critical roles of muscle-secreted angiogenic factors in...

23
Critical Roles of Muscle-Secreted Angiogenic Factors in Therapeutic Neovascularization Kaoru Tateno,* Tohru Minamino,* Haruhiro Toko, Hiroshi Akazawa, Naomi Shimizu, Shinichi Takeda, Takeshige Kunieda, Hideyuki Miyauchi, Tomomi Oyama, Katsuhisa Matsuura, Jun-ichiro Nishi, Yoshio Kobayashi, Toshio Nagai, Yoichi Kuwabara, Yoichiro Iwakura, Fumio Nomura, Yasushi Saito, Issei Komuro Abstract—The discovery of bone marrow– derived endothelial progenitors in the peripheral blood has promoted intensive studies on the potential of cell therapy for various human diseases. Accumulating evidence has suggested that implantation of bone marrow mononuclear cells effectively promotes neovascularization in ischemic tissues. It has also been reported that the implanted cells are incorporated not only into the newly formed vessels but also secrete angiogenic factors. However, the mechanism by which cell therapy improves tissue ischemia remains obscure. We enrolled 29 “no-option” patients with critical limb ischemia and treated ischemic limbs by implantation of peripheral mononuclear cells. Cell therapy using peripheral mononuclear cells was very effective for the treatment of limb ischemia, and its efficacy was associated with increases in the plasma levels of angiogenic factors, in particular interleukin-1 (IL-1). We then examined an experimental model of limb ischemia using IL-1– deficient mice. Implantation of IL-1– deficient mononuclear cells improved tissue ischemia as efficiently as that of wild-type cells. Both wild-type and IL-1– deficient mononuclear cells increased expression of IL-1 and thus induced angiogenic factors in muscle cells of ischemic limbs to a similar extent. In contrast, inability of muscle cells to secrete IL-1 markedly reduces induction of angiogenic factors and impairs neovascularization by cell implantation. Implanted cells do not secret angiogenic factors sufficient for neovascularization but, instead, stimulate muscle cells to produce angiogenic factors, thereby promoting neovascularization in ischemic tissues. Further studies will allow us to develop more effective treatments for ischemic vascular disease. (Circ Res. 2006;98:1194-1202.) Key Words: angiogenesis interleukins muscles P eripheral vascular disease (PVD), mainly caused by atherosclerosis, leads to obstruction of the blood supply to the lower or upper extremities. PVD is known to affect 10% to 15% of the adult population in developed countries and is often associated with coronary artery disease. 1 Arte- riosclerosis obliterans (ASO) is the most common cause of PVD affecting the lower limbs. Peripheral ischemia can also result from various types of vasculitis, including thromboan- giitis obliterans (TAO) or Buerger’s disease, which affects small- and medium-sized arteries and is related to tobacco use and male sex but not to other coronary risk factors. The 2 cardinal symptoms of limb ischemia are intermittent claudi- cation and rest pain: the latter symptom occurs in patients with critical limb ischemia and coincides with ischemic ulceration and gangrene. The treatments of PVD include pharmacotherapy, percutaneous transluminal angioplasty, and vascular surgery and are chosen depending on the severity of the symptoms and the arteries involved. 2 How- ever, as many as 50% of patients with critical limb ischemia will undergo limb amputation within 1 year because of an insufficient response to the treatments. 1,2 Recent progress in understanding the mechanisms under- lying vascular formation in adults as well as during embry- ogenesis has opened up a therapeutic avenue for patients without any current options. 3 Initial therapeutic approaches were aimed at delivering angiogenic factors, such as vascular endothelial growth factors (VEGF) and fibroblast growth factor-2, to ischemic tissues by using recombinant proteins or vectors encoding these factors. 4,5 A number of preclinical studies reported improvement of perfusion by such methods in animal models of limb ischemia. 6,7 Although the initial nonrandomized clinical trials showed beneficial effects, the results of controlled clinical trials have not been consistent. 7 More recently, bone marrow– derived circulating endothelial Original received November 1, 2005; revision received March 9, 2006; accepted March 22, 2006. From the Departments of Cardiovascular Science and Medicine (K.T., T.M., H.T., H.A., S.T., T.K., H.M., T.O., K.M., J.-i.N., Y. Kobayashi, T.N., Y. Kuwabara, I.K.), Clinical Cell Biology (N.S., Y.S.), and Molecular Diagnosis (F.N.), Chiba University Graduate School of Medicine, Japan; and Division of Cell Biology (Y.I.), Center for Experimental Medicine, Institute of Medical Science, University of Tokyo, Japan. *Both authors contributed equally to this study. Correspondence to Issei Komuro, MD, PhD, Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan. E-mail [email protected] © 2006 American Heart Association, Inc. Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/01.RES.0000219901.13974.15 1194 by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from by guest on June 27, 2018 http://circres.ahajournals.org/ Downloaded from

Upload: buicong

Post on 22-May-2018

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

Critical Roles of Muscle-Secreted Angiogenic Factors inTherapeutic Neovascularization

Kaoru Tateno,* Tohru Minamino,* Haruhiro Toko, Hiroshi Akazawa, Naomi Shimizu,Shinichi Takeda, Takeshige Kunieda, Hideyuki Miyauchi, Tomomi Oyama, Katsuhisa Matsuura,

Jun-ichiro Nishi, Yoshio Kobayashi, Toshio Nagai, Yoichi Kuwabara, Yoichiro Iwakura,Fumio Nomura, Yasushi Saito, Issei Komuro

Abstract—The discovery of bone marrow–derived endothelial progenitors in the peripheral blood has promoted intensivestudies on the potential of cell therapy for various human diseases. Accumulating evidence has suggested thatimplantation of bone marrow mononuclear cells effectively promotes neovascularization in ischemic tissues. It has alsobeen reported that the implanted cells are incorporated not only into the newly formed vessels but also secreteangiogenic factors. However, the mechanism by which cell therapy improves tissue ischemia remains obscure. Weenrolled 29 “no-option” patients with critical limb ischemia and treated ischemic limbs by implantation of peripheralmononuclear cells. Cell therapy using peripheral mononuclear cells was very effective for the treatment of limbischemia, and its efficacy was associated with increases in the plasma levels of angiogenic factors, in particularinterleukin-1� (IL-1�). We then examined an experimental model of limb ischemia using IL-1�–deficient mice.Implantation of IL-1�–deficient mononuclear cells improved tissue ischemia as efficiently as that of wild-type cells.Both wild-type and IL-1�–deficient mononuclear cells increased expression of IL-1� and thus induced angiogenicfactors in muscle cells of ischemic limbs to a similar extent. In contrast, inability of muscle cells to secrete IL-1�markedly reduces induction of angiogenic factors and impairs neovascularization by cell implantation. Implanted cellsdo not secret angiogenic factors sufficient for neovascularization but, instead, stimulate muscle cells to produceangiogenic factors, thereby promoting neovascularization in ischemic tissues. Further studies will allow us to developmore effective treatments for ischemic vascular disease. (Circ Res. 2006;98:1194-1202.)

