INTIMP-03357; No of Pages 7 International Immunopharmacology xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

International Immunopharmacology journal homepage: www.elsevier.com/locate/intimp

F

a

8

a r t i c l e

9 10 11 12 13

Article history: Received 7 April 2014 Received in revised form 2 July 2014 Accepted 27 July 2014 Available online xxxx

i n f o

a b s t r a c t

Mesenchymal stem cells (MSCs) were found to provide an effective therapeutic role in inflammatory diseases by modulating inflammatory responses and tissue regeneration by their differentiation ability. The present work sought to demonstrate the potential therapeutic use of MSCs in treating chronic inflammatory bowel disease (IBD) in mice. A new model to induce chronic IBD based on alternative administration periods of Dextran Sodium Sulfate (DSS). Mice were divided into 2 groups; one was treated with MSCs and the other was treated with phosphate-buffered saline (PBS). Assessment of therapeutic efficacy of MSCs was by measuring weight, stool scoring, histopathologic examination, and measuring the gene expression of inflammatory markers: Interleukin-23 (IL-23), Tumor necrosis factor-α (TNF-α), Interferon-γ (IFN-γ), and Intercellular adhesion molecule-1 (ICAM-1). The results showed that DSS administration causes bloody and watery stool, weight loss, and altered histopathologic picture. MSC treated mice showed a significant improvement in stool condition, weight gain, and normal histopathologic picture compared to the PBS treated mice. Moreover, gene expressions of inflammatory markers in the intestines of the MSC treated mice were also significantly lower than those of the PBS treated mice. In conclusion, the data here showed that MSCs have a clear potential efficacy in the treatment for IBD, as their immune modulation effects include inhibition in the expression of key inflammatory markers that each plays an important role in the pathogenesis of IBD. © 2014 Elsevier B.V. All rights reserved.

Keywords: DSS induced colitis Interleukin Tumor necrosis factors Interferon Adhesion molecules Bone marrow derived mesenchymal stem cell transplantation Immunomodulation

42 40 39

R

41

44

Q11 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37

R

38

43

R O

Department of Biochemistry, Faculty of Pharmacy, Misr International University, Cairo, Egypt Department of Biochemistry, Faculty of Medicine, Cairo University, Egypt Department of Biochemistry, Faculty of Pharmacy, Al Azhar University, Cairo, Egypt

P

c

E

Q1214 Q13 15 16 17 18 19 20 21 22

b

O

5 6Q6 7

D

4

Ahmed Gouda Abdel Rahman Abdel Salam a,⁎, Hazem M. Ata b, Tarek M. Salman c, Laila A. Rashed b, Dina Sabry b, Mona F. Schaalan a

E

Q53Q4

T

2

Potential therapeutic utility of mesenchymal stem cells in inflammatory bowel disease in mice

C

1

1. Introduction

U

N C O

Inflammatory bowel disease (IBD) is a chronic idiopathic disease 45 mainly represented by two major forms, ulcerative colitis (UC) and 46 Crohn's disease (CD) [1]. While the precise etiology of IBD is still 47 Q14 unknown, possible etiologies include the role of environmental factors 48 [2], genetics [3], microbial factors [4], and mucosal immune defects 49 [5]. Based on these etiologies, many treatment lines were developed. 50 The current range of treatments for IBD covers both conventional and 51 biological therapies. Conventional therapy includes the use of anti52 inflammatory drugs, immunosuppressive agents, antibiotics, and 53 probiotics; biological therapies mainly include the use of different 54 anti-TNF-α agents, and a plethora of other novel biological agents [6]. 55 The mechanism of DSS-induced colitis is mainly due to the direct 56 toxicity to the colonic epithelial cells, subsequently increasing the 57 permeability of the intestinal mucosa and allowing the transport of 58 luminal bacterial products from the bowel lumen to the submucosal 59 tissue [7]. This hypothesis is bolstered by the work of Leung et al. [8], ⁎ Corresponding author. Tel.: +20 1 589095. E-mail address: [email protected] (A.G.A.R.A. Salam).

who demonstrated that activated T cells in the colonic mucosa play a role in modulating the immunological reaction in DSS induced colitis in mice by interacting with lysed bacterial cell wall lipopolysaccharide and activated macrophages. It has been shown that CD4+ T cells play an important role in the development of DSS induced experimental colitis [9]. According to Yan et al. [10], DSS administration increases the gene expression of TNF-α, IFN-γ and IL-12 in the area of the distal colon due to massive infiltration of macrophages, T cells, B cells and neutrophils. Moreover, DSS causes an increase in the level of ICAM-1 that leads to the migration of leukocytes to the site of inflammation [7]. According to Dieleman et al. [11], acute DSS induced colitis is predominantly macrophage-driven colitis, while, Dieleman et al. [12] reported that chronic DSS induced colitis is mediated by Th1 and Th2 activation. Thus, it appears that activated macrophages alone lead to an acute colitis but that only activated macrophages interacting with activated T-cells lead to chronic disease [13]. Bone marrow transplantation (BMT) has improved not only hematological diseases such as leukemia, but also autoimmune diseases. However, BMT significantly affects the systemic immune system of recipients; whether it is effective for IBD remains controversial [14]. Bone marrow contains hematopoietic stem cells and non-hematopoietic

http://dx.doi.org/10.1016/j.intimp.2014.07.030 1567-5769/© 2014 Elsevier B.V. All rights reserved.

