rR

33
34 ADSCs. Bone marrow-derived MSCs (bmMSCs) have been shown to support the wound
35
36
healing of chronic skin wounds. Badiavas and Falanga (2003) demonstrated that chronic skin
37
ev

38
39 ulcers of patients with arterial and venous insufficiency were healed with complete wound
40
41 closure and less scar formation when treated with bmMSC-seeded grafts.21
iew

42
43
44
45
46 To characterize FmSCs relative to ADSCs, we analyzed the cytoskeletons and compositions
47
48 of the extracellular matrix as well as the mesenchymal phenotypes and differentiation
49
50
51 properties of both. The obtained data show for the first time that primary human dermal
52
53 fibroblasts in vitro share common characteristics with ADSCs, such as phenotype and
54
55 differentiation potential. Our results demonstrated that FmSCs fulfill the three main
56
57
58 characteristics of MSCs: they express all MSC-related surface antigens homogenously; their
59
60 cytoskeleton and matrix compositions are quite similar to that of MSCs; and they differentiate

along the adipogenic and osteogenic cell lineages.20

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 5 of 29 Rejuvenation Research

1
2
3
4
5
6 Both conditions, however, do not sufficiently explain the mechanism of endogenous stem cell
7
8 activation in the case of injury alone.
9
10
We therefore developed an in vitro model of trauma conditions by investigating the role of IL-
11
12
13 6, IL-1β and TNF-α on human skin stem cells, identified a receptor for Erythropoietin (EPO)
14
15 in these cells and investigated the role of EPO with and without the presence of the trauma
16
17
18
cytokines. The knowledge obtained from these studies was then transferred to three
19
20 independent and specific clinical cases: EPO was topically applied to a split-thickness skin
Fo

21
22 graft donor site, a pressure and a vascular ulcer.
23
24
rP

25
26
27 EPO has been used in clinical practice for a wide range of diseases22, obtaining systemic
28
29
ee

application via subcutaneous, intramuscular or intravenous route.23-28 A topical administration


30
31
32 to stem cells at the site of the wound injury would allow a more direct stimulatory response
rR

33
34 and would work only if an appropriate interference with site-specific mechanistic responses
35
36
occurred. The elucidation of such mechanisms and the development of a therapeutic potential
37
ev

38
39 has been the scope and success of this study.
40
41
iew

42
43
44 METHODS
45
46
47
48 Cell isolation
49
50
51 Human juvenile foreskin samples were obtained from four-year-old patients undergoing
52
53 circumcision after written consent was obtained. This study was approved by the ethics
54
55 commission of Leipzig University and was conducted in accordance with the Declaration of
56
57
58 Helsinki protocols.
59
60 Epidermal and dermal tissue were isolated by mechanical and enzymatic digestion, as

previously described by Ponec et al29. After removing the epidermis from the dermis, the

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 6 of 29

1
2
3 tissue was cut into small pieces and washed three times with sterile phosphate-buffered saline
4
5
6 (PBS) at room temperature. The pieces were then incubated with 0.075% collagenase type A
7
8 (Roche Diagnostics, Mannheim, Germany) for 12 h at 37°C with gentle agitation.
9
10
For FmSC suspensions, the enzymatic reaction was inactivated with DMEM/10% FBS
11
12
13 (Gibco/Invitrogen, Karlsruhe, Germany) and filtered through a 70-µm mesh. This cell
14
15 suspension was centrifuged at 600 x g for 5 min. The cell pellet was then gently resuspended
16
17
18
in DMEM/10% FBS, filtered through a 70-µm mesh and plated in conventional T75 tissue
19
20 culture flasks (BD Falcon, Heidelberg, Germany). Cells were cultured in DMEM
Fo

21
22 supplemented with 10% FBS, GlutaMAX-I, 4.5 g/L glucose and pyruvate (Gibco/Invitrogen).
23
24
rP

25
26
27 Cell proliferation
28
29
ee

FmSCs were seeded in six-well plates at passage 4. After one day of cultivation in
30
31
32 DMEM/10% FBS, the cells attached and adapted to start proliferation. On the following day,
rR

33
34 the cells were cultured with the same medium but without FBS to minimize serum-induced
35
36
effects. The next day, cytokine stimulation of the cells was initiated with cultivation in
37
ev

38
39 DMEM/10% FBS supplemented with or without 10 ng/mL EPO in combination with 10
40
41
iew

ng/mL IL-6, 10 ng/mL IL-1β or 10 ng/mL TNF-α. Controls were also performed with each
42
43
44
cytokine alone. At days 3, 5, 7 and 11, cells were trypsinized and viable cell numbers were
45
46 counted in a hemocytometer by trypan blue staining. All experiments were done in triplicate,
47
48 with three independent sets of patient materials.
49
50
51
52
53 Cell immunophenotyping
54
55 Immunophenotyping of FmSCs was performed as described previously by Lorenz et al19. The
56
57
58 following labeling reagents were used: fluorescein isothiocyanate (FITC)- or phycoerythrin
59
60 (PE)-conjugated mouse antibodies, anti-human CD31 (Biozol Diagnostica, Munich,

Germany), anti-human CD45 (Sigma-Aldrich, Seelze, Germany), anti-human CD90 and anti-

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 7 of 29 Rejuvenation Research

1
2
3 human CD105 (BD Biosciences, Heidelberg, Germany) and anti-human CD166 (Acris
4
5
6 Antibodies, Hiddenhausen, Germany). The monoclonal antibody mouse anti-human CD73
7
8 (BD Biosience) was unlabeled and combined with a secondary PE-labeled goat anti-mouse
9
10
antibody (Sigma-Aldrich). Incubation and flow cytometry analyses were performed according
11
12
13 to conventional techniques29. Isotype controls were equally concentrated, labeled or
14
15 unlabeled. The stained cells were analyzed on a FACS Calibur (BD Bioscience) using
16
17
18
CellQuest Pro (BD Bioscience). Fluorescence intensities were determined by flow cytometry
19
20 in a minimum of 1x104 cells.
Fo

21
22
23
24
Growth curves
rP

25
26
27 To record a growth curve, three individual tests were performed, each in triplicate. Cell
28
29
ee

counting was performed at day 0, 1, 3, 5 and 7 by trypsinization and trypan blue staining
30
31
32 using a Neubauer’s chamber. The cells were seeded at passage 6 with a density of 20,000
rR

