Marrow Stromal Cells for Cell-Based Therapy: The Role of Antiinflammatory Cytokines in Cellular Cardiomyoplasty

Similar documents
Paracrine Mechanisms in Adult Stem Cell Signaling and Therapy

Tissue repair. (3&4 of 4)

Journal Club Semmler Lorenz

Mesenchymal Stem Cells to Repair Vascular Damage after Chemotherapy: Past, Present and Future

Uncovering the mechanisms of wound healing and fibrosis

SUPPLEMENTAL MATERIAL. Supplementary Methods

Cell implantation after myocardial infarction: a 10 years experience from the ICREC laboratory

SUPPLEMENTARY INFORMATION

DOWNLOAD PDF CARDIAC REMODELING AND CELL DEATH IN HEART FAILURE

1. Cardiomyocytes and nonmyocyte. 2. Extracellular Matrix 3. Vessels שאלה 1. Pathobiology of Heart Failure Molecular and Cellular Mechanism

Research Article Cytological Evaluation of Hyaluronic Acid on Wound Healing Following Extraction

International Graduate Research Programme in Cardiovascular Science

Resident cardiac stem cells: how to find and use them

BIOCHEMICAL INVESTIGATIONS IN THE DIAGNOSTICS OF CARDIOVASCULAR DISORDERS. As. MARUSHCHAK M.I.

Histopathology: healing

renoprotection therapy goals 208, 209

Extracellular matrix Basic and translational science: Highlights of the congress

Tissue renewal and Repair. Nisamanee Charoenchon, PhD Department of Pathobiology, Faculty of Science

9/23/2017. Prof. Steven S. Saliterman. Department of Biomedical Engineering, University of Minnesota

שינויים מולקולאריים ומבניים באי ספיקת לב אפשרויות לטיפול עתידני

Promoting Fracture Healing Through Systemic or Local Administration of Allogeneic Mesenchymal Stem Cells

Prof. Sandrine Florquin Department of Pathology Academic Medical Center University of Amsterdam Amsterdam, The Netherlands. Slide 1.

Xenotransplant Cardiac Chimera: Immune Tolerance of Adult Stem Cells

Myocardial infarction

In Vivo Animal Models of Heart Disease. Why Animal Models of Disease? Timothy A Hacker, PhD Department of Medicine University of Wisconsin-Madison

Myocardial Infarction

With the recent advent in stem cell biology, it has been shown

LV FUNCTION ASSESSMENT: WHAT IS BEYOND EJECTION FRACTION

Imaging of Coronary Artery Disease: II

CT for Myocardial Characterization of Cardiomyopathy. Byoung Wook Choi, Yonsei University Severance Hospital, Seoul, Korea

An aneurysm is a localized abnormal dilation of a blood vessel or the heart Types: 1-"true" aneurysm it involves all three layers of the arterial

Healing & Repair. Tissue Regeneration

Pretargeting and Bioorthogonal Click Chemistry-Mediated Endogenous Stem Cell Homing for Heart Repair

Pathophysiology of heart failure with preserved ejection fraction. Extracellular matrix

Cell therapy: enhancing the therapeutic potential of cardiac progenitors for delivery post myocardial infarction. Rita Alonaizan

Evaluation of the wound healing response post deep dermal heating by fractional RF: INTRAcel

Mesenchymal Stem Cells and Cancer: Their Interplay

Therapeutic Potential of Human Umbilical Cord Derived Stem Cells in a Rat Myocardial Infarction Model

EARLY INFLAMMATORY RESPONSES TO VASCULAR DEVICES

Stem Cells. Keith Channon. Department of Cardiovascular Medicine University of Oxford John Radcliffe Hospital, Oxford

WIEF-AFF ROUNDTABLE Tokyo, Japan 26 May 2015

Devices are So Old School: The New World of Myocardial Regeneration

Cardiac Sarcoidosis. Millee Singh DO Non Invasive Cardiology First Coast Heart and Vascluar

The Cardiovascular System and Aging- Is it Built to Fail?

III. Results and Discussion

c Ischemia (30 min) Reperfusion (8 w) Supplementary Figure bp 300 bp Ischemia (30 min) Reperfusion (4 h) Dox 20 mg/kg i.p.

1) Severe, crushing substernal chest pain 2) radiate to the neck, jaw, epigastrium, or left arm. 3- rapid and weak pulse 4- nausea (posterior MI).

ΒΙΟΔΕΙΚΤΕΣ ΣΤΗΝ ΚΑΡΔΙΑΚΗ ΑΝΕΠΑΡΚΕΙΑ. ΔΗΜΗΤΡΙΟΣ ΤΟΥΣΟΥΛΗΣ Καθηγητής Καρδιολογίας

CD34 + VEGFR-3 + progenitor cells have a potential to differentiate towards lymphatic endothelial cells

Risk Stratification in Heart Failure: The Role of Emerging Biomarkers

In vivo bromodeoxyuridine (BrdU) incorporation was performed to analyze cell

Supplemental Table 1. Primer sequences for transcript analysis

SUPPLEMENTARY INFORMATION

Production of Exosomes in a Hollow Fiber Bioreactor

FOCUS ON CARDIOVASCULAR DISEASE

Exercise in Adverse Cardiac Remodeling: of Mice and Men

Exosomes/tricalcium phosphate combination scaffolds can enhance bone regeneration by activating the PI3K/Akt signalling pathway

Regenerative Medicine for Cardiomyocytes

Citation for published version (APA): Velde, S. V. D. (2006). Stem cell-mediated regeneration of the infarcted heart: inflammation rules? s.n.

Protocol. Progenitor Cell Therapy for the Treatment of Damaged Myocardium due to Ischemia

Recovery of Myocardial Infarction via Unique Modulation of the Cardiac Microenvironment

Evaluation of the wound healing response post - deep dermal heating by fractional RF: INTRACEL

Inflammation is Not the Enemy

Multimodality Imaging in Cardiac Stem Cell Research

Acute coronary syndrome. Dr LM Murray Chemical Pathology Block SA

Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features

Inflammation: Friend or Foe?