Please cite this article as: Salam AGARA, et al, Potential therapeutic utility of mesenchymal stem cells in inflammatory bowel disease in mice, Int Immunopharmacol (2014), http://dx.doi.org/10.1016/j.intimp.2014.07.030

60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80

Q7 Q8 Q9 Q10

103

2.1. Animals

104 105

110

Eight week female albino mice with an average weight of 18–25 g (provided from The Nile Company for Pharmaceutical Industries) were housed in the animal house facility at Misr International University and kept at 25 °C with normal light/dark cycle with free access to food and water for 7 days. 8 week male mice were housed in the same conditions and used for flushing of the bone marrow for isolation and culture of MSCs.

111

2.2. Development of DSS-induced colitis model in mice (pilot study)

112

According to Wirtz et al. [21], the appropriate cycle for the induction of chronic IBD was 5 ml DSS/day/mouse for 7 days, followed by replacing DSS on Day 8 with DSS-free drinking water for another 14 days, for three cycles. This approach was recommended because prolonged administration of DSS causes high mortality rates, and DSS-induced colitis (being a reversible model) requires administration of DSS and normal water alternatively for a specific period of time. Thus, a pilot study was undertaken in two parts: the first to determine the appropriate concentration of DSS to induce IBD, and the second to determine the proper DSS/ water cycle for our experimental purposes.

113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137

2.4. Administration of MSCs

154

2.2.1. Part A: assessment of determination of appropriate DSS concentration To determine the appropriate DSS concentration, 150 mice were placed in individual cages and randomly divided into three groups. In the first group, each mouse was given 5 ml of 2% DSS, in the second group 5 ml of 4% DSS, and in the third group 5 ml of 5% DSS for 7 days. 2.2.2. Part B: determination of appropriate DSS/water cycle To determine the appropriate DSS/water cycle to be used, 90 mice were placed in individual cages and randomly divided into three groups. In the first group, mice were administered 5 ml of 4% DSS for 7 days followed by 14 days of water administration. In the second group, mice were administered 5 ml of 4% DSS for 7 days followed by 10 days of water administration. In the third group, mice were administered 5 ml of 4% DSS for 7 days followed by 5 days of water administration. The suitable model to induce chronic colitis in mice using DSS is by administration of 4% DSS for 7 days followed by administration of water for 5 days for 3 consecutive cycles.

141 142 143 144 145 146 147 148 149 150 151 152 153

After 7 days of 4% DSS administration, mice were divided into two 155 groups; the first group, injected with PBS, and the second group, 156 injected intraperitoneally with MSCs [20] . 157 158

D

The treatment efficacy of the MSC was evaluated by three methods: 159 physical examination, histopathological examination, and measuring 160 the gene expression of inflammatory markers. 161 2.5.1. Macroscopic/microscopic evaluation of therapeutic efficacy of MSC on DSS-colitis The following indices of experimental colitis were monitored daily among the various experimental mice: (a) body weight, (b) food and water intake, and (c) stool condition. Daily, the mice were removed from their individual cages and weighed. The amount of food present and the level of water in their cage bottle were also noted. Loss of food, due to powdering, and water, due to any seeping/forced loss (i.e., if bedding was pushed up at the bottle), was noted when apparent. Stool condition was subjectively scored from grades 0 to 3 as described: grade 0 indicates a normal-to-semi-solid stool and no blood; grade 1 indicates a normal-to-semi-solid stool and blood-tinged; grade 2 indicates a semi-solid-to-fluid stool with definite evidence of blood; and, grade 3 indicates a bloody fluid stool [20].

162 163

2.5.2. Histopathological examination of intestine tissue Necropsy samples were taken from the intestine of mice in different groups and fixed in 10% formalin for 24 h. After rinsing in tap water, the samples were serially dehydrated in alcohol (methanol, ethanol, and then absolute ethanol). Specimens were then cleared in xylene, and embedded in paraffin. Samples were then sectioned at 4-μm thickness using a microtome. The tissue sections were collected on glass slides, de-paraffinized, stained with hematoxylin and eosin, and then subjected to blinded examination under a light microscope [20].

176 177

2.5.3. Inflammation by measuring the gene expression of inflammatory markers The mRNA expression of inflammatory mediators such as TNF-α, IFN-γ, IL-23, and ICAM-1 in the colon was measured by real-time reverse transcription-polymerase chain reaction (RT-PCR). Local mRNA expression was measured in the rectum and distal colon (slightly orally to the rectum) samples. Total RNA was extracted from the tissues using QIAamp RNA mini kit (Qiagen, Cairo, Egypt) according to the manufacturer's instructions. PCR primers and Taqman probes for each mediator were used. Real-time quantitative RT-PCR analyses were performed. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH)

185 186

T

108 109

C

106 107

139 140

2.5. Efficacy of MSC in treating IBD

E

98 99

R

96 97

R

94 95

O

92 93

C

90 91

N

88 89

U

87

Bone marrow was harvested by flushing the tibiae and femurs of 8-wk-old male albino mice (18–25 g) with Dulbecco's Modified Eagle's Medium (DMEM) supplemented with 10% fetal bovine serum (each from Gibco, Grand Island, NY). Nucleated cells were isolated with a density gradient [Ficoll/Histopaque (Pharmacia, Uppsala, Sweden)] and re-suspended in a complete culture medium supplemented with 1% penicillin–streptomycin (Gibco). Cells were incubated at 37 °C in 5% humidified CO2 for 12–14 days; media were changed every 2–3 days. When large colonies developed (80–90% confluence), cultures were washed twice with PBS (pH 7.4) and cells were trypsinized with 0.25% trypsin/1 mM EDTA solution for 5 min at 37 °C. After centrifugation, cell pellets were re-suspended with serumsupplemented medium and incubated in a 50-cm2 culture flask (Falcon, Cairo, Egypt). The resulting cultures were referred to as first-passage cultures [19].