33
34 cells/well (9.6 cm2) in a six-well plate (Falcon) two days before stimulation (day 0). The
35
36
exchange of media one day before stimulation from 10% FBS-containing DMEM to serum-
37
ev

38
39 free DMEM ensured the attachment of the cells during the last 24 hours and a minimal protein
40
41
iew

background from the FBS. The stimulation was made with or without the presence of IL-6,
42
43
44
IL-1β and TNF-α by adding EPO (NeoRecormon, Roche) and/or the cytokines (all
45
46 RELIAtech) to the media and performing a full media exchange. At days 3 and 5, half the
47
48 volume of the media was changed with media containing the initial concentrations of EPO
49
50
51 and/or cytokine. The cell scores of each sample and the average of all nine samples were
52
53 calculated, including the standard deviation. To compare the different growth curves, a
54
55 Student’s t-test was used to test the significance of the differences between the breakpoints.
56
57
58 For stimulation with cytokines, a concentration-dependent pretest was performed to identify
59
60 the minimum concentration needed for successful stimulation; this was determined to be 10

ng/mL.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 8 of 29

1
2
3
4
5
6 Clinical cases
7
8 In three highly different clinical situations, topical EPO application was used to support
9
10
wound healing. All patients provided informed consent based on the guidelines of the local
11
12
13 ethical committee and the national legal requirements in Germany. Topical treatment was
14
15 performed using a mixture of 3,000 IE erythropoietin-ß (NeoRecormon®, F. Hoffmann-La
16
17
18
Roche AG, Basel, Schweiz) and 20 g hydrogel (Varihesive®, ConvaTec, NJ, USA). In patient
19
20 A, the mixture was topically applied to a 0.3-mm deep split-thickness skin graft donor sites
Fo

21
22 measuring 8 x 24 cm directly after skin harvesting. The donor site at the thigh was
23
24
rP

25 subsequently closed with a polyurethane dressing (OpSite, Smith&Nephew, London, UK).


26
27 After three and six days, the mixture was again applied by puncturing the polyurethane film,
28
29
ee

30
which remained on the wound. At day 7, the film dressing was removed to evaluate the
31
32 reepithelialization. Another donor site of equal depth and dimension at the contralateral leg of
rR

33
34 patient A was treated similar but the mixture was replaced by hydrogel alone - without EPO.
35
36
37 Again the dressing rest in place for seven days followed by dressing removal and wound
ev

38
39 assessment. The standardized wound management provided an ideal comparability of both
40
41
iew

similar wounds.
42
43
44 Patient B had a non-healing pressure sore at the heel following urosepsis and Patient C had a
45
46 non-healing vascular ulcer. In both patients the wounds were surgically debrided and treated
47
48
in the same manner, using the same mixture of EPO and hydrogel. Moist wound management
49
50
51 was provided by covering both wounds with Varihesive® (Convatec, Skillman, NJ, USA). A
52
53 total of five dressing changes with new EPO applications were performed in both patients to
54
55
56
prepare the wound for skin grafting. The poor general condition of patient B and C did not
57
58 allow extensive reconstructional procedures. Therefore, the aim of local EPO application was
59
60 to prepare the wound bed in a manner that enables subsequent successful skin grafting. And to

avoid lower leg amputation by a minor surgical procedure.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 9 of 29 Rejuvenation Research

1
2
3 RESULTS
4
5
6 Sequencing the EPO receptor in human FmSCs
7
8 Cells were characterized for their expression of CD31, CD45, CD73, CD90, CD105 and
9
10
CD166. The expression of CD34, CD71 and CD133 was also examined. Fig. 1a shows the
11
12
13 mRNA expression profile of the EPO receptor in FmSCs. Sequencing of the PCR product for
14
15 the mRNA of the EPO receptor showed 90-98% sequence homology.
16
17
18
Figure 1
19
20 Figure 2
Fo

21
22 Figure 3
23
24
rP

25
26
27
28
29
ee

30
31
32 Switch from inhibitory to stimulatory effects of EPO on stem cell proliferation
rR

33
34 Cells were cultured from human biopsies and grown to the 4th passage in vitro. Stimulation of
35
36
EPO alone in the absence of any cytokines showed an inhibitory effect on stem cell growth
37
ev

38
39 (Fig. 1b). Cells under control conditions grew up to 1.545 million cells. In contrast, we
40
41
iew

observed a dramatic decrease in cell proliferation when the FmSCs were stimulated with
42
43
44
EPO. Cell proliferation was minimized by 32%, to a total cell number of 1.053 million cells.
45
46 IL-6 stimulation of FmSCs also resulted in decreased growth activity relative to the control
47
48 cells, but this was reversed in the presence of EPO (Fig. 1c). This indicates both an inhibitory
49
50
51 role of EPO (Fig. 1b) in the absence of cytokines or in the late phase of trauma and a
52
53 supportive, boosting activity of EPO in the presence of IL-6 (Fig. 1c). TNF-α was a strong
54
55 stimulator of FmSC proliferation, and the presence of EPO did not influence this. Cell
56
57
58 proliferation was elevated most with TNF-α. IL-1β had an inhibitory effect on the
59
60 proliferation of the stem cells compared to controls cells, both with and without EPO.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 10 of 29

1
2
3 Effect of stimulation on the expression of stem cell markers
4
5
6 To characterize the FmSCs, surface antigens were analyzed by flow cytometry. FmSCs
7
8 homogenously expressed CD90, CD73, CD105 and CD166. (Fig. 2, Fig. 3) In contrast,
9
10
expression of endothelial cell surface markers such as CD31 was not detected. In addition,
11
12
13 hematopoietic cell subpopulations positive for surface antigens such as CD45, CD14 and
14
15 CD133 were not observed.
16
17
18
During cultivation and stimulation of FmSCs with inflammatory cytokines in combination
19
20 with EPO, we did not detect any changes in the surface antigen expression of MSC markers.
Fo

21
22 Except when cultivating FmSCs with IL-6 in combination with EPO, we found changes in the
23
24
expression of CD90. This suggests that the unselected stem cell population was stimulated
rP

25
26
27 differently, and thus two different CD90-expressing cell populations were detected. (Fig. 3)
28
29
ee

30
31
32 In vivo experiments
rR

33
34 The more complex in vivo situation is characterized by an intricate interplay of cytokine
35
36
profiles, consisting of mixtures and time-sequence variations with respect to availability.
37
ev

38
39
40
41
iew

In Patient A, complete and stable reepithelialization of the split-thickness skin graft donor
42
43
44
site, topically treated with EPO, was achieved seven days after the operation. The wound
45
46 surface was closed, dry and clearly looked pale. (Fig.4a) In contrast, the donor site at the
47
48 contralateral thigh that was treated without EPO showed incomplete reepithelialization at this
49
50
51 time point, indicated by secretion and a dark-red wound surface. (Fig.4a)
52
53
54
55 In Patient B, sufficient granulation tissue formation was obtained after five local treatment
56
57
58 sessions with EPO, which provided an highly vascularized wound bed for sucessful split-
59
60 thickness skin grafting. The ideal prepared wound bed enabled salvage of the limb without

extensive reconstructive procedures (Fig. 4b).

