• 검색 결과가 없습니다.

G. BrdU-labeled cell count in SVZ and ischemic border zone

IV. DISCUSSION

A. Mode of actions of hMSCs

Compared with rats subjected to MCAO alone, IV injection of hMSCs 1 day after stroke significantly improved functional outcome according to the adhesive-remove test, mNSS, and rotarod test. Controversy surrounds the question of whether true transdifferentation (Cogle et al., 2004; Tondreau et al., 2004), spontaneous cell fusion (Terada et al., 2002; Ying et al., 2002), or trophic supports (Chen et al. (a), 2001) are the primary cause for the improvement in the functional outcome after application of MSCs in animal model for ischemic stroke. Our immunohistochemisty results indicate that endogeneous neuronal progenitor cells (BrdU positive), but not hMSCs (NuMA positive), expressed neuronal or glial phenotype. Our data suggest that, rather than transdifferentiation, upregulation of endogenous recovery mechanism either at the peri-infarct area (neurogenesis) or at area that are remote from the infarct (neuronal plasticity) were important role of hMSCs in the functional recovery after ischemic stroke. This benefit may reflect production of growth factors, including neurotrophins that may promote repair of damaged parenchymal cells, reduce apoptosis in the ischemic boundary zone, and enhance proliferation and differentiation of endogenous neural stem and progenitor cells in the SVZ after stroke in rats (Li et al., 2002). Supporting this hypothesis, a recently study reported that hMSCs have the capacity to secrete BDNF, NGF, VEGF and HGF in culture and that hMSCs are sensitive to the normal and ischemic injured brain environment and

13

respond through an increased production of these neurotrophins and angiogenic factors (Chen et al., 2002). The ability of MSCs to secrete multiple growth factors and cytokines is suggestive of their critical trophic roles in a plethora of cellular and physiological functions (Majumdar et al., 1998 and 2000; Wieczorek et al., 2003).

B. Passage of hMSCs as a Factor Affecting the Efficacy of Stem Cell Therapy

1. Passage of hMSCs and neurogenesis

Our data demonstrate that hMSC + P2 hMSC treatment promotes neurogenesis in the SVZ and IBZ and that such effect was more pronounced in the earlier passage MSCs (P2) than in the later passage (P6). Although there have been several studies concerning the growth pattern of MSCs (Zhang et al., 2005; Liu et al., 2003), Zhang et al. recently reported that the population doubling time of P4 rat MSCs was prolonged to nearly 3-fold compared to that on P1 and that, in contrast to the rapid proliferation phase in P1 rat MSCs, P4 rat MSCs entered a growth arrest phase (Zhang et al., 2005). The similar growth pattern of rat MSC, such as rapid proliferation at P1, slow growth at P3, and growth arrest at P4, were observed by Liu et al (Liu et al., 2003). These results suggest that the growth properties of different passage hMSCs might be one of the factors determining the neurogenesis in the ischemic rat brain, But there was no report comparing the effects of MSCs on neurogenesis after stroke depending on their passage.

14

Our hypothesis was that the characteristics of hMSCs (such as passage numbers) may be important in consideration of hMSCs for enhancement of the ischemia-induced neurogenesis. Our data showed that neurogenesis was enhanced by the application of hMSCs and that such effects were more prominent in earlier passage of hMSCs than in the later passage. Thus it is conceivable that in the clinical application of hMSCs it might be better to apply a smaller dose of early passage of hMSCs rather than delay to achieve more numbers of hMSCs of late passage with further ex vivo cultivation.

However, further studies comparing smaller dose of early passages and the larger dose of late passages because it was reported that animals with ischemia-induced brain damage infused with a high dose of MSCs (3×106) recovered better than did control animals infused with a low dose of MSCs (1×106) (Chen et al.(a), 2001). In this present study, no significant difference was noted in the behavioral recovery of animals that P2 and P6 hMSC injected MCAo rats. Further studies are needed to decide the ideal passage and numbers of hMSCs.

