• No results found

Bone marrow derived cells in collateral formation : studies toward therapeutic arteriogenesis Hellingman, A.A.

N/A
N/A
Protected

Academic year: 2021

Share "Bone marrow derived cells in collateral formation : studies toward therapeutic arteriogenesis Hellingman, A.A."

Copied!
19
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

toward therapeutic arteriogenesis

Hellingman, A.A.

Citation

Hellingman, A. A. (2011, September 15). Bone marrow derived cells in collateral formation : studies toward therapeutic arteriogenesis. Retrieved from https://hdl.handle.net/1887/17838

Version: Corrected Publisher’s Version

License: Licence agreement concerning inclusion of doctoral thesis in the Institutional Repository of the University of Leiden

Downloaded from: https://hdl.handle.net/1887/17838

Note: To cite this publication please use the final published version (if applicable).

(2)

Chapter 5

T-cell-pre-stimulated monocytes promote neovascularization in a murine hind limb ischemia model

A.A. Hellingman1, J.J. Zwaginga2,3, R.T. van Beem4, M.A.

Lijkwan1, L. Seghers1, M.R. de Vries1, P.J. van den Elsen2,5, A.J. van Zonneveld6,7, C.E. van der Schoot4, J.F. Hamming1, W.E. Fibbe2, P.H.A. Quax1,7 and S.B. Geutskens2,7

1Dept. of Vascular Surgery, Leiden University Medical Center, Leiden

2Dept. of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden

3Dept. Research and Education, Sanquin Blood Bank SW, Leiden.

4Dept. of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam

5Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands

6Dept. of Nephrology, Leiden University Medical Center, Leiden

7Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden

Eur J Vasc Endovasc Surg. 2011. In press

(3)

ABSTRACT

Aim

Monocytes play a significant role in neovascularization. The stimuli that differentiate monocytes along a pro-angio/arteriogenic-supporting pathway are currently unclear.

We investigated whether pre-stimulation of human monocytes with soluble T-cell- derived factors improve re-vascularization in murine hind limb ischemia as a new option for therapeutic angio- and arteriogenesis.

Design

Human monocytes were cultured with or without soluble T-cell-derived factors. Un- stimulated and pre-stimulated monocytes were transfused after induction of hind limb ischemia in nude mice.

Methods

Blood flow was measured with Laser Doppler Perfusion Imaging. Collaterals were visu- alized by immunohistochemistry and angiography. Monocytes were characterized by flowcytometry and Bio-plex assays.

Results: Transfusion of T-cell-pre-stimulated monocytes significantly improved blood flow recovery after hind limb ischemia and increased collateral size and collateral and capillary number in the post-ischemic paw. Pre-stimulated monocytes produced a wide variety of factors that support neo-vascularization such as platelet-derived growth factor-BB, vascular-endothelial growth factor, interleukin-4 and tumor necrosis factor- a. Few transfused human cells were detected in the muscle tissue, suggesting that paracrine rather than direct effects appear responsible for the enhanced recovery of bloodflow observed.

Conclusion

These results show a beneficial role for T-cell-pre-stimulated monocytes in neovascular- ization, rendering the monocyte a potential candidate for regenerative cell therapy that promotes revascularization in peripheral arterial disease patients.

(4)

INTRODUCTION

Collateral artery formation (arteriogenesis) may prevent tissue damage caused by arterial stenosis or occlusion and is crucially important to limit the consequences of peripheral arterial disease (PAD). Despite improvements of vascular- and endovascular techniques, major amputation is still described in one third of patients with critical limb ischemia, underscoring the urgent need for new therapeutic alternatives1.

Cell therapy is a promising novel therapeutic approach for the treatment of patients with PAD. Selection of the most efficient cell type for therapeutic purposes is a central issue.

Much attention has been given to the use of endothelial progenitor cells (EPCs) that may augment revascularization by direct incorporation into the vessel wall. Transfusion of EPCs isolated from circulating mononuclear cells or the increase of circulating EPC numbers via bone marrow (BM) mobilization have been described to promote neovascularization in different animal models2,  3. However, outcomes of clinical studies in which BM-derived cells (including EPCs) were transplanted into ischemic limbs of patients showed only minor improvements4-8. Moreover, the controversy about the nature of human blood EPC complicated the isolation procedure and therefore the development of a cellular therapy.