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 11 of 29 Rejuvenation Research

1
2
3
4
5
6 In Patient C, improved healing and sufficient granulation tissue formation was achieved
7
8 following five local applications of EPO. (Fig.4c) After subsequent skin grafting at days 21
9
10
and 56, the wound healed well and has remained stable for more than 12 months (Fig. 4c).
11
12
13
14
15 DISCUSSION
16
17
18
19
20 Erythropoietin is a type I cytokine that was approved by the US Food and Drug
Fo

21
22 Administration (FDA) in 1989 for the treatment of the anemia of end-stage renal disease.
23
24
Thereafter, erythropoietin has been used for the treatment of a diverse range of diseases,
rP

25
26
27 including cancer28,31,32, heart care erythropoiesis33-37, malaria38, ischemic and degenerative
28
29
ee

damage of neurons40, retinopathy40,41 and diabetic retinopathy42-45. EPO plays a crucial role in
30
31
32 the process of endochondral ossification in bone repair in mice via EPO-receptor expression46.
rR

33
34 Gough (2008) 47 supported the concept that understanding the EPO receptors by which EPO
35
36
signaling contributes to organ development provides information on the differentiation of
37
ev

38 48
39 erythrocytes. Interestingly, Foster et al. (2004) found increased EPO-R protein levels in
40
41
iew

dynamically growing canine lungs after pneumonectomy, suggesting a paracrine role for EPO
42
43
44
signaling in lung growth and remodeling. This hypothesis may be applicable to other types of
45
46 organ repair since EPO and EPO-R are expressed in several organs (e.g., kidney, brain, heart,
47
48 muscle and endothelial cells) 49. However, it is now known that EPO and EPO-R are local
49
50
51 products in a wide range of cells that specifically protect other cells from potentially cytotoxic
52
53 events and metabolic stress.
54
55
56
57 50
58 Adding to the evidence of Bodo et al. (2007) that normal human skin expresses EPO and
59
60 functional EPO-R, our study showed that skin stem cells specifically are the responsive

elements in normal skin containing the receptor for EPO and thus show a special readiness for

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 12 of 29

1
2
3 action in the case of traumatic skin injuries. In the case of injury and ischemic trauma,
4
5
6 cytokines IL-6, IL-1β and TNF-α are alerted. We demonstrated that the trauma cytokine IL-6
7
8 and EPO synergistically up-regulated stem cell growth in the case of hypoxic skin conditions.
9
10
In contrast, without trauma, EPO exerted an inhibitory effect on in vitro skin stem cells. This
11
12
13 effect reverted to stimulation in the presence of IL-6 and TNF- α. Only IL-1β maintained its
14
15 inhibitory function with or without EPO. Neither the trauma cytokines nor EPO grossly
16
17
18
changed the phenotype of the fibroblast precursor cells.
19
20 Our findings agree with the observations of Paus et al. (2009)51 that the oxygen sensing skin
Fo

21
22 response is mediated by skin EPO. In situations of low oxygen, skin EPO and IL-6 are
23
24
alarmed to react to the site-specific injury. This finding compliments the relevance of our data
rP

25
26
27 as a physiological and potentially highly relevant therapeutic strategy for endogenous stem
28
29
ee

cell activation in the case of trauma.


30
31
32
rR

33
34 In the human scalp, it was shown that hair follicles expressed EPO at the mRNA and protein
35
36
levels, up-regulated EPO transcription under hypoxic conditions and expressed transcripts of
37
ev

38
39 EPO-R and the EPO stimulatory transcriptional cofactor hypoxia-inducible factor-1α. These
40
41
iew

findings are in line with recent research results that showed that hair follicle-derived
42
43
44
keratinocytes were a major cell source for reepithelialization during wound healing52 and that
45
46 the hair follicle connective tissue sheath was a source of granulation tissue formation53.
47
48 Boutin et al. (2008)54 revealed the EPO-connected oxygen sensing functions of the skin and
49
50
51 elucidated how mammalian cells adapt to low oxygen levels by recruiting the skin as a central
52
53 coordinator of the systemic response to hypoxia. Hair follicles are able to detect insufficient
54
55 oxygen levels, a crucial mechanism of the extremely fast renewing and proliferating cell
56
57
58 population, to regulate its metabolic balance. Using transgenic mice studies, Kochling (1998)
59
60 revealed that the hypoxia response elements are located upstream (between 9.5 and 14 kb) of

the EPO gene in the kidney and downstream (within 0.7 kb) of the EPO gene in the liver. It

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 13 of 29 Rejuvenation Research

1
2
3 has been shown that the circulating levels of EPO may increase up to 1,000-fold in response
4
5
6 to hypoxia in the kidney53.
7
8
9
10
In the absence of trauma cytokines, EPO down-regulates the proliferation of skin stem cells in
11
12
13 vitro. In the case of traumatic skin hypoxia, IL-6 and TNF-α activate stem cells. Specifically,
14
15 in the presence of IL-6, the inhibitory role of EPO is reversed to increase stem cell
16
17
18
proliferation. This represents an adequate response to a pathophysiological need. The
19
20 stimulatory effects of TNF-α are not diminished by the previously inhibitory role of EPO.
Fo

21
22 Among the trauma cytokines studied here, only IL-1β also exhibited an inhibitory function
23
24
that did not interfere with the original inhibitory role of EPO. In vivo, we observed the net
rP