F

2. Materials and methods

85 86

138

O

102

83 84

2.3. Preparation of BM-derived MSCs

P

100 101

MSCs. These MSCs are isolated from the adult bone marrow and expanded ex vivo in order to differentiate into several types of cells, such as osteocytes, chondrocytes, adipocytes, hematopoietic supporting stroma, and endothelial cells [15]. Therefore, these MSCs may be useful in tissue regeneration [16]. Moreover, MSCs have shown to modulate allogeneic immune cell responses from a pro-inflammatory toward an antiinflammatory state by affecting dendritic cells, T-lymphocytes, and natural killer (NK) cells [17]. MSCs also appear to regulate the immune function in inflamed tissues by affecting the formation and secretion of pro-inflammatory cytokines and chemokines. In fact, the in vivo exogenous administration of MSCs imparts an anti-inflammatory action and improves the health of tissues that had become inflamed in response to injury induction [18]. Although, MSCs have shown an anti-inflammatory effect, their cellular mechanisms to pursue such function should be examined. In the present study, we sought to examine the anti-inflammatory potential of MSC exogenously administered to mice that were suffering from DSS induced colitis. Assessment of therapeutic efficacy of MSCs was by measuring weight, stool scoring, histopathologic examination, and measuring the gene expression of inflammatory markers: IL-23, TNF-α, IFN-γ, and ICAM-1 in colonic tissues recovered from the mice.

E

81 82

A.G.A.R.A. Salam et al. / International Immunopharmacology xxx (2014) xxx–xxx

R O

2

Please cite this article as: Salam AGARA, et al, Potential therapeutic utility of mesenchymal stem cells in inflammatory bowel disease in mice, Int Immunopharmacol (2014), http://dx.doi.org/10.1016/j.intimp.2014.07.030

164 165 166 167 168 169 170 Q15 171 172 Q16 173 174 175

178 179 180 181 182 183 184

187 188 189 190 191 192 193 194 195

A.G.A.R.A. Salam et al. / International Immunopharmacology xxx (2014) xxx–xxx

199

2.6. Statistical analysis

200

Data are expressed as mean ± SE. Significant differences were determined using independent t-test and SPSS for Windows Version 15.0 Computer Software (SPSS, Chicago, IL, USA). Non-parametric analyses were used for evaluating all the colitis indices. Results were considered significant at P-values b 0.01. 3. Results

206

3.1. Development of DSS-induced colitis model in mice (pilot study)

207

3.1.1. Part A: determination of appropriate DSS concentration To determine the appropriate model of induction of IBD by DSS, different DSS concentrations (2% [21], 4% and 5% [20]) were administered for 1 week. Assessment of IBD degree was performed using stool scoring, weight loss, and histopathologic examination.

216 217 218 219 220 221 222 223 224 225 226 227 228

3.1.1.2. Average body weight. Fig. 2 shows that through one week of DSS administration, 2% DSS administered mice gain weight as an indication to normal bowel condition, 4% DSS administered mice gradually lose weight as an indication to deterioration of bowel condition, and 5% DSS administered mice lose weight with 100% mortality rate.

3.1.1.3. Histopathologic examination. Fig. 3(a, b, c) reveals that the administration of 2% DSS did not cause any harm to the intestinal mucosa, while, the administration of 4% and 5% DSS causes almost the same degree of inflammation. 3.1.2. Part B: determination of appropriate DSS/water cycle To determine the appropriate DSS/water cycle, three cycles were examined. Assessment of IBD degree was performed using stool scoring, weight loss, and histopathologic examination.

N C O

229 3.1.2.1. Stool score. Fig. 4 shows that the administration of 4% DSS for 230 Q17 7 days followed by 5 days of normal drinking water ensures a continuous

state of inflammation.

232 233

3.1.2.2. Average body weight. Fig. 5 shows that the administration of 4% DSS for 7 days followed by 5 days of normal drinking water leads to mild improvement in the body weight.

U

231

234

3.5

stool scores

3 2.5

Day 1

Day2

Day3

Day4

Day5

Day6

Day7

DSS 2%

20.3

22.1

25

26.3

27

29

31

DSS 5%

24.6

22

19.5

17.3

0

0

0

DSS 4%

25

24.3

23.8

23.1

22.6

21.9

20.8

O

235

3.1.3. Assessment of MSC potential therapeutic effect on IBD in tested groups Efficacy of MSCs in treating IBD in the tested groups was assessed by stool scoring, average body weight, histopathologic examination, and measuring the gene expression of the inflammatory markers.

239 240

R O

3.1.2.3. Histopathologic examination. As per previous observations, histopathologic examination was performed for three cycles (7 days 4% DSS + 5 days water) in mice. Fig. 6 reveals a continuous inflammation state even after 5 days of normal drinking water.

236 Q18 237 238

241 242 243

3.1.3.1. Assessment of stool scoring in the studied groups. Fig. 7 shows that 244 stool condition was significantly improved by the administration of 245 MSCs. 246 3.1.4. Average body weight Fig. 8 shows that the average body weight of the mice in the studied groups in the indicated cycle intervals and revealed a significant increase in the body weight of treated group.