Key Words: angiogenesis � interleukins � muscles

Peripheral vascular disease (PVD), mainly caused byatherosclerosis, leads to obstruction of the blood supply

to the lower or upper extremities. PVD is known to affect10% to 15% of the adult population in developed countriesand is often associated with coronary artery disease.1 Arte-riosclerosis obliterans (ASO) is the most common cause ofPVD affecting the lower limbs. Peripheral ischemia can alsoresult from various types of vasculitis, including thromboan-giitis obliterans (TAO) or Buerger’s disease, which affectssmall- and medium-sized arteries and is related to tobacco useand male sex but not to other coronary risk factors. The 2cardinal symptoms of limb ischemia are intermittent claudi-cation and rest pain: the latter symptom occurs in patientswith critical limb ischemia and coincides with ischemiculceration and gangrene. The treatments of PVD includepharmacotherapy, percutaneous transluminal angioplasty,and vascular surgery and are chosen depending on the

severity of the symptoms and the arteries involved.2 How-ever, as many as 50% of patients with critical limb ischemiawill undergo limb amputation within 1 year because of aninsufficient response to the treatments.1,2

Recent progress in understanding the mechanisms under-lying vascular formation in adults as well as during embry-ogenesis has opened up a therapeutic avenue for patientswithout any current options.3 Initial therapeutic approacheswere aimed at delivering angiogenic factors, such as vascularendothelial growth factors (VEGF) and fibroblast growthfactor-2, to ischemic tissues by using recombinant proteins orvectors encoding these factors.4,5 A number of preclinicalstudies reported improvement of perfusion by such methodsin animal models of limb ischemia.6,7 Although the initialnonrandomized clinical trials showed beneficial effects, theresults of controlled clinical trials have not been consistent.7

More recently, bone marrow–derived circulating endothelial

Original received November 1, 2005; revision received March 9, 2006; accepted March 22, 2006.From the Departments of Cardiovascular Science and Medicine (K.T., T.M., H.T., H.A., S.T., T.K., H.M., T.O., K.M., J.-i.N., Y. Kobayashi, T.N., Y.

Kuwabara, I.K.), Clinical Cell Biology (N.S., Y.S.), and Molecular Diagnosis (F.N.), Chiba University Graduate School of Medicine, Japan; and Divisionof Cell Biology (Y.I.), Center for Experimental Medicine, Institute of Medical Science, University of Tokyo, Japan.

*Both authors contributed equally to this study.Correspondence to Issei Komuro, MD, PhD, Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine,

1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan. E-mail [email protected]© 2006 American Heart Association, Inc.

Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/01.RES.0000219901.13974.15

1194

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 2: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

progenitors have been identified in the peripheral blood8,9 andhave been shown to contribute to both physiological andpathological angiogenesis in adults.10,11 These findings haveled to the development of therapeutic neovascularizationtechniques using endothelial progenitors. Preclinical studiesindicated that implantation of bone marrow mononuclearcells (BM-MNC), which contain endothelial progenitors, intoischemic limbs was very effective.12–14 Consequently, thistherapeutic strategy was promptly tried for “no-option” pa-tients with critical limb ischemia. The results of the firstclinical trial showed that implantation of BM-MNC signifi-cantly improved the tissue oxygen concentration and bloodflow in ischemic limbs, resulting in a decrease of rest painand the involution of ischemic ulcers.15 Although promisingresults have been obtained, the mechanism by which celltherapy improves limb ischemia is largely unknown. Becausedirect incorporation of implanted cells into newly formedvessels is reported to be relatively rare, it has been assumedthat angiogenic factors secreted by implanted cells are re-sponsible for the efficacy of cell therapy.16,17

In the present study, we investigated the efficacy of celltherapy using peripheral blood mononuclear cells (PB-MNC)for the treatment of ischemic limbs. The results of our clinicaltrial showed that increased levels of angiogenic cytokinescorrelated with the response to treatment. Implantation ofmononuclear cells increased the production of the angiogeniccytokines in muscle cells. A deficiency of the angiogeniccytokines in muscle cells blunted the ability of implantedcells to increase vascularization, suggesting that muscle cellsbut not mononuclear cells were important as a source of theangiogenic cytokines. Accordingly, we propose a novelmechanism whereby implanted cells promotes neovascular-ization in ischemic tissues.

Materials and MethodsAnimalsGeneration and genotyping of interleukin (IL)-1�–deficient micehave been previously described.18 C57BL/6 mice were purchasedfrom SLC Japan. All mice used in this study were 8 to 12 weeks old.All experimental procedures were performed according to theguidelines of Chiba University for animal experiments and theprotocols were approved by our institutional review board.

Collection of Mouse PB-MNC and BM-MNCDonor mice were euthanized with a lethal dose of anesthetic, afterwhich whole blood or bone marrow were harvested. Mononuclearcells were subsequently separated using Histopaque 1083 (Sigma).To deplete putative endothelial progenitors, the mononuclear cellswere further incubated with a rat anti-mouse VEGF receptor-2antibody (eBiosciences) at a concentration of 0.5 �g per 1 millioncells. Then the population of cells negative for VEGF receptor-2 wasobtained using a magnetic cell sorter (Miltenyi Biotec) according tothe instructions of the manufacturer. The control cell population wasprepared using an isotype control anti-rat IgG2a antibody. Sortedcells were washed twice with PBS and resuspended in PBS at aconcentration of 1 million cells per 100 �L for the subsequentexperiments. In all experiments, the viability of the mononuclearcells was more than 96.0%, as judged by Trypan blue dye exclusion.

Hindlimb Ischemia ModelAfter the mice were anesthetized, the proximal part of the femoralartery and the distal portion of the saphenous artery were ligated andstripped out after all side branches were dissected free. After 24

hours (designated as day 1), either mononuclear cells (1.0�106 cellsin 100 �L of PBS) or PBS (100 �L) was injected into the ischemicmuscle. As a rescue model, some of the IL-1�–deficient mice weretreated with PB-MNC on day 1 and given an intramuscular injectionof 0.5 ng of mouse recombinant IL-1� (IBL) on days 3, 5, and 7 afterthe operation. The mice were euthanized at the indicated times.Before death, hindlimb perfusion was measured with a laser Dopplerperfusion analyzer (Moor Instruments).

Bone Marrow Transplantation ModelBefore bone marrow transplantation, 8- to 10-week-old male wild-type or IL-1�–deficient mice were exposed to total body irradiation(9 mGy). Bone marrow cell suspensions were isolated by flushingthe femurs and tibias harvested from wild-type or IL-1�–deficientmice. Bone marrow cell suspensions (1.5�107 cells) were injectedintravenously via the tail vein within 6 hours of irradiation. Thechimeric rate was more than 95%, as determined by fluorescence-activated cell sorting analysis of chimeric mice transplanted withbone marrow cells from the green fluorescent protein (GFP) trans-genic mice.

HistologyVastus and rectus femolis muscle tissues were removed from theischemic limbs after systemic perfusion with PBS and immediatelyembedded in OCT compound (Sakura Finetechnical). Then eachspecimen was snap frozen in liquid nitrogen and cut into 6-�msections. The sections were stained with antibodies for myosin(MF-20), CD31 (Pharmingen), or IL-1� (Santa Cruz Biotechnology)and counterstained with hematoxylin or 4�,6-diamidino-2-phenylindole (DAPI). Two transverse sections of the entire musclewere photographed digitally at a magnification of �100 (12 to 16photographs per mouse), and these photographs were reviewed in ablinded manner. Capillary endothelial cells were identified byimmunoreactivity for CD31 and quantified as the number persquare millimeter.

Cell CultureC2C12 myoblasts were cultured as described previously.19 Briefly,cells were maintained in DMEM supplemented with 20% FBS(growth medium) until use. In all experiments, the cells were seededinto 60-mm or 100-mm plastic culture dishes at a concentration of5�103 cells/cm2 (day 0) and were incubated at 37°C in a mixture of95% air and 5% CO2. After 24 hours (day 1), the growth medium waschanged to DMEM supplemented with 2% horse serum (differenti-ation medium). On day 4, the medium was replaced by freshdifferentiation medium with or without PB-MNC. On day 5, cellswere harvested after the mononuclear cells were completely removedby washing 5 times with PBS.