25
26
27 effect of cytokine and EPO stimulation in acute and chronic wound types. In both cases, the
28
29
ee

regenerative response was boosted qualitatively and quantitatively. By these mechanisms, the
30
31
32 skin trauma EPO system switches from its inhibitory function to a supportive role for
rR

33
34 boosting skin regeneration. The inflammatory activity of the wound itself represents a
35
36
permissive situation for the boosting activity of EPO, which seems to reduce the healing time
37
ev

38
39 at split-skin graft donor sites from 10 to 7 days. In the previously non-healing wounds, EPO
40
41
iew

assumes an enabling role that shifts the balance from non-healing to healing and triggers the
42
43
44
formation of granulation tissue. The expression of EPO-R on the stem cells suggests that this
45
46 could be a normal role of EPO that permits the human body to recover from site-specific
47
48 tissue damage or injury in any area of the human body. This mode of action has been
49
50
51 demonstrated clinically by the clearly accelerated reepithelialization of the EPO treated donor
52
53 site in direct comparison to the non-EPO treatet donor site at the same patient.
54
55
56
57
58 Non-healing chronic wounds are at the opposite end of the spectrum of acute wound-healing
59
60 mechanisms, progressing toward healing at a different rate. In the case of diabetic patients,

this represents a critical situation for surgical practice, as approximately 22 million diabetic

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 14 of 29

1
2
3 patients suffer from chronic wounds57,58, with many of them suffering from non-healing
4
5
6 chronic wounds59. There are approximately 5.2 million pressure ulcers and 7.6 million venous
7
8 ulcers in the world that require treatment every year.
9
10
11
12
13 Fibroblasts form granulation tissue via hyper-proliferation. This leads to a normal process of
14
15 not only cellular rebuilding of lost dermal tissue but also reconstitution of the physical barrier
16
17
18
of the basal lamina and the scaffold for revascularization. Large-area wounds do not heal
19
20 within a short time, so the risk of infection and dehydration rises dramatically. Our results
Fo

21
22 suggest that we can completely alter the wound-healing landscape and have a major impact on
23
24
the care of both acute and chronic wounds. This study provides mechanistic evidence to
rP

25
26
27 support the hypothesis that this novel treatment modality physically modifies the wound
28
29
ee

microenvironment and thereby promotes wound healing in clinical relevant manner.


30
31
32
rR

33
34 In a few clinical applications, fibroblasts were used to treat diabetic or venous ulcers 60-62, but
35
36
this methodology remains controversial. We report a mechanism explaining how endogenous
37
ev

38
39 stem cells can be activated locally at the site of a severe wound without necessitating the
40
41
iew

transplantation of exogenous cells. All clinical cases, although diverse in their


42
43
44
pathophysiology, were dramatic successes with respect to their respective healing responses.
45
46
47
48 Brines & Cerami described63 that EPO is locally produced in the immediate surrounding area
49
50
51 of a tissue injury to counteract the destructive effects of cytokines such as TNF-α by
52
53 preventing cell apoptosis, thus the development of secondary, proinflammatory cytokine-
54
55 induced injury can be reduced. However, a delicate balance in tissue injury exists between
56
57
58 EPO and proinflammatory cytokines such as TNF-α. Therefore, compensatory EPO
59
60 production by nearby tissue balances the effects of inflammatory mediators and prevents the

further spread of damage.63 Hamed et al. reported that treatment with topical EPO improves

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 15 of 29 Rejuvenation Research

1
2
3 the defect repair of excised wounds in diabetic rats.64 They suggested that vascular VEGF-
4
5
6 induced angiogenesis, enhanced collagen deposition and reduced apoptosis in the diabetic
7
8 wound bed are among the mechanisms that underlie the effects of topical EPO. This work by
9
10
Hamed et al. 64 was the first to investigate the use of topical EPO treatment for wound healing.
11
12
13 However we are the first of topical EPO treatment for acute and chronic wounds patients.
14
15 Although other cytokines such as tumor growth factor-β, monocyte chemoattractant protein-1
16
17
18
and colony-stimulating factor-1 are released from the invading inflammatory cells to the
19
20 wound bed upon skin injury and in chronic wounds65, we selected these cytokines (IL-6 ,
Fo

21
22 TNF- α and IL-1ß ) because these are leading cytokines which is associated with organ
23
24
trauma injury including chronic wound.66
rP

25
26
27
28
29
ee

Despite the beneficial effect on wound repair, one has to assume that doses of EPO are rather
30
31 67
32 high within the defect wound and low systemically. Rezaeian et al demonstrated that a
rR

33
34 triplicate intraperitoneal dose of 500IU EPO/kg bw over 48 hours did not influence RBC
35
36 68
count and Hematocrit, whereas Galeano et al observed a significant increase in RBC count
37
ev

38
39 and hemoglobin after 12 days of daily subcutaneous administration of 400IU EPO / kg bw. In
40
41
iew

compared to this, the EPO concentration of our present clinical study is 50 U (one time) by
42
43
44
topical application of the hydrogel containing EPO in the patients. This concentration (50U) is
45
46 75 times less than other existing dose of various other experimental or clinical models. Using
47
48 this concentration, we are conducting multicenter clinical trials for actute and choric wound
49
50
51 pateints. Everytime fresh hydrogel is prepared and half life of EPO is 48 hours and stable in
52
53 gel up to 12 weeks. There is no systemic effect of treated patients which is main advantages
54
55 of this topical application. We measured red blood cell (RBC) count and haemoglobin,
56
57
58 leukocyte and platelet count of the patients before and after EPO treatment but there were no
59
60 any difference.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 16 of 29

1
2
3 Our present investigation may provide a standard supplemental therapy for reducing the
4
5
6 mortality and morbidity associated with chronic wounds, especially in the elderly, the
7
8 disabled and those with diabetes. Especially large-area burn injuries, where the wound closure
9
10
is a race against time, may benefit from the healing accelerating characteristics of EPO. Not
11
12
13 only the burned, debrided and grafted areas but also the skin graft donor sites have to heal in a
14
15 limited time frame. Frequently, donor sites do not rejuvenate for reharvesting as fast as
16
17
18
needed, resulting in graft deficiencies that may lead to further extensive complications with
19
20 fatal outcomes. The targeted clinical areas will improve in the assistance toward accelerating
Fo

21
22 regeneration of acute and chronic wounds, and endogenous stem cell activation may reduce
23
24
the need for skin grafting of burns.
rP

