• Non ci sono risultati.

Key words. Smooth muscle cell. Atherosclerosis, Progenitor, Bone Mar-

N/A
N/A
Protected

Academic year: 2022

Condividi "Key words. Smooth muscle cell. Atherosclerosis, Progenitor, Bone Mar-"

Copied!
11
0
0

Testo completo

(1)

Circulating Progenitor Cells

Masataka Sata and Ryozo Nagai

Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan

Summary. Atherosclerosis is responsible for more than half of all deaths in western countries. Numerous studies have reported that exuberant ac- cumulation of smooth muscle cells play a principal role in the pathogenesis of vascular diseases. It has been assumed that smooth muscle cells derived from the adjacent medial layer migrate, proliferate and synthesize ex- tracellular matrix. Although much effort has been devoted, targeting mi- gration and proliferation of medial smooth muscle cells, no effective ther- apy to prevent occlusive vascular remodeling has been established. Re- cently, we reported that bone marrow cells substantially contribute to the pathogenesis of vascular diseases, in models of post-angioplasty resteno- sis, graft vasculopathy and hyperlipidemia-induced atherosclerosis. It was suggested that bone marrow cells may have the potential to give rise to vascular progenitor cells that home in the damaged vessels and differenti- ate into smooth muscle cells or endothelial cells, thereby contributing to vascular repair, remodeling, and lesion formation. This article overviews recent findings on circulating vascular precursors and describes potential therapeutic strategies for vascular diseases, targeting mobilization, hom- ing, differentiation and proliferation of circulating progenitor cells.

Key words. Smooth muscle cell. Atherosclerosis, Progenitor, Bone Mar-

row, Endothelial cell

(2)

Smooth muscle cells and vascular remodeling

Exuberant accumulation of smooth muscle cells (SMCs) plays a principal role in the pathogenesis of vascular diseases (Ross R, 1999). In athero- sclerotic plaques, SMCs proliferate and synthesise extra cellular matrixes, thereby contributing to lesion formation. Percutaneous coronary interven- tions (PCIs) have been widely adopted for treatment of coronary athero- sclerosis. However, a significant number of these procedures fail due to post-angioplasty restenosis. Although the increasing use of new devices for dilatation of stenosed arteries has lowered the incidence of acute com- plications, restenosis still limits the long-term outcome of percutaneous interventions. SMC hyperplasia is a major cause of post coronary bypass surgery occlusion, too. Furthermore, graft vasculopathy, a leading cause of graft failure and retransplantation after the first postoperative year, also results fi-om SMC hyperplasia. Therefore, much effort has been devoted to understanding the molecular pathways that regulates SMCs hyperplasia to prevent vascular diseases (Ross R, 1999). However, the pathogenesis re- mains largely unknown and, consequently, no effective therapy has been established.

There is a widely accepted view that atherosclerotic lesions result from an excessive, inflammatory-fibroproliferative response to various forms of insult to the endothelium and smooth muscle of the artery wall. In brief, upon endothelial injury, inflammatory cells infiltrate and secrete various cytokines. Cytokine triggers dedifferentiation of adjacent medial SMCs which serve to regulate vascular tone and blood flow under physiological conditions (Ross R, 1999). Dedifferentiated SMCs are characterized by a large cell body containing synthetic and secretary organelles (Ross R, 1999). It has been hypothesized that dedifferentiated SMCs migrate into the subendothelial space, proliferate, and synthesize extracellular matrix.

Similarly, it has been assumed that all of the neointimal cells in

post-angioplasty restenosis and graft vasculopathy are derived from adja-

cent medical smooth muscle cells. Thus, numerous pharmacological and

gene therapies have been proposed targeting dedifferentiation, migration,

proliferation of medial smooth muscle cells. However, there are several

phenomena that could not be explained in accordance with this hypothesis

(3)

(Sata M, 2003). Evidence in support of this hypothesis was provided in several models of vascular diseases (Saiura A, et al. 2001; Sata M. 2003;

Sata M, et al. 2002; Tanaka K, et al. 2003) (Fig. 1).

Atherosclerosis

IjÄ. V*' ^iZ. X ;.'

