Pathogens and Disease Advance Access published April 15, 2015

1

Host gene expression for Mycobacterium avium subsp. paratuberculosis

2

infection in human THP-1 macrophages

3

Running title: Potential host-response for MAP infection in THP-1 cell

4 5 6

Min-Kyoung Shin1,2, Seung Won Shin1, Myunghwan Jung1, Hongtae Park1, Hyun-Eui Park1,

7

& Han Sang Yoo1, 3

8 9 10 11 12 13 14

1

Department of Infectious Diseases, College of Veterinary Medicine, Seoul National

University, Seoul, 151-742 Korea 2

Dairy and Swine Research and Development Centre, Agriculture and Agri-Food Canada,

Sherbrooke, QC J1M 1Z3, Canada; 3

Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang,

232-916, Korea

15 16

Correspondence: Han Sang Yoo

17

Department of Infectious Diseases, College of Veterinary Medicine, Seoul National

18

University, Seoul, 151-742 Korea

19

Phone: +82-2-880-1263

20

Fax: +82-2-874-2738

21

E-mail: [email protected]

22

1

1 2

Abstract

3

Mycobacterium avium subsp. paratuberculosis (MAP) is the causative agent of Johne’s

4

disease, which causes considerable economic loss in the dairy industry and has a possible

5

relationship with Crohn’s disease (CD) in humans. As MAP has been detected in retail

6

pasteurized milk samples, its transmission via milk is of concern. Despite its possible role in

7

the etiology of CD, there have been few studies examining the interactions between MAP and

8

human cells. In the current study, we applied Ingenuity Pathway Analysis to the transcription

9

profiles generated from a murine model with MAP infection as part of a previously

10

conducted study. Twenty one genes were selected as potential host immune responses,

11

compared to the transcriptional profiles in naturally MAP-infected cattle, and validated in

12

MAP-infected human monocyte-derived macrophage THP-1 cells. Of these, the potential

13

host responses including up-regulation of genes related to immune response (CD14, S100A8,

14

S100A9, LTF, HP and CHCIL3), up-regulation of Th 1-polarizing factor (CCL4, CCL5,

15

CXCL9 and CXCL10), down-regulation of genes related to metabolism (ELANE, IGF1,

16

TCF7L2, and MPO), and no significant response of other genes (GADD45a, GPNMB,

17

HMOX1, IFNG, and NQO1) in THP-1 cell infected with MAP.

18

Keywords: Mycobacterium avium subsp. paratuberculosis, host responses, THP-1,

19

2

1 2

Introduction

3

Mycobacterium avium subsp. paratuberculosis (MAP) causes Johne’s disease (JD), a

4

chronic granulomatous enteropathy of ruminants such as cattle, sheep, deer, bison and elk

5

(Motiwala et al., 2006). Concerns have been raised about JD over economic losses in the

6

dairy industry as well as the possible relationship with a chronic inflammatory bowel disease

7

(IBD) in humans called Crohn’s disease (CD) (Thayer et al., 1984, Naser et al., 2004, Yoo &

8

Shin, 2012). It has been suggested that human IBD may be caused by an autoimmune

9

dysregulation and imbalance of T cells or host responsiveness against a microbial trigger

10

(Mizoguchi et al., 2003, Singh et al., 2007). In addition, many studies report that MAP or

11

MAP-specific serum antibodies are detected in the intestine, blood and breast milk of Crohn’s

12

disease patients (Naser et al., 2000, Naser et al., 2000, Schwartz et al., 2000, Naser et al.,

13

2004, Sechi et al., 2005). MAP DNA has been detected in retail pasteurized milk samples,

14

even commercially pasteurized milk in the United Kingdom. As transmission could occur

15

through milk and other dairy products, further research into the mechanisms underlying MAP

16

pathogenesis in human is needed (Millar et al., 1996, Ellingson et al., 2005). Despite its

17

possible role in the etiology of CD, there have been few studies examining the interactions

18

between MAP and human cells. Different gene expression patterns or cytokine responses

19

(TNF-α, IL-1β, and IL-10) were observed in human monocyte-derived macrophage THP-1

20

cells by different MAP strains (Motiwala et al., 2006, Borrmann et al., 2011).

21

In the current study, we applied a microarray-based approach to discover potential immune

22

response markers in a model of murine MAP infection and naturally MAP-infected cattle

23

followed by validation of the genes in human cells. In our previous study, the host responses

24

to MAP infection in mice and cattle were characterized by transcriptional analysis. 3

1

Significantly regulated genes were filtered and selected via the Ingenuity Pathways Analysis

2

(IPA) and evaluated in vitro using MAP-infected THP-1 cells.