Statistical AnalysisData are shown as mean�SEM unless otherwise noted. Changes ofthe rest pain scale and walking distance over time were analyzed bythe rank sum test. Differences of patient background factors betweenresponders and nonresponders were analyzed with the �2 test orFisher’s exact test. Comparison between multiple groups was per-formed by 1-way ANOVA followed by the Bonferroni procedure forcomparison of mean values. Comparisons between two groups weredone with the two-tailed Student’s t test or 2-way ANOVA. For theseanalyses, of P�0.05 was considered statistically significant.

An expanded Materials and Methods section is available in theonline data supplement at http://circres.ahajournals.org.

ResultsTherapeutic Neovascularization Using PB-MNCfor Critical Limb IschemiaBecause most patients with critical limb ischemia haveconcomitant coronary artery disease and cerebrovasculardisease, the collection of bone marrow cells, which requires

Tateno et al Mechanism of Cell Therapy 1195

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 3: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

general anesthesia,15 could cause fatal events in such patients.Given that angiogenic factors secreted from BM-MNC play amajor role in cell therapy, PB-MNC may be more beneficialbecause collection of PB-MNC is much safer and lessexpensive. To examine the effects implanting PB-MNC, weproduced a model of hindlimb ischemia in C57BL/6 mice andimplanted PB-MNC into the ischemic limbs of these animals.Implantation of PB-MNC significantly increased perfusioncompared with that in the control group (PBS) as assessed bylaser Doppler analysis (Figure 1A). The capillary density inischemic limbs was significantly greater in the PB-MNCgroup than in the PBS group (Figure 1B). Although BM-MNC has been reported to contain much more (�100-fold)endothelial progenitors than PB-MNC, there was no signifi-cant difference of neovascularization between the PB-MNCgroup and the BM-MNC group (Figure 1A and 1B), indicat-ing that implantation of PB-MNC is as efficient as BM-MNCfor the treatment of limb ischemia and suggesting thatendothelial progenitors do not play a pivotal role in thetreatment of limb ischemia at least by PB-MNC.

After obtaining approval from the ethical committee ofChiba University Graduate School of Medicine, we started a

pilot clinical trial to investigate whether therapeutic neovas-cularization by implantation of PB-MNC is feasible andeffective for ischemic limbs. We enrolled 29 patients withcritical limb ischemia caused by ASO or TAO who had notreatment options. More than 80% of the patients wererecommended to undergo major amputation by their doctorsbefore enrollment because of unhealed ischemic ulcers organgrene despite conventional therapy (Tables 1 and 2).Approximately half of the patients had chronic renal failureand were on dialysis 3 times weekly (Table 1). Apart from 5TAO subjects, most of the patients (82.7%) had 1 or morecomplications (Table 1). We implanted PB-MNC into theischemic limbs of these patients twice within a 1-monthperiod and estimated their response at 2 months, 6 months,and 1 year after treatment (as described in the online datasupplement). Rest pain was significantly decreased and wasnearly normalized by 1 year after treatment (Figure 1C). Themaximum walking distance also improved significantly, andthis improvement was preserved for a year (Figure 1D). Threepatients (11.1%) underwent major amputation, which wasmuch less frequent compared with the reported annual rate ofamputation in patients with critical limb ischemia (�50%).2

Figure 1. Effect of implanting PB-MNC. A, Limb perfusion measured by a laser Doppler analyzer at 2 weeks after treatment (photo-graphs). Scale bar�100 �m. The graph shows the ratio of ischemic limb (left) to nonischemic limb (right) blood flow. Data are shown asmean�SEM. *P�0.05, **P�0.01 vs PBS-treated group (n�8). B, Immunohistochemistry for CD31 (brown) in ischemic limbs at 2 weeksafter treatment with PBS, PB-MNC, or BM-MNC. Scale bar�100 �m. The number of CD31-positive cells per square millimeter isshown. Data are shown as mean�SEM. *P�0.05, **P�0.01 vs PBS-treated group (n�8). C, Changes of the rest pain scale afterimplantation of PB-MNC. D, Improvement of the maximum walking distance after implantation. Data are shown as mean�SEM.*P�0.001 vs 0 month; #P�0.05, ##P�0.001 vs 2 months (n�29). E, Nonhealing ischemic ulcer (left) is completely healed by 6 monthsafter implantation (right).

1196 Circulation Research May 12, 2006

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 4: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

Improvement of ischemic ulcers was observed 16 of 24patients (66.7%) (Figure 1E), whereas only 31% of thepatients showed substantial improvement of the ankle–bra-chial pressure index (ABPI), presumably because of under-estimation of improvement in this index of hemodialysispatients. There were no major complications related to thetreatment, such as death or coronary events. Three patientsdied of unrelated causes, including pneumonia and chronicrenal failure (11.1%), which was lower than the reportedannual mortality rate of patients with critical limb ischemia(�25%).1 These results indicate that implantation of PB-MNC was a safe and effective treatment for critical limbischemia.

Factors Associated With the Responseto TreatmentWe next investigated the factors related to the response totreatment. We divided the patients into a group of responders(n�21) and a group of nonresponders (n�8) according to aresponse score estimated from the improvement of rest pain,ischemic ulcers, walking distance, and ABPI. Then wecompared their background factors and laboratory data. Therewere no significant differences of patient background factors,including age, gender, and coronary risk factors, betweenresponders and nonresponders (Table 2). The serum level ofC-reactive protein, a marker of inflammation induced bycytokines, was significantly higher in the responders afterimplantation (Table 2). Therefore, we examined the plasma

levels of various angiogenic cytokines in the patients at days1, 3, 7, and 14 after implantation. We found that the peaklevels of IL-1�, IL-6, and VEGF, but not tumor necrosisfactor-� or granulocyte colony-stimulating factor, were mark-edly higher in responders than in nonresponders (Figure 2A;Figure I in the online data supplement). In contrast, therewere no significant differences of the number of implantedmononuclear cells and CD34-positive cells between the 2groups (Figure 2A). Consistent with these results, deprivationof putative endothelial progenitors (VEGF receptor-2–posi-tive cells) did not affect the ability of implanted mononuclearcells to promote neovascularization in a mouse model ofhindlimb ischemia (Figure 2B). Thus, we speculated that the

TABLE 1. Patient Background

Age, y 61.8�11.3

Gender

Male 24 (82.8%)

Female 5 (17.2%)

Diagnosis

ASO 19 (65.5%)

TAO 10 (34.5%)

Ischemic status

Fontaine 3 4 (13.8%)

Fontaine 4 25 (86.2%)

Previous treatment

PTA 7 (24.1%)

Bypass surgery 2 (6.9%)

ABPI 0.79�0.26

Rest pain scale 2.90�1.11

Complications

CRF on HD 14 (48.3%)

CAD 13 (44.8%)

CVD 8 (27.6%)

Diabetes 16 (55.2%)

Hyperlipidemia 16 (55.2%)

Hypertension 13 (44.8%)

Any complications 24 (82.7%)

Data are shown as mean�SD or the no. of patients (%). PTA indicatespercutaneous transluminal angioplasty; CRF, chronic renal failure; HD, hemo-dialysis; CAD, coronary artery disease; CVD, cerebrovascular disease.

