重大发现

1 构建基于干细胞的种子细胞用于骨组织再生修复

 

In the field of bone tissue engineering and regenerative medicine, generating patient-specific stem cells has been a long-standing goal. Human bone marrow mesenchymal stem cells(hBMSCs) are the current gold-standard cell source for many tissue-engineering therapies and have been successfully applied in clinics. However, hBMSCs are hampered by an invasive harvesting procedure, limited availability, and loss of potency in

seniors or patients with certain diseases and disorders. Human umbilical cord MSCs (hUCMSCs) are capable of differentiating into mesenchymal lineages. They are easily and abundantly available, with robust proliferation and self-renewal capability due to their origin in neonatal tissues . The main obstacle of hUCMSCs is immunogenic concerns when used heterologously. For autologous applications, the umbilical cord has to be properly cryopreserved from childbirth for an extended period of time.

1.1 将脐带间充质干细胞,骨髓间充质干细胞,胚胎干细胞和iPS等分别种植在具有良好的生物相容性的多孔骨水泥中

The harvested hiPSC–MSCs, hUCMSCs and hBMSCs were used in the following experiments.

Cells were seeded onto the RGD-biofunctionalized CPC disks in 24-well plates at a density of

3 ×105 cells/well . On the second day, the medium was changed into osteogenic medium which

 consisted of the MSC growth medium supplemented with 100 nM dexamethasone, 10 mM

ß-glycerophosphate, 0.05 mM ascorbic acid, and 10 nM 1a,25-dihydroxyvitamin (Sigma) .

 

At 1, 7 and 14 days, live/dead staining (Molecular Probes, Eugene, OR) was used to test cell

viability on CPC. Disks were washed with PBS and incubated with 4 mM ethidium homodimer-1

(EthD-1) and 2 mM calcein-AM in PBS for 20 min. The disks were examined using

epifluorescence microscopy (Eclipse TE2000-S, Nikon, Melville, NY). The percentage of live

cells P and the live cell density D were calculated as previously described . P = number of live

cells/(number of live cells + number of dead cells). D = number of live cells in the image/the

image area. Two randomly-chosen images for each sample were analyzed with three disks

per condition, yielding six images per group at each time point (technical replicates n = 6).

The test was independently repeated three times (biological replicates = 3) on three

different days by seeding new batches of cells on new batches of CPC disks.

 

 

Fig. 1. Cell viability when seeded on CPC scaffold. Live/dead staining of cells at 1 day and 14 days are shown in (A–F). In all three groups, live cells were abundant, and dead cells were few. Percentage of live cells on CPC was around 90% (G). All groups exhibited increasing live cell density (H). Bars with dissimilar letters indicate significantly different values (p < 0.05). Each value is mean ± sd (technical replicates n = 6). The test was independently repeated three times (biological replicates = 3).

 

Reference:

  • Zhou H, Weir MD, Xu HH. Effect of cell seeding density on proliferation and osteodifferentiation of umbilical cord stem cells on calcium phosphate cement-fiber scaffold. Tissue Eng Part A 2011;17:2603–13.
  • Thein-Han W, Liu J, Xu HH. Calcium phosphate cement with biofunctional agents and stem cell seeding for dental and craniofacial bone repair. Dent Mater 2012;28:1059–70.
  • Chen W, Liu J, Manuchehrabadi N, Weir MD, Zhu Z, Xu HH. Umbilical cord and bone marrow mesenchymal stem cell seeding on macroporous calcium phosphate for bone regeneration in rat cranial defects. Biomaterials 2013;34:9917–25.

1.2 探索上述干细胞在骨组织再生中的作用(体外)

For osteogenic differentiation, at 1, 7 and 14 days, RNA was extracted from six cell-seeded disks per group per time point. Two disks were pooled together as an individual RNA sample to have sufficient amount of RNA (technical replicates n = 3). The test was independently repeated

three times (biological replicates = 3) on three different days by seeding new batches of cells on new batches of CPC disks. The total cellular RNA was extracted with TRIzol reagent (Invitrogen) and PureLink RNA Mini Kit (Invitrogen), and then reverse-transcribed into cDNA by a High-capacity cDNA Reverse Transcription kit (Applied Biosystems, Foster City, CA). TaqMan gene expression kits were used to quantify targeted genes on human alkaline phosphatase (ALP, Hs00758162_m1), Runt-re-lated transcription factor (Runx2, Hs00231692_m1), collagen type I (COL1, Hs00164004), osteocalcin (OC, Hs00609452_g1), and glyc-eraldehyde 3-phosphate dehydrogenase (GAPDH, Hs99999905).Relative expression was evaluated using the 2DDCt method and normalized by the Ct of the housekeeping gene GAPDH. The Ct val-ue of hiPSC–MSCs, hUCMSCs, hBMSCs cultured on CPC scaffolds in growth medium for 1 day served as the calibrator .

 

For cell mineralization, at 1, 7 and 14 days, disks were fixed with 10% formaldehyde and stained with Alizarin Red S (ARS, Mil-lipore, Billerica, MA) for 1 h. The disks were then rinsed by deion-ized water four times to visualize the presence of calcified deposition by the cells (technical replicates n =6) . An Osteo-genesis Quantitation Kit (Millipore, ECM 815) was used to extract the stained minerals and measure the ARS concentration, following the manufacturer’s instructions (n = 6). Control CPC scaffolds with the same compositions, but without cells, were measured at the same time periods; they were subjected to the same culture medi-um and incubation conditions as the cell-seeded disks (n = 6). The control’s ARS concentration was subtracted from that of the cell-seeded scaffolds to yield the net mineral concentration synthesized by the cells . The test was independently repeated three times (biological replicates = 3).