The monocyte was highlighted as an alternative candidate for cell therapy that may facilitate vascular regeneration. Monocytes are pluripotent progenitor cells that de- pending on the stimulus encountered may differentiate into immune effectors such as macrophages or dendritic cells. Notably, a still growing number of studies additionally emphasizes the involvement of the monocyte or its mature progeny in angio- and ar- teriogenic processes. The beneficial effects of monocyte transfusion were underscored by several recent studies. Herold et al.9 demonstrated increased blood flow recovery after ligation-induced ischemia by transfusion of autologous monocytes that were engineered by adenoviral transduction to express Granulocyte/Macrophage-colony- stimulating factor (GM-CSF), possibly by stimulating mobilization of EPCs from the BM.

Interestingly, transfusion of untransduced autologous monocytes stimulated ex vivo with GM-CSF was ineffective. The importance of a stimulus to mature monocytes along a certain pathway was emphasized by a study of Urbich et al.10 Here, human blood- derived mononuclear cells, CD14- or CD14+-monocyte-enriched populations that were stimulated in endothelial medium with vascular endothelial growth factor (VEGF) to generate EPCs improved blood flow recovery in a hind limb ischemia model, whereas CD14-- or CD14+-populations that were not stimulated before infusion did not.

Besides monocytes, T-lymphocytes play an eminent role in adult vascular repair as was exemplified by the hampered blood flow recovery and reduced collateral density upon hind limb ischemia in various murine models that lack CD4+-T-helper lymphocytes11, 12. Moreover, significantly lower numbers of monocytes/macrophages accumulated in the ischemic muscles of CD4+-deficient mice11, suggesting that T-cell/monocyte interactions

(5)

are involved in arteriogenesis. We previously reported that co-culture of monocytes with CD4+-T-cells was indispensable for the formation of colony-forming-unit (CFU)-Hill colonies13. CFU-Hill colonies are correlated to vascular reactivity and although they were initially thought to represent colony growth of EPCs14, it is now widely accepted that these colonies are of hematopoietic origin15, 16. Instead of EPC-derived colony growth, this in vitro culture actually represents the efficiency of cluster formation between au- tologous T-lymphocytes and naïve CD14+-monocytes. Enhanced cluster formation was observed upon prior activation of CD4+-T-cells and replacement of T-cells by the addition of activated CD4+-T-cell-derived soluble factors similarly stimulated cluster formation13.

The aim of this study was to further investigate the phenotype of the T-cell-pre- stimulated monocytes and to examine whether pre-stimulation of naïve human CD14+- monocytes with T-cell-derived soluble factors promotes their capacity to improve vascular regeneration in a murine hind limb ischemia model for PAD.

MATERIALS AND METHODS

Isolation and culture of human primary cells

Human CD14+-monocytes or CD4+-T-cells-enriched fresh apheresis mononuclear cell preparations from healthy donors were obtained at Sanquin Bloodbank NW Amsterdam after informed consent and with approval of the Medical Ethical Committee of the Aca- demic Medical Centre, Amsterdam, The Netherlands. T-cell and monocyte isolation as well as CD4+-T-cell activation was performed as described previously13. Distinct CD14+/ CD16-, CD14+/CD16+ and CD14lo/CD16+ monocyte subsets were sorted with a MoFlow cell sorter (Beckman Coulter).

Total CD14+-monocytes or monocyte-subsets were cultured for 48 hours in fibronec- tin-coated plates (BD Biosciences) at a density of 1x106 cells/ml in EndocultTM medium (Stem Cell Technologies) or stimulated with a 4:1 mixture of EndocultTM medium and pooled T-cell-conditioned medium (TCCM) from 3-4 individual donors. For macrophage or dendritic cell (DC) cultures, CD14+-monocytes were similarly cultured for 48h in En- docultTM medium supplemented with 10 ng/ml macrophage-colony stimulating factor (M-CSF) (Peprotech) or GM-CSF (1000 U/ml) + interleukin (IL)-4 (2000 U/ml) (Invitrogen), respectively.

For transfusion in vivo, loosely adherent cells were harvested after 48 hours by pipet- ting, washed twice with PBS and infused into the mice within 1 hour.

Monocyte-conditioned medium was prepared by removal of culture media after 48 hours, followed by three washes with PBS and addition of fresh endocult medium that was subsequently harvested after 24, 48 or 72 hours. The production of secreted proteins was determined using a Bio-Plex Cytokine-assays (Bio-Rad).

(6)

Expression analysis of cell surface proteins by flowcytometry

Expression of leukocyte surface molecules was determined using directly conjugated antibodies that were added to the cells after 15 minutes of pre-incubation with 10 µg/ml purified human IgG to block non-specific binding. Non-specific isotype-matched anti- bodies were used as negative controls. Mean fluorescence intensity (MFI) of the labelled cells was measured using a flowcytometer (FACScan; Becton Dickinson) and analyzed with Cellquest software (Becton Dickinson) and WinMDI.