Detection and Assessment of MI: Use of Imaging Methods. Robert O. Bonow, M.D.

Index. Note: Page numbers of article titles are in boldface type.

Cellular cardiomyoplasty (CCM) have been the focus of

Cardiac MRI: Clinical Application to Disease

C57BL/6 Mice are More Appropriate. than BALB/C Mice in Inducing Dilated Cardiomyopathy with Short-Term Doxorubicin Treatment

Progenitor Cell Therapy for the Treatment of Damaged Myocardium due to Ischemia. Original Policy Date

Enhanced CABG is in Your Hands

Patterns of Left Ventricular Remodeling in Chronic Heart Failure: The Role of Inadequate Ventricular Hypertrophy

Supplementary Materials for

Lymphoid System: cells of the immune system. Answer Sheet

As outlined under External contributions (see appendix 7.1), the group of Prof. Gröne at the

Diagnosis and Management of Acute Myocardial Infarction

After this presentation and discussion, the participants should be able to:

Cordis EXOSEAL Vascular Closure Device

Mesenchymal Stem Cells

Left ventricular hypertrophy: why does it happen?

Regulation of the IGF axis by TGF-b during periosteal chondrogenesis: implications for articular cartilage repair

Postn MCM Smad2 fl/fl Postn MCM Smad3 fl/fl Postn MCM Smad2/3 fl/fl. Postn MCM. Tgfbr1/2 fl/fl TAC

Pathology of Cardiovascular Interventions. Body and Disease 2011

Pathology of Coronary Artery Disease

E. Cervio, P. Danieli, C. Ciuffreda, F. Pisano, M. Roccio, M. Gnecchi. The authors have no financial disclosures to declare

VWF other roles than hemostasis. Summary 1: VWF & hemostasis synthesis 11/4/16. Structure/function relationship & functions kDa.

Cardiovascular manifestations of HIV

Analysis on the mechanism of reduced nephron number and the pathological progression of chronic renal failure in Astrin deficient rats

Cellular cardiomyoplasty is a cell therapy approach

Supplementary material page 1/10

Supporting Information. Calculation of the relative contributions of myocyte proliferation, stem cell. Supporting Information Fig 1 (page 9)

Supplementary Material

PhD THESIS Epigenetic mechanisms involved in stem cell differentiation

Review Article Present and Future Perspectives on Cell Sheet-Based Myocardial Regeneration Therapy

Regenerative Tissue Matrix in Treatment of Wounds

Significant Improvement of Heart Function by Cotransplantation of Human Mesenchymal Stem Cells and Fetal Cardiomyocytes in Postinfarcted Pigs

Transcription:

Marrow Stromal Cells for Cell-Based Therapy: The Role of Antiinflammatory Cytokines in Cellular Cardiomyoplasty Guangyong Chen, MD, Madhur Nayan, BS, Minh Duong, BS, Juan-Francisco Asenjo, MD, Yin Ge, BS, Ray C-J Chiu, MD, PhD, and Dominique Shum-Tim, MD, MS Divisions of Cardiac Surgery and Surgical Research and Department of Anesthesia, McGill University Health Center, Montreal, Quebec, Canada Background. The mechanism by which marrow stromal cells (MSCs) improve cardiac function after myocardial infarction (MI) is still unclear. Because MI patients with lower circulating proinflammatory/antiinflammatory cytokine ratios have been reported to have a better prognosis and in vitro studies showed that MSCs express antiinflammatory cytokines, we hypothesized that changes in cytokine ratios in the infarct microenvironment after MSC therapy may play a role in improving early cardiac function after MI. Methods. Sixty-three rats that survived left coronary artery ligations were injected with culture media (group M) or MSCs (group C). Cardiac functional changes were assessed with echocardiography. Cytokine gene expressions of interleukin (IL)-1, IL-6, IL-8, (proinflammatory) and IL-10 (antiinflammatory) were quantified by real-time polymerase chain reaction. Extracellular matrix deposition, injury score, and the matrix metallopeptidase 2/tissue inhibitor of metallopeptidase 1 ratio were also analyzed. Results. The ratio of proinflammatory/antiinflammatory cytokine gene expression was decreased in group C at various times, particularly in the early postoperative period. In group C, the matrix metallopeptidase 2/tissue inhibitor of metallopeptidase 1 gene expression ratio was significantly lower than group M at the early phase (12 hours), which in group C was translated into significantly lower extracellular matrix deposition at 24 hours, 1, and 2 weeks. Functional recovery was also significantly better in cell therapy group C. Conclusions. Our data demonstrate that MSC therapy decreases the proinflammatory/antiinflammatory cytokine ratio in the microenvironment early after MI. This is associated with subsequent less scar formation and improved cardiac function. (Ann Thorac Surg 2010;90:190 8) 2010 by The Society of Thoracic Surgeons Accepted for publication Feb 26, 2010. Address correspondence to Dr Shum-Tim, Division of Cardiothoracic Surgery, McGill University Health Center, 1650 Cedar Ave, Ste C9-169, Montreal, QC H3G 1A4, Canada; e-mail: dshumtim@yahoo.ca. Autologous marrow stromal cells (MSCs) transplanted into infarcted myocardium can differentiate into cells of a cardiomyocyte phenotype and improve left ventricular function [1 3]. To date, however, the precise mechanisms of such cardiac functional improvement remain controversial. The postulated mechanisms include stem cell transdifferentiation into cardiomyocytes or cell fusion between the transplanted stem cells and residual cardiomyocytes [4]. The functional improvement has far exceeded the extent of the neocardiomyocytes identified in the periinfarcted areas, which raises the further question of whether cell differentiation has any important role, at least during the early posttherapy phase. In addition, the improvement in cardiac function appeared to precede the time required for cell transdifferentiation. It has also been suggested that the MSCs could improve cardiac function through a paracrine or immunomodulatory effect, such as angiogenesis factor, secretion of various cytokines, and extracellular matrix (ECM) deposition [5 9]. A clinical study showed that a reduced ratio of proinflammatory to antiinflammatory cytokines inferred a better prognosis in patients with recent myocardial infarction (MI) [10]. Tögel and colleagues [11] suggested the role of antiinflammatory cytokines from implanted MSCs in early improvement of renal function after acute renal failure. On the basis of these observations, we sought to explore whether changes in the proinflammatory/ antiinflammatory cytokine gene expression ratio in the infarct microenvironment after MSC implantations were associated with early improved cardiac function. Material and Methods All procedures in this study were in compliance with the Guide for the Care and Use of Laboratory Animals (National Institutes of Health publication No. 85-23) and the Guide to the Care and Use of Experimental Animals of the Canadian Council on Animal Care. 2010 by The Society of Thoracic Surgeons 0003-4975/$36.00 Published by Elsevier Inc doi:10.1016/j.athoracsur.2010.02.074