247 248

3.1.5. Histopathological examination of tested groups' intestine The gross picture of control group intestine revealed that circumscribed round ulceration was observed in the inner mucosa associated with congestion and redness all over the wall and that there was no gross finding observed in the mucosa and outer surface all over the intestine wall. Moreover, focal desquamation was observed in the mucosal lining epithelial cells associated with a massive number of inflammatory cell infiltration mainly neutrophils, macrophages and lymphocytes with congestion of the blood vessels in the underlying lamina propria, and the muscularis showed that oedema with inflammatory cell infiltration was observed in both caecum and colon, (Fig. 9a) which shows histopathological confirmation of IBD. On the other hand, the gross picture of treated group intestine revealed that there was no gross finding observed in the mucosa and outer surface all over the intestine wall. Moreover, no histopathological alteration was observed in the mucosal layer with the lamina propria as well as in the submucosal and muscular layers, (Fig. 9b), which shows a histopathologically normal intestine.

251 252

3.1.6. Assessment of body weight in the studied groups As illustrated in Fig. 10, the average body weights of the treated groups are significantly higher (P b 0.01), compared with the control group. This significant weight elevation is obvious starting from the second DSS administration cycle on days 19, 24 and 31.

269

3.1.7. The inflammatory markers in control and treated groups Fig. 11 illustrates the relative gene expression of pro-inflammatory markers TNF-α, and IL-23, IFN-γ, and ICAM-1 were measured in both control and treated groups, showing that, the relative gene expression

274

249 250

253 254 Q19 255 256 Q20 257 258 259 260 261 262 263 264 265 Q21 266 267 268

2 1.5 1 0.5 0

Q1

Mice diceased 0

T

215

10 5

3.1.1.1. Stool scoring. Fig. 1 shows that through one week of administration; 2% DSS causes no effect on the stool, 4% DSS causes gradual deterioration in stool condition, and 5% DSS causes severe deterioration in stool condition with 100% mortality.

C

213 214

E

212

R

210 211

15

Fig. 2. Pilot study (part A): weight loss.

R

208 209

20

F

205

25

P

203 204

30

D

201 202

35

weight in grams

198

was used as an internal standard with Taqman GAPDH control reagents (PE Applied Biosystems, CA, USA). Levels of mRNA expression were expressed as percentages of controls.

E

196 197

3

Mice diceased Day 1

Day2

Day3

Day4

Day5

Day6

Day7

DSS 2%

0

0

0

0

0

0

0

DSS 5%

0

3

3

3

0

0

0

DSS 4%

0

1

2

2

3

3

3

Fig. 1. Pilot study (part A): stool scoring.

Please cite this article as: Salam AGARA, et al, Potential therapeutic utility of mesenchymal stem cells in inflammatory bowel disease in mice, Int Immunopharmacol (2014), http://dx.doi.org/10.1016/j.intimp.2014.07.030

270 271 272 273

275 276 277

4

A.G.A.R.A. Salam et al. / International Immunopharmacology xxx (2014) xxx–xxx

b

c

F

a

4. Discussion

281 282

N

C

O

R

R

E

C

T

The application of MSC therapy has opened a new horizon for the treatment of IBD, as most of the pharmacological approaches haven't 283 yet reached the desired efficacy and safety outcomes. MSCs are 284 multipotent cells found in various species, including mice, and are 285 often used for cellular transplantation therapy [22]. Most previous 286 studies have focused on these attractive regenerative properties 287 of MSCs [22,23], indicating that MSCs can promote regeneration 288 of wounded or injured tissues by a differentiation into vascular 289 smooth muscle cells, endothelial cells, pericytes, or epithelial cells 290 [24]. 291 Chemical induction of IBD by DSS was considered an efficient and 292 easily applied model, however, choosing the correct dose to induce 293 colitis is a variable [21]. In the present study, the 4% DSS model was 294 used as administration of 2% and 5% DSS did not give the desired 295 outcome. In the first part of the pilot study the 2% DSS failed to induce 296 inflammation, which is in contrast to Wirtz et al. [21], who showed 297 that the administration of 2% DSS for 7 days had produced acute colitis. 298 However, using 5% DSS showed 100% mortality in accordance to Wirtz 299 et al. [21]. Concerning the second part of the pilot study, our results 300 showed a cycle of 7 days of 4% DSS followed by 5 days of normal drink301 Q22 ing water which is optimum for the induction of chronic IBD, in contrast 302 to Wirtz et al. [21] who induced a chronic model of colitis by the admin303 istration of DSS for 7 days followed by 14 days of water.