3 4

Materials and Methods

5

Selection of potential host response genes using transcriptional profiles in MAP-

6

infected mice and naturally MAP-infected cattle

7

We previously submitted a dataset of MAP infection to the GEO database (GSE62836) for

8

murine study (submmitted to another journal) and GEO database (GSE62835) for bovine

9

study (Shin et al., 2015). For murine study, briefly, five-week-old C57BL/6 female mice

10

were inoculated intraperitoneally with 300 μL PBS (control group) or a suspension of MAP

11

ATCC 19698 (3 × 107 cells/mouse, MAP-infected group). Spleen samples were taken from

12

five mice in each group, sacrificed at 3 and 6 weeks post infection (p.i.). Microarray analysis

13

was performed using total RNA from harvested spleen cells and the data were used for

14

identification of differentially expressed genes in MAP-infected mice compared to control

15

mice. Differentially expressed genes were filtered for a log2-fold change >2 with p < 0.05 by

16

Biomarker Filter and then analyzed by Biomarker Discovery both of IPA (Ingenuity Systems

17

Inc., Redwood, CA). The genes were further clarified in the IPA biomarker filter module

18

based on the following criteria: fluid – 〈〈all〉〉, disease – 〈〈Infectious Disease, Inflammatory

4

1

response, Immunological disease〉〉, and species – 〈〈mouse〉〉. Genes selected based on the

2

above criteria were compared with bovine transcriptional profiles and verified in THP-1 cells

3

following infection with MAP using quantitative real-time RT PCR.

4 5

Bacterial strain, cell line culture, and infection

6

MAP ATCC 19698 was cultured as previously described (Cha et al., 2013). For infection of

7

cells, MAP ATCC 19698 was suspended in phosphate-buffered saline (PBS) and used at a

8

dilution of 1 × 109 cells/mL.

9

The THP-1 human leukemic monocyte cell line was obtained from KCLB (Korea Cell Line

10

Bank, Seoul, Korea). THP-1 cells were cultured in RPMI 1640, supplemented with 10% heat-

11

inactivated fetal bovine serum (FBS), and Antibiotic-Antimycotic (all from Gibco Invitrogen,

12

Karlsruhe, Germany) agent at 37°C in humidified air with 5% CO2. THP-1 cells were

13

differentiated into macrophages by stimulation with phorbol-12-myristate-13-acetate (PMA;

14

Sigma Co., St. Louis, MO, USA) (50ng/ml) for 72h, washed with FBS-free RPMI 1640

15

medium and incubated with 5% FBS-RPMI 1640 medium without antibiotics for 24h before

16

the experiments. Differentiated THP-1 macrophage cells (1 × 106 cells/ml) were inoculated

17

with MAP 19698 at a multiplicity of infection (MOI) of 10:1 and incubated for 0, 6, 24, and

18

48h. Total RNA was extracted from cells collected at 0, 6, 24, and 48h post infection (p.i.).

19 5

1

Quantitative real-time PCR

2

The total RNA was isolated with an RNeasy Mini Kit (Qiagen, Valencia, USA) according to

3

the manufacturer's instructions. Two micrograms of total RNA from each sample were

4

reverse-transcribed into cDNA using the Quantitect® Reverse Transcription kit (Qiagen).

5

Real time qRT-PCR reactions were performed with 2 μl of cDNA using the Rotor-Gene

6

SYBR Green PCR kit (Qiagen) and Rotor-Gene Q real time PCR cycler (Qiagen). Cycling

7

parameters were as follows: 95°C for 5 minutes for 1 cycle, 95°C for 20 seconds, and then

8

60°C for 45 seconds for 40 cycles. Primers used in real-time PCR are shown in Table 1. The

9

gene expression level was normalized by the 2-ΔΔCt method using β-actin as an internal

10

control gene. The relative expression level was compared to the zero-time control to

11

determine the changes in expression-fold of each gene.

12 13

Statistical analysis

14

The data were expressed as mean ± standard error of the mean (SEM), and statistical

15

significance was analyzed by student’s t-test using IBM Statistical Package for Social

16

Sciences software (SPSS, version 21, SPSS Inc., Chicago, IL, USA). Differences were

17

considered significant if a value of p ≤ 0.05 was obtained.