2. Passage of hMSCs and chemokine expression

The chemokine SDF-1 (also known as CXCL12) and its receptor CXCR4 have been implicated in homing of stem cells to the bone marrow and the homing of bone marrow-derived cells to sites of injury. It was reported that SDF-1 protein expression was as detected by anti-SDF-1 is clearly upregulated in the infarcted hemisphere within 24 hours and it is maintained through at least 30 days post-occlusion (Hill et

15

al., 2004). Thus, with respect to the recipient brain condition, earlier application of hMSCs may be important because there exist the possibility that the homing effects of SDF-1 to injured area diminish with time (Abbott et al., 2004).

Our results of the prominent neurogenetic potentials of earlier passage of MSCs compared to that of the later passage MSCs are unlikely caused by the difference in the chemokine levels; our immunohistochemical study of SDF-1 showed that the chemokine levels of the rat were not different between the P2- and P6 MSCs received rat.

C. Conclusions

In conclusion, we demonstrate that MSCs derived from adult bone marrow can enhance neurogenesis in the ischemia rat brain and this potential is restricted by proliferation ability and cell passages.

16

Table 1. Modified neurologic severity score test.

Motor tests Points

• Raising the rat by the tail 1=Flexion of forelimb

1=Flexion of hindlimb

1=Head moved >10 degree vertical axis within 30s

3

1=proprioceptive(deep sensation, pushing paw against the table edge to stimulate limb muscles)

2

• Beam balance test (normal=1;maximum=6) 0= balance with steady posture

1= balance with steady posture grasps side of beam 2= hugs the beam & 1 limb falls down from the beam 3= 2 limbs fall down from the beam or spins on the beam 4= attempts to balance on the beam but falls off>40s 5= attempts to balance on the beam but falls off>20s 6=falls off: no attempt to balance or hang on the beam<20

6

• Reflexes and abnormal movements 1=pinna reflex(a head shake when touching the auditory meatus)

1=corneal reflex(an eye blink when lightly ouching the cornea with cotton) 1=startle reflex(a motor response to a brief noise from snapping a clipboard paper)

1=seizures, myoclonus, myodystrophy

4

Maximum points 18 One point is awarded for the inability to perform the tasks or for the lack of a tested reflex.13-18 =sever injury; 7-12 = moderate injury; 1-6 = mild injury.

17

Figure 1. TTC stained coronal brain sections. The images of brain section were captured by scanner. Brain slides (2mm Thick) stained by the TTC solution 1 day after MCAo in adult rat. The lesion of cerebral infarct was visualized as the white arer in contrast to the normal red-stained area. The development of the MCAo was limited in the MCA territory.

Figure 2. hMSC labeled in 14d after ischemia rat. Marked increase in NuMA-positive MSCs (B) and PKH2-labeled hMSC (C) in the ischemic border zone. Scar bar 20 ㎛ in B and 100 ㎛

18

Figure 3. Fifteen days after post-MCAo brain section labeled for SDF-1 in the border zone. SDF-1 labeled in the ischemia border zone (A and C) than in the contralateral zone (B). Scar bar 100㎛. B. scar bar 20㎛

Figure 4. Results of behave functional tests.

(A) Adhesive-remove test ; (B) Modified neurologic severity score [mNSS] test; and (C) Rotarod test before and after middle cerebral artery occlusion (MCAo).

Group 1: MCAo alone(n=4); group 2: intravenous infusion P2 hMSC; or group 3:

intravenous infusion P6 hMSC.

19

Figure 5. Neurogenesis.

BrdU immunostaining in the SVZ of the ipsilateral hemisphere at the 14d after each group (Left); (A) Sham operation, (B) tMCAO + PBS, (C) tMCAO + MSC P2, and (D) tMCAO + MSC P6.

BrdU-labeled cells (Right); In the SVZ and penumbra zone, the numbers of labeled cells were largest in P2 hMSC.

20

Figure 6. Phenotype of BrdU-labeled cells in border zone at 14d after MCAO.