P

280

Experiments done to investigate the therapeutic effect of MSCs in IBD are very few; the first attempt was done by Tanaka et al. [20] that induced DSS acute colitis and proved that the exogenous administration of MSCs ameliorates acute DSS induced colitis. Tanaka et al. [20] assessed the MSC therapeutic effect by measuring the RNA expression of inflammatory mediators such as TNF-α, IL-1β, COX-2, basic fibroblast growth factor (b-FGF), hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF), all of which significantly decreased in MSC treated mice. The second attempt was done by Liang et al. [25] in which human umbilical cord mesenchymal stem cells (hUC-MSCs) were used to treat acute trinitrobenzene sulfonic acid (TNBS) induced IBD. The inflammatory markers such as IL-17, IL-23, IFN-γ, and IL-6 were measured to assess the therapeutic efficacy of hUC-MSCs and were shown to be significantly decreased in the treated mice. Another attempt was done by Gonzalez et al. [26] in which acute colitis was induced by TNBS and adipose tissue derived MSCs were injected to the mice. Gonzalez et al. assessed the therapeutic effect of MSCs by measuring inflammatory cytokines such as TNF-α, IFN-γ, IL-6, and IL-1β, and chemokines such as macrophage inhibitory protein II which were significantly decreased in treated mice. In the current work, the MSCs were injected once after the first cycle of chronic DSS-induced colitis. We assessed the gene expression of pro-inflammatory cytokine levels (TNF-α, IFN-γ, IL-23 and ICAM-1) in an attempt to investigate their association with MSC therapy in IBD. The results of this work indicated that the level of TNF-α gene expression was significantly decreased after MSC administration. This result is supported by recent studies that have shown that MSCs can also modulate the systemic immune systems [17,18]. Ortiz et al. [18]

D

of the four inflammatory markers is significantly decreased (P b 0.01) in the treated group compared to the control group.

E

279

278

R O

O

Fig. 3. (a) shows that that through one week of DSS administration, 2% DSS administration causes a colon of mice with intact histological structure of mucosal layer (mu), lamina propria (Lp), submucosa (s), and muscularis (m) mice. (b) revealed that 4% DSS administration causes caecum with the massive number of inflammatory cell infiltration in the lamina propria (f) with congestion in blood vessels (v) and desquamation of the lining mucosal epithelium (m). (c) 5% DSS administration causes acute colitis extended to the muscular layer revealing caecum of the mice with oedema and inflammatory cell infiltration in the muscularis (m).

U

3.5 3 2.5 2 1.5 1 0.5 0

stool score in first day of the cycle

stool score aer 7 days 4% DSS

stool score in last day of the cycle

cycle 1: 7days DSS+ 14 days water

0

3

0

cycle 2: 7 days DSS+ 10 days water

0

3

0

cycle 3: 7 days DSS+ 5 days water

0

3

2

stool score

Fig. 4. Pilot study (part B): stool score.

Please cite this article as: Salam AGARA, et al, Potential therapeutic utility of mesenchymal stem cells in inflammatory bowel disease in mice, Int Immunopharmacol (2014), http://dx.doi.org/10.1016/j.intimp.2014.07.030

304 305 306 307 308 309 310 311 Q23 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331

A.G.A.R.A. Salam et al. / International Immunopharmacology xxx (2014) xxx–xxx

studied groups: stool score 3.5

25 20 15 10

0

349 350 351 352

Day 19

Day 24

Day 31

F

O

R O

et al. [26] provided further confirmation to our findings as in their experiment AMSC injected mice showed reduced levels of TNF-α in comparison to untreated mice. The results revealed that the expression of IFN-γ was significantly decreased in MSC treated mice. This finding was confirmed by Gonzalez et al. [26] who showed reduced levels of IFN-γ in AMSC injected mice in comparison to untreated mice and related the effect of AMSCs to their ability to suppress Th1 responses and induce T regulatory cells. Liang et al. [25] also confirmed our results when they found that hU-MSC significantly decreased the level of IFN-γ. Recent findings reported that IFN-γ has a dual role in the immunosuppressive effect of MSCs; the first is stated before as MSCs suppress Th1 and induce T regulatory cells. The second effect is that MSCs inhibited the proliferation of B cells only in the presence of IFN-γ, and the capacity of IFN-γ to induce the suppressive activity of MSCs is not dependent on indoleamine 2,3 deoxygenase (IDO) but is dependent on cell to cell contact mediated by the interaction between programmed death 1 (PD-1) and PD ligand 1 (PDL-1) [28]. The most possible mechanism supporting this theory is that the expression of PD-1 and PDL-1 from both MSCs and B cells displays a significant increase in the presence of IFN-γ. These results suggest that PD-1/PDL-1 interaction between MSCs and B lymphocytes inhibits the signal transduction pathways triggered by productive B cell stimulation by antigen and affects B cell expansion. Another mechanism was supported by Krampera et al. [29] who supposed that MSCs inhibit the proliferation of B cells only in the presence of IFN-γ which implies that IFN-γ causes MSCs to produce IDO, which catalyzes the conversion of tryptophan to kynurenine which in turn suppressed the proliferation of B cells. Spaggiari et al. [30] highlighted another role of IFN-γ in MSC

P

R

E

C

D

E

T

indicated that MSCs have an anti-inflammatory property and reduce inflammation and lung injury induced by bleomycin. In addition, Aggarwal and Pittenger [17] showed that human MSCs can modulate anti-inflammatory effects in co-cultures with purified subpopulations of immune-associated cells, such as dendritic cells (DCs), naive and effector T cells (T helper 1, Th1 and T helper 2, Th2), regulatory T cells (Tregs), and natural killer (NK) cells. MSCs can down-regulate inappropriate inflammatory responses by inhibiting pro-inflammatory Th1, DC1, and NK signaling and by promoting anti-inflammatory Th2 and/or suppressive Treg signaling. MSCs may thus orchestrate a shift from Th1 (pro-inflammatory) toward Th2 (anti-inflammatory) [17]. Accordingly, it is possible that MSCs could prove efficacy in the treatment of Th1-mediated inflammatory diseases. As for the pathogenesis of CD, a systemic and/or local inflammatory bowel disease, Th1 cytokines are known to have a more important role in pathogenesis than Th2 cytokines in local inflammatory tissues [27]. Recently, infliximab (anti-TNF-α antibody) as well as conventional anti-inflammatory agents are often used clinically to treat CD by attenuating inflammation. As for experimental colitis, chronic DSS-induced mucosal injury has the cytokine profile of Th1 responses [13]. Furthermore, Tanaka et al. [20] have reported that the exogenous administration of MSCs significantly decreased the level of TNF-α, IL-1β, and COX-2. Moreover, Gonzalez

U

353

Day 12

0

Fig. 7. Studied groups: stool score.

