You are on page 1of 13

MESENCHYMAL STEM CELL THERAPY TO PROMOTE LIMB

TRANSPLANT FUNCTIONAL RECOVERY


EMILIE B. FITZPATRICK, M.D.,1 MARY J. DEHART, B.S.,2 TOMMY A. BROWN, M.D.,1 and SHASHIKUMAR K. SALGAR, Ph.D.2*

Background: Limb transplantation is a viable option for reconstruction after traumatic limb loss; however, functional recovery can be sub-
optimal. The aim of this study was to determine whether mesenchymal stem cell (MSC) administration can improve limb transplant func-
tional recovery. Methods: Orthotopic syngeneic hindlimb transplants were performed in Lewis rats, followed by topical and intravenous
injections of syngeneic MSCs (5 3 106) or vehicle. Transplanted limb sensory and motor functions were tested by cutaneous pain reaction
and walking track analysis, respectively. Results: MSCs expanded ex vivo were CD291, CD312, CD342, CD441, CD45low, CD901, MHC
Class-I1, Class-II2, and pluripotent. Greater than 90% of limb transplants survived. At 4 weeks post-transplantation, the mean sensory
nerve (tibial, peroneal, or sural) function in MSC (n 5 9) and vehicle (n 5 9) groups was <0.3 on a scale of Grades 0–3 (0 5 No function;
3 5 Normal). By 8 weeks, the sensory scores for tibial, peroneal, and sural nerves were 2.2 6 0.7, 1.2 6 0.5, and 1.7 6 0.9 in the vehicle,
and 2.6 6 0.4, 1.0 6 0.9, and 1.7 6 0.9 in the MSC group, respectively (n 5 9/group). At 4, 8, 16, and 24 weeks, the overall sensory func-
tion was higher in MSC group (7/group). Sciatic Function Index (SFI), a measure of motor function, could not be calculated because of
poor foot prints; therefore, a novel grading system was developed. Bone fusion/vascularization as determined by X-ray films/laser Doppler
(2 week post-transplantation) were normal (n 5 3/group). Gastrocnemius muscle was atrophied (P < 0.05), and flexion contractures were
evident by 24 weeks. Conclusions: Bone marrow-derived MSC therapy appears to improve sensory function recovery in a rat limb trans-
plant model. V C 2016 Wiley Periodicals, Inc. Microsurgery 00:000–000, 2016.

Vascularized composite allotransplantation (VCA), or been promising in terms of graft (transplant) survival, func-
composite tissue allotransplantation (CTA), is an expand- tional recovery, and psychosocial impact.6–8 Lower limb
ing practice among subspecialty transplant centers in the transplantation is generally contraindicated due to assumed
reconstruction of complex tissue defects, to improve risks that outweigh the benefits; nonetheless, there are suc-
functional as well as cosmetic outcomes. VCA involves cessful reports.7,9,10 Lower limb prostheses are not feasible
transfer of composite tissue grafts (bone, muscle, skin, for all patients; therefore, limb transplantation may be the
nerve, vessel, etc.) from one individual to another, as a only alternative to those patients.10
single functional unit. It is being increasingly utilized in Unlike visceral solid organ transplants (e.g., liver), VCA
the reconstruction of traumatic injuries, such as amputa- requires optimal nerve regeneration and reinnervation of
tions and severe burns. This emerging therapy has partic- graft motor and sensory targets, to attain intrinsic muscle
ular relevance in the military population: as of February function and sensory input.11–13 The mammalian nervous
2013, there were 1,715 reported battle-injury amputations system has a limited axonal regeneration capacity following
sustained during the conflicts in Afghanistan and Iraq. injury.14,15 Functional recovery after peripheral nerve injury
Most of these (1,493) were classified as major limb loss.1 requires axonal outgrowth, myelination, and targeted rein-
The quality of life of a patient who has lost a limb can nervation.16–18 Microsurgical techniques have advanced
be enhanced with VCA, which is an elective procedure.2 peripheral nerve repair considerably, allowing precise epi-
Since the first successful hand transplantation in 19983 neural or perineural suture approximation. Novel cellular
and face transplantation in 2005,4 >100 upper limb and therapies are sought to enhance nerve regeneration and func-
30 face transplantations have been performed world- tion, including stem cell transplantation.14,16,19–24 The bene-
wide.5,6 Outcomes of upper limb and face transplants have ficial effect of exogenous stem cells on peripheral nerve
regeneration is not completely understood. However, several
1
reports suggest that cell replacement, trophic factor pro-
Department of Surgery, Madigan Army Medical Center, Tacoma, WA
98431 duction, axon guidance/sorting, remyelination, immune
2
Department of Clinical Investigation, Madigan Army Medical Center, modulatory effects etc., of stem cells may all contribute to
Tacoma, WA 98431
peripheral nerve regeneration.14,23–32
Disclosure: The authors declare no conflicts of interest, financial or otherwise.
The views expressed are those of the author(s) and do not reflect the official Mesenchymal stem cells (MSCs) are pluripotent progen-
policy of the Department of the Army, the Department of Defense or the U.S. itor cells found in many peripheral tissues, including bone
Government.
*Correspondence to: S. K. Salgar, Ph.D., Lead Research Physiologist, Chair, marrow. They can be readily expanded ex vivo, and have the
Scientific Review Committee, Department of Clinical Investigation, Madigan potential to self-renew and differentiate into several lineages,
Army Medical Center, 9040 Fitzsimmons Drive, Tacoma, WA 98431-1100.
E-mail: Shashikumar.k.salgar.civ@mail.mil including neuronal cell types such as Schwann cells.14,33,34
Received 2 December 2015; Revision accepted 31 March 2016; Accepted MSCs have been shown to have therapeutic benefit in animal
22 April 2016
Published online 00 Month 2016 in Wiley Online Library
models of Parkinson’s disease, multiple sclerosis, stroke, spi-
(wileyonlinelibrary.com). DOI: 10.1002/micr.30068 nal cord injury and peripheral nerve damage.35–38

Ó 2016 Wiley Periodicals, Inc.