(Upper lane) BrdU labeled cells were merged with NF-H( C ), NeuN (F),DCX (I) and GFAP (L). (Lower lane) No NuMA positive cells expressed GFAP (O).

Section were stained for NF-H, NeuN, DCX and GFAP(A,D,G,J) and BrdU (B,E,H,K); NuMA( N) and GFAP(M) were examined by confocal microscopy.

A-O Scale bar 20㎛.

21

22

REFERENCES

Abbott JD, Huang Y, Liu D, Hickey R, Krause DS, Giordano FJ. Stromal cell-derived factor-1alpha plays a critical role in stem cell recruitment to the heart after myocardial infarction but is not sufficient to induce homing in the absence of injury. Circulation. 2004 Nov 23;110(21):3300-5.

Bang O. Y., Lee, J. S., Lee, P. H., and Lee, G.. Autologous mesenchymal stem cell transplantation in stroke patients. Ann Neurol 57: 874-882. 2005.

Bang O.Y. Reply. Ann Neurol 58: 654-655. 2005.

Borlongan, C. V., Randall, T. S., Cahill, D. W., and Sanberg, P. R. Asymmetrical motor behavior in rats with unilateral striatal excitotoxic lesions as revealed by the elevated body swing test. Brain Res 676: 231-234. 1995.

Callaway, J. K., Knight, M. J., Watkins, D. J., Beart, P. M., Jarrott, B., and Delaney, P.

M. A novel, rapid, computerized method for quantitation of neuronal damage in a rat model of stroke. J Neurosci Methods 102: 53-60. 2000.

Chen, H., Chopp, M., Zhang, Z. G., and Garcia, J. H. The effect of hypothermia on transient middle cerebral artery occlusion in the rat. J Cereb Blood Flow Metab 12: 621-628. 1992.

Chen, J., Li, Y., and Chopp, M. Intracerebral transplantation of bone marrow with BDNF after MCAo in rat. Neuropharmacology 39: 711-716. 2000.

Chen, J., Li, Y., Wang, L., Lu, M., Zhang, X., and Chopp, M. Therapeutic benefit of intracerebral transplantation of bone marrow stromal cells after cerebral ischemia in rats. J Neurol Sci 189: 49-57. 2001. (b)

Chen, J., Li, Y., Wang, L., Zhang, Z., Lu, D., Lu, M., and Chopp, M. Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats. Stroke 32: 1005-1011. 2001. (a)

Chen, J., Li, Y., Zhang, R., Katakowski, M., Gautam, S. C., Xu, Y., Lu, M., Zhang, Z., and Chopp, M. Combination therapy of stroke in rats with a nitric oxide donor and human bone marrow stromal cells enhances angiogenesis and neurogenesis.

Brain Res 1005: 21-28. 2004.

Chen, J., Wang, C., Lu, S., Wu, J., Guo, X., Duan, C., Dong, L., Song, Y., Zhang, J., Jing, D., Wu, L., Ding, J., and Li, D. In vivo chondrogenesis of adult bone-marrow-derived autologous mesenchymal stem cells. Cell Tissue Res 319:

429-23

438. 2005.

Chen, J., Zhang, Z. G., Li, Y., Wang, L., Xu, Y. X., Gautam, S. C., Lu, M., Zhu, Z., and Chopp, M. Intravenous administration of human bone marrow stromal cells induces angiogenesis in the ischemic boundary zone after stroke in rats. Circ Res 92: 692-699. 2003.

Chen, X., Li, Y., Wang, L., Katakowski, M., Zhang, L., Chen, J., Xu, Y., Gautam, S.

C., and Chopp, M. Ischemic rat brain extracts induce human marrow stromal cell growth factor production. Neuropathology 22: 275-279. 2002.

Cogle CR, Yachnis AT, Laywell ED, et al. Bone marrow transdifferentiation in brain after transplantation: a retrospective study. Lancet;363:1432–1437. 2004.

Deng J, Petersen BE, Steindler DA, Jorgensen ML, Laywell ED. Mesenchymal Stem Cells Spontaneously Express Neural Proteins in Culture, and Are Neurogenic After Transplantation. Stem Cells.; [Epub ahead of print] 2005.