Animals

To avoid immune reactivity against transfused human cells, immunodeficient nude C57Bl6 (no functional T and B-lymphocytes) and NOD-scid-IL2Rgamma(null) (addition- ally lack NK-cell activity) mice (male, 10-12 weeks, The Jackson Laboratory, USA) were used for in vivo experiments. Experiments were approved by the committee on animal welfare of our institute.

Surgical procedure to induce hind limb ischemia and injection of cells

Before surgery, mice were anesthetized with intraperitoneal (i.p.) injection of a combi- nation of Midazolam (5mg/kg, Roche), Medetomidine (0.5mg/kg, Orion) and Fentanyl (0.05mg/kg, Janssen). The effect of transfusion of human monocytes was tested in 2 models of hind limb ischemia: a total excision model and a double electro-coagulation model17.

For unilateral total excision of the femoral artery, ligatures were placed proximal (inguinal ligament) and distal (popliteal artery). After dissecting all side branches, the whole artery was removed after cutting the femoral artery between the ligatures. For the double coagulation model, both common iliac artery and femoral artery were uni- lateral electro-coagulated. First an electro-coagulation of the common iliac artery was performed and directly afterwards an electro-coagulation of the femoral artery. After surgery, the skin was closed with Ethilon 6.0 sutures. Twenty-four hours after surgery, mice were intravenously (i.v.) injected into the tail vein with PBS, unstimulated control or pre-stimulated monocytes.

Laser Doppler Perfusion Imaging

Blood flow was measured before ischemia induction, immediately after ischemia induc- tion and 3, 7, 14, 21 and 28 days after surgery in the ischemic and non-ischemic paws, using Laser Doppler Perfusion Imaging (Moor Instruments). Perfusion was expressed as a ratio of the ischemic to non-ischemic limb, as described previously18.

(7)

Histological analysis

Animals were sacrificed 28 days after ischemia induction and calf and adductor muscles (from ischemic and non-ischemic paw) were removed and fixed with 4% formaldehyde and paraffin-embedded. Serial 5µm cross-sections were generated. Sections were re- hydrated and endogenous peroxidase activity was blocked for 20 minutes in methanol containing 0.3% hydrogen peroxide. Capillaries and collaterals were visualized using antibodies recognizing CD31 on endothelial cells or a-smooth muscle actin (SMA) in smooth muscle cells, respectively.

For CD31 labeling, sections were pre-incubated with trypsin (30 minutes/ 37°C), incu- bated overnight with primary antibody (BD Biosciences) followed by a biotin-conjugated secondary antibody (AbCam). The reaction was enhanced by tyramine amplification and the avidin-biotin horseradish-peroxidase (HRP) system (DakoCytomation) and visualized by NovaRED (Vector laboratories).

For SMA labeling, tissue sections were incubated overnight with anti-αSMA (Dako) without antigen retrieval. Labeling was followed by an HRP-conjugated secondary antibody (Dako). Anti-human CD45 was used to identify human cells in frozen tissue sections fixed with ice-cold acetone. Labeling was visualized using the above described HRP-labeling procedure. All sections were counterstained with hematoxylin. Isotype control antibodies were used as controls. Quantification of labeled tissue sections was performed using ImageJ (9 sections per mouse were analyzed to obtain the mean per animal, 10 animals per group were measured).

Distribution of human primary cells

Blood was harvested and single cell suspensions were made of the spleen, liver and the non-ischemic femur at 24 hours (n=2), 48 hours (n=3) or 7 days (n=3) after transplan- tation with pre-stimulated monocytes. Adductor and calf muscles of both paws were frozen and cross-sectioned for immunohistochemistry. Human cells were visualized using flowcytometry with a directly conjugated human-specific anti-CD45 antibody (Becton-Dickinson).

Statistical analysis

Results are expressed as mean ± SEM. For in vivo and ex vivo experiments comparisons between means were performed using a One-Way-ANOVA-test. For in vitro protein mea- surements comparisons between means were performed using a paired t-test. P-values

<0.05 were considered statistically significant. Calculations were performed in SPSS 16.0.

(8)

RESULTS

Spindle-shaped monocyte clusters are formed from both classical and non- classical monocyte subsets

Typical spindle-shaped clusters formed upon culture of CD14+-monocytes with soluble T-cell-derived factors (Figure 1A, lower row), but not when cultured without soluble T-cell-derived factors (Figure 1A, upper row) or upon addition of soluble T-cell-derived factors of unstimulated CD4+T-cells (not shown). Upon culture of total CD14+-selected cells, control and pre-stimulated monocytes both express CD14 and CD16, similar to un- cultured non-classical monocytes (Figure 1B). We therefore questioned whether cluster formation was related to a distinct monocyte subset. Using FACSsorting we separated uncultured CD16neg (R1), CD16med (R2), and CD16hi (R3) human CD14+-monocyte-subsets and observed that all subsets are able to form spindle-shaped clusters upon pre- stimulation (Figure 1C).