2 Fitzpatrick et al.

Furthermore, MSCs have anti-inflammatory and immuno- flow cytometry (Guaua Technologies, Millipore, CA,
suppressive properties.39,40 Due to the unique combination USA). The representative MSC population of interest was
of proregenerative and immunomodulatory properties, MSC gated and analyzed for cell surface markers (Fig. 1A). Ex
therapy in the setting of VCA could potentially enhance vivo expanded MSCs (passage  8; n  4) were studied for
limb transplant function as well as promote tolerance. their in vitro differentiation potential in to multilineages
Our objectives in this study were: (1) to examine whe- using osteogenesis (A10072-01), adipogenesis (A10070-
ther bone marrow derived MSC administration can improve 01), and chondrogenesis (A10071-01) differentiation kits
sensory and motor nerve function recovery in limb trans- (GIBCO, Life Technologies, NY) as per manufacturer’s
plants; and (2) to successfully establish a rat orthotopic hind instructions. Osteocyte (red stained calcium deposits), adi-
limb transplant model at our institution for the first time. pocyte (red stained lipid granules), and chondrocyte (blue
stain indicating synthesis of proteoglycans by chondrocytes
MATERIALS AND METHODS and developing chondrogenic pellets) differentiation are
shown in Figure 1B.
Animals
Ten- to 12-week-old inbred male Lewis (RT1l) rats, MSC Transplantation
weighing 300 g, were purchased from Harlan Sprague Daw- Briefly, ex vivo expanded MSCs (passage 8) were
ley (Indianapolis, IN). All experiments were conducted follow- suspended in cold phosphate buffered saline (calcium and
ing animal protocol approval by the Madigan Army Medical magnesium free), filtered (40–70 mm pore size) to create
Center Animal Care and Use Committee, and as per institu- a homogeneous solution, and kept on ice until injected
tional guidelines. Animals involved in this study were main- (<2 h). These measures were to prevent cell aggregation
tained in accordance with the ‘Guide for the Care and Use of and decrease the risk of animal death due to pulmonary
Laboratory Animals’ published by the National Research embolism. Topical MSC administration: following limb
Council/Institute of Laboratory Animal Research (ILAR). transplantation, MSCs (5 3 106 cells per animal in
0.5 mL saline) or vehicle were infused locally at the
Mesenchymal Stem Cell Isolation and Expansion
sites of bone fusion, vascular anastomosis and nerve
Rats were euthanized by injecting sodium pentobarbitol repair prior to muscle approximation and skin closure.
(40–80 mg/rat) intraperitoneally. Bone marrow cells This was done only once on the day of surgery. Intrave-
(BMCs) were isolated from femur and tibia, and suspended nous administration: immediately after surgery and clos-
at 5–10 3 107 cells/mL in MSC complete or growth ing the surgical wound, MSCs (5 3 106 cells per animal
medium (Dulbecco’s Modified Eagle’s medium2low glu- in 1–1.5 mL of saline) or vehicle were injected (Injection
cose, glutamax, pyruvate, 10% fetal bovine serum, penicil- 1) intravenously (IV) over 2 min via the dorsal penile
lin [100 U/mL], and streptomycin [100 mg/mL], Gibco/ vein. Three more intravenous MSC (5 3 106 cells) or
Life Technologies, NY), as described previously.41 BMCs vehicle injections (Injections 2, 3, and 4) were repeated
were then plated at a density of 5 3 105 cells/cm2 and cul- at weekly intervals.
tured at 378C in 5% CO2 for 72 h in complete medium.
After 72 h, nonadherent cells in the supernatant were Surgical Procedure
removed completely and the medium was replaced with The general surgical techniques used for orthotopic
fresh medium. Adherent cells were propagated for approxi- limb transplantation are as previously described.42,43
mately 2–4 weeks, subcultured, and frozen (passage 3) in Briefly, the animal was anesthetized with intraperitoneal
freezing medium. Approximately 1–2 weeks prior to injec- (IP) injection of ketamine (40–80 mg/kg) and xylazine
tion, frozen cells were thawed and expanded. (5–10 mg/kg), and anesthesia was maintained with inhal-
ant 1–2% isoflurane. The surgical site was prepared and
Mesenchymal Stem Cell Characterization
a skin incision was made around the circumference of
and Differentiation the right hind limb at the level of the inguinal ligament.
The monoclonal antibodies [mAbs (clones)] used for Beginning laterally, the skin of the upper thigh was
MSC characterization were as follows: antirat CD34 mobilized to expose the biceps femoris. The biceps fem-
(ICO115) (Santa Cruz Biotechnology, Santa Cruz, CA); oris was then divided near the distal attachments to the
anti-rat CD11b/c (OX-42), CD29 (Ha2-5), CD31 stifle and tibia, leaving a sufficient edge of residual tissue
(TLD3A12), CD44H (OX-49), CD45 (OX-1), CD90 (OX- for later repair. The biceps was reflected to expose the
7), class I-RT1.A (OX-18), class II-RT1.B (OX6), and iso- sciatic nerve within the stifle fossa. The sciatic nerve
type control antibodies (BD Biosciences, San Jose CA). All was dissected out proximally to the point of emergence
antibodies used were fluorochrome labeled mAbs. The cells from below the gluteus muscle, preserving mesoneurial
obtained from independent cultures (passage 8; n  4) tissue. The sciatic nerve was transected proximally after
were sorted using forward and side scatter properties by a tag suture of 10-0 nylon was placed. The femoral

Microsurgery DOI 10.1002/micr


Stem Cells and Limb Transplantation 3

Figure 1. Mesenchymal stem cell (MSC): A. Characterization—MSC (passage 8; n  4) were stained with fluorescently labeled antigen
specific monoclonal antibodies and analyzed by FACS (fluorescence activated cell sorter) using flow cytometer. The MSCs were highly
positive for CD29, CD90, CD44 markers, moderately positive for RT1.A (Class I), and negative for CD31 (data not shown), CD34, CD45,
and RT1.B (Class II); representative histograms are shown in the figure. B. MSC differentiation—MSC (passage 8; n  4) were cultured
with osteogenesis, adipogenesis and chondrogenesis differentiation medium and stained with 2% Alizarin red, 0.5% Oil O Red, and 1%
Alcian Blue, respectively. The red calcium deposits are indicative of osteocyte, red lipid droplets are indicative of adipocyte, and blue carti-
laginous pellets are indicative of chondrocyte differentiation.