TABLE 2. Clinical Response and Patient Background

Nonresponders(n�8)

Responders(n�21) P

Age, y 60.6�11.7 62.3�11.3 NS

Gender

Male 7 (87.5%) 17 (81.0%) NS

Female 1 (12.5%) 4 (19.0%)

Diagnosis

ASO 6 (75.0%) 13 (61.9%) NS

TAO 2 (25.0%) 8 (38.1%)

Ischemic status

Fontaine 3 1 (12.5%) 3 (14.3%) NS

Fontaine 4 7 (87.5%) 18 (85.7%) NS

Previous revascularization 3 (37.5%) 5 (23.8%) NS

Duration of illness (month) 19.6�24.0 52.1�55.8 NS

ABPI 0.85�0.20 0.77�0.28 NS

Rest pain scale 2.90�1.40 2.90�1.00 NS

Need for major amputation 8 (80.0%) 13 (61.9%) NS

Complications

CRF on HD 4 (50.0%) 10 (47.6%) NS

CAD 4 (50.0%) 9 (42.9%) NS

LVEF (%) 48.1�28.4 48.2�28.1 NS

CVD 3 (37.5%) 5 (23.8%) NS

Diabetes 6 (75.0%) 10 (47.6%) NS

Hyperlipidemia 4 (50.0%) 12 (57.1%) NS

Hypertension 4 (50.0%) 9 (42.9%) NS

MBP (mm Hg) 81.0�34.1 86.8�31.9 NS

Laboratory data

FBS (mg/dL) 139.6�45.1 133.2�49.3 NS

HbA1c (%) 5.6�0.9 5.8�0.9 NS

T-Cho (mg/dL) 169.8�33.2 184.6�37.2 NS

LDL-Cho (mg/dL) 105.6�24.6 114.8�32.7 NS

HDL-Cho (mg/dL) 43.9�10.0 49.7�10.7 NS

CRP (mg/dL) 2.9�3.8 5.4�11.0 0.018

Data are shown as mean� SD or the no. of patients (%). CRF indicateschronic renal failure; HD, hemodialysis; CAD, coronary artery disease; LVEF, leftventricular ejection fraction; CVD, cerebrovascular disease; MBP, mean bloodpressure; FBS, fasting blood sugar; T-Cho, total cholesterol; Cho, cholesterol;CRP, C-reactive protein. Patients with osteomyelitis were excluded for analysisof CRP data.

Tateno et al Mechanism of Cell Therapy 1197

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 5: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

increase of angiogenic factors secreted by implanted mono-nuclear cells might influence the efficacy of this therapy.

Increased Expression of IL-1� in Ischemic LimbsIs Crucial for NeovascularizationBecause IL-1� is known to be a potent angiogenic cytokineand induces a number of angiogenic factors includingVEGF,20,21 we examined the role of IL-1� on neovascular-ization induced by mononuclear cell implantation in a modelof hindlimb ischemia. When PB-MNC from wild-type micewere implanted into the ischemic limbs of wild-type mice,blood flow and capillary density were significantly increasedcompared with those in a PBS-treated group (Figure 3A and3B). Unexpectedly, PB-MNC derived from IL-1�–deficientmice18 increased the blood flow of the ischemic limbs inwild-type mice as efficiently as wild-type PB-MNC (Figure3A). Likewise, implantation of IL-1�–deficient PB-MNCsignificantly increased capillary density compared with thePBS-treated group (Figure 3B). In contrast, implantation ofwild-type PB-MNC into ischemic limbs of IL-1�–deficientmice had no effect on neovascularization (Figure 3A and 3B),suggesting that increased expression of IL-1� in ischemiclimbs is more important for neovascularization than expres-sion by implanted cells. Expression of VEGF in the ischemiclimbs of wild-type mice was significantly elevated by im-plantation of IL-1�–deficient PB-MNC as well as wild-typePB-MNC compared with the PBS-treated group (Figure 3C).

However, implantation of wild-type PB-MNC failed to in-duce VEGF expression in the ischemic limbs of IL-1�–deficient mice (Figure 3C). The pattern of IL-1� expressionin ischemic limbs was very similar to that of VEGF (Figure3D). A significant increase of IL-1� expression was observedin the ischemic limbs of wild-type mice treated with wild-type or IL-1�–deficient PB-MNC, whereas there was noinduction of IL-1� in the ischemic limbs of IL-1�–deficientmice after implantation of wild-type PB-MNC (Figure 3D).These results suggest that implantation of PB-MNC inducesIL-1� expression in ischemic limbs and promotes neovascu-larization by the induction of angiogenic factors such asVEGF.

Previous studies demonstrated that infiltration of macro-phages plays a critical role in IL-1�–induced neovasculariza-tion.22,23 To test this, we injected PB-MNC from GFPtransgenic mice into the ischemic limbs of wild-type orIL-1�–deficient mice and performed histological analyses at5 days after treatment. The number of infiltrating host-derived (GFP-negative) macrophages was significantly fewerin the ischemic limbs of IL-1�–deficient mice than in thoseof wild-type mice (supplemental Figure II). PB-MNC implan-tation significantly induced macrophage infiltration com-pared with PBS-treated mice (supplemental Figure II). Therewas no difference in infiltration of macrophages into nonis-chemic limbs between wild-type and IL-1�–deficient mice

Figure 2. Factors associated with the response to treatment. A, Results are shown as box plots representing median, 25th and 75thpercentiles as boxes and the range of data as bars. *P�0.05 vs nonresponders (responders, n�21; nonresponders, n�8). B, Depletionof VEGF receptor 2 (VEGFR2)-positive mononuclear cells did not affect the ability of implanted cells to promote increases of perfusion-(upper) and capillary density (lower) at 1 week after implantation. Scale bar�100 �m. R2(–): VEGFR2-depleted mononuclear cells.R2(�): control mononuclear cells. Data are shown as mean�SEM. *P�0.05, **P�0.01 vs PBS-treated group (n�8).

1198 Circulation Research May 12, 2006

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 6: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

(supplemental Figure II). Thus, infiltration of macrophagesmay be also involved in the mechanisms underlying IL-1�-induced neovascularization.

To investigate whether host-derived hematopoietic cellscontribute to induction of IL-1� by mononuclear cell implan-tation, we produced chimeric mice. When PB-MNC fromIL-1�–deficient mice were implanted into the ischemic limbsof wild-type mice with IL-1�–deficient bone marrow, bloodflow and capillary density were significantly increased com-pared with those in a PBS-treated group (Figure 3E and 3F).In contrast, injection of IL-1�–deficient PB-MNC into theischemic limbs of IL-1�–deficient mice with wild-type bonemarrow did not enhance neovascularization (Figure 3A, 3B,3E, and 3F). Consistent with these results, PB-MNC implan-tation markedly induced IL-1� expression in the ischemiclimbs of wild-type mice with IL-1�–deficient bone marrowcompared with the limbs of IL-1�–deficient mice with

wild-type bone marrow (Figure 3G), which suggests a minorrole of host-derived hematopoietic cells in the production ofIL-1� after treatment.

Critical Roles of Muscle-SecretedAngiogenic FactorsTo determine the type of cells in which IL-1� expression isinduced by implantation of PB-MNC, we performed immu-nohistochemical analysis of ischemic limbs after implanta-tion. Expression of IL-1� was localized to the skeletal musclecells, as identified by staining for sarcomeric myosin, andwas particularly detected in the regenerating tissues (Figure4A). Likewise, in situ hybridization revealed that IL-1� waspredominantly expressed by muscle cells and, to a lesserextent, by other types of cells such as vascular cells andinfiltrating leukocytes (Figure 4B and data not shown). Totest whether PB-MNC could increase the expression of

Figure 3. Impairment of PB-MNC-induced neovascularization in IL-1�–deficient mice. A, The graph shows relative blood flow in theischemic limbs of wild-type mice treated with PBS (PBS3Wild), wild-type PB-MNC (Wild3Wild), or IL-1�–deficientPB-MNC (KO3Wild) and the limbs of IL-1�–deficient mice treated with PBS (PBS3KO) or wild-type PB-MNC (Wild3KO). B, Immuno-histochemistry for CD31 in ischemic limbs. C, Expression of VEGF in ischemic limbs of the same mice at 1 week after treatment wasanalyzed by the ribonuclease protection assay (RPA). Relative expression to that of L32 (the internal control) is shown. D, Expression ofIL-1� in ischemic limbs of the same mice at 1 week after implantation was analyzed by RPA and relative expression to that of L32 isshown. ND indicates not detected. Data are shown as mean�SEM. *P�0.05, **P�0.01 vs wild-type mice treated with PBS (n�5). E, F,and G, The graph shows blood flow (E), capillary density (F), and expression of IL-1� (G) in the ischemic limbs of wild-type mice withIL-1�–deficient bone marrow treated with PBS (PBS3Wild�KO-BM) or IL-1�–deficient PB-MNC (KO3Wild�KO-BM) and the limbs ofIL-1�–deficient mice with wild-type bone marrow treated with IL-1�–deficient PB-MNC (KO3KO�Wild-BM). The values are shown asdescribed in the legend of Figure 1A for blood flow and capillary density. Data are shown as mean�SEM. *P�0.05, **P�0.01 vsPBS3Wild�KO-BM (n�5).