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Reference:

  • Chen W, Liu J, Manuchehrabadi N, Weir MD, Zhu Z, Xu HH. Umbilical cord and bone marrow mesenchymal stem cell seeding on macroporous calcium phosphate for bone regeneration in rat cranial defects. Biomaterials 2013;34:9917–25.

Fig. 2. Osteogenic differentiation of cells on CPC. RT-PCR results for ALP (A), Runx2 (B), COL1 (C), and OC (D). Each value is mean ± sd (technical replicates n = 3). The test was independently repeated three times (biological replicates = 3). Osteogenic markers were upregulated in all groups. OC, the late osteogenic marker, decreased in hiPSC-MSCs group, indicating that hiPSC–MSCs did not progress completely to mature osteoblasts in the in vitro induction. Mineral synthesis by cells was detected by Alizarin Red staining (E). The bone mineral matrix became denser and darker red with increasing time. Cell-synthesized mineral concentration was measured by the osteogenesis assay (F)(technical replicates n = 6 for mineralization). The test was independently repeated three times (biological replicates = 3). Bars with dissimilar letters indicate significantly different values (p < 0.05).

1.3 比较不同的干细胞与骨水泥复合材料促进颅骨缺损大鼠病灶骨组织再生的效率

Critical-sized cranial defects were created in male athymic nude rats (Hsd:RH-Fox1mu, 8 weeks old, weighing 200–250 g, Harlan, Indianapolis, IN) in accordance with the protocol approved by the University of Maryland (IACUC # 0909014) and NIH guidelines. Under general anesthesia of 75 mg/kg body weight of ketamine and 10 mg/kg of xylazine, a skin incision was made on the midline of cranium. The periosteum was ablated, and a full-thickness stan-dardized trephine defect, 8 mm in diameter, was made in the cal-varium under continuous saline buffer irrigation. Cell-seeded CPC scaffolds were maintained in osteogenic media for 14 days before implantation. Cell-free CPC was used as control. Rats were random-ly divided into four groups for: hiPSC–MSC–CPC scaffold, hUCMSC–CPC scaffold, hBMSC–CPC scaffold, and CPC control without cells, with six rats per group (technical replicates n = 6). The grafts were harvested at 12 weeks (w) and fixed in 10% formalin.

 

Samples harvested at 12 weeks were scanned with micro-CT (lCT40, Scanco Medical, Bassersdorf, Switzerland). The parameters were set at a resolution of 18 lm, I = 114 lA, E = 70 kVp, with inte-gration time of 300 ms. Specimens were decalcified and embedded in paraffin. The cen-tral region of the implant and defect was cut into 5 lm thick sec-tions and stained with hematoxylin and eosin (H&E) and Masson’s Trichrome. Six samples per group were evaluated and scored with a quan-titative grading scale (Table 1) . Samples were assessed for:(1) hard tissue response at the bone-scaffold interface; (2) bone bridging at the dura side of the defect; and (3) bone formation within the scaffold pores.

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Fig. 3. Representative sagital micro-CT images of hiPSC–MSC–CPC (A), hUCMSCs (C), hBMSCs (D), and CPC control (E). Profile lines are shown for hiPSC–MSC–CPC (B), and CPC control (F). New bone is marked by arrows. A profile line was draw from the peripheral host bone, across the defect area to the other side of the defect boundary. The gray values on profile line were demonstrated in a curve diagram. Cell-seeded scaffolds exhibited higher signals (high intensity areas) than peripheral host bone. In contrast, CPC control had no such distinguishable differences in bone density, indicating less new bone.

Fig. 4. Representative H&E images. New bone areas were stained in pink red (arrows). The white area was due to slight detachment of the tissue or decalcification of CPC. The dark, dark purple and light purple areas were residual CPC. The periosteal side is on the top side, while the dura side is at the bottom. New bone (arrows) was mainly found on the dura side, and some new bone was deposited within the macropores of CPC. There was more new bone in cell-seeded groups than cell-free control.

Fig. 5. Representative Masson’s trichrome staining images. In cell-seeded groups, mineralized (deep blue areas) and nonmineralized osteoid (orange-red) mainly deposited on the dura side, and some new bone was formed within the pores of the scaffold. CPC control group had much less new bone.

 

Reference:

  • Chen W, Liu J, Manuchehrabadi N, Weir MD, Zhu Z, Xu HH. Umbilical cord and bone marrow mesenchymal stem cell seeding on macroporous calcium phosphate for bone regeneration in rat cranial defects. Biomaterials 2013;34:9917–25.
  • Cheng YL, Lin YT, Shih KS. Rapid prototyping mandible model for dental implant surgery simulation. Comput Aided Des Appl 2012;9:177–85.
  • Ohgushi H, Goldberg VM, Caplan AI. Repair of bone defects with marrow cells and porous ceramic. Experiments in rats. Acta orthop Scand 1989;60:334–9.
  • Patel ZS, Young S, Tabata Y, Jansen JA, Wong ME, Mikos AG. Dual delivery of an angiogenic and an osteogenic growth factor for bone regeneration in a critical size defect model. Bone 2008;43:931–40.

fig.3

fig.5

fig.4

赵亮教授

.......................................................................................................................................................................

 

............................

南方医科大学南方医院

 

广东省广州市白云区广州大道北1838号南方医院新外科楼16楼关节与骨病外科

邮编:510515

南方医科大学南方医院临床研究中心

 

广东省广州市白云区广州大道北1838号南方医院新实验楼7楼关节与骨病外科

邮编:510515

主页      动态      研究方向      课题      发表论文      团队成员      年鉴      授课      联系我们

粤ICP备15088577号 Site designed by Dr. Zhao's  Lab

地址:广东省广州市白云区广州大道北1838号  邮编:510515