Post-PCI restenosis Graft Vasculopathy

D

Progenitors

Bone Marrow

Fig. 1. Bone marrow derived vascular progenitors contribute to vascular remodeling in hyperlipidemia-induced atherosclerosis, post percutaneous coronary intervention (PCI) restenosis, and graft vasculopathy (Sata M, et al. 2002). Reproduced with per- mission from Nature Publishing Company

Contribution of bone marrow cells to transplant-associated arteriosclerosis

The contribution of bone marrow cells was first investigated in graft vas- culopathy, a robust form of atherosclerosis which develops in transplanted organs rapidly, leading to failure of the allograft (Sata M, 2003). In this study, heterotopic cardiac transplantation was performed between wild-type mice and ROSA26 mice (Saiura A, et al. 2001; Sata M, et al.

2002), which are knock-in mice ubiquitously expressing LacZ

(LacZ-mice). Four weeks after transplantation, the allografts were har-

vested and stained with X-gal (5-bromo-4-chloro-3-indolyl

ß-D-galactopyranoside). The lumen of large epicardial coronary arteries,

their smaller branches and arterioles were found to have narrowed owing

(4)

to concentric neointimal hyperplasia. The majority of the neointima was composed of recipient cells expressing LacZ (Saiura A, et al. 2001; Sata M, et al. 2002). It was also observed that some of the medial SMCs as well as endothelial cells (ECs) had been replaced by recipient cells. Im- munofluorescence studies revealed that LacZ-positive cells in the neoin- tima expressed various markers for SMCs, including myosin heavy chain, calponin, h-caldesmon, and a-smooth muscle actin (a-SMA) (Sata M, et al.

2002). Conversely, when LacZ-positive hearts were transplanted into wild-type mice, LacZ-negative neointima developed on the LacZ-positive coronary arteries. Furthermore, in situ hybridization of the allografts from female to male mice revealed that most of the cells corresponded to cells from the recipients (Saiura A, et al. 2001). These results indicated that the majority of the neointimal cells derived from the recipient cells, but not from the medial cells of donor origin. Consistent with these observations, others independently reported that recipient cells are a major source of graft vasculopathy in the aortic transplantation model (Hillebrands J, et al.

2001; Hu Y, et al. 2002; Shimizu K, et al. 2001). Moreover, it was re- ported that most of the neointimal cells and ECs were derived

from recipients in human transplant-associated arteriosclerosis after renal transplantation (Grimm P C, et al. 2001).

To identify the potential source of recipient cells that contribute to al- lograft vasculopathy, bone marrow transplantation (BMT) was performed form LacZ mice to wild-fype mice (BMT^^'^^^""''' mice). After four to eight weeks, wild-type hearts were transplanted into the g^jLacz^wiid ^j^^g p^^j.

weeks after cardiac transplantation, most of the neointimal cells were

found to be LacZ-positive. Similarly, when wild-type hearts were trans-

planted into wild-type mice whose bone marrow had been reconstituted

with that of transgenic mice that ubiquitously expressed enhanced green

fluorescent protein (GFP mice) (BMT^''^^'""'^ mice) (Sata M, et al. 2002),

it was observed that GFP-positive cells accumulated on the luminal side of

the graft coronary arteries under a fluorescence illuminator. Immunofluo-

rescence study revealed that some of the GFP-positive cells in graft vas-

culopathy expressed a-SMA. These results indicate that recipient bone

marrow cells may substantially contribute to neointimal formation in trans-

planted grafts.

(5)

Contribution of bone marrow cells to vascular remodeling after mechanical injury

Bone marrow cells can also contribute to the pathogenesis of lesion forma- tion after mechanical vascular injury (Sata M, et al. 2002; Tanaka K, et al.

2003). The bone marrow of wild-type mice were replaced with that of LacZ-mice

(BMT'''"^^^""'''

mice). It was found that transplanted LacZ bone marrow cells had settled in bone marrow, spleen, and thymus, whereas no LacZ-positive cell was detected in uninjured femoral arteries of EMT^^*^^^

*'''' mice. Four to eight weeks after BMT, a large wire was inserted into the femoral artery of gMx^acz^wiid j^j^-g^ using a new method of vascular in- jury that resembles balloon angioplasty (Sata M, et al. 2000). This injury

led to complete endothelial denudation and marked enlargement of the lu- men (Sata M, et al. 2003, Sata M, et al. 2001, Sata M, et al. 2000). The cellularity of the medial layer decreased owing to acute onset of SMC apoptosis (Sata M, et al. 2000). One week after the injury, the artery re- mained dilated. X-gal staining revealed that LacZ-positive cells attached to the luminal side of the injured vessels. LacZ-positive ceils did not express a marker for SMCs (a-SMA) or that for endothelial cells (ECs, CD31).