Tateno et al Mechanism of Cell Therapy 1199

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 7: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

angiogenic cytokines by muscle cells, we coincubated mouseembryonic muscle cells (C2C12) with PB-MNC from wild-type mice for 24 hours under differentiating conditions andharvested muscle cells after intensive wash with PBS toeliminate PB-MNC from the samples. Coincubation withPB-MNC increased the expression of various angiogeniccytokines such as IL-1�, IL-6, and VEGF by C2C12 cells(Figure 4C). C2C12 cells treated with PB-MNC as well asnontreated cells expressed myosin (Figure 4D), whereas ex-pression of CD31 was not detected in these cells (Figure 4C),indicating that contamination of PB-MNC was negligible.Coincubation of C2C12 cells with human PB-MNC alsoinduced expression of mouse cytokines as determined byRPA using a mouse-specific probe (supplemental Figure III).We, therefore, concluded that implantation of mononuclearcells promotes neovascularization by stimulating muscle cellsto increase the production of angiogenic cytokines such asIL-1�.

We then examined whether treatment with IL-1� restoresthe efficacy of PB-MNC implantation in the ischemic limbsof IL-1�–deficient mice. We observed that neovasculariza-tion occurred in the ischemic tissues at 1 week and that theappearance of the skeletal muscles became normal by 3weeks after implantation (Figure 4E). Implantation of wild-type PB-MNC did not induce neovascularization in theischemic limbs of IL-1�–deficient mice at 1 week, and, thus,necrosis occurred by 3 weeks after implantation (Figure 4E).In contrast, administration of IL-1� in conjunction withimplantation of wild-type PB-MNC significantly increasedneovascularization (P�0.01 versus IL-1�– deficient micetreated with wild-type PB-MNC; n�3) and prevented necro-sis of the ischemic limbs in IL-1�–deficient mice at 3 weeks(Figure 4E). Consistent with the results of histological anal-yses, improvement of blood flow by PB-MNC implantationin wild-type mice was significantly better than in IL-1�–deficient mice (Figure 3A and supplemental Figure IV).

Figure 4. Mononuclear cellimplantation increases the pro-duction of angiogenic factors bymuscle cells. A, Left photographshows immunohistochemistry forIL-1� (brown) in ischemic limbs at5 days after implantation. Rightphotographs indicate immuno-fluorescence for IL-1� (red),myosin (green), and the nucle-i (blue). Scale bar�50 �m. B, Insitu hybridization for IL-1� (arrow)in ischemic limbs at 5 days afterimplantation. AS indicates anti-sense probe; S, sense probe; HE,hematoxylin/eosin staining. C,C2C12 cells were cultured underdifferentiating conditions with orwithout PB-MNC (2.5�105 cells/mL) for 24 hours, and expressionof angiogenic cytokines wasexamined by RPA. Glyceralde-hyde-3-phosphate dehydrogenas-e (GAPDH) was used as the con-trol. D, C2C12 cells with orwithout PB-MNC were examinedfor myosin expression by Westernblot analysis. Actin was used asthe loading control. Undifferenti-ated C2C12 cells were used as anegative control (N). E, Immuno-histochemistry for CD31 in theischemic limbs of wild-type micetreated with wild-typePB-MNC (Wild3Wild) and thelimbs of IL-1�–deficient micetreated with wild-typePB-MNC (Wild3KO), or wild-typePB-MNC plus IL-1� (Wild�IL-1�3KO) at 1 week (left) and 3weeks (right) after implantation.Scale bar�100 �m.

1200 Circulation Research May 12, 2006

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 8: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

Moreover, administration of IL-1� in conjunction with im-plantation of wild-type PB-MNC partially improved theeffect of PB-MNC implantation on blood flow in IL-1�–deficient mice (supplemental Figure IV). However, adminis-tration of IL-1� without PB-MNC implantation did notimprove limb ischemia in wild-type mice (supplemental Fig-ure V). Thus, it is conceivable that induction of IL-1� isprerequisite for the efficacy of PB-MNC implantation, but anIL-1� only treatment is insufficient for limb salvage.

DiscussionThe present study provided a possible mechanism for theprocess of neovascularization during cell therapy for limbischemia. The results of our clinical trial showed that angio-genic cytokines, especially IL-1�, were associated with theresponse to treatment. Many previous studies have suggestedthat angiogenic factors secreted by implanted cells play acritical role in therapeutic neovascularization.16,17 In contrast,our in vitro and in vivo studies demonstrated that theimplanted mononuclear cells did not secrete cytokines suffi-cient for neovascularization but, instead, stimulated musclecells to produce IL-1�. This is consistent with our observa-tion that most of the implanted cells disappeared from theischemic tissues as early as 3 days after implantation, whichwas before reconstruction of the vascular system started(K.T., T.M., I.K., unpublished data, 2006). Thus, it is likelythat muscle cells but not implanted cells are a major source ofangiogenic cytokines in ischemic limbs.

We noted that IL-1� was predominantly expressed bymyocytes with central nuclei in ischemic limbs, indicatingthat these cells are regenerating myocytes (Figure 4A). Wealso observed that implantation of mononuclear cells in-creased the number of central nucleated myocytes in ischemiclimbs (K.T., T.M., I.K., unpublished data, 2006). We there-fore propose a novel model of neovascularization, in whichimplanted mononuclear cells enhance muscle regenerationand the increased expression of IL-1� by regenerating myo-cytes promotes neovascularization via induction of angio-genic factors, thereby contributing to limb salvage. Implan-tation of wild-type mononuclear cells increased the numberof regenerating myocytes in the ischemic limbs of IL-1�–deficient mice compared with PBS-treated IL-1�–deficientmice (Figure 4E and supplemental Figure VI). However, theregeneration of myocytes was not accompanied by neovas-cularization at 1 week, and, thus, necrosis occurred by 3weeks after implantation (Figure 4E). These results suggestthat implantation of mononuclear cells promotes muscleregeneration and that the regenerated muscle-secreted IL-1�induces neovascularization, leading to suitable conditions forfurther muscle regeneration. Thus, although expression ofIL-1� by muscle cells is required for mononuclear cellspromoting neovascularization, administration of IL-1� couldnot obviate the need for injection of mononuclear cells.