Pre-stimulated monocytes do not develop into endothelial-like cells and are phenotypically distinct from classical macrophages and DC

Considering the controversy in the monocyte-related origin of EPC we examined whether pre-stimulated CD14+-monocytes express endothelial cell (EC)-specific markers upon culture. Both control and pre-stimulated monocytes retain cell surface expression of the leukocyte-specific b2-integrin CD11b and CD45 at the cell surface and show com- parable expression of the b1-integrins CD49f and CD49d or the shared hematopoietic/

EC marker CD31 (Figure 2A). Expression of the hematopoietic stem cell/EC-specific CD34 was not observed upon culture.

T-cell-derived soluble factors can induce differentiation of monocytes into immature DCs and also increase expression of the co-stimulatory molecules CD40, CD83 and CD8619. To investigate whether our culture procedure induced a distinct differentiation program we compared cell surface expression of co-stimulatory and subset-specific molecules with that of classical monocyte progeny such as macrophages or DC.

No differences in the surface expression levels of the abovementioned co-stimulatory molecules were observed between pre-stimulated and GM-CSF+IL-4-cultured mono- cytes (Figure 2B) whereas expression levels of the typical macrophage marker CD115 and the DC-marker CD209 (DC-Sign) were considerably lower on pre-stimulated monocytes as compared to M-CSF-generated macrophages or GM-CSF+ IL-4-generated DC (Figure 2B). These differences suggest an alternative route of differentiation via pre-stimulation with T-cell-derived soluble factors.

(9)

Figure 1. Spindle-shaped monocyte clusters are formed from classical CD16- and non-classical CD16+

monocyte subsets.

A. Total CD14+-monocytes were cultured in EndocultTM medium without (upper row) or pre-stimulated with soluble T-cell-derived factors (lower row) and imaged after 24 hours (left), 48 hours (middle) or 5 days (right). Magnification 100x.

B. Flow cytometry was used to determine the cell surface expression of CD14 and CD16 on naïve uncultured monocytes (left dot plot), unstimulated monocytes cultured for 48 hours (middle) or pre- stimulated monocytes cultured for 48 hours (right dot plot).

C. The histograms depict the number of clusters formed by monocyte subsets that were sorted based on their CD14 and CD16 expression using regions R1-R3 as indicated in Figure 1B. Sorted cells were cultured for 5 days in EndocultTM medium supplemented with soluble T-cell-derived factors. A representative donor out of 2 different donors is shown.

(10)

Figure 2 Pre-stimulated CD14+-monocytes do not develop into endothelial-like cells and are phenotypically distinct from classical macrophages and DC.

A. Histograms show cell surface expression as determined by flowcytometry of the indicated hematopoietic or endothelial cell-associated markers in black lines and isotype controls as filled grey histograms of unstimulated control monocytes or pre-stimulated monocytes cultured for 48 hours.

B. Histograms show cell surface expression as determined by flowcytometry of the indicated co- stimulatory or subset-specific markers in black lines and isotype controls as filled grey histograms of pre-stimulated, M-CSF-stimulated or GM-CSF+IL-4-stimulated monocytes that were cultured for 48 hours.

A representative donor/culture is shown. Results were confirmed in at least 3 different donors.

(11)
(12)

Figure 3 Transfusion of pre-stimulated CD14+-monocytes improves blood flow restoration after total excision of the femoral artery.

A. The graph depicts the mean±SEM of blood flow restoration in hind limb of nude C57Bl6 mice after transfusion with PBS (n=10), unstimulated control monocytes (n=10) (control) or pre-stimulated monocytes (n=10). Blood flow was monitored for 4 weeks and expressed as ratio between the operated and non-operated limb measured before and after surgery at day 3, 7, 14, 21 and 28. The residual bloodflow was determined by measuring the bloodflow directly after surgery. *P<0.05.

Images on the right depict representative LDPI images of paws of mice transfused with PBS (upper), unstimulated monocytes (middle) or pre-stimulated monocytes (lower) at 14 days after induction of hind limb ischemia.

B. Histograms depict mean±SEM of the number of collaterals (left) or mean±SEM of the collateral surface (right) in the post-ischemic adductor muscle as quantified by smooth muscle actin (SMA) labelling.