Germano, A. F., Dixon, C. E., d'Avella, D., Hayes, R. L., and Tomasello, F.

Behavioral deficits following experimental subarachnoid hemorrhage in the rat. J Neurotrauma 11: 345-353. 1994.

Hayashi, T., Iwai, M., Ikeda, T., Jin, G., Deguchi, K., Nagotani, S., Zhang, H., Sehara, Y., Nagano, I., Shoji, M., Ikenoue, T., and Abe, K.. Neural precursor cells division and migration in neonatal rat brain after ischemic/hypoxic injury. Brain Res 1038: 41-49. 2005.

Hill WD, Hess DC, Martin-Studdard A, Carothers JJ, Zheng J, Hale D, Maeda M, Fagan SC, Carroll JE, Conway SJ. SDF-1 (CXCL12) is upregulated in the ischemic penumbra following stroke: association with bone marrow cell homing to injury. J Neuropathol Exp Neurol. 63(1):84-96. 2004.

Jin, H. K., Carter, J. E., Huntley, G. W., and Schuchman, E. H.. Intracerebral transplantation of mesenchymal stem cells into acid sphingomyelinase-deficient mice delays the onset of neurological abnormalities and extends their life span. J Clin Invest 109: 1183-1191. 2002.

Khan, S. H., Baziany, A., Banigesh, A., Hemmings, S. J., and Shuaib, A. Evaluation of an optimal temperature for brain storage in delayed 2, 3,5-triphenyltetrazolium chloride staining. J Neurosci Methods 98: 43-47. 2000.

Krause, D. S., Theise, N. D., Collector, M. I., Henegariu, O., Hwang, S., Gardner, R., Neutzel, S., and Sharkis, S. J. Multi-organ, multi-lineage engraftment by a single

24

bone marrow-derived stem cell. Cell 105: 369-377. 2001.

Kurozumi, K., Nakamura, K., Tamiya, T., Kawano, Y., Kobune, M., Hirai, S., Uchida, H., Sasaki, K., Ito, Y., Kato, K., Honmou, O., Houkin, K., Date, I., and Hamada, H. BDNF gene-modified mesenchymal stem cells promote functional recovery and reduce infarct size in the rat middle cerebral artery occlusion model. Mol Ther 9: 189-197. 2004.

Li, Y., Chen, J., Chen, X. G., Wang, L., Gautam, S. C., Xu, Y. X., Katakowski, M., Zhang, L. J., Lu, M., Janakiraman, N., and Chopp, M. Human marrow stromal cell therapy for stroke in rat: neurotrophins and functional recovery. Neurology 59: 514-523. 2002.

Liu, Y., Song, J., Liu, W., Wan, Y., Chen, X., and Hu, C. Growth and differentiation of rat bone marrow stromal cells: does 5-azacytidine trigger their cardiomyogenic differentiation? Cardiovasc Res 58: 460-468. 2003.

Majumdar, M. K., Thiede, M. A., Haynesworth, S. E., Bruder, S. P., and Gerson, S. L.

Human marrow-derived mesenchymal stem cells (MSCs) express hematopoietic cytokines and support long-term hematopoiesis when differentiated toward stromal and osteogenic lineages. J Hematother Stem Cell Res 9: 841-848. 2000.

Majumdar, M. K., Thiede, M. A., Mosca, J. D., Moorman, M., and Gerson, S. L.

Phenotypic and functional comparison of cultures of marrow-derived mesenchymal stem cells (MSCs) and stromal cells. J Cell Physiol 176: 57-66.

1998.

Markgraf, C. G., Green, E. J., Hurwitz, B. E., Morikawa, E., Dietrich, W. D., McCabe, P. M., Ginsberg, M. D., and Schneiderman, N. Sensorimotor and cognitive consequences of middle cerebral artery occlusion in rats. Brain Res 575: 238-246.

1992.