18

19

Results

20

Host gene expression in MAP-infection mice and naturally MAP-infected cattle

21

In our previous study, C57BL/6 mice infected with MAP showed most severe

22

immunopathologic changes at 3 and 6 weeks p.i. by transcriptional profiling and 6

1

histopathological analysis. As the first step of data mining process, the “up-regulated” genes

2

were selected using the cut-off value (log2-fold change > 2 and p < 0.05). In the second, the

3

genes were imported to the Biomarker Discovery module of IPA and filtered by the criteria,

4

like: excreted in all biological fluids, related to infectious disease, inflammatory response, or

5

immunological disease. The twenty one eligible markers selected by IPA were related to

6

immune system processes (IFNG, CCL4, CCL5, CD14, CXCL9, CXCL10, HP, S100A8,

7

S100A9, MPO), metabolic processes (ELANE, CD68, CHI3L1, HP, S100A8, S100A9, IGF1,

8

TCF7L2, MPO, LTF, HMOX1), localization (CD68, IGF1, LTF, TFRC), and others

9

(GADD45, GPNMB, NQO1) as classified by GO biological processes (Table 1). These genes

10

were compared with the transcriptional profiles in naturally MAP-infected cattle (Table 1).

11

Naturally MAP-infected cattle were divided into three groups as follows: Test1, fecal PCR

12

positive; Test2, MAP-ELISA positive; and Test3, fecal PCR positive and MAP-ELISA

13

positive as previously described (Shin et al., 2015). The selected genes showed various

14

expression according to the groups in cattle, while all of the genes were up-regulated in

15

MAP-infected mice. The selected genes were divided into three groups based on comparison

16

of the results of mice and cattle. Group A was occupied by up-regulated genes in cows with

17

MAP-specific antibodies (Test2 and Test3). Group B was occupied by down-regulated genes

18

in cows as fecal shedder (Test1). The genes, which were up-regulated in MAP-infected mice,

19

but no significant response in MAP-affected cows, were Group C.

20 21

Validation of biomarker candidates in THP-1 by qPCR

22

The twenty one selected candidates were evaluated in human THP-1 cells at various time

23

points following MAP infection. No significant change in expression was observed in 7 of

24

these genes (CD68, GADD45, GPNMB, HMOX1, IFNG, NQO1 and TCF7L2) at any time 7

1

point relative to the uninfected control. The expression of CCL4 (3.5-fold up-regulation),

2

CCL5 (2.4-fod up-regulation), CXCL9 (3.5-fold up-regulation), and TFRC (2.1-fold up-

3

regulation) genes were increased at 6h p.i. but not at later time points and the expression of

4

CD14 (4.1- and 4.8-fold up-regulation at 24h and 48h p.i.), CXCL10 (1.8- and 3.4-fold up-

5

regulation at 24h and 48h p.i.), LTF (1.7- and 1.4-fold up-regulation at 24h and 48h p.i.),

6

S100A8 (6.2- and 6.7-fold up-regulation at 24h and 48h p.i.), and S100A9 (2.7- and 3.9-fold

7

up-regulation at 24h and 48h p.i.) genes were increased at 24h and 48h p.i. (Figure 1).

8

CHI3L1 (2.6-fold up-regulation), and HP (4.3-fold up-regulation) genes showed increased

9

expression only at 48h p.i. after MAP infection (Figure 1), while ELANE (1.7- and 1.8-fold

10

down-regulation at 24h and 48h p.i.), IGF1 (2.3- and 1.5-fold down-regulation at 24h and

11

48h p.i.), and MPO (2.1- and 1.8-fold down-regulation at 6h and 48h p.i.) genes showed

12

down-regulation. Therefore, among the selected genes from the transcriptomes of MAP-

13

infected mice, the expression of fourteen of these genes were also significantly up- or down-

14

regulated in human THP-1 cells following MAP infection.

15 16

Discussion

17

Although the oral route is the natural infection route of MAP in cattle, the intraperitoneal

18

(IP) injection has been frequently utilized in MAP infection of mice and our previous murine

19

study also was challenged MAP intraperitoneally, because earlier studies demonstrated that

20

low infectivity rates are the main problem of the oral administration of MAP in mouse model

21

(Mutwiri et al., 1992; Veazey et al., 1995). In the previous study, we analyzed transcriptional

22

profiles induced in the murine spleen, wherein the innate and adaptive immune systems

23

combined, following MAP infection. The expression patterns of twenty one candidate genes

24

were analyzed at various time points post infection. CD68 and CD14, both macrophage 8

1

markers, showed different expression patterns. CD68 was not significantly altered by MAP

2

infection in THP-1 cells. However, in these experiments PMA-stimulated THP-1 macrophage

3

cells were used and CD68 was already expressed in these cells prior to infection which may

4

explain why no further change was observed. The expression of CD14 increased at 24h and

5

48h post infection. A role for CD14 has been reported in the regulation of

6

monocyte/macrophage apoptosis so up-regulation of CD14 may have an effect on levels of

7

apoptosis during infection (McClure et al., 1987).