The dilated lumen gradually narrowed due to neointimal hyperplasia, which was primarily composed of SMCs. A significant amount of neoin- timal and medial cells were LacZ-positive as determined by X-gal staining (Sata M, et al. 2002; Tanaka K, et al. 2003). Immunofluorescence double staining documented that some bone marrow-derived LacZ positive cells in the neointimal lesions expressed a-SMA or CD31 (Tanaka K, et al.

2003). These results indicate that bone marrow cells may give rise to vas- cular cells, thereby contributing to arterial remodeling after wire-mediated endovascular injury.

Bone marrow-derived SMC-like cells in atherosclerotic plaques To evaluate the potential source of SMCs observed in atherosclerotic plaques (Ross R, 1993), the bone marrow of ApoE -/- mice was replaced with that of GFP-mice

(BMT^^^^^P""^"'"

mice) or ROSA26-mice (BMT^^'^^

ApoE-/- jj^jjjg-) (Sata M, et al. 2002). The recipient mice were fed a west-

ern-type diet for eight weeks, starting at four weeks after BMT.

(6)

GFP-positive cells accumulated in atherosclerotic plaques developing in the aorta of

B]V[XGFP~*APOE-/- ^[^Q

Immunofluorescence study on the cross sections revealed that some of the GFP-positive cells expressed markers for smooth muscle cells (a-SMA). Similarly, in the atherosclerotic lesions of the BMxLacz^ApoE-/- j^jj,g^ some of the LacZ-positive cells expressed a-SMA. Furthermore, immunogold-labeling for LacZ identified bone marrow-derived SMC-like cells having muscle fibers in the atherosclerotic plaques of

BMT^"=^^'^P''^"'"

mice. The SMC-like cells displayed typical

"synthetic" phenotype. These results suggest that some of the SMC-like cells observed in hyperlipidemia-induced atherosclerotic plaques may originate from bone marrow.

New mechanism for SMC accumulation in vascular lesions Coronary angioplasty causes vessel wall injury and induces SMCs prolif- eration with subsequent abundant production of extracellular matrix.

Transplant-associated arteriosclerosis is also considered a consequence of an immunological attack against the allograft by the recipient (Billingham ME, 1987). Similarly, various atherogenic substances, such as oxidized LDL, homocysteine, angiotensin II and lipopolysaccharides, have been reported to induce vascular cell apoptosis, presumably initiating the earli- est phase of lesion development in atherosclerosis (Tricot O, et al. 2000).

Therefore, neointima formation appears to be similar to the healing process in response to vascular injuries. In addition to the conventional assumption that damaged tissues are repaired by individual parenchymal cells, an ac- cumulating body of evidence indicates that there exist somatic stem cells that are mobilized to remote organs, differentiate into required lineages and participate in organ repair and regeneration (Asahara T, et al. 1997;

Hill J M, et al. 2003). Bone marrow might be an additional source of vas-

cular cells that contribute to vascular repair and pathological remodeling in

models of post-angioplasty restenosis, transplant-associated arteriosclero-

sis and hyperlipidemia-induced atherosclerosis.

(7)

Cell fusion as a possible mechanism of bone marrow

"differentiation" into SMC

Recent evidence suggests that somatic stem cells or adult stem cells remain even in the adult body. Many animal experiments documented that adult stem cells can transdifferentiate into other lineages. In gender-mismatched human bone marrow transplantation, it was reported that donor-derived hematopoietic stem cells participated in organ regeneration. On the other hand, recent papers suggest that adult stem cells adopt tissue-specific phe- notype by cell fusion in vitro (Terada N, et al. 2002) and in vivo (Wang X, et al. 2003) but not by transdifferentiation. Consistent with this notion, previous reports documented polyploidization of vascular SMCs in re- sponse to mechanical and humoral stimuli (Campbell J H, et al. 1991).

Thus, it is possible that cell fusion can account for, at least in part, the ac- cumulation of bone marrow-derived SMC-like cells in vascular lesions , although spontaneous cell fusion between recipient and donor-derived cells seems to be a rare event in a murine model of cardiac transplantation (Saiura A, et al. 2003).