Ann Thorac Surg CHEN ET AL 2010;90:190 8 MARROW STROMAL CELLS FOR CARDIOMYOPLASTY 191 Table 1. Cytokine Gene Expression of the Whole Heart Cytokine Group 12 Hours, Folds 24 Hours, Folds 1 Week, Folds 2 Weeks, Folds IL-1 M 2.498 0.663 0.120 0.045 1.097 0.993 3.942 1.305 C 19.309 12.015 a 0.837 0.508 b 1.813 0.598 4.165 2.948 0.0004 0.000001 0.000008 IL-6 M 0.325 0.205 0.154 0.146 0.257 0.140 1.757 1.693 C 1.172 0.736 c 0.317 0.180 0.412 0.396 2.673 5.743 0.374 0.079 0.942 IL-8 M 0.509 0.266 0.473 0.272 4.083 1.912 2.605 1.470 C 0.846 0.371 0.831 0.346 5.453 3.780 2.031 1.978 0.446 0.000001 0.567 IL-10 M 1.333 0.392 1.089 0.830 0.534 0.351 0.468 0.283 C 12.866 8.317 d 29.294 28.111 e 0.887 0.610 1.506 1.654 0.001 0.002 0.004 a p 0.005; b p 0.005; c p 0.012; d p 0.006; e p 0.025. IL interleukin. Isolation, Culture, and Labeling of Rat MSCs Rat MSC cultures were prepared according to Caplan s method, and then MSCs were transfected as previously described [12] with pmfg-lacz plasmid containing the -galactosidase gene for identification of the transplanted cells in the myocardium. The resulting MSCs expressing lacz were expanded until transplantation. Experimental Design Immunocompetent female syngenic Lewis rats (200 to 250 g, Charles River Canada, St. Constance, Que) were used. Proximal left coronary artery ligations were done in 72 rats and 9 rats died within 24 hours. The remaining 63 were randomly assigned to two groups, 32 to group C and 31 to group M. In group M, 150 L of medium was directly injected into three different sites around the periinfarct area of the myocardium 10 minutes after coronary ligation. In group C, isogenic MSCs (3 10 6 ) suspended in medium were similarly injected after ligation. The injection of MSCs at the periinfarction area was essential because a previous study suggested that cell-tocell contact was necessary for the phenotypic expression of MSCs in a coculture in vitro model [13]. Rats were sacrificed at various predetermined time points. Cardiac Functional Assessment Transthoracic echocardiography was performed on all animals in groups M and C before operation (baseline), at 12 and 24 hours, and at 1 and 2 weeks after ligation and treatment. All animals were sedated in a quite room and in a right lateral decubitus position to ensure similar views in all echocardiographic assessments. A commercial echocardiographic system was obtained that used a 12-MHz probe with a small footprint (SonoSite, Seattle, WA). Scanning was performed as previously described [12]. The heart was scanned in longitudinal view in 2-dimensions and M-mode, where it is usually possible to see a 2-chamber view. Then the probe was turned 90 to get the best possible cross-sectional view of the left ventricle (LV) just above the papillary muscles. To minimize the intraobserver error in measurements, three consecutive measurements were averaged. The LV end-diastolic (LVEDD) and end-systolic (LVESD) diameters were measured according to the American Society of Echocardiology leading-edge method [12]. Fractional shortening (FS) was determined as [(LVEDD LVESD)/LVEDD]. The ejection fraction (EF) was estimated as [(LVEDV LVESV)/LVEDV]. To ensure reproducibility, all before and after intervention measurements were performed by one experienced observer (J.F.A.) who was blinded to the treatment groups. Harvesting of Hearts and Tissue Processing The recipient rats were sacrificed at 12 and 24 hours, and at 1 and 2 weeks after cell therapy and echocardiography. Their hearts were harvested and dissected from the Table 2. Proinflammatory/Antiinflammatory Cytokine Gene Expression Ratios Cytokine Group 12 Hours, Folds 24 Hours, Folds 1 Week, Folds 2 Weeks, Folds IL-1 /IL-10 M 2.117 1.231 0.129 0.043 2.081 0.769 10.150 4.674 C 1.610 0.741 0.047 0.029 a 2.677 1.232 5.219 4.965 0.056 0.000001 0.017 IL-6/IL-10 M 0.295 0.251 0.175 0.184 0.597 0.366 4.983 5.689 C 0.110 0.054 0.020 0.018 b 0.446 0.207 2.023 3.105 0.126 0.0002 0.232 IL-8/IL-10 M 0.432 0.308 0.584 0.219 9.768 7.391 5.684 2.256 C 0.094 0.094 c 0.064 0.063 d 7.167 4.607 2.432 2.020 e 0.066 0.000001 0.575 a p 0.001; b p 0.033; c p 0.027; d p 0.00003; e p 0.019. IL interleukin.