Microsurgery DOI 10.1002/micr


4 Fitzpatrick et al.

vessels were isolated from the inguinal ligament to the was manually restrained and the transplanted limb was
level of the epigastric takeoff. Tag sutures of 10-0 nylon gently and repeatedly manipulated through the normal
were placed proximally on the femoral vessels. All mus- range of motion.43 Each session lasted up to 5 min, as
cle groups were then sharply divided slightly proximal to long as the animal tolerated it well.
the level of the mid-femur. Heparin (300 mL [50 IU])
was administered via the tail vein for anticoagulation. Experimental Design
The femoral vessels were then clamped above the previ- Group A (n 5 9 rats) received syngeneic MSCs, and
ously placed suture tags and transected. Using a 22- Group B (n 5 9 rats) received vehicle (saline control).
gauge angiocatheter, approximately 5–10 mL of ice-cold Both groups underwent right hind limb transplantation,
TM
Plegisol solution (Hospira, Lake Forest, IL) was per- followed by MSCs or vehicle administration. Starting 1
fused through the femoral artery, until the venous efflu- week post-transplantation, all animals received manual
ent was clear. The osteotomy was performed at the mid physiotherapy for the transplanted limb (5 min, 1–2
times per week) as described previously.43 The following
R
femur using a rotary saw (DREMELV 7300-N/8 Mini-
Mite 4.8-V, Robert Bosch Tool Corporation, Racine, WI) parameters were studied: cutaneous pain reaction test for
with a stainless steel saw blade. Once detached, the sensory nerve function was done on a weekly basis and
donor limb (graft) was wrapped in moist gauze and walking track analysis for motor function every two
placed on ice, until the recipient animal was prepared weeks. Randomly selected animals (2 week post-
and ready for transplantation. The donor animal was transplantation; n 5 3 from each group) underwent laser
euthanized. Doppler imaging to assess vascularization of the trans-
The recipient animal received a preoperative antibi- planted limb, and radiologic analysis to assess fusion of
otic, cefazolin (25 mg/kg body weight) subcutaneously the donor and native femoral ends. Animals were eutha-
(SQ), and was anesthetized and prepared for surgery. The nized at 26–32 weeks post-transplantation and gastrocne-
right hind limb was removed in the same fashion as for mius muscle weights (transplanted limb and contralateral
the donor, with division of the femoral vessels, sciatic native limb) were recorded. In addition, donor and recipi-
nerve, and muscle groups occurring further distally to ent animal weights at the time of transplantation, recipi-
preserve adequate tissue for approximation with the ent weight at euthanasia, and transplant limb ischemic
donor limb. The donor limb was transplanted by per- time were recorded.
forming osteosynthesis of the femur using an intramedul-
lary pin, as described previously43 and orthopedic bone ASSESSMENT OF LIMB FUNCTIONAL RECOVERY
cement (Simplex P, Stryker, Mahway, NJ), to achieve a
Cutaneous Pain Reaction (the Flexor
rigid fixation. The vascular anastomoses were performed
“Withdrawal” Spinal Reflex)
using the vascular cuff technique, as described previ-
ously.42,44 Briefly, thin-walled polyimide tubing (River- The somatosensory reinnervation of the transplanted
tech Medical LLC, Chattanooga, TN) was used to limb was assessed by cutaneous pain reaction in response
fashion arterial (0.724 mm inside diameter, 0.025 mm to a painful stimulus applied to the distal limb, as described
wall thickness) and venous (1.151 mm inside diameter, previously.43 Testing began 2 weeks post-transplantation
0.025 mm wall thickness) cuffs approximately 2.5 mm in and continued at weekly intervals until the end of the study
length. The sciatic nerve was repaired using 2–4 simple period. Animals were not sedated or anesthetized for this
interrupted epineurial sutures of 10-0 nylon (Figs. 2A analysis. Briefly, animals were restrained by a gentle hand-
and 2B). MSCs or vehicle was administered topically at hold with the hindlimbs in suspension, and testing began
the sites of the nerve repair and vascular anastomoses, once the animal was relaxed. Atraumatic forceps were used
and injected at the site of muscle repair and bone fusion. to apply a brief painful stimulus in selected areas of the
All muscle groups were approximated with 6-0 prolene. foot, innervated by the tibial, peroneal, sural and saphenous
The skin was closed with interrupted 4-0 nylon sutures nerves (Fig. 3A), as described previously.43,45 The stimulus
and stainless steel clips. was first applied to the native (left) hind limb, and the
response was observed. The stimulus was then applied in
Postoperative Management the same area on the transplanted hindlimb (right), and the
Lactated ringers solution (5 mL, SQ), buprenorphine response was graded in comparison to the native limb. The
(0.02–0.05 mg/kg, SQ every 12 h as needed) and cefazo- withdrawal reflex was graded using a subjective scale as
lin (20 mg/kg, SQ every 12 h for 3 days) was adminis- described previously46: 0, No response; 1, Mild response;
tered postoperatively. Daily/weekly body weights were 2, Moderate response; 3, Strong response (normal). Each of
monitored and animals were closely observed for signs the four nerve territories was tested three times per session
of pain or distress. Physiotherapy was provided 1–2 times and each response graded separately (Fig. 3). It should be
per week, beginning 1–2 weeks after surgery. The animal noted that sensory testing used in this study is a standard

Microsurgery DOI 10.1002/micr


Stem Cells and Limb Transplantation 5

Figure 2. Rat hindlimb transplantation: A. Schematic of donor and recipient surgeries; B. Transplant surgery—under general anesthesia
(B1) medial (B2) and lateral (B3) aspects of the thigh exposed show femoral vessels and sciatic nerve, respectively. The limb was
removed at the mid femur from the donor animal (B4). The recipient animal was prepared by removing the right hind limb (B4) and the
donor limb was attached to the recipient stabilizing with intramedullary pin and bone cement (B4–5). Medially, femoral vessels were anas-
tomosed using vascular cuff technique (B6–8). Laterally, the donor and recipient sciatic nerve ends were approximated surgically with
10.0 silk sutures (B9–10). The opposing respective muscles/tissues of the donor and recipient were aligned and sutured. Skin was
approximated using clips (B11). C. Laser Doppler analysis—two or more weeks following limb transplantation laser Doppler analysis
revealed normal vascularization. D. Radiograph analysis—six to eight weeks following transplantation, radiographs confirmed significant
bone fusion and intramedullary pin (rod) in proper position. a, Artery; v, vein; D, donor; R, recipient; DSN, donor sciatic nerve; RSN, recip-
ient sciatic nerve; SNR, sciatic nerve repair; Tx, transplanted limb; N, native limb; IMP, intramedullary pin.

Microsurgery DOI 10.1002/micr


6 Fitzpatrick et al.

Figure 3. Cutaneous pain reaction assessment: A. Sensory nerve function—was assessed by pinch technique in the territories of the tibial
(T), peroneal (P), sural (Sur), and saphenous (S) nerves as described, previously.42 Withdrawal/vocal response was scored in comparison
to the normal limb (0 5 no response, 1 5 slight, 2 5 moderate, 3 5 normal). L, lateral; M, medial. B–E. Mean sensory score of individual
nerves in vehicle and mesenchymal stem cell (MSC) treated groups up to 26 weeks post-transplantation. Tibial nerve function was first
recovered followed by sural, peroneal, and saphenous nerves. F. Overall sensory nerve function—there was no significant (P > 0.05) dif-
ference between vehicle and MSC treated groups. However, sensory function recovery was slightly better in MSC group than in vehicle
group.

technique used by several investigators in the past, and is time. Animals underwent conditioning trials 3–5 days prior to
subjective in nature; investigator in this study was very transplantation. Testing began 2 weeks post-transplantation
well trained to perform such a testing. and continued at 2-week intervals until the end of study
period. The sciatic function index (SFI), a conventional mea-
Walking Track Analysis sure used to assess hindlimb motor function, 47requires mea-
Motor function of the transplanted limb was assessed by surement of three print characteristics: print length [distance
walking track analysis, as described previously.47–50 Briefly, between the heel and 3rd toe], toe spread [distance between
animals were tested in a confined walkway (approximately the 1st and 5th toe] and intermediary toe spread [distance
10 cm wide 3 10 cm high 3 70 cm long), which was lined between the 2nd and 4th toe]. Due to the poor quality of foot
with white paper and led into a dark shelter. Water-soluble prints obtained in this study, the SFI could not be calculated.
black ink was applied directly to the plantar surfaces of the Therefore, a novel grading scale was developed based on the
hind feet, and the animal was placed at the end of the walk- presence of a heel print and the number of individual toe
way and allowed to walk down the corridor into the shelter; prints. The prints were graded from 0 (no print) to 4 (complete
this was repeated three times, with fresh ink applied each print) as shown in Figure 4.