*P<0.05, ** P<0.01. Representative photographs of SMA-labelling are shown 28 days after transfusion with PBS (left), unstimulated monocytes (middle) and pre-stimulated monocytes (right). Magnification 100x.

C. The histogram depicts mean±SEM of the number of capillaries in the post-ischemic calf muscle as quantified by anti-mouse-specific CD31 labelling (9 sections per muscle were analyzed of 10 animals/

treatment group). *P<0.05, ** P<0.01. Representative photographs of CD31-labelling are shown 28 days after transfusion with PBS (left), unstimulated monocytes (middle) and pre-stimulated monocytes (right).

Magnification 100x.

D. Representative angiographs were made 14 days after the surgical procedure of post-ischemic paws transfused with PBS (left), unstimulated monocytes (middle) and pre-stimulated monocytes (right).

In contrast to mice transfused with PBS or non-stimulated monocytes, the adductor muscle of mice transfused with pre-stimulated monocytes showed typical cork-screw like collaterals as is indicated with the black arrow.

Figure 4 Pre-stimulated CD14+-monocytes improve blood flow restoration after double electro-coagulation.

The graphs depicts the mean±SEM of blood flow restoration in hind limbs of NOD-SCID-IL2Rgamma(null) mice after transfusion with PBS (n=10), unstimulated control monocytes (n=6) or pre-stimulated monocytes (n=4). *P<0.05. Images on the right depict representative LDPI images of paws of mice transfused with PBS (upper), unstimulated monocytes (middle) or pre-stimulated monocytes (lower) 14 days after induction of hind limb ischemia. Blood flow was monitored for 4 weeks and expressed as ratio between the operated and non-operated limb measured before and after surgery at day 3, 7, 14, 21 and 28. The residual bloodflow was determined by measuring the bloodflow directly after surgery. *P<0.05.

(13)

Figure 5 Pre-stimulated CD14+-monocytes produce various pro-angio-/arteriogenic factors.

A. Images show isotype-control (left) and anti-humanCD45-specific (right) labelling of the ischemic adductor muscle, 24 hours upon transfusion of pre-stimulated human monocytes. Magnification 400x.

B. Histograms show the mean±SEM (n=3) of indicated growth factors and cytokines determined in media conditioned for 24, 48 or 72 hours by unstimulated control or pre-stimulated monocytes after the removal of initial culture media used for stimulation (48 hours) and vigorous washing with PBS. *P<0.05, ** P<0.01.

Pre-stimulation of total CD14+-monocytes induces the capacity to improve blood flow recovery in vivo

Directly after total excision of the femoral artery, blood flow in the treated hind limb decreased to less than 10% of the blood flow of the untreated hind limb (Figure 3A).

Blood flow recovery progressed gradually in all groups (n=10/ group). Nude C57Bl6 mice transfused with pre-stimulated monocytes showed significantly improved blood flow recovery from day7 until day21 after ischemia as compared to mice transfused with PBS or unstimulated control monocytes (Figure 3A). Fourteen days after surgery, restoration of blood flow was complete in mice transfused with pre-stimulated monocytes, whereas only 70% restoration of blood flow was observed in the treated hind limb of control

(14)

mice. Importantly, transfusion of unstimulated control monocytes did not have any beneficial effect on blood flow recovery.

Increase in collateral and capillary density in post-ischemic skeletal muscle of mice treated with pre-stimulated monocytes

Mice treated with pre-stimulated monocytes showed a significantly higher number of a- smooth muscle actin (SMA)-expressing collaterals in the post-ischemic adductor muscle as compared to controls (Figure 3B). Furthermore, significantly larger collaterals were observed in the adductor muscle of mice transfused with pre-stimulated monocytes as compared to controls (Figure 3B). The collateral density or cross-sectional surface (diameter) of SMA-expressing collaterals of the non-operated hind limb did not show any differences between groups (data not shown). In addition, transfusion with pre- stimulated monocytes significantly increased the CD31+-capillary density of the post- ischemic calf muscles of mice as compared to controls (Figure 3C). Again, analyses of capillary density of the non-operated paw did not show differences between the groups (data not shown).

To visualize collateral growth in the post-ischemic hind limb, angiographs were made 14 days after hind limb ischemia induction. Solid collaterals were observed in the an- giographs of post-ischemic adductor muscles of mice transfused with pre-stimulated monocytes (Figure 3D), confirming the results of the SMA-staining.

Improved blood flow recovery after transfusion of pre-stimulated monocytes in a double electro-coagulation hind limb ischemia mouse model

To confirm general application of the above described findings, we injected pre- stimulated CD14+-monocytes in NOD-scid IL2Rgamma(null) mice after double electro- coagulation of both the femoral artery and the iliac artery. Again, transfusion with pre-stimulated monocytes significantly improved blood flow recovery after ischemia as compared to PBS, 21 and 28 days after ischemia induction (Figure 4).