25
26
27
28
29
ee

References
30
31
32
rR

33
34 1. Singer AJ, Clark RA. Cutaneous wound healing. N Engl J Med 1999;341:738-46.
35
36
2. Müller T, Ander L, Kolf K, Woitalla D, Muhlack S. Comparison of 200 mg retarded
37
ev

38
39 release levodopa/carbidopa - with 150 mg levodopa/carbidopa/entacapone application:
40
41
iew

pharmacokinetics and efficacy in patients with Parkinson's disease. J Neural Transm


42
43
44
2007;114:1457-1462.
45
46 3. Bobis S, Jarocha D, Majka M. Mesenchymal stem cells: characteristics and clinical
47
48 applications. Folia Histochem Cytobiol 2006;44:215-230.
49
50
51 4. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA,
52
53 Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal
54
55 stem cells. Science 1999;284:143-147.
56
57
58 5. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK,
59
60 Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol

Biol Cell 2002;13:4279-4295.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 17 of 29 Rejuvenation Research

1
2
3 6. Hauner H, Schmid P, Pfeiffer EF. Glucocorticoids and insulin promote the differentiation
4
5
6 of human adipocyte precursor cells into fat cells. J Clin Endocrinol Metab 1987;64:832-835.
7
8 7. Grigoriadis AE, Heersche JN, Aubin JE. Differentiation of muscle, fat, cartilage, and bone
9
10
from progenitor cells present in a bone-derived clonal cell population: effect of
11
12
13 dexamethasone. J Cell Biol 1988;106:2139-2151.
14
15 8. Wakitani S, Saito T, Caplan AI. Myogenic cells derived from rat bone marrow
16
17
18
mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve 1995;18: 1417-1426.
19
20 9. Toma JG, Akhavan M, Fernandes KJ, Barnabe-Heider F, Sadikot A, Kaplan DR, Miller
Fo

21
22 FD. Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell
23
24
Biol 2001;3:778-784.
rP

25
26
27 10. Crigler L, Kazhanie A, Yoon TJ, Zakhari J, Anders J, Taylor B, Virador VM. Isolation of
28
29
ee

a mesenchymal cell population from murine dermis that contains progenitors of multiple cell
30
31
32 lineages. Faseb J 2007;21:2050-2063.
rR

33
34 11. Toma JG, Akhavan M, Fernandes KJ, Barnabe-Heider F, Sadikot A, Kaplan DR, Miller
35
36
FD. Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell
37
ev

38
39 Biol 2001;3:778-784.
40
41
iew

42
43
44
12. French MM, Rose S, Canseco J, Athanasiou KA. Chondrogenic differentiation of adult
45
46 dermal fibroblasts. Ann Biomed Eng 2004;32:50-56.
47
48 13. Hirata K, Tsukazaki T, Kadowaki A, Furukawa K, Shibata Y, Moriishi T, Okubo Y,
49
50
51 Bessho K, Komori T, Mizuno A, Yamaguchi A. Transplantation of skin fibroblasts expressing
52
53 BMP-2 promotes bone repair more effectively than those expressing Runx2. Bone
54
55 200;32:502-512.
56
57
58 14. Krebsbach PH, Gu K, Franceschi RT, Rutherford RB. Gene therapy-directed
59
60 osteogenesis: BMP-7-transduced human fibroblasts form bone in vivo. HumGene Ther.

2000;11:1201-1210.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 18 of 29

1
2
3 15. Rutherford RB, Moalli M, Franseschi RT, Wang D, Gu K, Krebsbach PH. Bone
4
5
6 morphogenetic protein-transduced human fibroblasts convert to osteoblasts and form bone in
7
8 vivo. Tissue Eng 2002;8:441-452.
9
10
16. Lysy PA, Smets F, Sibille C, Najimi M, Sokal EM. Human skin fibroblasts:From
11
12
13 mesodermal to hepatocyte-like differentiation. Hepatology 2007;46:1574-1585.
14
15 17. Chen FG, Zhang WJ, Bi D, Liu W, Wei X, Chen FF, Zhu L, CuiL, Cao Y. Clonal analysis
16
17
18
of nestin(-) vimentin(+) multipotent fibroblasts isolated from human dermis. J Cell Sci
19
20 2007;120:2875-2883.
Fo

21
22 18. Toma, JG, McKenzie IA, Bagli D, Miller FD. Isolation and characterization of
23
24
multipotent skin-derived precursors from human skin. Stem Cells 2005;23:727-737.
rP

25
26
27 19. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP,
28
29
ee

Hedrick MH. Multilineage cells from human adipose tissue: implications for cell-based
30
31
32 therapies. Tissue Eng 2001;7:211-228.
rR

33
34 20. Lorenz K, Sicker M, Schmelzer E, Rupf T, Salvetter J, Schulz-Siegmund M, Bader A.
35
36
Multilineage differentiation potential of human dermal skin-derived fibroblasts. Exp Dermatol
37
ev

38
39 2008b;17:925-932.
40
41
iew

42
43
44
21. Badiavas EV, Falanga V. Treatment of chronic wounds with bone marrowderived cells.
45
46 Arch Dermatol 2003;139:510-516.
47
48 22. Noguchi CT, Wang L, Rogers HM, Teng R, Jia Y. Survival and proliferative roles of
49
50
51 erythropoietin beyond the erythroid lineage. Expert Reviews in Molecular Medicine
52
53 2008;1:36.
54
55 23. Amarguellat F, Gogusev J, and Drueke TB. Direct effect of erythropoietin on rat vascular
56
57
58 smooth-muscle cell via a putative erythropoietin receptor. Nephrol Dial Transplant
59
60 1996;11:687-692.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 19 of 29 Rejuvenation Research

1
2
3 24. Alvarez Arroyo MV, Castilla MA, Gonzalez Pacheco FR, Tan D, Riesco A, Casado S,
4
5
6 Caramelo C. Role of vascular endothelial growth factor on erythropoietinrelated endothelial
7
8 cell proliferation. Journal of American Society on Nephrology 1998;9:1998-2004.
9
10
25. Buemi M, Marino D, Floccari F, Ruello A, Nostro L, Aloisi C, Marino MT, Di Pasquale
11
12
13 G, Corica F, Frisina N. Effect of interleukin 8 and ICAM-1 on calcium-dependent outflow of
14
15 K+ in erythrocytes from subjects with essential hypertension. Current Medical Research and
16
17
18
Opinion 2004;20:19-24.
19
20 26. Buemi M, Vaccaro M, Sturiale A, Galeano MR, Sansotta C, Cavallari V, Floccari F,
Fo