8

Increased expression of S100A8 and S100A9 (also termed myeloid-related proteins (MRP)8

9

and MRP14) was observed, consistent with data demonstrating release of these proteins by

10

activated phagocytes during mycobacterial infection both in vitro and in vivo (Pechkovsky et

11

al., 2000, Frosch et al., 2004). Cows with MAP-specific antibodies showed up-regulation of

12

MRP8 and MRP14 (Shin et al., 2015). They form stable complexes (MRP8/14) and are found

13

in a variety of inflammatory conditions, including rheumatoid arthritis, allograft rejections,

14

and inflammatory bowel and lung diseases (Kerkhoff et al., 1998). Therefore, their potential

15

role in inflammatory disorders including CD has already been highlighted (Lugering et al.,

16

1995, Foell et al., 2004). LTF (lactoferrin) gene was also slightly up-regulated in THP-1 cell

17

infected with MAP. Serum lactoferrin was strongly and inversely associated with increasing

18

bacterial index of M. leprae in leprosy patients (Muruganand et al., 2004). Of group A,

19

TCF7L2 gene was down-regulated at 6 h p.i., but there was not statistical significant

20

responses. TCF7L2 was suggested to modulate hepatic glucose metabolism in mammals, thus

21

contributing to the development of type 2 diabetes (Oh et al., 2012).

22

Although expression of th1 type chemokines and IFNG was not observed in naturally MAP-

23

infected cows, exception of down-regulation of CCL4 and CCL5 in Test1 of cows, th1 type

24

chemokines were increased at 6 h p.i. in THP-1 cell infected with MAP, especially 9

1

expression of CXCL10 was gradually increased in course of time. Th1 type chemokines and

2

chemokine receptors (CCL5 and CXCR3) show increased infiltration in granulomas of CD

3

patients (Oki et al., 2005), and a role for CCL5 and related chemokine receptors was

4

suggested in the pathogenesis of JD in THP-1 cells stimulated with MAP isolates (Motiwala

5

et al., 2006). Our results which indicate up-regulation of Th1 type chemokines (CCL4, CCL5,

6

CXCL9, and CXCL10) in MAP-infected THP-1 cells are consistent with previous data and

7

suggest a relationship between Th1 type chemokines and their receptors and the host

8

response to MAP infection. Interferon-inducible protein 10 (IP-10, CXCL10) has been

9

reported to be up-regulated in some Th1-mediated inflammatory disorders in humans

10

including type 1 diabetes mellitus (Cossu et al., 2013), rheumatoid arthritis (Patel et al.,

11

2001), multiple sclerosis (Balashov et al., 1999), sarcoid granulomatous disease (Beard et al.,

12

1999) and IBD (Zwick et al., 2002). CXCL10 has been described as an alternative or adjunct

13

biomarker to IFN-γ, showing high expression in serum of tuberculosis and tuberculosis-HIV

14

co-infected patients (Jordao et al., 2008). Detection of CXCL9 and CXCL10 by qPCR was

15

also reported as a potential method to enhance diagnostic sensitivity in human tuberculosis

16

(Flynn & Chan, 2003, Magee et al., 2012). Monokine induced by interferon-γ (MIG, CXCL9)

17

was also induced in a murine model of IBD (McKinney et al., 2000, Ito et al., 2006). This

18

suggests that Th1 type chemokines may be valuable biomarkers for MAP infection.

19

Haptoglobin (HP), an acute phase protein, has been demonstrated to chemoattract

20

monocytes, to modulate the immune response, and to have anti-inflammatory activities

21

(Beard et al., 2000, Madsen et al., 2001, Moura et al., 2012). HP was suggested as an obesity

22

and inflammatory marker in its role as a novel monocyte chemoattractant (Beard et al.,

23

2000). Furthermore, serum HP was used within a disease activity index in a model of murine

24

IBD (Ito et al., 2006) and proposed as an independent prognostic factor in epithelial ovarian 10

1

cancer patients (Hines et al., 2014). Elevated serum levels of Chitinase 3-Like-1 (CHI3L1)

2

have been correlated with increased disease severity and a poorer prognosis for many

3

diseases, including cancers, autoimmune diseases, and chronic inflammatory conditions

4

(Ravva & Stanker, 2005). Our findings of up-regulation of HP and CHI3L1 at 48h p.i. in

5

MAP-infected THP-1 also suggest that these genes could provide a useful measure in