Microsurgery DOI 10.1002/micr


Stem Cells and Limb Transplantation 7

Figure 4. Walking track analysis: (A) Progression of walking track (foot prints)—normal and transplanted limb foot prints were obtained as
described previously.47,50 In both vehicle and MSC treated animals, the transplant limb foot prints became less distinct with time due to
progressive foot flexion contractures in the transplanted limb (4D). (B) Grading system for foot print analysis—we developed a novel grad-
ing system (grades 0–4) based on the quality of foot prints available to us. (C) Walking track analysis—there was no significant (P > 0.05)
difference between vehicle and MSC treated groups until 26 weeks post-transplantation. (D) Foot flexion contracture—all animals devel-
oped some degree of foot contracture by 20 weeks post-transplantation despite manual physiotherapy.

Laser Doppler and Radiologic Analyses grammed to scan both hind limbs. The images were ana-
Animals randomly selected 2 weeks post-transplantation lyzed using the laser Doppler software, and the native limb
underwent laser Doppler analysis to assess vascularization served as the control. For radiologic analysis, animals were
of the transplanted limb. Under isoflurane anesthesia, the anesthetized and placed in the Kodak in vivo Imaging
hind limbs were shaved and the animal was placed in dorsal System-FX (Carestream Health Inc., CA) in dorsal or ven-
recumbency in the laser Doppler imager, which was pro- tral recumbency. Both hindlimbs were included in the

Microsurgery DOI 10.1002/micr


8 Fitzpatrick et al.

Table 1. Animal Weight, Ischemic Time, and Gastrocnemius Muscle Weight (Mean 6 SD) in Limb Transplanted Animals
P value between
columns (vehicle vs. P value between P value between
Group Vehicle MSC MSC) rows (vehicle) rows (MSC)
Donor weight (g) 288.5 6 19.7 297.4 6 38.9 P > 0.05 Donor vs. recipient Donor vs. recipient
(n 5 9)a (n 5 9)a weight P > 0.05b P > 0.05b
Recipient weight (g) 300.5 6 19.7 312.6 6 48.4 P > 0.05
(n 5 9)a (n 5 9)a
Ischemic time (min) 167.7 6 14.3 170.8 6 31.4 P > 0.05
(n 5 9)a (n 5 9)a
Weight at euthanasia (g) 441.6 6 24.3 460.4 6 63.1 P > 0.05
(n 5 7)a (n 5 8)a
Normal limb gastrocnemius 2.3 6 0.2 2.4 6 0.15 P > 0.05 Normal vs. transplant Normal vs. transplant
muscle weight (g) (n 5 7)a (n 5 8)a limb weight P < 0.05c limb weight
Transplanted limb gastrocnemius 1.52 6 0.3 1.37 6 0.2 P > 0.05 P < 0.05c
muscle weight (g) (n 5 7)a (n 5 8)a
MSC, mesenchymal stem cells.
a
Common superscripts (‘a’) between columns (vehicle and MSC) indicate no significant (P > 0.05) difference.
b
Body weight between donor and recipient animals at the time of transplantation in vehicle or MSC group did not vary significantly (P > 0.05).
c
Transplant gastrocnemius muscle weight was significantly (P < 0.05) reduced in both vehicle and MSC treated groups compared to their contralateral normal
limbs.

image field. The images were analyzed to assess fusion of CD45 (hematopoietic stem cell-specific markers). Ex vivo
the donor and native femoral ends. cultured MSCs (passage 8) were CD291, CD901,
CD441, CD31–, CD34–, CD45low, CD11b/clow, MHC
Gastrocnemius Muscle Mass class I (RT1.A)1, and MHC class II (RT1.B) (Fig. 1A).
After euthanasia, the gastrocnemius muscles of the The MSC markers CD29 and CD90 were abundantly
transplanted and native hind limbs were carefully dis- expressed (>90%), and hematopoietic stem cell markers
sected, harvested, and weighed; the mean gastrocnemius (CD31, CD34, CD45) were least expressed (<1%). The
muscle weights were compared between the groups. class I (RT1.A) was moderately expressed (<50%), and
Class II (RT1.B) was least expressed (<1%).
Statistical Analysis
Statistical analysis was performed with SPSS software MSC differentiation
version PASW Statistics18 (SPSS Inc., Chicago, IL). The MSCs cultured in osteogenesis, adipogenesis, and
data between two groups were compared by Student t chondrogenesis differentiation media formed calcium
test or ANOVA with Bonferroni correction. All P values deposits (stained red) produced from osteocytes, lipids
were two-tailed and values <0.05 were considered to (stained red) produced in adipocytes, and chondrogenic
indicate statistical significance. pellets (stained blue) containing proteoglycans produced
from chondrocytes, respectively, as shown in Figure 1B.
RESULTS

Animal Characteristics Limb Transplant Surgery


The donor rat body weight was 288.5 6 19.7 and Overall, the limb transplant surgery was highly suc-
297.4 6 38.9 g in vehicle and MSC treated groups, cessful, with >90% limb transplant survival at 2 weeks
respectively; the recipient rat weight was 300.5 6 19.7 post-transplantation. The operative time for donor surgery
and 312.6 6 48.4 g in vehicle and MSC treated groups, was 75 min, and for recipient surgery (limb removal
respectively. There was no significant difference and transplantation) was 120 min. The ischemic time
(P > 0.05) between donor and recipient weights in either (time from donor limb vascular clamping to reperfusion)
vehicle or MSC treated groups (Table 1). was 167.7 6 14.3 and 170.8 6 31.4 min in vehicle and
MSC treated groups, respectively, and did not differ sig-
Mesenchymal Stem Cell Characteristics and nificantly (P > 0.05) (Table 1).
Differentiation Postoperatively, all animals developed edema in their
MSCs were confirmed on the basis of previously transplanted limbs that persisted for approximately 1
accepted cell surface markers,51 including the presence week. Animals bore weight (partially) on the transplanted
of CD29 and CD90 (stromal cell-specific markers), and limb by 1–2 weeks, and started moving around actively
the absence or low expression of CD34, CD31, and by 3–4 weeks.