Few human cells were found in the post-ischemic muscle tissue

The distribution of pre-stimulated monocytes was assessed in blood, spleen, liver and BM after 24 hours, 48 hours or 7 days using flowcytometry in combination with an a- humanCD45-specific antibody. Human cells were not observed in any of the tissues/

time-points examined by FACS analysis of the tissue extracts (data not shown). However, human cells were observed upon total sectioning of the ischemic adductor tissue after 24 hours (Figure 5A), indicating that human monocytes can be recruited to the murine muscle tissue upon ischemia induction.

(15)

Pre-stimulated monocytes produce a wide variety of pro-angio-/arteriogenic factors

We next examined whether pre-stimulated monocytes produce pro-angio- and/or pro-arteriogenic growth factors. Increased levels of platelet-derived growth factor (PDGF)-BB, VEGF, IL-6, IL-8, IL10 and tumor-necrosis factor (TNF)-a were observed in me- dium conditioned by pre-stimulated monocytes as compared to unstimulated control monocytes after 24, 48 and 72 hours. Significant increases in expression levels were found after 72 hours for granulocyte-CSF, IL-4, IL-13, IL-17 and interferon(IFN)g, whereas the levels of fibroblast growth factor(FGF), GM-CSF and IL12p70 did not differ between media conditioned by unstimulated or pre-stimulated monocytes (Figure 5B).

DISCUSSION

There is ample evidence that monocytes play a crucial role in neovascularization and have the potential to augment collateral artery growth. For that reason transfusion of CD14+-monocytes has been highlighted as cell therapy that promotes revasculariza- tion in adults. Previous studies showed that proper stimulation of monocytes is critical to exploit this capacity9, 10. In the present study we show that pre-stimulation of total CD14+-monocytes with activated CD4+-T-cell-derived soluble factors enhanced their ca- pacity to promote revascularization and significantly improved restoration of blood flow recovery after hind limb ischemia as compared to untreated mice or mice transfused with unstimulated monocytes.

Monocyte transfusion was first examined in a total excision model using nude C57Bl6 mice and additionally tested in a double electro-coagulation model using NOD-scid- IL2Rgamma(null) mice. Total excision of the femoral artery disrupts all connections to the pre-existing collateral bed. To form functional collaterals in this model, the pre-existing vessels need not only to enlarge their diameter (arteriogenesis), the disrupted connec- tions also require repair and sprouting of new capillaries (angiogenesis). In contrast, all side branches and pre-existing connections to the vasculature are kept intact upon double electro-coagulation of the vascular tree in the second model, which mimics the clinical situation of multi-level occlusions in PAD at best. Pre-stimulation of monocytes significantly improved revascularization in both models, which emphasizes the capacity of pre-stimulated monocytes to improve vascular regeneration.

In addition to LDPI monitoring, immunohistochemical analysis was used to examine neovascularization at the tissue level. In accordance with the LDPI data, SMA staining of the post-ischemic adductor muscle showed an increase in collateral density and collateral diameter upon transfusion of pre-stimulated monocytes as compared to un- stimulated monocytes or PBS. Moreover, a three-fold higher CD31+-capillary density was

(16)

observed after pre-stimulated monocyte-transfusion as compared to controls, which underscores the capacity of pre-stimulated monocytes to support both arterio- as well as angiogenesis in the total excision model.

As sentinels of the innate immune system monocytes efficiently communicate with T-lymphocytes. T-lymphocytes are immune cells that belong to the adaptive immune system but they also play an evident role in adult vascular repair11,  12. Hampered monocyte-T-cell interactions, as exemplified by reduced formation of CFU-Hill clusters, appear to be correlated with cardiovascular disease14. Although the extrapolation to in vivo mechanisms is still unresolved, CFU-Hill cluster formation is regarded as a marker of vascular regeneration. CFU-Hill cluster formation is efficiently mimicked by purified monocytes that are stimulated by soluble T-cell-derived factors13. In this study we show the effectiveness of this pre-stimulation to direct monocyte differentiation in a pro- angio/arteriogenic-supporting pathway. Cell surface expression profiles of lineage- and subset-specific markers indicate that these pro-angio/ arteriogenic monocytes do not develop into EPC or EC-like cells. Moreover, they phenotypically differ from macrophages and DC.