21
22 D’Amico F, Torre V, Calapai G, Frisina N, Guarneri F, Vermiglio G. Recombinant human
23
24
erythropoietin influences revascularization and healing in a rat model of random ischaemic
rP

25
26
27 flaps. Acta Derm Venereol 2002;82:411-417.
28
29
ee

27. Vaccaro M, Magaudda L, Cutroneo G, Trimarchi F, Barbuzza O, Guarneri B. Changes in


30
31
32 the distribution of laminin a1 chain in psoriatic skin. Immunohistochemical study using
rR

33
34 confocal laser scanning microscopy British Journal of Dermatology 2002;146:392-398.
35
36
28. Bennett CL, Silver SM, Djulbegovic B, Samaras AT, Blau CA, Gleanson KJ, Barnato SE,
37
ev

38
39 Elvermann KM, Courtney DM, McKoy JM, Edwards BJ, Tigue CC, Raisch DW, Yarnold PR,
40
41
iew

Dorr DA, Kuzel TM, Tallman MS, Trifilio SM, West DP, Lai SY, Henke M. Venous
42
43
44
thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin
45
46 administration for the treatment of cancer-associated anemia. JAMA 2008;299:914-924.
47
48 29. Ponec M, Weerheim A, Kempenaar J, Mommaas AM, Nugteren DH. Lipid composition
49
50
51 of cultured human keratinocytes in relation to their differentiation. J Lipid Res 1988;29:949-
52
53 61.
54
55 30. Brouty-Boye D, Raux H, Azzarone B, Tamboise A, Tamboise E, Beranger S, Magnien V,
56
57
58 Pihan I, Zardi L, Israel L. Fetal myofibroblast-like cells isolatedfrom post-radiation fibrosis in
59
60 human breast cancer. Int J Cancer 1991;47:697-702.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 20 of 29

1
2
3 31. Jelkmann W, Bohlius J, Hallek M, Sytkowski AJ. The erythropoietin receptor in normal
4
5
6 and cancer tissues. Critical Review on Oncology and Hematology 2008;67:39-61.
7
8 32. Littlewood TJ, Bajetta E, Nortier JW, Vercammen E, Rapoport B, Epoietin Alpha Study
9
10
Group. Effects of epoetin alfa on hematologic parameters and quality of life in cancer patients
11
12
13 receiving nonplatinum chemotherapy: results of a randomized, double-blind, placebo-
14
15 controlled trial. Journal of Clinical Oncology 2001;19:2865-2874.
16
17
18
33. Burger D, Lei M, Geoghegan-Morphet N, Lu X, Xenocostas A, Feng Q. Erythropoietin
19
20 protects cardiomyocytes from apoptosis via up-regulation of endothelial nitric oxide synthase.
Fo

21
22 Cardiovascular Research 2006;72:51-59.
23
24
34. Calvillo L, Latini R, Kajstura J, Leri A, Anversa P, Ghezzi P, Salio M, Cerami A, Brines
rP

25
26
27 M. Recombinant human erythropoietin protects the myocardium from ischemia-reperfusion
28
29
ee

injury and promotes beneficial remodeling. Proclaimed National Acadamie of Sciences, USA
30
31
32 2003;100:4802-4806.
rR

33
34 35. Moon C, Krawczky M, Ahn D, Ahmet I, Paik K, Lakatta EG, Talan MI. Erythropoietin
35
36
reduces myocardial infarction and left ventricular functional decline after coronary artery
37
ev

38
39 ligation in rats. Proclaimed National Acadamie of Sciences, USA 2003;100:11612-11617.
40
41
iew

42
43
44
36. Rui T, Cepinskas G, Feng Q, Kvietys PR. Delayed preconditioning in cardiac myocytes
45
46 with respect to development of a proinflammatory phenotype: role of SOD and NOS.
47
48 Cardiovascular Research 2003;59:901-911.
49
50
51 37. Tada H, Kagaya Y, Takeda M, Ohta J, Asaumi Y, Satoh K, Ito K, Karibe A, Shirato K,
52
53 Minegishi N, Shimokawa H. Endogenous erythropoietin system in non-hematopoietic lineage
54
55 cells play a protective role in myocardial ischemia/reperfusion. Cardiovascular Research
56
57
58 2006;71:466-477.
59
60 38. Bienvenu AL, Ferrandiz J, Kaiser K, Latour C, Picot S. Artesunateerythropoietin

combination for murine cerebral malaria treatment Acta Trop 2008;106:104-108.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 21 of 29 Rejuvenation Research

1
2
3 39. Digicaylioglu M, Lipton SA, Erythropoietin-mediated neuroprotection involves cross-talk
4
5
6 between Jak2 and NF-kappaB signalling cascades. Nature 2001;412:601-602.
7
8 40. Chen J, Connor KM, Aderman CM, Willett KL, Aspegren OP, Smith LE. Supression of
9
10
retinal neovascularization by erythropoietin siRNA in a mouse model of proliferative
11
12
13 retinopathy. Invest Ophthalmol Vis Sci 2009;50:1329- 1335.
14
15 41. Morita M, Ohneda O, Yamashita T, Takahashi S, Suzuki N, Nakajima O, Kawauchi S,
16
17
18
Ema M, Shibahara S, Udono T, Tomita K, Tamai M, Sogawa K, Yamamoto M, Fujii-
19
20 Kuriyama Y. HLF/HIF-2alpha is a key factor in retinopathy of prematurity in association with
Fo

21
22 erythropoietin. EMBO Journal
23
24
2003;22:1134-1146.
rP

25
26
27 42. Friedman EA, Brown CD, Berman DH. Erythropoietin in diabetic macular edema and
28
29
ee

renal insufficiency. American Journal of Kidney Diseases 1995;26:202-208.