192 CHEN ET AL Ann Thorac Surg MARROW STROMAL CELLS FOR CARDIOMYOPLASTY 2010;90:190 8 Fig 1. Infarct scar area and the total area of left ventricular myocardium are shown at 12 hours and extending to 2 weeks. The red represents extracellular matrix deposition in scar tissue, and the grey area represents myocardium (original magnification 4). Compared with group M, group C had a significantly less area of extracellular matrix deposition. surrounding tissue. The LV was cut transversely in half, evenly along the largest circumference of the infarcted area. One half was fixed and stained for -galactosidase activity using X-gal staining, as described previously [12]. The remaining half of the heart specimen was finely minced and put into a tube with the RNALater solution (Qiagen, Valencia, CA) and stored at 4 C for RNA extraction. Quantitative Real-Time Polymerase Chain Reaction Total RNA was isolated with an RNeasy kit (Qiagen) according to the manufacturer s instructions. Total RNA (5 g) was subjected to first-strand complimentary DNA synthesis with QuantiTect Reverse Transcription Kit (Qiagen). Quantitative real-time polymerase chain reaction (RT- PCR) was performed to compare the changes in expression of different inflammatory cytokines using the following primers: -Actin: F 5=-CACGCCATCCTGCGTCTGGA-3=; R5=-GCACCGTGTTGGCGTAGAG-3= IL-1 : F5=-TCAGGCAGATGGTGTCTGTC-3=; R5=-GGTCTATATCCTCCAGCTGC-3= IL-6: F 5=-AACGCCTGGAAGAAGATGCC-3=; R5=-CTCAGGCTGAACTGCAGGAA-3= IL-8: F 5=-TTTCTGCAGCTCTCTGTGAGG-3=; R5=-CTGCTGTTGTTGTTGCTTCTC-3= IL-10: F 5=-GCGACTTGTTGCTGACCGG-3=; R5=-GAACCTTGGAGCAGATTTTG-3= Fig 2. Histologic examination of the hearts shows marrow stromal cell retention in the left ventricle but a decrease in number over time. X-gal staining (original magnification 200) confirmed successful engraftment of marrow stromal cells (blue cells) at various times points (arrows).

Ann Thorac Surg CHEN ET AL 2010;90:190 8 MARROW STROMAL CELLS FOR CARDIOMYOPLASTY 193 Table 3. Matrix Metallopeptidase 2/Tissue Inhibitor of Matrix Metallopeptidase 1 Gene Expression Ratio Group 12 Hours, Folds 24 Hours, Folds 1 Week, Folds 2 Weeks, Folds M 2.072 0.937 0.344 0.146 9.571 15.653 1.053 0.703 C 0.872 0.721 a 0.322 0.657 8.832 10.414 0.564 0.623 0.720 0.001 0.996 a p 0.015. The My-iQ system (Bio-Rad, Hercules, CA) was used to monitor RT-PCR amplification using SYBR Green I. Reaction conditions were as follows: hot start for 120 seconds at 95 C, melting at 95 C for 15 seconds, annealing at 56 C for 12 seconds, and amplification at 72 C for 15 seconds. Samples were run in duplicate, and the average crossing point value was used for calculations by the method of 2- CT. The relative quantitation value of a target gene, normalized to -actin as the internal control gene, was expressed as a number, which indicated the relative expression compared with that gene. The possibility of amplifying contaminating DNA and nonspecific amplification were avoided with precautions [11]. Conventional PCR and Gel Bands Semiquantification Using the same samples as in quantitative RT-PCR assay, the complimentary DNA was also used in regular PCR to compare changes in the genes expressing matrix metallopeptidase 2 (MMP-2) and tissue inhibitor of metallopeptidase 1 (TIMP-1). We mixed the primers of -actin and TIMP-1 together in one tube for each running, and MMP-2 was done separately with the following thermal protocol: 50 C for 30 minutes, 95 C for 15 minutes, 30 cycles at 94 C for 30 seconds, 55 C for 30 seconds, 72 C for 1 minute, and the final extension step at 72 C for 10 minutes. Primers used for amplification were: MMP-2: F 5=-AGGACAAGTGGTCCGCGTAAAG-3=; R5=- CACTTCCGGTCATCATCGTAGT-3= TIMP-1: F 5=-ACTAAGATGCTCAAAGGATTCG-3=; R5=-ATCGCTCTGGTAGCCCTTCT-3= -Actin: F 5=-CACGCCATCCTGCGTCTGGA-3=; R5=-AGCACCGTGTTGGCGTAGAG-3= Images of the PCR ethidium bromide-stained agarose gels were acquired with an image system (Cohu Inc, Poway, CA), and quantification of the bands was performed by ImageJ-1.41 software (National Institutes of Health, Bethesda, MD). Band intensity was expressed as relative absorbance units normalized to -actin. The ratio between the samples was determined and compared. Collagen Content and Tissue Injury Measurement Five consecutive 5- m sections were prepared from the midportion of the infracted area in all samples. Sections were stained with the collagen-specific dye Sirius red 3BA in a saturated picric acid solution to allow clear discrimination between cardiomyocytes and ECM. The slides were scanned at 2400 dpi with 4 magnification, and the pictures were red-green-blue split to the green color layer for measurement. In the digital images, scar area and the total area of myocardium were traced manually by a blinded observer and measured automatically by the computer by the ImageJ-1.41 software. Infarct size, expressed as a percentage, was calculated by dividing the sum of infarct areas by the total sum of LV areas, including those without infarct scar, and multiplying by 100. The following morphologic criteria were applied in a blinded fashion for tissue injury assessment: score 0, no damage; score 1 (mild), interstitial edema and focal necrosis; score 2 (moderate), diffuse myocardial cell swelling and necrosis; score 3 (severe), necrosis with presence of contraction bands and neutrophil infiltrate; score 4 (highly severe), widespread necrosis with presence of contraction bands, neutrophil infiltrate, and hemorrhage. Statistical Analysis All data are expressed as mean standard deviation. Data were analyzed using SPSS 16.0 software (SPSS Inc, Chicago, IL). The t test and two-way repeated analysis of variance (ANOVA) were used to make comparisons and test the differences for continuous variables. A value of p 0.05 was considered statistically significant. Table 4. Scar Area of the Left Ventricle Group 12 Hours, % 24 Hours, % 1 Week, % 2 Weeks, % M 2.01 1.37 1.95 0.91 19.30 10.28 24.46 10.97 C 1.97 0.41 0.75 0.44 a 9.36 4.96 b 7.57 5.81 c 0.000001 0.000001 0.000001 a p 0.0001; b p 0.030; c p 0.000008.