Studied groups: average body weight 40 35 30 Average Body Weight

347 348

Day 7

0

treated

R

345 346

0

control

N C O

344

1

Day 1

Fig. 5. Pilot study (part b): Average body weight.

342 343

2

1 0

cycle 3: 7 days DSS+ 5 days water

340 341

2

1.5 0.5

cycle 2: 7 days DSS+ 10 days water

338 339

3

2

5

cycle 1: 7days DSS+ 14 days water

336 337

3

2.5

Average Average Average weight in weight aer weight in last first day of 7 days 4% day of the the cycle DSS cycle

334 335

3

3 stool score

weight in grams

30

332 333

5

25 20 15 10 5 0 Day 1

Day 7

Day 12

Day 19 control

Day 24

Day 31

Treated Q2

Fig. 6. The figure shows a colon of mice with desquamation of the lining of the epithelium (m) with inflammatory cell infiltration in the underlying lamina propria (f).

Fig. 8. Studied groups: average body weight.

Please cite this article as: Salam AGARA, et al, Potential therapeutic utility of mesenchymal stem cells in inflammatory bowel disease in mice, Int Immunopharmacol (2014), http://dx.doi.org/10.1016/j.intimp.2014.07.030

354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 Q24 370 371 372 373 374 375 376 377 378 379 380 Q25 381

6

A.G.A.R.A. Salam et al. / International Immunopharmacology xxx (2014) xxx–xxx

403 404 405

R O

P

T

Studied groups: average body weight 40 35 30 25 20

Tested groups: gene expression of inflammatory markers 120 99.23186875 100 Relative Quanitification

401 402

C

399 400

E

397 398

R

395 396

R

393 394

O

391 392

Regarding the decreased the gene expression of ICAM-1 in MSC treated mice, this is the first attempt to prove that MSC immunosuppressive effect includes the suppression of ICAM-1 expression. In contrast to our findings, Ren et al. [35] showed that MSCs do express ICAM-1 in order to perform the immunosuppressive effect. Generally, adhesion molecules play a dual role in the immunosuppressive effect of MSCs; first, Chamberlain et al. [36] stated that inflamed endothelial tissue releases proinflammatory cytokines, chemokines, and adhesion molecules in order to attract macrophages, lymphocytes, and other inflammatory cells. Second, the release of pro-inflammatory cytokines, chemokines, and adhesion molecule plays a major role in the homing of MSCs to the site of injury. Interestingly, Segers et al. [37] stated that ICAM-1 and VCAM-1 are considered markers for stem cells; the observation that confirms that MSCs can express adhesion molecules. According to Ren et al. [35] T cells secrete several inflammatory cytokines, including IFN-γ, TNF-α, and IL-1 that leads to striking upregulation in the expression of two adhesion molecules: ICAM-1 and VCAM-1 that are responsible for the increased adhesion between MSCs and T cells. Importantly, blocking of the function of adhesion molecules significantly reversed the immunosuppressive effect of MSCs in vitro and in vivo. Summing up, ICAM-1 plays a dual role as it facilitates the homing of MSCs to the site and allows the cell to cell contact, the cornerstone MSC immunosuppressive effect. To correlate these observations with our results we can conclude that in the chronic model of IBD the administration of MSCs will significantly increase the expression of ICAM-1 in the first few days of injection. However, after 26 days of administration and the severity of inflammation has decreased and most of the ICAM-1 has been consumed during the

C

389 390

N

387 388

U

385 386

cell-mediated lyses upon incubation of MSCs with IFN-γ. Moreover, in this work, the gene expression of IL-23 was significantly decreased in MSC treated mice. Our result is supported by the study of Liang et al. [25] in which the level of IL-23 was significantly lower in the colon tissues of hUC-MSC treated mice. IL-23 performs its antiinflammatory function mainly by induction of Th17 that releases IL-17 [31]. Although the precise mechanism by which IL-23 maintains Th17 responses in vivo is still not well understood, recent studies have shown that Th17 cell lineage commitment is driven by TGF-β in the presence of pro-inflammatory cytokines, whereas IL-23 seems to expand or maintain Th17 cell populations [32]. IL-23 is a type 1 heterodimeric IL-12 like cytokine, comprising of a 19-kD 4-fold helical core α subunit (IL-23p19), and a disulfide linked to an additional 40-kD distinct β subunit (IL-12p40). IL-23 and IL-12 share a common subunit in their receptor complex due to the common p40 subunit [33]. Gonzalez et al. [26] reported that colons of AMSC treated mice contained a reduced level of IL-12 in comparison with untreated IBD induced colitis mice. Moreover, MSCs inhibit the differentiation of monocytes into dendritic cells by inhibiting the response of the former to maturation signals, reducing the expression of co-stimulatory molecules and hampering the ability of the former to stimulate naïve T cell proliferation and IL-12 secretion [34]. Furthermore, the level of IL-17 was significantly decreased in the colon tissue of hU-MSC treated mice [25].