30
31
32 43. Lee IG, Chae SL, Kim JC. Involvement of circulating endothelial progenitor cells and
rR

33
34 vasculogenic factors in the pathogenesis of diabetic retinopathy. Eye 2006;20:546-552.
35
36
37
ev

38
39 44. Tong Z, Yang Z, Patel S, Chen H, Gibbs D, Yang X, Hau VS, Kaminoh Y, Harmon J,
40
41
iew

Pearson E, Buehler J, Chen Y, Yu B, Tinkham NH, Zabriskie NA, Zeng J, Luo L, Sun JK,
42
43
44
Prakash M, Hamam RN, Tonna S, Constantine R, Ronquillo CC, Sadda S, Avery RL, Brand
45
46 JM, London N, Anduze AL, King GL, Bernstein PS, Watkins S, Genetics of Diabetes and
47
48 Diabetic Complication Study Group, Jorde LB, Li DY, Aiello LP, Pollak MR, Zhang K.
49
50
51 Promoter polymorphism of the erythropoietin gene in severe diabetic eye and kidney
52
53 complications. Proclaimed National Acadamie of Sciences, USA 2008;105:6998-7003.
54
55
56
57
58 45. Manzoni P, Maestri A, Gomirato G, Takagi H, Watanabe D, Matsui S. Erythropoietin as a
59
60 retinal angiogenic factor in proliferative diabetic retinopathy. New England Journal of

Medicine 2005;353:782-792.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 22 of 29

1
2
3 46. Holstein JH, Menger MD, Scheuer C, Meier C, Culemann U, Wirbel RJ, Garcia P,
4
5
6 Pohlemann T. Erythropoietin (EPO): EPO-receptor signaling improves early endochondral
7
8 ossification and mechanical strength in fracture healing. Life Science 2007;80:893-900.
9
10
47. Gough NR. Making Sense of EPO Receptors. Science 2008;320:1397 48. Foster DJ, Moe
11
12
13 OW, Hsia CCW Upregulation of erythropoietin receptor during postnatal and
14
15 postpneumonectomy lung growth. Am J Physiol 2004;287:L1107–L1115.
16
17
18
49. Brines M, Cerami A. Emerging boilogicala roles for erythropoietin in the nervous system.
19
20 National Review on Neuroscience 2005;6:484-894.
Fo

21
22 50. Bodó E, Kromminga A, Funk W, Laugsch M, Duske U, Jelkmann W, Paus R. Human hair
23
24
follicles are an extrarenal source and a nonhematopoietic target of erythropoietin. FASEB J
rP

25
26
27 2007; 21:3346-54.
28
29
ee

51. Paus R, Bodó E, Kromminga A, Jelkmann W. Erythropoietin and the skin: a role for
30
31
32 epidermal oxygen sensing? Bioessays 2009; 31:344-8.
rR

33
34
35
36
52. Ito M, Liu Y, Yang Z, Nguyen J, Liang F, Morris RJ, Cotsarelis G. Stem cells in the hair
37
ev

38
39 follicle bulge contribute to wound repair but not to homeostasis ofthe epidermis. Nature
40
41
iew

Medicine 2005;111351-1354.
42
43
44
53. Maxwell PH, Ferguson DJ, Nicholls LG, Johnson MH, Ratcliffe PJ. Theinterstitial
45
46 response to renal injury: fibroblast-like cells show phenotypic changes and have reduced
47
48 potential for erythropoietin gene expression.
49
50
51 Kidney International 1997;52:715-724.
52
53 54. Boutin AT, Weidemann A, Fu Z, Mesropian L, Gradin K, Jamora C, Wiesener M, Eckardt
54
55 KU, Koch CJ, Ellies LG, Haddad G, Haase VH, Simon MC, Poellinger L, Powell FL,
56
57
58 Johnson RS. Epidermal sensing of oxygen is essential for systemic hypoxic response. Cell
59
60 2008;133:223-34.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 23 of 29 Rejuvenation Research

1
2
3 55. Köchling J, Curtin PT, Madan A. Regulation of human erythropoietin gene induction by
4
5
6 upstream flanking sequences in transgenic mice. Br. J. Haematol 1998;103:960–8.
7
8 56. Higley HR, Ksander GA, Gerhardt CO, Falanga V. Extravasation of macromolecules and
9
10
possible trapping of transforming growth factor-beta in venous ulceration. British Journal of
11
12
13 Dermatology 1995;132:79-85.
14
15 57. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates
16
17
18
for the year 2000 and projections for 2030. Diabetes Care 2004; 27:1047-1053,
19
20 58. Brem H, Tomic-Canic M. Cellular and molecular basis of wound healing in diabetes. J
Fo

21
22 Clin Invest. 2007;117:1219-1222.
23
24
59. Falanga V. Wound healing and its impairment in the diabetic foot. Lancet 2005;
rP

25
26
27 366:1736-1743.
28
29
ee

60 . Langer A, Rogowski W.Systematic review of economic evaluations of human cell-


30
31
32 derived wound care products for the treatment of venous leg and diabetic foot ulcers. BMC
rR

33
34 Health Serv Res. 2009 ;9:115.
35
36
61. Hanft JR, Surprenant MS. Healing of chronic foot ulcers in diabetic patients treated with a
37
ev

38
39 human fibroblast-derived dermis. J Foot Ankle Surg. 2002 ;41:291-299.
40
41
iew

62. A. Hjerppe, M. Hjerppe, V. Autio, R. Raudasoja, A. Vaalasti, . Treatment of Chronic


42
43
44
Leg Ulcers with a Human Fibroblast-Derived Dermal Substitute: A Case Series of 114
45
46 Patients. Wounds. 2004;16(3) © 2004 Health Management Publications, Inc
47
48 63. Brines M, Cerami A.Erythropoietin-mediated tissue protection: reducing collateral
49
50
51 damage from the primary injury response. J Intern Med. 2008;264:405-432.
52
53 64. Hamed S, Ullmann Y, Masoud M, Hellou E, Khamaysi Z, Teot L.Topical erythropoietin
54
55 promotes wound repair in diabetic rats. J Invest Dermatol. 2010;130287-94.
56
57
58 65. Rappolee DA, Mark D, Banda MJ, Werb Z. Wound macrophages express TGF-alpha and
59
60 other growth factors in vivo: analysis by mRNA phenotyping. Science. 1988 ;241:708-712.