194 CHEN ET AL Ann Thorac Surg MARROW STROMAL CELLS FOR CARDIOMYOPLASTY 2010;90:190 8 Table 5. Tissue Injury Group 12 Hours 24 Hours 1 Week 2 Weeks M 1.22 0.46 1.65 0.53 2.15 0.41 1.70 0.42 C 1.73 1.08 3.16 0.78 a 1.71 0.38 b 0.98 0.46 c 0.072 0.000001 0.000001 a p 0.000004; b p 0.014; c p 0.0004. Results Inflammatory Cytokines Gene Expression Changes in the LV The fresh heart tissue slices for the total RNA extraction included infarcted area, periinfarcted area, and normal heart tissue from the LV; therefore, the gene expression results reflected the global gene expression changes of the entire LV. Group C showed more of an increase than group M in gene expressions for all the measured proinflammatory and antiinflammatory cytokines IL-1, IL-6, and IL-10 from 12 hours to 2 weeks and for IL-8 from 12 to 24 hours. The increase in cytokines was significant, especially during the first 12 to 24 hours after MI and gradually tapered over time. In particular, the increase in antiinflammatory IL-10 was much more exaggerated during this early phase (Table 1). Repeated measures ANOVA revealed that differences were significant over time within groups in the expression of IL-1, IL-8, and IL-10. The ratios of proinflammatory/antiinflammatory cytokine gene expression reflected the relationship of proinflammatory vs antiinflammatory cytokines changes in Table 1 and were significantly lower in group C, especially during the first 12 to 24 hours after cell therapy. The significance gradually tapered off over time, except for the IL-8/IL-10 ratio at 2 weeks postoperatively (Table 2). All of the ratios were also significantly different over time within groups, as was demonstrated by repeated measures ANOVA. The proinflammatory/antiinflammatory cytokine ratio suggested that MSC therapy modified the cytokine profiles in the infarct microenvironment in favor of an antiinflammatory effect early after cell implantation. MMP-2 and TIMP-1 Gene Expressions in LV The PCR analysis of MMP-2 and TIMP-1 gene expression indicated that MSC injections caused a reduction in the expression of MMP-2 at 12 hours, 24 hours, 1 week, and 2 weeks (0.50-, 0.57-, 0.95, 0.80-fold, respectively), and an increase of TIMP-1 at corresponding times (1.24-, 1.09-, 1.02-, 2.05-fold, respectively). The ratio of MMP-2/ TIMP-1 was significantly reduced in group C especially during early phase at 12 hours, and a similar trend was observed at 24 hours, 1 week, and 2 weeks (Table 3). A significant difference over time within groups was found with repeated measures ANOVA. ECM Deposition Measurement Histopathologic analysis revealed a significant reduction of scar ECM deposition in the cell therapy group at 24 hours, 1 week, and at 2 weeks. Repeated measures ANOVA demonstrated that scar ECM deposition was very significantly reduced over time in group C (Fig 1 and Table 4). Histopathologic Evaluation Histologic examination of serial sections of every heart in group C confirmed the successful engraftment of laczlabeled MSCs (blue cells) at 12 hours, 24 hours, and 1 week after cell implantation, but labeled MSCs were scarce at 2 weeks (Fig 2). Tissue injury and inflammatory cell infiltration early after MI and cell therapy at 12 and 24 hours were actually higher in group C than in group M (Table 5). However, subacute myocardial injury and inflammatory cell infiltration at 1 and 2 weeks showed a significant decrease in group C compared with group M. The difference over time within groups was significant, as was demonstrated by repeated measures ANOVA. Cardiac Functional Assessment The baseline EF and FS were not significantly different between groups. Compared with group M, group C showed improvement in EF as early as 12 hours after treatment (group M 0.599 0.069 vs group C 0.651 0.112, p 0.069), and reached significance at 24 hours (group M 0.674 0.074 vs group C 0.750 0.111, p 0.028), 1 week (group M 0.600 0.131 vs group C 0.758 0.058, p 0.0004), and 2 weeks (group M 0.450 0.112 vs group C 0.762 0.124, p 0.0002). Repeated measures ANOVA revealed that EF differences were significant over time within groups (p 0.003) and between groups (p 0.000001). FS showed a similar trend with improvement in group C at 12 hours (group M 0.283 0.044 vs group C 0.322 0.082, p 0.051) and was significantly increased at 24 hours (group M 0.333 0.055 vs group C 0.391 0.090, p 0.038), 1 week (group M 0.282 0.037 vs group C 0.419 0.119, p 0.001), and 2 weeks (group M 0.198 0.061 vs group C 0.455 0.111, p 0.001), respectively. Repeated measures ANOVA also revealed that FS differences were significant over time within groups (p 0.006) and between groups (p 0.000001). Taken together, these data suggest that the beneficial effects of stem cell therapy actually occurred quite early after myocardial injury. Comment Recent studies have reported that transplanting MSCs improves cardiac function and that MSCs could survive