Average Body Weight

383 384

D

382 Q26 immunosuppressive activity which is the decreased susceptibility to NK

O

Fig. 9. (a) reveals histopathologic findings in the control group; this figure shows the inflammatory cell infiltration (f), oedema (o), and dilated blood vessels (v) in the submucosal layer. (H&E ×80). (b) reveals histopathological findings in the treated group. this figure shows a normal histological structure of the mucosa (m) and muscularis (ml) with serosa (s) (H&E ×64).

E

Q3

b

F

a

15 10

82.30847612

80 60 40

29.54936333

25.88068514

5 20 0.633109875

0 Day 1

Day 7

Day 12

Day 19

Day 24

control Treated Fig. 10. Studied groups: average body weight.

Day 31

0.065108

0.0859065

3.22869375

0 ICAM-1

IFN-γ

IL-23

TNF-α

Control Treated Fig. 11. Tested groups: gene expression of inflammatory markers.

Please cite this article as: Salam AGARA, et al, Potential therapeutic utility of mesenchymal stem cells in inflammatory bowel disease in mice, Int Immunopharmacol (2014), http://dx.doi.org/10.1016/j.intimp.2014.07.030

406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433

A.G.A.R.A. Salam et al. / International Immunopharmacology xxx (2014) xxx–xxx

C

453 454

E

451 452

R

F

449 450

R

447 448

N C O

445 446

U

443 444

O

[1] Podolsky DK. Inflammatory bowel disease. N Engl J Med 2002;347:417–29. [2] Shanahan F. Crohn's disease. Lancet 2002;359:62–9. [3] Rioux JD, Xavier RJ, Taylor KD, Silverberg MS, Goyette P, Huett A, et al. Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis. Nat Genet 2007;39:596–604. [4] Strober W, Fuss I, Mannon P. The fundamental basis of inflammatory bowel disease. J Clin Invest 2007;117:514–21. [5] Hue S, Ahern P, Buonocore S, Kullberg MC, Cua DJ, McKenzie BS, et al. Interleukin-23 drives innate and T cell-mediated intestinal inflammation. J Exp Med 2006;203: 2473–83. [6] Triantafillidis JK, Merikas E, Georgopoulos F. Current and emerging drugs for the treatment of inflammatory bowel disease. Drug Des Devel Ther 2011;5:185–210. [7] Ni J, Chen SF, Hollander D. Effects of dextran sulphate sodium on intestinal epithelial cells and intestinal lymphocytes. Gut 1996;39:234–41. [8] Leung FW, Heng MC, Allen S, Seno K, Leung JW, Heng MK. Involvement of luminal bacteria, heat shock protein 60, macrophages and gammadelta T cells in dextran sulfate sodium-induced colitis in rats. Dig Dis Sci 2000;45:1472–9. [9] Shintani N, Nakajima T, Okamoto T, Kondo T, Nakamura N, Mayumi T. Involvement of CD4+ T cells in the development of dextran sulfate sodium-induced experimental colitis and suppressive effect of IgG on their action. Gen Pharmacol 1998;31:477–81. [10] Yan Y, Kolachala V, Dalmasso G, Nguyen H, Laroui H, Sitaraman SV, et al. Temporal and spatial analysis of clinical and molecular parameters in dextran sodium sulfate induced colitis. PLoS One 2009;4:e6073. [11] Dieleman LA, Ridwan BU, Tennyson GS, Beagley KW, Bucy RP, Elson CO. Dextran sulfate sodium-induced colitis occurs in severe combined immunodeficient mice. Gastroenterology 1994;107:1643–52. [12] Dieleman LA, Palmen MJ, Akol H, Bloemena E, Pena AS, Meuwissen SG, et al. Chronic experimental colitis induced by dextran sulphate sodium (DSS) is characterized by Th1 and Th2 cytokines. Clin Exp Immunol 1998;114:385–91. [13] Egger B, Bajaj-Elliott M, MacDonald TT, Inglin R, Eysselein VE, Buchler MW. Characterisation of acute murine dextran sodium sulphate colitis: cytokine profile and dose dependency. Digestion 2000;62:240–8. [14] Sonwalkar SA, James RM, Ahmad T, Zhang L, Verbeke CS, Barnard DL, et al. Fulminant Crohn's colitis after allogeneic stem cell transplantation. Gut 2003;52:1518–21. [15] Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002;418:41–9.