Microsurgery DOI 10.1002/micr


Stem Cells and Limb Transplantation 9

0.16 6 0.25
1.52 6 0.47
1.91 6 0.57
2.25 6 0.34
2.39 6 0.33
2.68 6 0.21
MSC, mesenchymal stem cell injected group; Tx, transplantation. Sensory nerve function was graded from 0 to 3; 0, no response; 1, mild; 2, moderate; and 3, normal (see “ Materials and Methods” ). Sen-
sory response to individual nerve (tibial, saphenous, peroneal, sural) stimuli significantly (P < 0.05) improved over time in both vehicle and MSC groups. However, there was no significant (P > 0.05) differ-
ence between vehicle and MSC treated groups at any time point post-Tx tested. Interestingly, the total sensory score (determined by averaging individual nerve scores) in MSC treated group was better
Osteosynthesis of the femur using an intramedullary

MSC
rod and bone cement provided adequate fixation (Fig.
2B; 4, 5). Furthermore, radiographs demonstrated proper
Total bone fusion (Fig. 2D). The “cuff” technique employed
for vascular anastomosis (Fig. 2B; 7, 8) was highly effi-
cient; none of the transplants showed signs of vascular
0.21 6 0.20
1.35 6 0.33
1.73 6 0.36
2.16 6 0.27
2.47 6 0.24
2.36 6 0.36
Table 2. Sensory Nerve Function Score (Mean 6 SD) as Determined by Cutaneous Pain Reaction (Pinch Reflex) Test in the Transplanted Foot

Vehicle

leak or cuff failure during the entire post-transplantation


period. The sciatic nerve repair is shown in Figure 2B;
9, 10. The surgical wounds healed well, and sutures were
removed by 10–12 days. Several animals partially
removed the skin clips, which required replacements.
1.70 6 0.89
1.63 6 0.77
2.24 6 0.74
2.62 6 0.49
2.67 6 0.30
0.0401 6 1

One animal was euthanized due to self-mutilation of the


MSC

transplanted limb. The laser Doppler imaging of trans-


planted limb confirmed adequate vascularization (Fig.
Sural

2C), in addition to the visual assessment of pink toes for


blood supply.
0.33 6 0.71
1.67 6 0.94
1.78 6 0.80
2.00 6 0.37
2.54 6 0.56
2.46 6 0.40
Vehicle

ASSESSMENT OF LIMB FUNCTIONAL RECOVERY

Cutaneous Pain Reaction Test


Normal innervation results in an immediate with-
0.07 6 0.22
1.00 6 0.91
1.33 6 0.73
1.52 6 0.54
1.81 6 0.57
2.33 6 0.56

drawal response, with or without vocalization which was


MSC

graded 0–3 (see “Materials and Methods”). The mean


sensory function scores in the territories of individual
Peroneal

nerves (Fig. 3A) are presented in Table 2 and Figures


3B–3F. Sensory recovery was earliest in the distribu-
0.30 6 0.51
1.17 6 0.47
1.19 6 0.90
1.67 6 0.96
2.00 6 0.56
2.33 6 0.62

tion of the tibial nerve, followed by the sural, peroneal,


Vehicle

and saphenous nerve territories. At 4 weeks, post-


transplantation, the overall sensory nerve function was
grade 0.21 6 0.20 and 0.16 6 0.25 in vehicle and MSC
treated animals, respectively. However, by 6–8 weeks it
0.00 6 0.00
0.81 6 0.94
1.75 6 1.28
2.29 6 0.91
2.190 6 .79
2.780 6 .40

was significantly (P < 0.05) higher and clearly recogniz-


MSC

able, grade 1.0 (Fig. 3F), in both vehicle and MSC


treated groups. The overall sensory functional recovery
Saphenous

improved over time, reaching a maximum of grade 2.5.


There was no significant difference (P > 0.05) in overall
sensory nerve function recovery between the vehicle and
0.00 6 0.00
0.42 6 0.58
1.22 6 0.97
2.00 6 0.87
2.42 6 0.43
1.67 6 0.93
Vehicle

MSC treated groups. However, MSC administration


enhanced sensory functional recovery marginally (Fig.
3F; Table 2).
than vehicle group but was not significant (P > 0.05).

Walking Track Analysis


0.52 6 0.78
2.56 6 0.41
2.92 6 0.15
2.95 6 0.13
2.95 6 0.13
2.94 6 0.14

Using the grading system developed in this study (see


MSC

“Materials and Methods”), unexpectedly, the walking


track analysis revealed progressively poorer foot prints
Tibial

with time (up to 26 weeks) in both vehicle and MSC


treated groups (Fig. 4A). The grading system (grades 0–
0.22 6 0.44
2.17 6 0.69
2.85 6 0.34
2.96 6 0.11
2.92 6 0.15
3.00 6 0.00

4) is presented in Figure 4B. There was no significant


Vehicle

(P > 0.05) difference in mean print grades between the


vehicle and MSC treated groups (Fig. 4C). The cause for
poor prints was progressive flexion–contractures of the
foot (Fig. 4D). The severity of flexion contracture
post-Tx
Week

increased with the time post-transplantation; ultimately,


12
16
20
24

the motor function could not be assessed effectively.


4
8

Microsurgery DOI 10.1002/micr


10 Fitzpatrick et al.

Gastrocnemius Muscle Mass Time of sensory nerve function recovery (4–6 weeks)
The gastrocnemius muscle mass was significantly and significant progress observed with time (up to 26
(P < 0.05) reduced in the transplanted limb (1.37 6 0.2 g) weeks) in vehicle and MSC treated groups are in agree-
compared to normal limb (2.4 6 0.15 g) in MSC treated ment with the previous reports on limb transplanta-
group; and a similar finding was observed in vehicle tion.43,46 The poor foot prints obtained in walking track
treated group (Table 1). There was no difference analysis in the present study was mainly due to progres-
(P > 0.05) in gastrocnemius muscle mass between normal sive foot flexion–contractures. This feature has been
limbs (left) of vehicle and MSC treated animals, and well-documented.48,54 Several procedures to remedy the
between transplanted limbs (right) of vehicle and MSC development of foot flexion–contractures have been
treated animals. attempted in limb transplantation and crushed/transected
sciatic nerve injury models with modest success. Stras-
DISCUSSION berg and co-workers55 used wire mesh floor to house the
rats in addition to manual physiotherapy following sciatic
We examined whether bone marrow derived MSC nerve repair that reduced foot flexion–contractures and
administration can improve sensory and motor function improved recovery. Endo and co-workers54 used tread-
recovery in an orthotopic rat hind limb transplant model. mill training protocol for hind limb transplanted rats that
Our limb transplant surgical procedure included utilization improved plantar surface contact and foot prints. How-
of the vascular cuff technique to anastomose donor and ever, in the present study, none of the above methods
recipient vessels, intramedullary pin and bone cement for were combined with MSC therapy. Nonetheless, in the
stable bone fixation, and sciatic nerve repair with epineurial present study animals were given 5 min manual physio-
sutures. Our findings include restoration of limb transplant therapy 1–2 times per week.
sensory function (>75%), which was slightly improved Yeh and co-workers43 used distal nerve anastomosis
with MSC therapy, incomplete motor function recovery techniques (tibial, peroneal, sural) which enhanced motor
secondary to flexion–contractures, and highly successful function and prevented flexion–contractures. It has been
surgical technique with >90% limb transplant survival and well documented since the early development of periph-
<3 h of ischemia time. The follow-up period post- eral nerve repair techniques that outcomes are directly
transplantation in this study was about 26 weeks. related to the level of the nerve injury; distal injuries
MSC therapy appeared to improve sciatic nerve func- have superior functional recovery to proximal injuries.
tion recovery, but was not significant. In our model, The triple nerve repair technique, though it requires more
MSCs were applied to the nerve repair after the donor time and is technically more challenging, may provide
and recipient sciatic nerves were approximated with sur- superior outcomes in this model.56–58
gical sutures. Goel and co-workers14 transplanted bone- Because of poor foot prints we were unable to calcu-
marrow derived mononuclear cells between the proximal late Sciatic Function Index (SFI), a conventional measure
and distal ends of transected sciatic nerve in a nerve to assess limb motor function.49 According to our novel
transection model and found accelerated and enhanced grading system (grades 0–4) there was a significant
nerve regeneration. This effect may be due to stem cell reduction in print scores with time in both vehicle and
trophic factors causing axonal growth and stem cell dif- MSC treated groups, opposite to what we anticipated.
ferentiation into Schwann-like cells, leading to myelin Low print scores due to foot flexion–contractures is an
reformation. In another study, transplanted Schwann cells inherent limitation of walking track analysis, particularly
(derived from bone marrow MSCs) into transected sciatic in the setting of limb transplant. While SFI has been reli-
nerve resulted in vigorous nerve regeneration with myelin ably used in models of sciatic nerve injury, more sophis-
synthesis in rats.35 There are other reports that demon- ticated gait analysis techniques, such as the CatWalkTM
strated the ability of bone marrow MSCs in differentiat- XT system (Noldus, Wageningen, The Netherlands), may
ing into Schwann cell-like cells both in vivo and in vitro be better suited for studies in limb transplantation.
and induce myelination of regenerated nerve fibers after The surgical procedures in orthotopic-hindlimb trans-
sciatic nerve injury.28,52,53 It should be noted that none plantation are extensive and time-consuming. Strong
of the above studies described were in a limb transplant bone fixation is vital for the success of limb transplanta-
model. We believe a similar mechanism should be tion. Osteosynthesis performed provided stable fixation
expected in a limb transplant model. To our knowledge, and prevented torsion as confirmed by our X-ray imaging
there are no reports on the efficacy of bone marrow- is in agreement with previous report.43,46 The cuff tech-
derived MSCs on limb nerve function recovery in an nique44 used to anastomose femoral vessels was less time
orthotopic rat limb transplant model. Ours was the first consuming (compared to conventional suture technique),
attempt in which MSC administration appeared to have effective, and had no vascularization problems. Short
some benefit (sensory nerve function recovery). vascular anastomosis time (<20 min) reduced warm