Ann Thorac Surg CHEN ET AL 2010;90:190 8 MARROW STROMAL CELLS FOR CARDIOMYOPLASTY 195 from several days to beyond 6 months [1 3]. There have been controversies about whether stem cells actually differentiate into functioning myocytes and whether the number of engrafted cells could account for the improved function [14 18]. In an acute renal failure model, Tögel and colleagues [11] observed significant renal functional recovery within 3 days after MSC therapy, a period in which none of the administered MSCs had differentiated into kidney tubular or endothelial cell phenotypes. In the current study, we also demonstrated that isogeneic MSCs provided a significant functional benefit because group C showed a significant improvement in cardiac function as early as 24 hours after cell therapy and up to 2 weeks. Although few studies have evaluated the early cardiac functional changes after MSC treatment, Francis and colleagues [14] showed improved cardiac function as early as 1 hour after transplanting MSCs, suggesting that mechanisms other than cell differentiation or cell fusion might be responsible at different stages of cell-based therapy. Tang and colleagues [19] also raised doubts about stem cell differentiation as a sole contributor to myocardial regeneration and subsequent functional recovery because very few engrafted MSCs were shown to express specific cardiac markers, such as connexin-43 and cardiac troponin I. The small number of engrafted cells in proportion to the functional improvement suggested that increased myocardial mass as a sole mechanism for improved cardiac function appeared less plausible. Some reports have suggested other mechanisms, among which the secretion of various mediators through a paracrine effect from the transplanted MSCs has gained increasing attention [6, 20]. Persistent proinflammatory cytokine elevation has been associated with a worse clinical outcome in patients suffering recent MI. Induction and release of the proinflammatory cytokines IL-1, tumor necrosis factor- (TNF- ), IL-6, and IL-8 are consistently found in experimental models and clinical cases of MI [21, 22]. There is also evidence showing a dramatic reduction in infarct size with the use of specific antiinflammatory strategies [23 25]. Our current study suggests that transplanted MSCs up-regulated both proinflammatory and antiinflammatory gene expressions in the infracted myocardium early after implantation. This cytokine profile was contrary to our original hypothesis that MSCs might inhibit the expression of proinflammatory cytokines while increasing the antiinflammatory cytokine activity. Our findings, however, are consistent with a clinical study by Kilic and colleagues [10], who showed that serum concentrations of IL-1 and IL-6 were significantly higher in patients with new coronary events. On the other hand, Tögel and colleagues [11] showed that at 24 hours after cell therapy, the expression of proinflammatory/antiinflammatory cytokines reversed significantly in favor of antiinflammatory IL-10, basic fibroblastic growth factor, transforming growth factor-, and B-cell lymphoma 2 up-regulation in MSC treated kidneys. This was associated with measurable recovery in renal function too early to be related to neoglomerular cell formation. Interestingly, such renoprotection was not obtained with syngenic fibroblasts. Histologic results of our study also demonstrated that acute myocardial injury caused fluxes of inflammatory cells infiltration, cytokine mediators, and tissue injury in groups M and C within the first 24 hours. However, the antiinflammatory cytokines surge exceeded that of the proinflammatory counterparts, leading to more favorable ECM remodeling and functional improvement in longer follow-up. These findings suggested that the transplanted MSCs did not change or inhibit the inflammatory reaction histopathologically after acute MI. Reactive neutrophil cells and damaged endothelial cells of blood vessels classically produce the proinflammatory cytokines, and therefore, it is not expected that MSCs implantation after MI would reverse this phenomenon immediately. The tissue trauma associated with MSC implantation per se may perhaps further aggravate the inflammatory response. On the other hand, these findings could also reflect the insufficient number of retained MSCs to completely suppress the acute inflammation, because many injected cells might have been lost early after injection due to mechanical leakage or washout, as suggested by Teng and colleagues [26]. By 12 to 24 hours after MSC implantation, however, the retained stem cells may respond and subsequently reversed the proinflammatory/antiinflammatory ratio of the microenvironment. In the current study, IL-10 gene expression was remarkably up-regulated relative to the proinflammatory cytokines early after MSC therapy and showed evidence of strong cytoprotective effects on the cardiomyocytes. This was associated with decreased MMP-2 and increased TIMP-1 gene expression during the repair process after MSC therapy. MMP-mediated matrix degradation is crucial in infarct healing and the pattern of left ventricular remodeling, leading to various LV functional changes in patients with heart failure. Berry and colleagues [27] showed that MSC injections limited cell apoptosis and fibrosis and thus improved LV function and reduced dilatation. Recent work by Guo and colleagues [28] examined the cytokines and functional parameters 4 weeks after cell therapy and revealed a decrease in deposition of collagen types I and III and decreased gene and protein expression of MMP-1 and TIMP-1. These results correlated with attenuated ventricular dilatation and thickening. Interestingly, the authors also reported decreased expression of proinflammatory cytokines such as TNF-, IL-1, and IL-6, suggesting a possible role for inflammatory mediators in the remodeling process [28, 29]. IL-1 and TNF- have been shown to increase MMP-2 and MMP-9 matrix degradation and activity [30], whereas IL-10 and transforming growth factor- enhanced TIMP synthesis, regulating the matrix degradation and producing a favorable myocardial remodeling for better functional recovery. Therefore, changes in the cytokine milieu might play a key role in regulating ECM after myocardial injury. In our study, the analysis of antiinflammatory IL-10 gene expression, ECM formation, and functional recovery cor-