6

progression of MAP infection. Expression of TFRC gene was increased at 6h p.i. and

7

maintained at 24h and 48h post infection. TFRC is known as an early phagosome marker, and

8

its expression may be affected by the interference of MAP in the phagosome maturation

9

process (Ruhl & Collins, 1995). TFRC was also up-regulated at 6 h p.i. in Raw 264.7 cell

10

infected with MAP (Cha et al., 2013). In addition, fecal shedder of naturally infected cows

11

showed down-regulation of MPO (Myeloperoxidase) gene (Test1 and Test3, Table1), and the

12

gene was also down-regulated in THP-1 infected with MAP. MPO gene was known to

13

generates reactive oxidants and that has been implicated in mycobacterial killing (Borelli et

14

al., 1999). Down-regulation of MPO gene could be suspected to be associated with resistance

15

of MAP against host defense response. In the present study, THP-1 cell treated with PMA for

16

differentiation before infection with MAP. Even though phenotype of THP-1 differentiated

17

using modified protocol, which was incubated for further 5 days in fresh media after

18

treatment with PMA, but further 1 day incubation in our case, shared characteristics of

19

primary monocyte-derived macrophages (Daigneault et al., 2010), the responses of natural

20

MAP infection in human macrophages could be different from those in THP-1.

21

In the present study, we can see the potential host responses including up-regulation of

22

genes related to immune response (CD14, S100A8, S100A9, LTF, HP and CHCIL3), up-

23

regulation of Th 1-polarizing factor (CCL4, CCL5, CXCL9 and CXCL10), down-regulation

24

of genes related to metabolism (ELANE, IGF1, TCF7L2, and MPO), and no significant 11

1

response of other genes (GADD45a, GPNMB, HMOX1, IFNG, and NQO1) in human

2

macrophage cells infected with MAP, which were selected by an in silico genomic

3

technique (Biomarker Discovery of IPA) in MAP-infected mice.

4 5

Supporting information

6

Supplementary table S1. Primers used for qRT-PCR

7

8

9 10

Acknowledgements This study was supported by the Rural Development Administration (PJ00897001) and the Research Institute for Veterinary Science, Seoul National University, South Korea.

11

12

1 2

3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49

References Balashov KE, Rottman JB, Weiner HL & Hancock WW (1999) CCR5(+) and CXCR3(+) T cells are increased in multiple sclerosis and their ligands MIP-1alpha and IP-10 are expressed in demyelinating brain lesions. Proc Natl Acad Sci U S A 96: 6873-6878. Beard PM, Rhind SM, Sinclair MC, Wildblood LA, Stevenson K, McKendrick IJ, Sharp JM & Jones DG (2000) Modulation of gammadelta T cells and CD1 in Mycobacterium avium subsp. paratuberculosis infection. Vet Immunol Immunopathol 77: 311-319. Beard PM, Henderson D, Daniels MJ, et al. (1999) Evidence of paratuberculosis in fox (Vulpes vulpes) and stoat (Mustela erminea). Vet Rec 145: 612-613. Borelli V, Banfi E, Perrotta MG & Zabucchi G (1999) Myeloperoxidase exerts microbicidal activity against Mycobacterium tuberculosis. Infect Immun 67: 4149-4152. Borrmann E, Mobius P, Diller R & Kohler H (2011) Divergent cytokine responses of macrophages to Mycobacterium avium subsp. paratuberculosis strains of Types II and III in a standardized in vitro model. Vet Microbiol 152: 101-111. Cha SB, Yoo A, Park HT, Sung KY, Shin MK & Yoo HS (2013) Analysis of transcriptional profiles to discover biomarker candidates in Mycobacterium avium subsp. paratuberculosis-infected macrophages, RAW 264.7. J Microbiol Biotechnol 23: 1167-1175. Cossu A, Ferrannini E, Fallahi P, Antonelli A & Sechi LA (2013) Antibodies recognizing specific Mycobacterium avium subsp. paratuberculosis's MAP3738c protein in type 1 diabetes mellitus children are associated with serum Th1 (CXCL10) chemokine. Cytokine 61: 337-339. Daigneault M, Preston JA, Marriott HM, Whyte MK, Dockrell DH (2010) The identification of markers of macrophage differentiation in PMA-stimulated THP-1 cells and monocyte-derived macrophages. PLoS One 5:e8668. Ding QR, Jin T, Wang ZZ & Chen Y (2007) Catalase potentiates retinoic acid-induced THP-1 monocyte differentiation into macrophage through inhibition of peroxisome proliferator-activated receptor gamma. J Leukocyte Biol 81: 1568-1576. Ellingson JL, Anderson JL, Koziczkowski JJ, Radcliff RP, Sloan SJ, Allen SE & Sullivan NM (2005) Detection of viable Mycobacterium avium subsp. paratuberculosis in retail pasteurized whole milk by two culture methods and PCR. J Food Prot 68: 966-972. Flynn JL & Chan J (2003) Immune evasion by Mycobacterium tuberculosis: living with the enemy. Curr Opin Immunol 15: 450-455. Foell D, Frosch M, Sorg C & Roth J (2004) Phagocyte-specific calcium-binding S100 proteins as clinical laboratory markers of inflammation. Clinica Chimica Acta 344: 37-51. Frosch M, Vogl T, Waldherr R, Sorg C, Sunderkotter C & Roth J (2004) Expression of MRP8 and MRP14 by macrophages is a marker for severe forms of glomerulonephritis. J Leukoc Biol 75: 198206. Hines ME, 2nd, Turnquist SE, Ilha MR, et al. (2014) Evaluation of novel oral vaccine candidates and validation of a caprine model of Johne's disease. Front Cell Infect Microbiol 4: 26. Ito R, Shin-Ya M, Kishida T, et al. (2006) Interferon-gamma is causatively involved in experimental inflammatory bowel disease in mice. Clin Exp Immunol 146: 330-338. Jordao L, Lengeling A, Bordat Y, Boudou F, Gicquel B, Neyrolles O, Becker PD, Guzman CA, Griffiths G & Anes E (2008) Effects of omega-3 and -6 fatty acids on Mycobacterium tuberculosis in macrophages and in mice. Microbes Infect 10: 1379-1386. Kerkhoff C, Klempt M & Sorg C (1998) Novel insights into structure and function of MRP8 (S100A8) and MRP14 (S100A9). BBA Mol Cell Res 1448: 200-211. Lugering N, Stoll R, Kucharzik T, Schmid KW, Rohlmann G, Burmeister G, Sorg C & Domschke W (1995) Immunohistochemical Distribution and Serum Levels of the Ca2+-Binding Proteins Mrp8, Mrp14 and Their Heterodimeric Form Mrp8/14 in Crohns-Disease. Digestion 56: 406-414. 13