441 442

R O

457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 560

440

P

References

438 439

[16] Matsumoto R, Omura T, Yoshiyama M, Hayashi T, Inamoto S, Koh KR, et al. Vascular endothelial growth factor-expressing mesenchymal stem cell transplantation for the treatment of acute myocardial infarction. Arterioscler Thromb Vasc Biol 2005;25: 1168–73. [17] Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005;105:1815–22. [18] Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N, et al. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A 2003;100:8407–11. [19] Alhadlaq A, Mao JJ. Mesenchymal stem cells: isolation and therapeutics. Stem Cells Dev 2004;13:436–48. [20] Tanaka F, Tominaga K, Ochi M, Tanigawa T, Watanabe T, Fujiwara Y, et al. Exogenous administration of mesenchymal stem cells ameliorates dextran sulfate sodiuminduced colitis via anti-inflammatory action in damaged tissue in rats. Life Sci 2008;83:771–9. [21] Wirtz S, Neufert C, Weigmann B, Neurath MF. Chemically induced mouse models of intestinal inflammation. Nat Protoc 2007;2:541–6. [22] Khalil PN, Weiler V, Nelson PJ, Khalil MN, Moosmann S, Mutschler WE, et al. Nonmyeloablative stem cell therapy enhances microcirculation and tissue regeneration in murine inflammatory bowel disease. Gastroenterology 2007;132:944–54. [23] Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol 2008;8:726–36. [24] Komori M, Tsuji S, Tsujii M, Murata H, Iijima H, Yasumaru M, et al. Involvement of bone marrow-derived cells in healing of experimental colitis in rats. Wound repair and regeneration: official publication of the Wound Healing Society [and] the European Tissue Repair. Society 2005;13:109–18. [25] Liang L, Dong C, Chen X, Fang Z, Xu J, Liu M, et al. Human umbilical cord mesenchymal stem cells ameliorate mice trinitrobenzene sulfonic acid (TNBS)-induced colitis. Cell Transplant 2011;20:1395–408. [26] Gonzalez MA, Gonzalez-Rey E, Rico L, Buscher D, Delgado M. Adipose-derived mesenchymal stem cells alleviate experimental colitis by inhibiting inflammatory and autoimmune responses. Gastroenterology 2009;136:978–89. [27] Monteleone I, Vavassori P, Biancone L, Monteleone G, Pallone F. Immunoregulation in the gut: success and failures in human disease. Gut 2002;50(Suppl. 3):III60–4. [28] Schena F, Gambini C, Gregorio A, Mosconi M, Reverberi D, Gattorno M, et al. Interferongamma-dependent inhibition of B cell activation by bone marrow-derived mesenchymal stem cells in a murine model of systemic lupus erythematosus. Arthritis Rheum 2010;62:2776–86. [29] Krampera M, Cosmi L, Angeli R, Pasini A, Liotta F, Andreini A, et al. Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells 2006;24:386–98. [30] Spaggiari GM, Capobianco A, Becchetti S, Mingari MC, Moretta L. Mesenchymal stem cell-natural killer cell interactions: evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-cell proliferation. Blood 2006;107:1484–90. [31] Kastelein RA, Hunter CA, Cua DJ. Discovery and biology of IL-23 and IL-27: related but functionally distinct regulators of inflammation. Annu Rev Immunol 2007;25: 221–42. [32] Bettelli E, Oukka M, Kuchroo VK. T(H)-17 cells in the circle of immunity and autoimmunity. Nat Immunol 2007;8:345–50. [33] Beadling C, Slifka MK. Regulation of innate and adaptive immune responses by the related cytokines IL-12, IL-23, and IL-27. Arch Immunol Ther Exp (Warsz) 2006; 54:15–24. [34] Nauta AJ, Kruisselbrink AB, Lurvink E, Willemze R, Fibbe WE. Mesenchymal stem cells inhibit generation and function of both CD34+-derived and monocytederived dendritic cells. J Immunol 2006;177:2080–7. [35] Ren G, Zhao X, Zhang L, Zhang J, L'Huillier A, Ling W, et al. Inflammatory cytokineinduced intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in mesenchymal stem cells are critical for immunosuppression. J Immunol 2010; 184:2321–8. [36] Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells 2007;25:2739–49. [37] Segers VF, Van Riet I, Andries LJ, Lemmens K, Demolder MJ, De Becker AJ, et al. Mesenchymal stem cell adhesion to cardiac microvascular endothelium: activators and mechanisms. Am J Physiol Heart Circ Physiol 2006;290:H1370–7.

D

456

436 437

T

455

anti-inflammatory process of MSCs, resulting in a significant decrease of ICAM-1 gene expression at the end of experiment. In conclusion, induction of chronic IBD in mice showed an increase in the gene expression of proinflammatory markers such as TNF-α, IFN-γ, IL-23, and ICAM-1. The control group (IBD-induced mice injected by sterile PBS) showed no decrease in the gene expression level of proinflammatory markers. Upon treating IBD induced mice with bone marrow derived MSCs, the gene expression of proinflammatory marker was significantly decreased. These conclusions highlighted many of the pathophysiological cascades of inflammatory bowel disease. The inflammation induced by DSS was significantly associated with the production of ICAM-1 that leads to migration and infiltration of macrophages and Th1, producing thereby TNF-α, IFN-γ and IL-23. Interestingly, ICAM-1 has a dual role in stem cell immunomodulatory effect; the first role is that high levels of ICAM-1 in inflammation site lead to the migration of MSCs because stem cells have surface receptors similar to those on the surface of macrophages, T cells, and B cells. The second role is that the high levels of TNF-α and IFN-γ in the inflamed tissue provoke MSCs to produce ICAM-1 that attracts inflammatory cells to adhere to the stem cell surface to exert its immunomodulatory effect. Finally, we can conclude that bone marrow derived MSCs can be used as an effective treatment of IBD.

E

434 435

7

Please cite this article as: Salam AGARA, et al, Potential therapeutic utility of mesenchymal stem cells in inflammatory bowel disease in mice, Int Immunopharmacol (2014), http://dx.doi.org/10.1016/j.intimp.2014.07.030

494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559

Potential therapeutic utility of mesenchymal stem cells in inflammatory bowel disease in mice.

Mesenchymal stem cells (MSCs) were found to provide an effective therapeutic role in inflammatory diseases by modulating inflammatory responses and ti...
2MB Sizes 0 Downloads 8 Views