196 CHEN ET AL Ann Thorac Surg MARROW STROMAL CELLS FOR CARDIOMYOPLASTY 2010;90:190 8 related with result reported by others and suggests a potential mechanism of stem cell therapy in an MI model. The temporal and spatial discrepancies of various cytokine expressions during different scenarios of tissue organ injuries have been suggested to be more important in determining the outcome rather than the individual mediator per se. One study showed IL-6 expression was enhanced early during the first 6 hours after reperfusion of the previously ischemic myocardium [31]. Others have suggested that after 24 hours of reperfusion, the IL-6 mrna expression was down-regulated in the same ischemic segments in which IL-10 mrna up-regulation was found [11]. On the other hand, in a non-reperfused infarct model, IL-6 over-expression lasted as long as 24 hours [32], but in such a permanent coronary occlusion model, both IL-10 and IL-6 mrna over-expression persisted simultaneously [33]. These findings suggested that although a surge in proinflammatory cytokines invariably occurred after injury, the expression of antiinflammatory mediators reflected a counter-regulatory response that is important for survival and favorable organ function recovery. Recent studies have supported the notion that a lack of an appropriate antiinflammatory counter-balance in response to a certain proinflammatory reaction, rather than the individual proinflammatory or antiinflammatory mechanism, might be the culprit of the increased coronary risk [34, 35]. In our study, proinflammatory and antiinflammatory cytokine gene expressions were both up-regulated after MSC therapy. However, the LV functional recovery suggested that the two opposing cytokine forces had an unbalanced effect, such that the antiinflammatory effects predominated and resulted in more favorable histopathologic remodeling and subsequent improved cardiac functional improvement. This possible paracrine mechanism of MSC therapy may at least partly explain the early functional improvement after MI after stem cell therapy, before transdifferentiation or other more protracted mechanisms take place at a later period. The current study has some limitations that merit further investigations. First, it is possible that MSCs act through other collective multifactorial mechanisms by many complex mediators. The role of inflammatory cytokines may only be a fraction of the overall mechanisms of stem cell therapy in this MI model. It is, however, interesting that a brief and possibly small amount of retained MSCs might exert profound longer protective effects on the injured myocardium, as many others have demonstrated. Second, we did not determine the fate, the subtype, and the actual number of MSCs required to produce the beneficial effects. This is, however, a question that is actively pursued by many others in the field, and no agreement has yet been reached at present. Third, the current rodent model is far from clinical relevance, and a study in larger animals should be considered. On the other hand, the use of small rodents is a well accepted and more preferable model in the study of molecular mechanisms of stem cell therapy. Better understanding of the mechanisms of stem cell therapy will undoubtedly lead to future larger-animal studies and provide information for evidence-based research trials in the clinical setting. The present study proposes a new mechanism implicating antiinflammatory cytokines in the early benefit of MSC implantation after MI. Fourth, this study did not address the relative contribution of various cytokines that surge from damaged tissue or injected MSCs, and further investigation using a knock-out rodent model will help to answer this question. However, we confirmed that the presence of MSCs modified these mediators and resulted in better functional recovery. Also, whether injection of other cell sources would produce the observed benefit is a pertinent question that we did not address. Tögel and colleagues [11], however, showed that the administration of fibroblasts did not reproduce the renoprotective effects in a renal injury model. Finally, the cell therapy was injected 10 minutes after coronary ligation, which may not necessarily simulate clinical settings. However, the current model maximizes the effect of inflammatory cytokines after acute MI. Further study in a large-animal model will allow the introduction of MI in a noninvasive fashion, followed by cell therapy. This will overcome the current limitation with our rodent model in which creation of MI and reintervention for MSC injection at a later time will produce prohibitively high surgical mortality. In summary, our study shows that isogeneic MSCs injected into periinfarcted regions of ischemic myocardium after coronary ligation reduces the proinflammatory/ antiinflammatory cytokine gene expression ratio that is correlated with improved cardiac function. The ratio of cytokine gene expressions favoring antiinflammation with less scar formation suggests that a paracrine mechanism may at least partly explain the early functional improvement. These findings open up a new and potentially important mode of action for MSCs, which also provides further impetus to study the role of IL-10 in the pathophysiology of MI. We thank Dr James Hanley, Department of Epidemiology and Statistics, McGill University, for his assistance with statistics. This research was supported in part by the Natural Sciences and Engineering Research Council (NSERC) and Fonds de la Recherche en Sante du Quebec (FRSQ). References 1. Amado LC, Saliaris AP, Schuleri KH, et al. Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarction. Proc Natl Acad Sci U S A 2005;102:11474 9. 2. Orlic D, Kajstura J, Chimenti S, et al. Bone marrow cells regenerate infarcted myocardium. Nature 2001;410:701 5. 3. Atoui R, Shum-Tim D, Chiu RC. Myocardial regenerative therapy: immunologic basis for the potential universal donor cells. Ann Thorac Surg 2008;86:327 34. 4. Xu M, Wani M, Dai Y, et al. Differentiation of bone marrow stromal cells into the cardiac phenotype requires intercellular communication with myocytes. Circulation 2004;110:2658 65.