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52

Madsen M, Graversen JH & Moestrup SK (2001) Haptoglobin and CD163: captor and receptor gating hemoglobin to macrophage lysosomes. Redox Rep 6: 386-388. Magee DA, Taraktsoglou M, Killick KE, et al. (2012) Global gene expression and systems biology analysis of bovine monocyte-derived macrophages in response to in vitro challenge with Mycobacterium bovis. PLoS One 7: e32034. McClure HM, Chiodini RJ, Anderson DC, Swenson RB, Thayer WR & Coutu JA (1987) Mycobacterium paratuberculosis infection in a colony of stumptail macaques (Macaca arctoides). J Infect Dis 155: 1011-1019. McKinney JD, Honer zu Bentrup K, Munoz-Elias EJ, Miczak A, Chen B, Chan WT, Swenson D, Sacchettini JC, Jacobs WR, Jr. & Russell DG (2000) Persistence of Mycobacterium tuberculosis in macrophages and mice requires the glyoxylate shunt enzyme isocitrate lyase. Nature 406: 735-738. Millar D, Ford J, Sanderson J, Withey S, Tizard M, Doran T & HermonTaylor J (1996) IS900 PCR to detect Mycobacterium paratuberculosis in retail supplies of whole pasteurized cows' milk in England and Wales. Appl Environ Microbiol 62: 3446-3452. Mizoguchi A, Mizoguchi E & Bhan AK (2003) Immune networks in animal models of inflammatory bowel disease. Inflamm Bowel Dis 9: 246-259. Motiwala AS, Janagama HK, Paustian ML, Zhu X, Bannantine JP, Kapur V & Sreevatsan S (2006) Comparative transcriptional analysis of human macrophages exposed to animal and human isolates of Mycobacterium avium subspecies paratuberculosis with diverse genotypes. Infect Immun 74: 60466056. Moura DF, de Mattos KA, Amadeu TP, Andrade PR, Sales JS, Schmitz V, Nery JA, Pinheiro RO & Sarno EN (2012) CD163 favors Mycobacterium leprae survival and persistence by promoting antiinflammatory pathways in lepromatous macrophages. Eur J Immunol 42: 2925-2936. Muruganand D, Daniel E, Ebenezer GJ, Rabboni SE, Segar P & Job CK (2004) Mycobacterium leprae infection and serum lactoferrin levels. Lepr Rev 75: 282-288. Mutwiri GK, Butler DG, Rosendal S, Yager J (1992) Experimental infection of severe combined immunodeficient beige mice with Mycobacterium paratuberculosis of bovine origin. Infect Immun 60:4074-4079. Naser SA, Schwartz D & Shafran I (2000) Isolation of Mycobacterium avium subsp paratuberculosis from breast milk of Crohn's disease patients. Am J Gastroenterol 95: 1094-1095. Naser SA, Ghobrial G, Romero C & Valentine JF (2004) Culture of Mycobacterium avium subspecies paratuberculosis from the blood of patients with Crohn's disease. Lancet 364: 1039-1044. Naser SA, Hulten K, Shafran I, Graham DY & El-Zaatari FA (2000) Specific seroreactivity of Crohn's disease patients against p35 and p36 antigens of M. avium subsp. paratuberculosis. Vet Microbiol 77: 497-504. Oh KJ, Park J, Kim SS, Oh H, Choi CS & Koo SH (2012) TCF7L2 modulates glucose homeostasis by