Microsurgery DOI 10.1002/micr


Stem Cells and Limb Transplantation 11

ischemia time to a minimum which is in agreement with In conclusion, the strategy used to improve limb
the previous report.44 The donor and recipient sciatic transplant function recovery utilizing bone marrow
nerve repair in the present study grossly looked well derived MSC administration is attractive, feasible and
aligned, yet nerve function recovery was suboptimal; this deserve further investigation. Physiotherapy seems to be
was possibly due to improper axon growth, improper vital for optimal limb transplant functional recovery.
apposition/alignment of the nerve fascicles14,15,17,18,21,52,59 When clear foot prints are not available to calculate sci-
or other unknown factors. atic function index (SFI), alternate methodology and
In the present study, tissue quality was maintained by grading system developed in this study could be used to
limiting exposure to desiccation and minimizing the total measure motor function recovery.
ischemic period (cold and warm) of donor limb. Cell atro-
phy, necrosis and fibrosis are proportionate to ischemic ACKNOWLEDGMENTS
times. It’s known that longer ischemic times result in more The authors acknowledge Mr. Juan Tercero, Ms. Joanna
severe morphological changes to the muscle. Tsuji and co- Dandeneau, and Mr. John Schaphorst, Department of
workers60 observed that cold ischemic period of <8 h and
Clinical Investigation, for their assistance with animal
warm ischemic period of <4 h would restore good function
care, anesthesia, and behavioral testing.
of major muscular extremities. The short ischemic time
(<3 h) in the present study prevented any major ischemic
damage to limb muscles.
The significant reduction in transplant gastrocnemius REFERENCES
muscle weight (muscular atrophy) was possibly due to poor 1. Fischer H. U.S. Military Casualty Statistics: Operation New Dawn,
peripheral nerve regeneration and reinnervation of the target Operation Iraqi Freedom, and Operation Enduring Freedom. Con-
muscles, compounded by prolonged disuse of the limb, gressional Research Service Report for Congress, February 5, 2013;
http://www.crs.gov, RS22452:1–12.
which is in agreement with previous reports.12,13 Self- 2. Jensen SE, Butt Z, Bill A, Baker T, Abecassis MM, Heinemann
mutilation of the transplanted limb occurs in certain strains AW, Cella D, Dumanian GA. Quality of life considerations in upper
of rats more commonly than in others.61 In the present study, limb transplantation: Review and future directions. J Hand Surg
2012;37:2126–2135.
one animal showed some symptoms of self-mutilation of the 3. Dubernard JM, Owen E, Herzberg G, Lanzetta M, Martin X, Kapila
transplanted limb and was euthanized humanely. H, Dawahra M, Hakim NS. Human hand allograft: Report on first 6
Advanced immunosuppressive drugs and novel surgical months. Lancet 1999;353:1315–1320.
procedures have enabled successful VCAs. Nonetheless, 4. Devauchelle B, Badet L, Lengele B, Morelon E, Testelin S,
Michallet M, D’ Hauthuille C, Dubernard JM. First human face allo-
there are many challenges to counter prior to its broad graft: Early report. Lancet 2006;368:203–209.
scale applications in humans: long-term complications 5. Kueckelhaus M, Fischer S, Seyda M, Bueno EM, Aycart MA,
such as chronic rejection, graft-versus-host disease, lack of Alhefzi M, ElKhal A, Pomahac B, Tullius SG. Vascularized com-
posite allotransplantation: Current standards and novel approaches to
optimal functional recovery, and poor understanding of prevent acute rejection and chronic allograft deterioration. Trans-
tolerance induction. Research to identify novel therapeutic plant Int 2015. doi:10.1111/Tri.12652.
modalities (stem cells and small molecules) is expected to 6. Siemionow M, Gharb BB, Rampazzo A. Successes and lessons
learned after more than a decade of upper extremity and face trans-
make a significant improvement in the field of VCA. plantation. Curr Opin Org Transplant 2013;18:633–639.
Following are the specific measures and recommenda- 7. Cavadas PC, Ibanez J, Thione A, Alfaro L. Bilateral trans-humeral
tions to overcome the limitations of the present study in the arm transplantation: Result at 2 years. Am J Transplant 2011;11:
future. (1) Due to equipment limitations, we provided pas- 1085–1090.
8. Diaz-Siso JR, Bueno EM, Sisk GC, Marty FM, Pomahac B, Tullius
sive, but not active, physiotherapy. Including wire-mesh SG. Vascularized composite tissue allotransplantation—State of the
therapy or tread mill exercises may reduce contractures and art. Clin Transplant 2013;27:330–337.
enhance functional recovery. (2) Motor function data was 9. Cavadas PC, Landin L, Ibanez J, Roger I, Nthumba P. Infrapopliteal
lower extremity replantation. Plast Reconstr Surg 2009;124:532–539.
limited in part by the use of the walking track analysis and 10. Cavadas PC, Thione A, Carballeira A, Blanes M. Bilateral transfe-
SFI as our endpoint. Alternative gait analysis techniques moral lower extremity transplantation: Result at 1 year. Am J Trans-
such as CATwalk XT system (Noldus Information Tech- plant 2013;13:1343–1349.
11. Yan Y, MacEwan MR, Hunter DA, Farber S, Newton P, Tung TH,
nology, Leesburg, VA), which automatically classifies and Mackinnon SE, Johnson PJ. Nerve regeneration in rat limb allografts:
analyzes footprints, will likely generate more reliable data. Evaluation of acute rejection rescue. Plast Reconstr Surg 2013;131:
(3) Nerve transection and repair at the level of the sciatic 499e–511e.
12. Hare GM, Evans PJ, Mackinnon SE, Best TJ, Bain JR, Szalai JP,
nerve might have resulted in poor target nerve innervations Hunter DA. Walking track analysis: A long-term assessment of
and poor motor function. Performing the nerve repair closer peripheral nerve recovery. Plast Reconstr Surg 1992;89:251–258.
to the target of innervations at the level of sciatic nerve 13. Totosy de Zepetnek JE, Zung HV, Erdebil S, Gordon T. Innervation
branches (tibial, sural, and peroneal nerves) might enhance ratio is an important determinant of force in normal and reinnervated
rat tibialis anterior muscles. J Neurophysiol 1992;67:1385–1403.
muscle reinnervation, reduce contractures, and improve 14. Goel RK, Suri V, Suri A, Sarkar C, Mohanty S, Sharma MC, Yadav
functional recovery. PK, Srivastava A. Effect of bone marrow-derived mononuclear cells