Ann Thorac Surg CHEN ET AL 2010;90:190 8 MARROW STROMAL CELLS FOR CARDIOMYOPLASTY 197 5. Kamihata H, Matsubara H, Nishiue T, et al. Implantation of bone marrow mononuclear cells into ischemic myocardium enhances collateral perfusion and regional function via side supply of angioblasts, angiogenic ligands, and cytokines. Circulation 2001;104:1046 52. 6. Besse A, Trimoreau F, Praloran V, et al. Effect of cytokines and growth factors on the macrophage colony-stimulating factor secretion by human bone marrow stromal cells. Cytokine 2000;12:522 5. 7. Xu X, Xu Z, Xu Y, et al. Effects of mesenchymal stem cell transplantation on extracellular matrix after myocardial infarction in rats. Coron Artery Dis 2005;16:245 55. 8. Molina EJ, Palma J, Gupta D, et al. Reverse remodeling is associated with changes in extracellular matrix proteases and tissue inhibitors after mesenchymal stem cell (MSC) treatment of pressure overload hypertrophy. J Tissue Eng Regen Med 2009;3:85 91. 9. Mannello F. Commentary: multipotent mesenchymal stromal cell recruitment, migration, and differentiation: what have matrix metalloproteinases got to do with it? Stem Cells 2006;24:1904 7. 10. Kilic T, Ural D, Ural E, et al. Relation between proinflammatory to anti-inflammatory cytokine ratios and long-term prognosis in patients with non-st elevation acute coronary syndrome. Heart 2006;92:1041 6. 11. Tögel F, Hu Z, Weiss K, Isaac J, Lange C, Westenfelder C. Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms. Am J Physiol Renal Physiol 2005;289:F31 42. 12. Atoui R, Asenjo JF, Chen G, et al. Marrow stromal cells as universal donor cells for myocardial regenerative therapy: their unique immune tolerance. Ann Thorac Surg 2008;85:571 9. 13. Fukuhara S, Tomita S, Yamashiro S, et al. Direct cell-cell interaction of cardiomyocytes is key for bone marrow stromal cells to go into cardiac lineage in vitro. J Thor Cardiovasc Surg 2003;125:1470 80. 14. Francis J, Chu Y, Johnson AK, et al. Acute myocardial infarction induces hypothalamic cytokine synthesis. Am J Physiol Heart Circ Physiol 2004;286:H2264 71. 15. Dai W, Field LJ, Rubart M, et al. Survival and maturation of human embryonic stem cell-derived cardiomyocytes in rat hearts. J Mol Cell Cardiol 2007;43:504 6. 16. Suzuki K, Murtuza B, Beauchamp JR, et al. Dynamics and mediators of acute graft attrition after myoblast transplantation to the heart. FASEB J 2004;18:1153 5. 17. Suzuki K, Murtuza B, Beauchamp JR, et al. Role of interleukin-1beta in acute inflammation and graft death after cell transplantation to the heart. Circulation 2004;110:II219 24. 18. Dow J, Simkhovich BZ, Kedes L, et al. Washout of transplanted cells from the heart: a potential new hurdle for cell transplantation therapy. Cardiovasc Res 2005;67:301 7. 19. Tang YL, Zhao Q, Zhang YC, et al. Autologous mesenchymal stem cell transplantation induces VEGF and neovascularization in ischemic myocardium. Regul Pept 2004;117:3 10. 20. Tang YL, Zhao Q, Qin X, et al. Paracrine action enhances the effects of autologous mesenchymal stem cell transplantation on vascular regeneration in rat model of myocardial infarction. Ann Thorac Surg 2005;80:229 36. 21. Compton SJ, Cairns JA, Holgate ST, et al. The role of mast cell tryptase in regulating endothelial cell proliferation, cytokine release, and adhesion molecule expression: tryptase induces expression of mrna for IL-1 beta and IL-8 and stimulates the selective release of IL-8 from human umbilical vein endothelial cells. J Immunol 1998;161:1939 46. 22. Wollert KC, Drexler H. The role of interleukin-6 in the failing heart. Heart Fail Rev 2001;6:95 103. 23. Lacraz S, Nicod LP, Chicheportiche R, et al. IL-10 inhibits metalloproteinase and stimulates TIMP-1 production in human mononuclear phagocytes. J Clin Invest 1995;96:2304 10. 24. Ono K, Matsumori A, Shioi T, et al. Cytokine gene expression after myocardial infarction in rat hearts possible implication in left ventricular remodeling. Circulation 1998;98:149 56. 25. Paoletti R, Gotto AM Jr, Hajjar DP. Inflammation in atherosclerosis and implications for therapy. Circulation 2004;109: III20 6. 26. Teng CJ, Luo J, Chiu RC, et al. Massive mechanical loss of microspheres with direct intramyocardial injection in the beating heart: implications for cellular cardiomyoplasty. J Thorac Cardiovasc Surg 2006;132:628 32. 27. Berry MF, Engler AJ, Woo YJ, et al. Mesenchymal stem cell injection after myocardial infarction improves myocardial compliance. Am J Physiol Heart Circ Physiol 2006;290: H2196 203. 28. Guo J, Lin GS, Bao CY, Hu ZM, Hu MY. Anti-inflammation role for mesenchymal stem cells transplantation in myocardial infarction. Inflammation 2007;30:97 104. 29. Cleutjens JP, Kandala JC, Guarda E, et al. Regulation of collagen degradation in the rat myocardium after infarction. J Mol Cell Cardiol 1995;27:1281 92. 30. Siwik DA, Chang DL, Colucci WS. Interleukin-1beta and tumor necrosis factor-alpha decrease collagen synthesis and increase matrix metalloproteinase activity in cardiac fibroblasts in vitro. Circ Res 2000;86:1259 65. 31. Deten A, Volz HC, Briest W, Zimmer HG, et al. Cardiac cytokine expression is up-regulated in the acute phase after myocardial infarction. Experimental studies in rats. Cardiovasc Res 2002;55:329 40. 32. Fuchs M, Hilfiker A, Kaminski K, et al. Role of interleukin-6 for LV remodeling and survival after experimental myocardial infarction. FASEB J 2003;17:2118 120. 33. Frangogiannis NG, Mendoza LH, Lindsey ML, et al. IL-10 is induced in the reperfused myocardium and may modulate the reaction to injury. J Immunol 2000;165:2798 808. 34. Tziakas D, Chalikias G, Kaski J, et al. Inflammatory and anti-inflammatory variable clusters and risk prediction in acute coronary syndrome patients: a factor analysis approach. Atherosclerosis 2007;193:196 203. 35. Dominguez-Rodriguez A, Abreu-Gonzalez P, Garcia- Gonzalez MJ, Kaski JC. Soluble CD40 ligand: interleukin-10 ratio predicts in-hospital adverse events in patients with ST-segment elevation myocardial infarction. Thromb Res 2007;121:293 9. INVITED COMMENTARY Despite significant medical advancements, myocardial ischemic disease is still a leading cause of death in the world. The regenerative capacity of the heart is now generally believed to exist; however, its capacity to regenerate an adequate amount of myocardial tissue during ischemic insults is insufficient. Subsequently, cardiomyocytes are lost as a result of myocardial ischemia, triggering cardiac dysfunction and eventual cardiac failure. One of the most promising novel therapies for cardiac failure is cellular cardiomyoplasty, which relies on transferring cells into the compromised myocardium to improve function. Although initial results have shown promise, the effect of cellular cardiomyoplasty has been limited due to the conflicting results seen in experimental animal models and clinical trials. Many questions remain unanswered in this approach to treating heart failure. First, the mechanism whereby cellular cardiomyoplasty improves myocardial performance is still unknown. Second, the optimal cell type skeletal myoblasts vs bone marrow-derived cells 2010 by The Society of Thoracic Surgeons 0003-4975/$36.00 Published by Elsevier Inc doi:10.1016/j.athoracsur.2010.05.011