regulating CREB- and FoxO1-dependent transcriptional pathway in the liver. PLoS Genet 8: e1002986. Oki M, Ohtani H, Kinouchi Y, Sato E, Nakamura S, Matsumoto T, Nagura H, Yoshie O & Shimosegawa T (2005) Accumulation of CCR5+ T cells around RANTES+ granulomas in Crohn's disease: a pivotal site of Th1-shifted immune response? Lab Invest 85: 137-145. Patel DD, Zachariah JP & Whichard LP (2001) CXCR3 and CCR5 ligands in rheumatoid arthritis synovium. Clin Immunol 98: 39-45. Pechkovsky DV, Zalutskaya OM, Ivanov GI & Misuno NI (2000) Calprotectin (MRP8/14 protein complex) release during mycobacterial infection in vitro and in vivo. FEMS Immunol Med Microbiol 29: 27-33. Ravva SV & Stanker LH (2005) Real-time quantitative PCR detection of Mycobacterium avium subsp paratuberculosis and differentiation from other mycobacteria using SYBR Green and TaqMan assays. J Microbiol Methods 63: 305-317. Ruhl A & Collins SM (1995) Enteroglial Cells (Egc) Are an Integral-Part of the Neuroimmune Axis in the Enteric Nervous-System (Ens). Gastroenterology 108: A680-A680. Schwartz D, Shafran I, Romero C, Piromalli C, Biggerstaff J, Naser N, Chamberlain W & Naser SA 14

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21

(2000) Use of short-term culture for identification of Mycobacterium avium subsp paratuberculosis in tissue from Crohn's disease patients. Clin Microbiol Infec 6: 303-307. Sechi LA, Scanu AM, Molicotti P, Cannas S, Mura M, Dettori G, Fadda G & Zanetti S (2005) Detection and isolation of Mycobacterium avium subspecies paratuberculosis from intestinal mucosal biopsies of patients with and without Crohn's disease in Sardinia. Am J Gastroenterol 100: 1529-1536. Shin MK, Park HT, Shin SW, Jung M, Im YB, Park HE, Cho YI & Yoo HS (2015) Whole-Blood Gene-Expression Profiles of Cows Infected with Mycobacterium avium subsp. paratuberculosis Reveal Changes in Immune Response and Lipid Metabolism. J Microbiol Biotechnol 25: 255-267. Singh UP, Singh S, Singh R, Karls RK, Quinn FD, Potter ME & Lillard JW, Jr. (2007) Influence of Mycobacterium avium subsp. paratuberculosis on colitis development and specific immune responses during disease. Infect Immun 75: 3722-3728. Thayer WR, Coutu JA, Chiodini RJ, Vankruiningen HJ & Merkal RS (1984) Possible Role of Mycobacteria in Inflammatory Bowel-Disease .2. Mycobacterial Antibodies in Crohns-Disease. Digest Dis Sci 29: 1080-1085. Veazey RS, Taylor HW, Horohov DW, Krahenbuhl JL, Oliver JL, 3rd, Snider TG, 3rd (1995) Histopathology of C57BL/6 mice inoculated orally with Mycobacterium paratuberculosis. J Comp Pathol 113:75-80. Yoo HS & Shin SJ (2012) Recent research on bovine paratuberculosis in South Korea. Vet Immunol Immunopathol 148: 23-28. Zwick LS, Walsh TF, Barbiers R, Collins MT, Kinsel MJ & Murnane RD (2002) Paratuberculosis in a mandrill (Papio sphinx). J Vet Diag Invest 14: 326-328.