Microsurgery DOI 10.1002/micr


12 Fitzpatrick et al.

on nerve regeneration in the transection model of the rat sciatic 33. Li M, Ikehara S. Bone-marrow-derived mesenchymal stem cells for
nerve. J Clin Neurosci 2009;16:1211–1217. organ repair. Stem Cells Int 2013;2013:132642.
15. Hall S. Nerve repair: a neurobiologist’s view. J Hand Surg 2001;26: 34. Kitada M. Mesenchymal cell populations: Development of the
129–136. induction systems for Schwann cells and neuronal cells and finding
16. Euler de Souza Lucena E, Guzen FP, Lopes de Paiva Cavalcanti JR, the unique stem cell population. Anat Sci Int 2012;87:24–44.
Galvao Barboza CA, Silva do Nascimento Junior E, Cavalcante Jde 35. Dezawa M, Takahashi I, Esaki M, Takano M, Sawada H. Sciatic nerve
S. Experimental considerations concerning the use of stem cells and regeneration in rats induced by transplantation of in vitro differentiated
tissue engineering for facial nerve regeneration: A systematic bone-marrow stromal cells. Eur J Neurosci 2001;14:1771–1776.
review. J Oral Maxillofac Surg 2014;72:1001–1012. 36. Coronel MF, Musolino PL, Brumovsky PR, Hokfelt T, Villar MJ.
17. Dedkov EI, Kostrominova TY, Borisov AB, Carlson BM. Survival Bone marrow stromal cells attenuate injury-induced changes in gala-
of Schwann cells in chronically denervated skeletal muscles. Acta nin, NPY and NPY Y1-receptor expression after a sciatic nerve con-
Neuropathol 2002;103:565–574. striction. Neuropeptides 2009;43:125–132.
18. Walsh S, Midha R. Use of stem cells to augment nerve injury repair. 37. Karussis D, Kassis I, Kurkalli BG, Slavin S. Immunomodulation and
Neurosurgery 2009;65:A80–A86. neuroprotection with mesenchymal bone marrow stem cells (MSCs):
19. Rochkind S, Geuna S, Shainberg A. Chapter 25: Phototherapy in A proposed treatment for multiple sclerosis and other neuroimmuno-
peripheral nerve injury: Effects on muscle preservation and nerve logical/neurodegenerative diseases. J Neurol Sci 2008;265:131–135.
regeneration. Int Rev Neurobiol 2009;87:445–464. 38. Tang Y, Yasuhara T, Hara K, Matsukawa N, Maki M, Yu G, Xu L,
20. Gu S, Shen Y, Xu W, Xu L, Li X, Zhou G, Gu Y, Xu J. Application Hess DC, Borlongan CV. Transplantation of bone marrow-derived
of fetal neural stem cells transplantation in delaying denervated mus- stem cells: A promising therapy for stroke. Cell Transplant 2007;16:
cle atrophy in rats with peripheral nerve injury. Microsurgery 2010; 159–169.
30:266–274. 39. Kuo YR, Chen CC, Goto S, Lin PY, Wei FC, Chen CL. Mesenchy-
21. Pan HC, Cheng FC, Chen CJ, Lai SZ, Lee CW, Yang DY, Chang mal stem cells as immunomodulators in a vascularized composite
MH, Ho SP. Post-injury regeneration in rat sciatic nerve facilitated allotransplantation. Clin Dev Immunol 2012;2012:854846.
by neurotrophic factors secreted by amniotic fluid mesenchymal 40. Plock JA, Schnider JT, Solari MG, Zheng XX, Gorantla VS. Per-
stem cells. J Clin Neurosci 2007;14:1089–1098. spectives on the use of mesenchymal stem cells in vascularized com-
22. Murakami T, Fujimoto Y, Yasunaga Y, Ishida O, Tanaka N, Ikuta posite allotransplantation. Front Immunol 2013;4:175.
Y, Ochi M. Transplanted neuronal progenitor cells in a peripheral 41. Manning E, Pham S, Li S, Vazquez-Padron RI, Mathew J, Ruiz P,
nerve gap promote nerve repair. Brain Res 2003;974:17–24. Salgar SK. Interleukin-10 delivery via mesenchymal stem cells: A
23. Pan HC, Chen CJ, Cheng FC, Ho SP, Liu MJ, Hwang SM, Chang novel gene therapy approach to prevent lung ischemia–reperfusion
MH, Wang YC. Combination of G-CSF administration and human injury. Hum Gene Ther 2010;21:713–727.
amniotic fluid mesenchymal stem cell transplantation promotes 42. Sucher R, Oberhuber R, Margreiter C, Rumberg G, Jindal R, Lee
peripheral nerve regeneration. Neurochem Res 2009;34:518–527. WP, Margreiter R, Pratschke J, Schneeberger S, Brandacher G.
24. Pan HC, Chin CS, Yang DY, Ho SP, Chen CJ, Hwang SM, Chang Orthotopic hind-limb transplantation in rats. J Vis Exp 2010;12:41.
MH, Cheng FC. Human amniotic fluid mesenchymal stem cells in 43. Yeh LS, Gregory CR, Theriault BR, Hou SM, Lecouter RA. A func-
combination with hyperbaric oxygen augment peripheral nerve tional model for whole limb transplantation in the rat. Plast Reconstr
regeneration. Neurochem Res 2009;34:1304–1316. Surg 2000;105:1704–1711.
25. Santiago LY, Clavijo-Alvarez J, Brayfield C, Rubin JP, Marra KG. 44. Sucher R, Oberhuber R, Rumberg G, Hautz T, Zelger B, Glodny B,
Delivery of adipose-derived precursor cells for peripheral nerve Jindal R, Pulikkottil B, Gorantla VS, Brandacher G, Margreiter R,
repair. Cell Transplant 2009;18:145–158. Andrew Lee WP, Schneeberger S. A rapid vascular anastomosis
26. Amoh Y, Kanoh M, Niiyama S, Hamada Y, Kawahara K, Sato Y, technique for hind-limb transplantation in rats. Plast Reconstr Surg
Hoffman RM, Katsuoka K. Human hair follicle pluripotent stem 2010;126:869–874.