Upon i.v. infusion the pre-stimulated monocytes were detected in the ischemic adduc- tor muscle, but did not incorporate into the vessel wall. Overall, few human cells were found suggesting that they are present transiently or act in a more systemic manner, most likely via the release of paracrine factors. In support of this idea, we show that pre- stimulated monocytes produce a wide variety of growth factors and cytokines that are known to promote neovascularization20-22 such as VEGF, PDGF-BB, TNFa, IL-1b, IL-4 and IL-13. Other factors such as G-CSF and IL-8 may additionally favor neovascularization in- directly by stimulating mobilization of leukocytes and EPC from the BM. Fortunately, no increase was observed of the pro-inflammatory cytokine IL12. However, the increased IFNg production warrants some precaution: IFNg positively affects myeloid cell prolif- eration, but has been shown to directly induce arteriosclerosis of the vessel wall23.

Although effective, in the current form this approach is not yet suitable for clinical use.

To be able to replace the allogeneic T-cell-derived factors we will need to delineate those factors that drive pro-angio/ arteriogenic monocyte differentiation. This additionally may open up vistas to pharmacological solutions that support pro-angio/ arteriogenic monocyte differentiation in vivo. Furthermore, in the hind limb ischemia models used it is presently not clear by which mechanism the transfused monocytes contribute to the improved neovascularization. The involvement of transfused cultured monocytes might differ from that of endogenous uncultured monocytes that normally circulate in the peripheral blood. To optimize the current strategy further studies will be required to identify the exact role of the transfused monocyte

(17)

Overall, our results support a beneficial role for pre-stimulated monocytes in vascular regeneration and render this pro-angio/ arteriogenic monocyte an interesting candi- date for regenerative cell therapy that promotes revascularization in patients with PAD.

ACKNOWLEDGEMENTS

The authors would like to thank E. Mul for technical assistance and Dr. J.J. Bajramovic for critically reading this manuscript. The authors gratefully acknowledge the financial support of the Translational Excellence in Regenerative Medicine (TeRM) Smart Mix Program of the Netherlands Ministry of Economic Affairs and the Netherlands Ministry of Education, Culture and Science.

Conflict-of-interest disclosure: The authors declare that they have no competing finan- cial interests.

(18)

REFERENCE LIST

1. Norgren L, Hiatt WR, Dormandy JA, Nehler MR, Harris KA, Fowkes FG. Inter-Society Consensus for the Management of Peripheral Arterial Disease (TASC II). J Vasc Surg 2007 Jan;45 Suppl S:S5-67.

2. Takahashi T, Kalka C, Masuda H, Chen D, Silver M, Kearney M, et al. Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nat Med 1999 Apr;5(4):434-8.

3. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, et al. Isolation of putative progeni- tor endothelial cells for angiogenesis. Science 1997 Feb 14;275(5302):964-7.

4. Van Tongeren RB, Hamming JF, Fibbe WE, van Weel V, Frerichs SJ, Stiggelbout AM, et al. Intra- muscular or combined intramuscular/intra-arterial administration of bone marrow mononuclear cells: a clinical trial in patients with advanced limb ischemia. J Cardiovasc Surg (Torino) 2008 Feb;49(1):51-8.

5. Bartsch T, Falke T, Brehm M, Zeus T, Kogler G, Wernet P, et al. [Transplantation of autologous adult bone marrow stem cells in patients with severe peripheral arterial occlusion disease]. Med Klin (Munich) 2006 Mar 22;101 Suppl 1:195-7.

6. Gu YQ, Zhang J, Guo LR, Qi LX, Zhang SW, Xu J, et al. Transplantation of autologous bone marrow mononuclear cells for patients with lower limb ischemia. Chin Med J (Engl ) 2008 Jun 5;121(11):963-7.

7. Matoba S, Tatsumi T, Murohara T, Imaizumi T, Katsuda Y, Ito M, et al. Long-term clinical outcome after intramuscular implantation of bone marrow mononuclear cells (Therapeutic Angiogenesis by Cell Transplantation [TACT] trial) in patients with chronic limb ischemia. Am Heart J 2008 Nov;156(5):1010-8.

8. Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S, Masaki H, et al. Therapeutic angiogenesis for patients with limb ischaemia by autologous transplantation of bone-marrow cells: a pilot study and a randomised controlled trial. Lancet 2002 Aug 10;360(9331):427-35.

9. Herold J, Pipp F, Fernandez B, Xing Z, Heil M, Tillmanns H, et al. Transplantation of monocytes:

a novel strategy for in vivo augmentation of collateral vessel growth. Hum Gene Ther 2004 Jan;15(1):1-12.

10. Urbich C, Heeschen C, Aicher A, Dernbach E, Zeiher AM, Dimmeler S. Relevance of monocytic features for neovascularization capacity of circulating endothelial progenitor cells. Circulation 2003 Nov 18;108(20):2511-6.