We recently reported a similar finding that granulocytecolony-stimulating factor acts directly on cardiomyocytes andpromotes neovascularization in ischemic myocardium byinducing the production of angiogenic factors.24 Our resultsalso coincide with recent evidence suggesting an importantrole for the interaction between macrophages and skeletal

muscle in the process of muscle regeneration after injury.25

During this process, muscle cells modulate macrophageinvasion by releasing cytokines such as monocyte chemoat-tractant protein-1 and VEGF.26 On the other hand, macro-phages release factors in vitro that promote the proliferationof muscle cells.27 Macrophages are also present in vivo atsites where muscle regeneration occurs,28 and macrophagedepletion significantly impairs muscle regeneration aftertransplantation of myogenic cells,29 suggesting a critical roleof macrophages in muscle repair. However, there are nodefinitive studies to show which factors (if any) are releasedby macrophages in vivo to influence the process of musclerepair. In our study, it is unclear how the implanted mono-nuclear cells stimulated muscle cells to proliferate and inducethe expression of angiogenic factors. It would be also inter-esting to determine whether the similar mechanisms areinvolved in other modes of therapeutic vascularization suchas cytokine therapy. Further studies on the roles of cellularcommunications will allow us to develop more effectivetreatments for ischemic vascular disease.

AcknowledgmentsThis work was supported by a grant-in-aid for scientific research anddevelopmental scientific research from the Ministry of Education,Science, Sports, and Culture, Health and Labor Sciences ResearchGrants (to I.K.) and grants from the Japan Research Foundation forClinical Pharmacology, NOVARTIS foundation, and the Ministry ofEducation, Science, Sports, and Culture of Japan (to T.M.).

References1. Braunwald E, Zipes DP, Libby P. Heart Disease: A Textbook of Cardio-

vascular Medicine. 6th ed. Philadelphia: Saunders; 2001.2. Dormandy JA, Rutherford RB. Management of peripheral arterial disease

(PAD). TASC Working Group. TransAtlantic Inter-Society Concensus(TASC). J Vasc Surg. 2000;31:S1–S296.

3. Carmeliet P. Mechanisms of angiogenesis and arteriogenesis. Nat Med.2000;6:389–395.

4. Takeshita S, Zheng LP, Brogi E, Kearney M, Pu LQ, Bunting S, FerraraN, Symes JF, Isner JM. Therapeutic angiogenesis. A single intraarterialbolus of vascular endothelial growth factor augments revascularization ina rabbit ischemic hind limb model. J Clin Invest. 1994;93:662–670.

5. Harada K, Grossman W, Friedman M, Edelman ER, Prasad PV, KeighleyCS, Manning WJ, Sellke FW, Simons M. Basic fibroblast growth factorimproves myocardial function in chronically ischemic porcine hearts.J Clin Invest. 1994;94:623–630.

6. Ferrara N, Alitalo K. Clinical applications of angiogenic growth factorsand their inhibitors. Nat Med. 1999;5:1359–1364.

7. Yla-Herttuala S, Alitalo K. Gene transfer as a tool to induce therapeuticvascular growth. Nat Med. 2003;9:694–701.

8. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T,Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitorendothelial cells for angiogenesis. Science. 1997;275:964–967.

9. Shi Q, Rafii S, Wu MH, Wijelath ES, Yu C, Ishida A, Fujita Y, KothariS, Mohle R, Sauvage LR, Moore MA, Storb RF, Hammond WP.Evidence for circulating bone marrow-derived endothelial cells. Blood.1998;92:362–367.

10. Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M,Kearne M, Magner M, Isner JM. Bone marrow origin of endothelialprogenitor cells responsible for postnatal vasculogenesis in physiologicaland pathological neovascularization. Circ Res. 1999;85:221–228.

11. Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation fororgan vascularization and regeneration. Nat Med. 2003;9:702–712.

12. Ikenaga S, Hamano K, Nishida M, Kobayashi T, Li TS, Kobayashi S,Matsuzaki M, Zempo N, Esato K. Autologous bone marrow implantationinduced angiogenesis and improved deteriorated exercise capacity in a ratischemic hindlimb model. J Surg Res. 2001;96:277–283.

Tateno et al Mechanism of Cell Therapy 1201

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 9: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

13. Shintani S, Murohara T, Ikeda H, Ueno T, Sasaki K, Duan J, Imaizumi T.Augmentation of postnatal neovascularization with autologous bonemarrow transplantation. Circulation. 2001;103:897–903.

14. Li TS, Hamano K, Suzuki K, Ito H, Zempo N, Matsuzaki M. Improvedangiogenic potency by implantation of ex vivo hypoxia prestimulatedbone marrow cells in rats. Am J Physiol Heart Circ Physiol. 2002;283:H468–H473.

15. Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S,Masaki H, Amano K, Kishimoto Y, Yoshimoto K, Akashi H, Shimada K,Iwasaka T, Imaizumi T. Therapeutic angiogenesis for patients with limbischaemia by autologous transplantation of bone-marrow cells: a pilotstudy and a randomised controlled trial. Lancet. 2002;360:427–435.

16. Urbich C, Dimmeler S. Endothelial progenitor cells: characterization androle in vascular biology. Circ Res. 2004;95:343–353.

17. Kinnaird T, Stabile E, Burnett MS, Epstein SE. Bone-marrow-derivedcells for enhancing collateral development: mechanisms, animal data, andinitial clinical experiences. Circ Res. 2004;95:354–363.

18. Horai R, Asano M, Sudo K, Kanuka H, Suzuki M, Nishihara M,Takahashi M, Iwakura Y. Production of mice deficient in genes forinterleukin (IL)-1alpha, IL-1beta, IL-1alpha/beta, and IL-1 receptor an-tagonist shows that IL-1beta is crucial in turpentine-induced fever devel-opment and glucocorticoid secretion. J Exp Med. 1998;187:1463–1475.

19. Takahashi A, Kureishi Y, Yang J, Luo Z, Guo K, Mukhopadhyay D,Ivashchenko Y, Branellec D, Walsh K. Myogenic Akt signaling regulatesblood vessel recruitment during myofiber growth. Mol Cell Biol. 2002;22:4803–4814.

20. Voronov E, Shouval DS, Krelin Y, Cagnano E, Benharroch D, IwakuraY, Dinarello CA, Apte RN. IL-1 is required for tumor invasiveness andangiogenesis. Proc Natl Acad Sci U S A. 2003;100:2645–2650.

21. El Awad B, Kreft B, Wolber EM, Hellwig-Burgel T, Metzen E, FandreyJ, Jelkmann W. Hypoxia and interleukin-1beta stimulate vascular endo-thelial growth factor production in human proximal tubular cells. KidneyInt. 2000;58:43–50.

22. Nakao S, Kuwano T, Tsutsumi-Miyahara C, Ueda S, Kimura YN,Hamano S, Sonoda KH, Saijo Y, Nukiwa T, Strieter RM, Ishibashi T,Kuwano M, Ono M. Infiltration of COX-2-expressing macrophages is aprerequisite for IL-1beta-induced neovascularization and tumor growth.J Clin Invest. 2005;115:2979–2991.

23. Saijo Y, Tanaka M, Miki M, Usui K, Suzuki T, Maemondo M, Hong X,Tazawa R, Kikuchi T, Matsushima K, Nukiwa T. Proinflammatory cy-tokine IL-1 beta promotes tumor growth of Lewis lung carcinoma byinduction of angiogenic factors: in vivo analysis of tumor-stromal inter-action. J Immunol. 2002;169:469–475.

24. Harada M, Qin Y, Takano H, Minamino T, Zou Y, Toko H, Ohtsuka M,Matsuura K, Sano M, Nishi J, Iwanaga K, Akazawa H, Kunieda T, ZhuW, Hasegawa H, Kunisada K, Nagai T, Nakaya H, Yamauchi-TakiharaK, Komuro I. G-CSF prevents cardiac remodeling after myocardialinfarction by activating the Jak-Stat pathway in cardiomyocytes. NatMed. 2005;11:305–311.

25. Tidball JG. Inflammatory processes in muscle injury and repair. Am JPhysiol Regul Integr Comp Physiol. 2005;288:R345–R353.