(hfPS) cells promote regeneration of peripheral-nerve injury: An 45. Wiesenfeld-Hallin Z. Partially overlapping territories of nerves to
advantageous alternative to ES and iPS cells. J Cell Biochem 2009; hindlimb foot skin demonstrated by plasma extravasation to anti-
107:1016–1020. dromic C-fiber stimulation in the rat. Neurosci Lett 1988;84:261–265.
27. Cheng FC, Tai MH, Sheu ML, Chen CJ, Yang DY, Su HL, Ho SP, 46. Song YX, Muramatsu K, Kurokawa Y, Kuriyama R, Sakamoto S,
Lai SZ, Pan HC. Enhancement of regeneration with glia cell line- Kaneko K, Taguchi T. Functional recovery of rat hind-limb allo-
derived neurotrophic factor-transduced human amniotic fluid mesen- grafts. J Reconstr Microsurg 2005;21:471–476.
chymal stem cells after sciatic nerve crush injury. J Neurosurg 2010; 47. Bain JR, Mackinnon SE, Hunter DA. Functional evaluation of com-
112:868–879. plete sciatic, peroneal, and posterior tibial nerve lesions in the rat.
28. Chen CJ, Ou YC, Liao SL, Chen WY, Chen SY, Wu CW, Wang Plast Reconstr Surg 1989;83:129–138.
CC, Wang WY, Huang YS, Hsu SH. Transplantation of bone mar- 48. Varejao AS, Meek MF, Ferreira AJ, Patricio JA, Cabrita AM. Func-
row stromal cells for peripheral nerve repair. Exp Neurol 2007;204: tional evaluation of peripheral nerve regeneration in the rat: Walking
443–453. track analysis. J Neurosci Methods 2001;108:1–9.
29. Pan H, Zhao K, Wang L, Zheng Y, Zhang G, Mai H, Han Y, Yang 49. Varejao AS, Melo-Pinto P, Meek MF, Filipe VM, Bulas-Cruz J.
L, Guo S. Mesenchymal stem cells enhance the induction of mixed Methods for the experimental functional assessment of rat sciatic
chimerism and tolerance to rat hind-limb allografts after bone mar- nerve regeneration. Neurol Res 2004;26:186–194.
row transplantation. J Surg Res 2010;160:315–324. 50. Brown CJ, Mackinnon SE, Evans PJ, Bain JR, Makino AP, Hunter
30. Carlson KB, Singh P, Feaster MM, Ramnarain A, Pavlides C, Chen ZL, DA, Hare GM. Self-evaluation of walking-track measurement using
Yu WM, Feltri ML, Strickland S. Mesenchymal stem cells facilitate a Sciatic Function Index. Microsurgery 1989;10:226–235.
axon sorting, myelination, and functional recovery in paralyzed mice 51. Nagaya N, Fujii T, Iwase T, Ohgushi H, Itoh T, Uematsu M,
deficient in Schwann cell-derived laminin. Glia 2011;59:267–277. Yamagishi M, Mori H, Kangawa K, Kitamura S. Intravenous admin-
31. Marconi S, Castiglione G, Turano E, Bissolotti G, Angiari S, istration of mesenchymal stem cells improves cardiac function in
Farinazzo A, Constantin G, Bedogni G, Bedogni A, Bonetti B. rats with acute myocardial infarction through angiogenesis and myo-
Human adipose-derived mesenchymal stem cells systemically genesis. Am J Physiol Heart Circ Physiol 2004;287:H2670–2676.
injected promote peripheral nerve regeneration in the mouse model 52. Shimizu S, Kitada M, Ishikawa H, Itokazu Y, Wakao S, Dezawa M.
of sciatic crush. Tissue Eng A 2012;18:1264–1272. Peripheral nerve regeneration by the in vitro differentiated-human
32. Kuo YR, Chen CC, Shih HS, Goto S, Huang CW, Wang CT, Chen bone marrow stromal cells with Schwann cell property. Biochem
CL, Wei FC. Prolongation of composite tissue allotransplant survival Biophys Res Commun 2007;359:915–920.
by treatment with bone marrow mesenchymal stem cells is corre- 53. Cuevas P, Carceller F, Garcia-Gomez I, Yan M, Dujovny M. Bone
lated with T-cell regulation in a swine hind-limb model. Plast marrow stromal cell implantation for peripheral nerve repair. Neurol
Reconstr Surg 2011;127:569–579. Res 2004;26:230–232.

Microsurgery DOI 10.1002/micr


Stem Cells and Limb Transplantation 13

54. Endo T, Ajiki T, Minagawa M, Hoshino Y, Kobayashi E. Treadmill 58. Grinsell D, Keating CP. Peripheral nerve reconstruction after injury:
training for hindlimb transplanted rats. Microsurgery 2007;27:220– A review of clinical and experimental therapies. Biomed Res Int
223. 2014;2014:698256.
55. Strasberg SR, Watanabe O, Mackinnon SE, Tarasidis G, Hertl MC, 59. Ide C. Peripheral nerve regeneration. Neurosci Res 1996;25:101–121.
Wells MR. Wire mesh as a post-operative physiotherapy assistive 60. Tsuji N, Yamashita S, Sugawara Y, Kobayashi E. Effect of pro-
device following peripheral nerve graft repair in the rat. J Peripher longed ischaemic time on muscular atrophy and regenerating nerve
Nerv Syst 1996;1:73–76. fibres in transplantation of the rat hind limb. J Plast Surg Hand Surg
56. Brown PW. Factors influencing the success of the surgical repair of 2012;46:217–221.
peripheral nerves. Surg Clin North Am 1972;52:1137–1155. 61. Shir Y, Zeltser R, Vatine JJ, Carmi G, Belfer I, Zangen A,
57. Hubbard JH. The quality of nerve regeneration. Factors independent Overstreet D, Raber P, Seltzer Z. Correlation of intact sensibility
of the most skillful repair. Surg Clini North Am 1972;52:1099– and neuropathic pain-related behaviors in eight inbred and outbred
1108. rat strains and selection lines. Pain 2001;90:75–82.

Microsurgery DOI 10.1002/micr

You might also like