11. van Weel V, Toes RE, Seghers L, Deckers MM, de Vries MR, Eilers PH, et al. Natural killer cells and CD4+ T-cells modulate collateral artery development. Arterioscler Thromb Vasc Biol 2007 Nov;27(11):2310-8.

12. Stabile E, Burnett MS, Watkins C, Kinnaird T, Bachis A, la Sala A, et al. Impaired arteriogenic re- sponse to acute hindlimb ischemia in CD4-knockout mice. Circulation 2003 Jul 15;108(2):205-10.

13. van Beem RT, Noort WA, Voermans C, Kleijer M, ten BA, van Ham SM, et al. The presence of ac- tivated CD4(+) T cells is essential for the formation of colony-forming unit-endothelial cells by CD14(+) cells. J Immunol 2008 Apr 1;180(7):5141-8.

14. Hill JM, Zalos G, Halcox JP, Schenke WH, Waclawiw MA, Quyyumi AA, et al. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med 2003 Feb 13;348(7):593- 600.

15. Rohde E, Malischnik C, Thaler D, Maierhofer T, Linkesch W, Lanzer G, et al. Blood monocytes mimic endothelial progenitor cells. Stem Cells 2006 Feb;24(2):357-67.

(19)

16. Yoder MC, Mead LE, Prater D, Krier TR, Mroueh KN, Li F, et al. Redefining endothelial progeni- tor cells via clonal analysis and hematopoietic stem/progenitor cell principals. Blood 2007 Mar 1;109(5):1801-9.

17. Hellingman AA, Bastiaansen AJ, de Vries MR, Seghers L, Lijkwan MA, Lowik CW, et al. Variations in Surgical Procedures for Hind Limb Ischaemia Mouse Models Result in differences in Collateral Formation. Eur J Vasc Endovasc Surg 2010;40(6):796-803.

18. van Weel V, Deckers MM, Grimbergen JM, van Leuven KJ, Lardenoye JH, Schlingemann RO, et al.

Vascular endothelial growth factor overexpression in ischemic skeletal muscle enhances myoglo- bin expression in vivo. Circ Res 2004 Jul 9;95(1):58-66.

19. Kato K, Takaue Y, Wakasugi H. T-cell-conditioned medium efficiently induces the maturation and function of human dendritic cells. J Leukoc Biol 2001 Dec;70(6):941-9.

20. Kano MR, Morishita Y, Iwata C, Iwasaka S, Watabe T, Ouchi Y, et al. VEGF-A and FGF-2 synergistically promote neoangiogenesis through enhancement of endogenous PDGF-B-PDGFRbeta signaling.

J Cell Sci 2005 Aug 15;118(Pt 16):3759-68.

21. Naldini A, Leali D, Pucci A, Morena E, Carraro F, Nico B, et al. Cutting edge: IL-1beta mediates the proangiogenic activity of osteopontin-activated human monocytes. J Immunol 2006 Oct 1;177(7):4267-70.

22. Fukushi J, Ono M, Morikawa W, Iwamoto Y, Kuwano M. The activity of soluble VCAM-1 in angio- genesis stimulated by IL-4 and IL-13. J Immunol 2000 Sep 1;165(5):2818-23.

23. Tellides G, Tereb DA, Kirkiles-Smith NC, Kim RW, Wilson JH, Schechner JS, et al. Interferon-gamma elicits arteriosclerosis in the absence of leukocytes. Nature 2000 Jan 13;403(6766):207-11.

Referenties

GERELATEERDE DOCUMENTEN

Cells from the bone marrow can go to the damaged kidney and directly differentiate into TEC (22) or myofibroblasts (12). There are multiple BMDC populations

License: Licence agreement concerning inclusion of doctoral thesis in the Institutional Repository of the University of Leiden Downloaded.

Molecular imaging of bone marrow mononuclear cell survival and homing in a murine model of peripheral artery disease. JACC

An alternative model of double electro-coagulation of both the femoral artery and iliac artery for test- ing new therapeutic approaches is studied, since the extremely fast blood

The role for NK cells in arteriogenesis was established when these cells were found to accumulate around collateral arteries and by the observation of impaired arteriogenesis

Blood flow recovery after a single femoral artery (distal anatomical level) electro-coagulation (n=3, green line) or a single iliac artery (proximal anatomical

Interestingly, we observed a relatively limited cell survival after intramuscular injec- tion in the adductor muscle. After a short-term post-MNC-transplantation increase in BLI

Although the present study showed that monocytes are present near collaterals in the post-ischemic adductor muscle of PAR2 -/- mice, impaired activated Ly6C lo monocytes in