Implications for regenerative medicine

There is an accumulating body of evidence that in the adult bone marrow

there are somatic stem cells that can differentiate into various lineages

(Blau H M, et al. 2001). Our results also suggest that we may be able to

construct biological artificial arteries using bone marrow-derived vascular

progenitors (Fig. 2)(Kaushal S, et al. 2001). Given the pluripotency of

adult stem cells, they may potentially differentiate into unfavorable

cell-types. In fact, there are a number of similarities between stem cells

and cancer cells (Reya T, et al. 2001). Our findings constitute evidence

that somatic stem cells do participate in pathological remodeling in remote

organs (Saiura A, et al. 2001; Sata M, et al. 2002). For clinical use of adult

stem cells, attention should be paid to potential adverse effects as well as

beneficial aspects caused by the transplanted stem cells.

(8)

I

I

e

Fig. 2. Functional artery might be engineered with bone marrow cells or circu- lating vascular progenitors, a In vitro differentiation method. Bone mar- row-derived endothelial cells (ECs) and smooth muscle cells (SMCs) are used to reconstitute the arterial wall, b Scaffold method. Circulating progenitors are attracted to the implanted scaffold to generate the arterial wall

Future Direction

Further studies are needed to identify and characterize putative vascular

progenitor cells. It was observed that bone marrow-derived cells were

negative for markers of SMCs and ECs when they attached to the luminal

side of the artery one week after mechanical injury (Sata M, et al. 2002). It

is likely that plastic immature cells may be mobilized to the injured ves-

sels, where they differentiate in response to mechanical and humoral stim-

uli. Consistently, it was reported that blood cells contain progenitors that

have the potential to differentiate into either ECs or SMCs in vitro accord-

ing to the composition of the culture medium (Simper D, et al. 2002). Ex-

periments that dissect molecular mechanisms by which progenitors are re-

cruited and differentiate at the site of injury are warranted.

(9)

Conclusions

In summary, our findings indicate that bone marrow cells, including he- matopoietic stem cells, contribute not only to the healing process of in- jured organs, but also to pathological remodeling. Our findings provide the

basis for the development of new therapeutic strategies for vascular dis- eases, targeting mobilization, homing, differentiation and proliferation of bone marrow-derived vascular progenitor cells.

References

Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzen- bichler B, Schatteman G, Isner JM (1997) Isolation of putative progenitor en- dothelial cells for angiogenesis. Science 275:964-967

Billingham ME (1987) Cardiac transplant atherosclerosis. Transplant Proc 19:19-25

Blau H M, BrazeIton T R, Weimann J M (2001) The evolving concept of a stem cell: entity or function? Cell 105:829-841

Campbell J H, Tachas G, Black M J, Cockerill G, Campbell G R (1991) Molecular biology of vascular hypertrophy. Basic Res Cardiol 86:3-11

Grimm P C, Nickerson P, Jeffery J, Savani R C, Gough J, McKenna R M, Stem E, Rush D N (2001) Neointimal and tubulointerstitial infiltration by recipient mesenchymal cells in chronic renal-allograft rejection. N Engl J Med 345:93-97

Hill J M, Zalos G, Halcox J P, Schenke W H, Waclawiw M A, Quyyumi A A, Finkel T (2003) Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med 348:593-600

Hillebrands J L, Klatter F A, van Den Hurk, B M, Popa E R, Nieuwenhuis P, Rozing J (2001) Origin of neointimal endothelium and alpha-actin-positive smooth muscle cells in transplant arteriosclerosis. J Clin Invest 107:1411- 1422

Hu Y, Davison F, Ludewig B, Erdel M, Mayr M, Url M, Dietrich H, Xu Q (2002)

Smooth muscle cells in transplant atherosclerotic lesions are originated from

recipients, but not bone marrow progenitor cells. Circulation 106:1834-1839

(10)

Kaushal S, Amiel G E, Guleserian K J, Shapira O M, Perry T, Sutherland F W, Rabkin E, Moran A M, Schoen F J, Atala A, Soker S, Bischoff J, Mayer J E, Jr (2001) Functional small-diameter neovessels created using endothelial pro- genitor cells expanded ex vivo. Nat Med 7:1035-1040

Reya T, Morrison S J, Clarke M F, Weissman 1 L (2001) Stem cells, cancer, and cancer stem cells. Nature 414:105-111

Ross R (1999) Atherosclerosis-An inflammatory disease. N Eng J Med 340:115-126

Ross R (1993) The pathogenesis of atherosclerosis: a perspective for the 1990s.