26. Chazaud B, Sonnet C, Lafuste P, Bassez G, Rimaniol AC, Poron F,Authier FJ, Dreyfus PA, Gherardi RK. Satellite cells attract monocytesand use macrophages as a support to escape apoptosis and enhancemuscle growth. J Cell Biol. 2003;163:1133–1143.

27. Massimino ML, Rapizzi E, Cantini M, Libera LD, Mazzoleni F, Arslan P,Carraro U. ED2� macrophages increase selectively myoblast prolif-eration in muscle cultures. Biochem Biophys Res Commun. 1997;235:754–759.

28. Pimorady-Esfahani A, Grounds MD, McMenamin PG. Macrophages anddendritic cells in normal and regenerating murine skeletal muscle. MuscleNerve. 1997;20:158–166.

29. Lescaudron L, Peltekian E, Fontaine-Perus J, Paulin D, Zampieri M,Garcia L, Parrish E. Blood borne macrophages are essential for thetriggering of muscle regeneration following muscle transplant. Neuro-muscul Disord. 1999;9:72–80.

1202 Circulation Research May 12, 2006

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 10: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

Nomura, Yasushi Saito and Issei KomuroJun-ichiro Nishi, Yoshio Kobayashi, Toshio Nagai, Yoichi Kuwabara, Yoichiro Iwakura, Fumio

Takeda, Takeshige Kunieda, Hideyuki Miyauchi, Tomomi Oyama, Katsuhisa Matsuura, Kaoru Tateno, Tohru Minamino, Haruhiro Toko, Hiroshi Akazawa, Naomi Shimizu, ShinichiCritical Roles of Muscle-Secreted Angiogenic Factors in Therapeutic Neovascularization

Print ISSN: 0009-7330. Online ISSN: 1524-4571 Copyright © 2006 American Heart Association, Inc. All rights reserved.is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231Circulation Research

doi: 10.1161/01.RES.0000219901.13974.152006;98:1194-1202; originally published online March 30, 2006;Circ Res. 

http://circres.ahajournals.org/content/98/9/1194World Wide Web at:

The online version of this article, along with updated information and services, is located on the

http://circres.ahajournals.org/content/suppl/2006/03/30/01.RES.0000219901.13974.15.DC1Data Supplement (unedited) at:

  http://circres.ahajournals.org//subscriptions/

is online at: Circulation Research Information about subscribing to Subscriptions: 

http://www.lww.com/reprints Information about reprints can be found online at: Reprints:

  document. Permissions and Rights Question and Answer about this process is available in the

located, click Request Permissions in the middle column of the Web page under Services. Further informationEditorial Office. Once the online version of the published article for which permission is being requested is

can be obtained via RightsLink, a service of the Copyright Clearance Center, not theCirculation Researchin Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions:

by guest on June 27, 2018http://circres.ahajournals.org/

Dow

nloaded from

Page 11: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

Online Data Supplement K Tateno, et al. Mechanism of cell therapy

1

Materials and Methods

Patients

We enrolled patients aged 20 years or older who suffered from ischemic rest pain and/or

non-healing ischemic ulceration due to ASO or TAO despite conventional therapy,

including nonsurgical and surgical revascularization. Before entry into this study, we

performed physical examination, laboratory tests, X-rays, electrocardiography,

echocardiography, carotid artery ultrasonography, cardiac scintigraphy, and coronary

angiography in all candidates. They also underwent arteriography of the lower

extremities, measurement of ABPI, a laser Doppler study, and thermography to confirm

the existence of PVD unsuitable for non-surgical or surgical revascularization. We

excluded patients with proliferative diabetic retinopathy or those with malignancy

during the previous 5 years. We also excluded patients with limb ischemia due to

causes other than ASO or TAO, such as collagen vascular disease. Patient enrolment

was completed by May 2004, and a total of 29 patients underwent therapeutic

neovascularization by implantation of PB-MNC. We obtained written informed

consent from all of these patients. The ethical committee of Chiba University

Graduate School of Medicine reviewed and approved the study protocol.

Study design

The primary outcome was the safety of the treatment based on the occurrence of

adverse events related to the treatment. Secondary outcomes were the incidence of

major amputation of treated limbs, improvement of rest pain, walking distance,

ischemic ulcers (more than 25% reduction in size), and ABPI (an increase of more than

0.1). Rest pain scale was determined as follows: +4, severe pain unsolved with

morphine, pentazocine, or non-steroidal anti-inflammatory drugs (NSAID); +3,

moderate pain NSAID necessary; +2, slight pain NSAID unnecessary; +1, very slight

Page 12: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

Online Data Supplement K Tateno, et al. Mechanism of cell therapy

2

pain; 0, complete resolved. Non-responder was defined as a patient who revealed no

improvement of rest pain, walking distance, ischemic ulcers, or ABPI. We performed

follow-up examinations at 2 months, 6 months, and 12 months after treatment. We

also monitored adverse events, including the progression of atherosclerosis, by the

examinations described above.

Procedure for clinical implantation of PB-MNC

PB-MNC were harvested by peripheral blood apheresis with a COBE Spectra Apheresis

System (Gambro). The system was operated in the manual mode with the target

haematosis being set at 2.0–3.0%. Patient underwent apheresis for 240 min while

awake and resting quietly. When the patient had severe anaemia (hemoglobin<7.4

g/dl) or left ventricular dysfunction (ejection fraction<45%), the apheresis time was

reduced to 180 min. The total leukocyte count and mononuclear cell count in the

apheresis product was 1.75×1010±0.73 (mean±SD) and 1.09×1010±0.39 (mean±SD),

respectively. Part of the product was subjected to FACS analysis to determine the

number of CD34-positive cells. The product was concentrated to 20 ml by

centrifugation at 2,000×g for 10 min and then was implanted on the same day.

PB-MNC were injected intramuscularly into the ischemic lower extremities at sites 1 to

3 cm apart, with each injection being 0.1 to 0.3 ml in volume. We examined the

plasma levels of VEGF, interleukin (IL)-1β, IL-6, tumor necrosis factorα (TNFα), and

granulocyte colony-stimulating factor (G-CSF) in the patients by using human-specific

ELISA (R & D systems). We measured these cytokines at 1, 3, 7, and 14 days after

treatment and used the peak levels of cytokines for a comparison between responder and

non-responder because the time point at which each cytokine reached its maximum

varied.

Page 13: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

Online Data Supplement K Tateno, et al. Mechanism of cell therapy

3

Western blot analysis

Samples were prepared in lysis buffer (10 mM Tris-HCL, pH 8, 140 mM NaCl, 5 mM

EDTA, 0.025% NaN3, 1% Triton X-100, 1% deoxycholate, 0.1% SDS, 1 mM PMSF, 5

µg/ml leupetin, 2 µg/ml aprotinin, 50 mM NaF, and 1 mM Na2VO3). The lysates (30

µg) were resolved by SDS polyacrylamide gel electrophoresis. Proteins were

transferred to a polyvinylidine difluoride membrane (Millipore) and incubated with the

primary antibody followed by a horseradish anti-mouse immunoglobulin G antibody

(Jackson). The primary antibodies used for Western blotting were anti-myosin

antibody (MF-20) and anti-actin antibody (Sigma).

Extraction of RNA and ribonuclease protection assay (RPA)

Total RNA was extracted from hindlimb muscle tissue or C2C12 cells by the

guanidinium thiocyanate-phenol chloroform method. The levels of IL-1β, IL-6, and

VEGF mRNA were examined with a multi-probe ribonuclease protection assay system

(BD Biosciences) according to the manufacturer’s instructions. The level of

expression of each of these genes was quantified by the Bio Imaging Analyzer System

(Fuji Photo Film).