15

1

Table 1. Expression levels of selected genes in MAP-infected mice and naturally MAP-infected cattle

Group3

A

Mouse spleen1

Gene Symbol

Gene Name

CD14

CD14 molecule

CD68

CD68 molecule

S100A8

S100 calcium binding protein A8

S100A9

S100 calcium binding protein A9

ELANE

elastase, neutrophil expressed

HMOX1

heme oxygenase (decycling 1)

LTF

lactotransferrin

TCF7L2

transcription factor 7-like 2 (T cell specific, HMG-box)

CCL4

chemokine (C-C motif) ligand 4

CCL5

chemokine (C-C motif) ligand 5

HP

haptoglobin

MPO

myeloperoxidase

CXCL9

Chemokine (C-X-C motif) ligand 9

CXCL10

Chemokine (C-X-C motif) ligand 10

IFNG

interferon, gamma

B

C

Location

Cow whole blood2

Family

3 weeks p.i.

6 weeks p.i.

Test1

Test2

Test3

transporter

5.04

2.05

-

2.04

2.5

other

2.3

2.61

-

Cytoplasm

other

64.76

5

-

1.6

1.9

Cytoplasm Extracellular Space Cytoplasm Extracellular Space

other

46.77

4.65

-

1.6

1.9

peptidase

12.99

5.61

-

2.0

1.8

enzyme

6.9

2.85

-

2.3

2.8

peptidase

64.27

7.68

-

3.8

2.9

transcription regulator

3.29

2.12

-

2.0

2.0

cytokine

2.03

2.6

-2.3

-

-

cytokine

2.84

3.02

-2.3

-

-

peptidase

23.85

11.88

-2.8

-

-

enzyme

39.33

7.85

-1.8

-

-1.6

cytokine

4.65

8.4

-

-

-

cytokine

4.94

4.58

-

-

-

cytokine

2.62

3.22

-

-

-

Plasma Membrane Plasma Membrane

Nucleus Extracellular Space Extracellular Space Extracellular Space Cytoplasm Extracellular Space Extracellular Space Extracellular Space 16

2.3

TFRC

transferrin receptor (p90, CD71)

GPNMB

glycoprotein (transmembrane)nmb

GADD45 CHI3L1 IGF1 NQO1

Plasma membrane Plasma Membrane

growth arrest and DNA-damageinducible protein chitinase 3-like 1 (cartilage glycoprotein- 39) insulin-like growth factor1 (somatomedin C)

Extracellular Space Extracellular Space

NAD(P)H dehydrogenase, quione1

Cytoplasm

Nucleus

transporter

11.13

9.12

-

-

-

enzyme

5.49

14.22

-

-

-

other

5.0

6.6

-

-

-

enzyme

12

7.36

-

-

-

growth Factor

2.47

2.35

-

-

-

enzyme

2.14

2.21

-

-

-

1

1

2

2

expression data from microarray after naturally MAP-infected cattle in previous study (Shin et al., 2015). The groups were divided into three groups as follows: Test1,

3

cows shed MAP by feces, Test2, cows with MAP-specific antibodies, and Test3, cows shed MAP by feces with MAP-specific antibodies. 3The selected genes were divided

4

into three groups based on comparison of the results of mice and cattle, A; up-regulated genes in both mice and cattle, B; up- and down-regulated genes in mice and cattle,

5

respectively,

These are gene expression data from microarray at 3 and 6 weeks after infection of mice with MAP in previous study (submitted to another journal). These are gene

C;

up-regulated

and

no

significant

17

changes

in

mice

and

cattle,

respectively.

1

18

1 2

Figure legend

3 4

Fig. 1. Gene expression changes between Map-infected THP-1 cells and uninfected 19

1

control. The selected genes were grouped into three groups as follows: A, up-regulated genes

2

in both mice and cattle; B, up- and down-regulated genes in mice and cattle, respectively; C,

3

up-regulated and no significant changes in mice and cattle, respectively. The relative

4

expression level was normalized with the

5

control by the 2-ΔΔCT method. Significance level set at p ≤ 0.05 (*, p ≤ 0.05; **, p ≤ 0.01; ***,

6

p ≤ 0.001).

-actin expression level relative to zero-time

7

20

Host gene expression for Mycobacterium avium subsp. paratuberculosis infection in human THP-1 macrophages.

Mycobacterium avium subsp. paratuberculosis (MAP) is the causative agent of Johne's disease, which causes considerable economic loss in the dairy indu...
671KB Sizes 1 Downloads 7 Views