Miller, M. W., and Nowakowski, R. S. Use of bromodeoxyuridine-immunohistochemistry to examine the proliferation, migration and time of origin of cells in the central nervous system. Brain Res 457: 44-52. 1988.

Savitz SI, Rosenbaum DM, Dinsmore JH, et al. Cell transplantation for stroke. Ann Neurol;52:266 –275. 2002

Schallert, T., Kozlowski, D. A., Humm, J. L., and Cocke, R. R. Use-dependent structural events in recovery of function. Adv Neurol 73: 229-238. 1997.

Shake, J. G., Gruber, P. J., Baumgartner, W. A., Senechal, G., Meyers, J., Redmond, J.

25

M., Pittenger, M. F., and Martin, B. J. Mesenchymal stem cell implantation in a swine myocardial infarct model: engraftment and functional effects. Ann Thorac Surg 73: 1919-1925; discussion 1926. 2002.

Strauer BE, Brehm M, Zeus T, et al. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans.

Circulation 106: 1913–1918. 2002

Tateishi-Yuyama E, Matsubara H, Murohara T, et al. Therapeutic Angiogenesis using Cell Transplantation (TACT) Study Investigators. Therapeutic angiogenesis for patients with limb ischaemia by autologous transplantation of bone-marrow cells: a pilot study and a randomised controlled trial. Lancet 360:

427–435. 2002

Terada N, Hamazaki T, Oka M, et al. Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion. Nature 416:542–545. 2002.

Tondreau T, Lagneaux L, Dejeneffe M, et al. Bone marrow-derived mesenchymal stem cells already express specific neural proteins before any differentiation.

Differentiation 72:319–326. 2004.

Tse HF, Kwong YL, Chan JK, et al. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet 361:47– 49. 2003.

Vacanti, V., Kong, E., Suzuki, G., Sato, K., Canty, J. M., and Lee, T. Phenotypic changes of adult porcine mesenchymal stem cells induced by prolonged passaging in culture. J Cell Physiol 205: 194-201. 2005.

Wada, K., Sugimori, H., Bhide, P. G., Moskowitz, M. A., and Finklestein, S. P. Effect of basic fibroblast growth factor treatment on brain progenitor cells after permanent focal ischemia in rats. Stroke 34: 2722-2728. 2003.

Wang, C. X., Yang, Y., Yang, T., and Shuaib, A. A focal embolic model of cerebral ischemia in rats: introduction and evaluation. Brain Res Brain Res Protoc 7: 115-120. 2001.

Wieczorek, G., Steinhoff, C., Schulz, R., Scheller, M., Vingron, M., Ropers, H. H., and Nuber, U. A. Gene expression profile of mouse bone marrow stromal cells determined by cDNA microarray analysis. Cell Tissue Res 311: 227-237. 2003.

Ying QL, Nichols J, Evans EP, Smith AG. Changing potency by spontaneous fusion.

Nature 416:545–548. 2002

26

Young, R. G., Butler, D. L., Weber, W., Caplan, A. I., Gordon, S. L., and Fink, D. J.

Use of mesenchymal stem cells in a collagen matrix for Achilles tendon repair. J Orthop Res 16: 406-413. 1998.

Zhang, F. B., Li, L., Fang, B., Zhu, D. L., Yang, H. T., and Gao, P. J. Passage-restricted differentiation potential of mesenchymal stem cells into cardiomyocyte-like cells. Biochem Biophys Res Commun 336: 784-792. 2005.

Zhang, R., Wang, L., Zhang, L., Chen, J., Zhu, Z., Zhang, Z., and Chopp, M. Nitric oxide enhances angiogenesis via the synthesis of vascular endothelial growth factor and cGMP after stroke in the rat. Circ Res 92: 308-313. 2003.

Zhang, R., Zhang, L., Zhang, Z., Wang, Y., Lu, M., Lapointe, M., and Chopp, M. A nitric oxide donor induces neurogenesis and reduces functional deficits after stroke in rats. Ann Neurol 50: 602-611. 2001.

27

28

관련 문서