In situ hypridization

In situ hybridization was carried out as previously described.1,2 A 570 bp Pst-I

fragment of the IL-1β cDNA (provided by Dr Yoichiro Iwakura)3 4 was subcloned into

pBluescript II SK (Stratagene), linearized by restriction enzymes and transcribed by

RNA polymerases (Roche), whereby the digoxigenin labeled the antisense and sense

probes. Muscle samples were fixed overnight in 4% paraformaldehyde (PFA) in

phosphate-buffered saline (PBS), then in sucrose and embedded with OCT compound.

Sections were cut, airdried and then fixed in 4% PFA for 15 minutes at room

Page 14: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

Online Data Supplement K Tateno, et al. Mechanism of cell therapy

4

temperature. After washing with PBS, sections were treated with proteinase K (5 µg/ml

for 15 minutes), refixed with PFA, washed with PBS, treated with 0.2 M hydrochloride

for 10 minutes and washed again with PBS. Samples were acetylated for 10 minutes,

washed with PBS and dehydrated with ethanol. Hybridization mixture (50% formamide,

10 mM Tris-HCl pH 7.6, 200 µg/ml tRNA, 1×Denhardt's solution, 10% dextran sulfate,

600 mM NaCl, 0.25% SDS, 1 mM EDTA, and the probe) was then applied on top of the

section, overlaid with parafilm, and incubated overnight at 50°C in a humidified

chamber. Sections were washed for 30 minutes with 2×SSC/50% formamide at 50°C,

treated with RNase for 30 minutes at 37°C, washed with 2×SSC for 20 minutes at 50°C

and washed twice with 0.2xSSC for 20 minutes each at 50°C. Sections were rinsed with

buffer 1 (100 mM Tris-HCl pH7.5, 150 mM NaCl), blocked with 10% sheep serum in

buffer 1 for 2 hours and incubated with alkaline phosphatase-conjugated sheep

polyclonal antidigoxigenin antibody (Roche) diluted 1:4000 in buffer 1 for 30 minutes.

After washing twice with buffer 1, alkaline phosphatase activity was detected in the

presence of NBT/BCIP (Roche).

References

1. Nishinakamura R, Matsumoto Y, Nakao K, Nakamura K, Sato A, Copeland NG,

Gilbert DJ, Jenkins NA, Scully S, Lacey DL, Katsuki M, Asashima M, Yokota T.

Murine homolog of SALL1 is essential for ureteric bud invasion in kidney

development. Development. 2001;128:3105-115.

2. Meltzer JC, Sanders V, Grimm PC, Stern E, Rivier C, Lee S, Rennie SL, Gietz

RD, Hole AK, Watson PH, Greenberg AH, Nance DM. Production of

digoxigenin-labelled RNA probes and the detection of cytokine mRNA in rat

spleen and brain by in situ hybridization. Brain Res Brain Res Protoc.

1998;2:339-351.

3. Gray PW, Glaister D, Chen E, Goeddel DV, Pennica D. Two interleukin 1 genes

Page 15: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

Online Data Supplement K Tateno, et al. Mechanism of cell therapy

5

in the mouse: cloning and expression of the cDNA for murine interleukin 1 beta.

J Immunol. 1986;137:3644-3648.

4. Horai R, Asano M, Sudo K, Kanuka H, Suzuki M, Nishihara M, Takahashi M,

Iwakura Y. Production of mice deficient in genes for interleukin (IL)-1alpha,

IL-1beta, IL-1alpha/beta, and IL-1 receptor antagonist shows that IL-1beta is

crucial in turpentine-induced fever development and glucocorticoid secretion. J

Exp Med. 1998;187:1463-1475.

Page 16: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

Online Data Supplement K Tateno, et al. Mechanism of cell therapy

6

Figure legends

Supplementary Figure 1 Factors associated with the response to treatment. Results

are shown as box plots representing median, 25th and 75th percentiles as boxes and the

range of data as bars. IL-6, Interleukin 6; TNFα, tumor necrosis factorα; G-CSF,

granulocyte colony-stimulating factor. *P=0.08 versus non-responders (responders,

n=21; non-responders, n=8).

Supplementary Figure 2 Immunohistochemistry for macrophages. Wild-type

PB-MNC was implanted into the ischemic limbs of wild-type (Wild→Wild) or

IL-1β-deficient mice (Wild→KO). The ischemic limbs of wild-type mice were also

treated with PBS (PBS→Wild) and served as a control. Five days after PB-MNC

implantation, the sections of the ischemic or non-ischemic limbs were stained with

anti-Mac3 antibody. The graph indicates the number of infiltrating macrophages into

the ischemic or non-ischemic limbs (Non-ischemic). *P<0.05 versus PBS→Wild,

#P<0.01 versus Wild→Wild (n=5).

Supplementary Figure 3 Mononuclear cell implantation increases the production of

angiogenic factors by muscle cells. C2C12 cells were cultured under differentiating

conditions with or without human PB-MNC (2.5×105 cells/ml) for 24 hr and expression

of mouse angiogenic cytokines was examined by RPA. Glyceraldehyde-3-phosphate

dehydrogenase (GAPDH) was used as the control.

Supplementary Figure 4 Laser Doppler analysis. The graph shows relative blood

flow in the ischemic limbs of wild-type mice treated with PBS (PBS→Wild), wild-type

PB-MNC (Wild→Wild) and the limbs of IL-1β-deficient mice treated with PBS

(PBS→KO), wild-type PB-MNC (Wild→KO) or wild-type PB-MNC plus IL-1β

(Wild+IL-1β→KO). Data are shown as mean±SEM. *P<0.05 versus PBS-treated

group (n=5).

Page 17: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

Online Data Supplement K Tateno, et al. Mechanism of cell therapy

7

Supplementary Figure 5 Effect of IL-1β treatment on limb ischemia. The graphs

show relative blood flow (A) and capillary density (B) in the ischemic limbs of

wild-type mice treated with PBS (PBS→Wild), IL-1β (IL-1β→Wild) or wild-type

PB-MNC (Wild→Wild). Data are shown as mean±SEM. *P<0.05, **P<0.01 versus

PBS-treated group (n=3-5).

Supplementary Figure 6 Immunohistochemistry for CD31 in the ischemic limbs of

IL-1β-deficient mice treated with wild-type PB-MNC. Scale bar: 100 µm.

Page 18: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

CBA

60

TN

F- α

(n

g/d

L)

G-C

SF

(n

g/d

L)

IL-6

(n

g/d

L)

40

20

0

150

100

50

0

60

40

20

0

Clinical response

++−−

Clinical response

++−−

Clinical response

++−−

Supplementary Figure 1

*P=0.08

Page 19: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

No

n-isch

emic W

ild

No

n-isch

emic K

O

#

*3,000

2,000

1,000

0

Mac

-3 p

osi

tive

cel

ls/m

Supplementary Figure 2

Wild

KO

PB

SW

ild

Wild

Wild

Page 20: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

VEGF

IL-6

GAPDH

IL-1β

PB-MNC −− ++

Supplementary Figure 3

Page 21: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

*

0.8

0.6

0.4

0.2

0

Blo

od

flo

w

Supplementary Figure 4

Wild

KO

PB

SW

ild

Wild

Wild

PB

SK

O

Wild

+ IL

-1βK

O

Page 22: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

BA

0.8

0.6

0.4

0.2

0

Blo

od

flo

w

1,000

800

600

0

400

200

***

CD

31-p

osi

tive

cel

ls/m

m2

PB

SW

ild

Wild

Wild

IL-1β

Wild

PB

SW

ild

Wild

Wild

IL-1β

Wild

Supplementary Figure 5

Page 23: Critical Roles of Muscle-Secreted Angiogenic Factors in ...circres.ahajournals.org/content/circresaha/98/9/1194.full.pdf · endothelial growth factors (VEGF) ... *Both authors contributed

PBS KO

Supplementary Figure 6