Nature 362:801-809

Saiura A, Sata M, Washida M, Sugawara Y, Hirata Y, Nagai R, Makuuchi M (2003) Little evidence for cell fusion between recipient and Donor-Derived cells. J Surg Res 113:222-227

Saiura A, Sata M, Hirata Y, Nagai R, Makuuchi M (2001) Circulating smooth muscle progenitor cells contribute to atherosclerosis. Nat Med 7:382-383 Sata M (2003) Circulating vascular progenitor cells contribute to vascular repair,

remodeling, and lesion formation. Trends Cardiovasc Med 13:249-253 Sata M, Tanaka K, Ishizaka N, Hirata Y, Nagai R (2003) Absence of p53 Leads to

Accelerated Neointimal Hyperplasia After Vascular Injury. Arterioscler Thromb Vase Biol 23:1548-1552

Sata M, Saiura A, Kunisato A, Tojo A, Okada S, Tokuhisa T, Hirai H, Makuuchi M, Hirata Y, Nagai R (2002) Hematopoietic stem cells differentiate into vas- cular cells that participate in the pathogenesis of atherosclerosis. Nat Med 8:403-409

Sata M, Sugiura S, Yoshizumi M, Ouchi Y, Hirata Y, Nagai R (2001) Acute and chronic smooth muscle cell apoptosis after mechanical vascular injury can occur independently of the Fas-death pathway. Arterioscler Thromb Vase Biol 21:1733-1737

Sata M, Maejima Y, Adachi F, Fukino K, Saiura A, Sugiura S, Aoyagi T, Imai Y,

Kurihara H, Kimura K, Omata M, Makuuchi M, Hirata Y., Nagai R (2000) A

mouse model of vascular injury that induces rapid onset of medial cell apop-

tosis followed by reproducible neointimal hyperplasia. J Mol Cell Cardiol

32:2097-2104

(11)

Shimizu K, Sugiyama S, Aikawa M, Fukumoto Y, Rabkin E, Libby P, Mitchell R N (2001) Host bone-marrow cells are a source of donor intimal smooth- mus- cle-like cells in murine aortic transplant arteriopathy. Nat Med 7:738-741 Simper D, Stalboerger P G, Panetta C J, Wang S, Caplice N M (2002) Smooth

muscle progenitor cells in human blood. Circulation 106:1199-1204

Tanaka K, Sata M, Hirata Y, Nagai R (2003) Diverse contribution of bone marrow cells to neointimal hyperplasia after mechanical vascular injuries. Circ Res 93:783-790

Terada N, Hamazaki T, Oka M, Hoki M, Mastalerz D M, Nakano Y, Meyer E M, Morel L, Petersen B E, Scott E W (2002) Bone marrow cells adopt the phe- notype of other cells by spontaneous cell fusion. Nature 416:542-545

Tricot O, Mallat Z, Heymes C, Belmin J, Leseche G, Tedgui A (2000) Relation between endothelial cell apoptosis and blood flow direction in human athero- sclerotic plaques. Circulation 101:2450-2453

Wang X, Willenbring H, Akkari Y, Torimaru Y, Foster M, Al-Dhalimy M, La-

gasse E, Finegold M, Olson S, Grompe M (2003) Cell fusion is the principal

source of bone-marrow-derived hepatocytes. Nature 422:897-901

Riferimenti

Documenti correlati

Herein, we investigated HIF-1a, VEGF and JunB expression in OC ES-2 and SKOV-3 cells treated by PDMSCs conditioned medium (CM) in order to verify our hypothesis that

In particular, it demonstrates how these qualities reach this objective: transparency can increase trust, clear benefits, if well communicated, can be perceived

Per poter inquadrare e delimitare l’area di studio si è proceduto al reperimento della cartografia più significativa esistente e riguardante la zona; in particolare sono

Le liberalità in questione non sono soggette alla forma prevista per la donazione, in quanto si applica la forma richiesta per i negozi effettivamente conclusi. Le

In this study, DOX was loaded in β-cyclodextrin based nanosponges (BNS-DOX), and the effect of DOX and BNS-DOX was compared in vitro in human and mouse breast cancer cell lines,

In this study, we validated a Physician Global Assessment (PGA) for extent in vitiligo based on an international work‐ shop including worldwide vitiligo experts.. This PGA can be

Figure 6: While standard test set validation usually favours over-fitted nonlinear PCA models, model validation based on the correctness of missing data estimation provides a