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 24 of 29

1
2
3 66. Mikhal'chik EV, Piterskaya JA, Budkevich LY, Pen'kov LY, Facchiano A, De Luca C,
4
5
6 Ibragimova GA, Korkina LG.Comparative study of cytokine content in the plasma and wound
7
8 exudate from children with severe burns. Bull Exp Biol Med. 2009 ;148:771-775.
9
10
67. Rezaeian F, Wettstein R, Amon M, Scheuer C, Schramm R, Menger MD, Pittet B, Harder
11
12
13 Y. Erythropoietin protects critically perfused flap tissue. Ann Surg. 2008 ;248:919-929.
14
15 68. Galeano M, Altavilla D, Cucinotta D, Russo GT, Calò M, Bitto A, Marini H, Marini R,
16
17
18
Adamo EB, Seminara P, Minutoli L, Torre V, Squadrito F. Recombinant human
19
20 erythropoietin stimulates angiogenesis and wound healing in the genetically diabetic mouse.
Fo

21
22 Diabetes. 2004 ;53:2509-2517.
23
24
rP

25
26
27
28
29
ee

30
31
32
rR

33
34
35
36
37
ev

38
39
40
41
iew

42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 25 of 29 Rejuvenation Research

1
2
3
4
5
6 Figure 1
7
8
Cells were cultured from human biopsies and grown to the 4th passage in vitro. (a) The
9
10 mRNA expression profile of the EPO receptor in FmSCs. Sequencing of the PCR product for
11
12 the mRNA of the EPO receptor showed 90-98% sequence homology. (b, c) Proliferation of
13
14
15 FmSCs under hypoxic conditions and under the influence of 10 ng/ml trauma cytokine
16
17 stimulation. (b) Stimulation with EPO alone in the absence of any additional cytokines under
18
19 otherwise identical cell culture conditions showed an inhibitory effect on stem cell growth. (c)
20
Fo

21
22 IL-6 stimulation of FmSCs also resulted in decreased growth activity in vitro. This was
23
24 reversed in the presence of EPO. Each point represents the mean ± SD of nine experiments
rP

25
26 and statistically significant difference (P < 0.005, student’s test).
27
28
29
ee

30
31 Figure 2
32 Phenotype of the FmSCs, determined using flow cytometry. EPO triggering did not change
rR

33
34 the phenotype at all; CD31, CD45, CD 73 remained stable with or without the presence of
35
36 trauma cytokines. No major population shift.
37
ev

38
39
Figure 3
40
41
iew

Phenotype of FmSCs did not change with CD105 and CD166 remained stable with or without
42
43 the presence of trauma cytokines. We did observe that cells expressing CD90 partially
44
45 switched to a non-CD90-expressing subpopulation in the presence of IL-6. This “IL-6 effect”
46
47
paralleled the growth curves in the presence of IL-6. No major population shift except this
48 (Control CD 166: EPO CD 166).
49
50
51
52
Figure 4
53
54
55 (a) Patient A was a 26 year male, who had suffered 25 % body surface flame burn injury,
56
57 requiring split skin grafting on day 7 after trauma.A 26-year-old patient with a 0.3-mm split-
58
59
60
thickness skin graft donor site seven days after three treatment sessions without (left) and with

local erythropoietin (right). The polyurethane film dressing was not changed until removal

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 26 of 29

1
2
3 seven days following surgery, which allowed a standardized wound management and a
4
5
6 comparability of the healing results. Note the closed, dry and pale-red wound surface of the
7
8 EPO treated side indicating a complete reepithelization as well as the secretion and the dark-
9
10
red surface of the non-EPO donor site as a sign of incomplete healing.
11
12
13
14
15 (b) Patient B was a 64 year lady with a pressure sore (Campbell stage VI including
16
17
18
deperiosted calcanear bone) of her right heel following urosepsis. Sural flap plasty had been
19
20 performed already with partial flap loss. The patient had refused further reconstructive
Fo

21
22 procedures but was focused on preventing amputation by all conservative means. A 64-year-
23
24
old diabetic patient with a partial necrotic heel after urosepsis providing poor granulation
rP

25
26
27 tissue following conventional treatment (left). Clinical results after five treatments with local
28
29
ee

EPO and subsequent split-thickness skin grafting (right). Note the almost complete wound
30
31
32 closure. The preoperative preparation of the wound allowed salvage of the limb by a minor
rR

33
34 surgical procedure.
35
36
37
ev

38
39 (c) Patient C was a 69 year male with peripheral arterial occlusive disease, stage IV with a
40
41
iew

single arterial supply for the lower leg, non suitable for interventional or surgical
42
43
44
macrovascular reconstruction. The patient had distal leg ulcer and partial necrosis of the
45
46 peroneal tendons since more than 6 months. A 69-year-old diabetic patient with a grade III
47
48 ulcer at the lateral malleolus based on a peripheral arterial disease grade IV. Exposed tendons
49
50
51 at the bottom of the wound and absence of granulation tissue formation subsequent
52
53 revascularization and conservative wound treatment (left). Clinical results after only five local
54
55 treatments with EPO optimizing the wound bed for subsequent split-thickness skin grafting
56
57
58 (right). Note the complete wound closure, which has been stable for more than 12 months.
59
60

Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 27 of 29 Rejuvenation Research

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
Fo
19
20
21
22
r
23
24
Pe

25
26
27
28
er

29
30
31
32
Re

33
34
35
36
vi

37
38
39
ew

40
41
42
43
44
45
46
47
209x297mm (150 x 150 DPI)
48
49
50
51
52
53
54
55
56
57
58
59
60
Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 28 of 29

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
Fo
19
20
21
22
r
23
24
Pe

25
26
27
28
er

29
30
31
32
Re

33
34
35
36
vi

37
38
39
ew

40
41
42
43
44
45
46
47
48
49 209x297mm (150 x 150 DPI)
50
51
52
53
54
55
56
57
58
59
60
Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Page 29 of 29 Rejuvenation Research

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
Fo
19
20
21
22
r
23
24
Pe

25
26
27
28
er

29
30
31
32
Re

33
34
35
36
vi

37
38
39
ew

40
41
42
43
44
45
46
47
48
49 209x297mm (150 x 150 DPI)
50
51
52
53
54
55
56
57
58
59
60
Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801
Rejuvenation Research Page 30 of 29

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
Fo
19
20
21
22
r
23
24
Pe

25
26
27
28
er

29
30
31
32
Re

33
34
35
36
vi

37
38
39
ew

40
41
42
43
44
45
46
47 146x202mm (72 x 72 DPI)
48
49
50
51
52
53
54
55
56
57
58
59
60
Mary Ann Liebert, Inc., 140 Huguenot Street, New Rochelle, NY 10801

You might also like