• Non ci sono risultati.

6 Animal Models of Inflammatory Bowel Diseases

N/A
N/A
Protected

Academic year: 2022

Condividi "6 Animal Models of Inflammatory Bowel Diseases"

Copied!
14
0
0

Testo completo

(1)

Diseases

M.F. Neurath

6.1 Inflammatory Bowel Diseases . . . . 119

6.2 Cytokine Profiles of Lamina Propria T Cells in IBD . . . . 121

6.3 Cytokine Signaling in IBD T Cells . . . . 122

6.4 Animal Models of IBD . . . . 123

6.5 Lessons from the Animal Models . . . . 125

6.6 Treatment of Experimental Colitis with Recombinant Cytokines or Anti-cytokine Antibodies . . . . 127

6.7 Therapeutic Modulation of Cytokines in IBD . . . . 129

References . . . . 130

Here we discuss animal models of chronic intestinal inflammation and their relevance and implications for inflammatory bowel dis- eases (IBD) in humans. Accordingly, we will characterize immuno- logical features in human IBD first and then focus on similarities in mouse models of experimental colitis. Special reference will be made to cytokines and cytokine signaling events in intestinal inflam- mation.

6.1 InflammatoryBowel Diseases

Alterations of the mucosal immune system have been shown to con-

tribute to the pathogenesis of IBD in humans that are defined as in-

flammations of the gastrointestinal tract not due to specific patho-

(2)

gens (Podolsky 1991; Schunk et al. 2000; Strober and Neurath 1995;

Strober et al. 1997; Neurath et al. 1996, 2002a). IBD comprise two major forms: ulcerative colitis (UC) and Crohn's disease (CD).

These diseases can be discriminated by clinical and immunological features. Whereas UC is characterized by a more superficial inflam- mation limited to the large bowel, CD manifests as a transmural granulomatous inflammation that can occur anywhere in the alimen- tary canal. Recent evidence by various groups suggests a fundamen- tal derangement of mucosal immunoregulation in patients with IBD (Neurath et al. 2001; Elson 2002). One aspect of this altered immu- noregulation is a hyperresponsiveness to mucosal antigens caused by a dysregulated immune response to otherwise less immunogenic or harmless products of the intestinal flora. For instance, it was shown that normal mucosal T cells mount far lower in vitro proliferative re- sponses to common microbial antigens than do peripheral T cells, whereas IBD mucosal T-cell responses are equal to that of peripher- al T cells (Duchmann and Zeitz 1998). Furthermore, clinical obser- vations show that IBD manifests itself most frequently in the termi- nal ileum and colon (i.e., those sites with the highest bacterial con- centrations in the intestine), that CD relapses are prevented by diver- sion of fecal stream, and that increases in gut permeability precede acute flares in CD (Duchmann and Zeitz 1998). Therefore, the emerging concept is that bacterial antigens from the commensal flora drive chronic intestinal inflammation in IBD patients.

Bacterial antigens cause an unbalanced activation of the mucosal immune system in IBD patients, thereby leading to chronic intestinal inflammation. In the normal human gut, the intestinal immune sys- tem comprises antigen presenting cells (APCs), epithelial cells, in- traepithelial lymphocytes, and lamina propria (LP) lymphocytes. The special activation and differentiation status of LP T cells is docu- mented by the finding that proliferative capacities and cytokine pro- files of LP lymphocytes differ significantly from those of the periph- eral blood. For instance, LP T cells are significantly more respon- sive to CD2 ligation than peripheral blood lymphocytes but produce less cytokines in response to TCR/CD3 receptor stimulation.

Furthermore, with regard to their proliferative capacities it has been

shown that LP T cells proliferate poorly in response to TCR/CD3 re-

ceptor stimulation compared with peripheral T cells, and somewhat

(3)

better but still poorly in response to CD2/CD28 pathway stimula- tion. In summary, normal human LP T cells are poor proliferators but vigorous lymphokine producers when stimulated via the CD2/

CD28 accessory stimulation pathway.

There is now increasing evidence that these LP T cells play a central role in the pathogenesis of IBD (Duchmann and Zeitz 1998).

This is underlined by the clinical finding that CD in HIV-infected patients is ameliorated or abrogated when CD4

+

T cell numbers in these patients drop. In addition, there is strong evidence that LP CD4

+

T cells mediate chronic intestinal inflammation in several ani- mal models of IBD, as discussed below. The key role of LP T cells in IBD is most likely based on their altered activation and differen- tiation status with consecutive pathologic cytokine responses and macrophage activation. This hypothesis is supported by the clinical effects of monoclonal anti-CD4

+

antibodies in patients with CD, and by the observation that reduction of T helper cell numbers by a con- comitant HIV infection suppresses disease activity in CD (Duch- mann and Zeitz 1998; Emmrich et al. 1991). Consistent with these observations, bone marrow transplantation because of concomitant leukemia can induce long-term remissions in CD, suggesting that the host immune dysregulation plays a role in the perpetuation of this disease that can be corrected by hematopoietic cell transplantation.

Finally, antibodies to TNF that are successfully used in treating CD patients induce rapid mucosal T-cell apoptosis (programmed cell death) within 2 days, indicating that the therapeutic efficacy of these antibodies could be due to the elimination of T effector cells in the gut (Hove et al. 2002). Finally, LP T lymphocytes are also a major source for cytokine production in patients with IBD.

6.2 Cytokine Profiles of Lamina Propria T Cells in IBD

Based on the above data there is considerable interest in understand-

ing the function of IBD T cells by determining their capacity to

produce various regulatory lymphokines. Since accessory pathway

stimulation is important for T-cell activation in mucosal immune

responses, it was of interest to determine cytokine production and T-

cell proliferation in IBD T lymphocytes under accessory pathway

(4)

stimulation. It was found that whereas IBD lamina propria T cells both from UC and CD patients gave lower proliferative responses than did control lamina propria T cells via the TCR/CD3 signaling pathway, proliferative responses via the CD2/CD28 pathway were relatively preserved. The production of IFN-c, the signature cytokine of Th1 T cells, has also been measured in IBD lamina propria T cells after stimulation with anti-CD2 plus anti-CD28 antibodies.

When purified lamina propria T cells from patients with CD were stimulated via accessory signaling pathways, IFN-c production was significantly increased compared to T cells from control patients, whereas IL-2 production was reduced (Fuss et al. 1996). Thus this lymphokine profile may represent a modified Th1 cytokine profile present in patients with CD. In contrast to CD, few data are avail- able on cytokine production by lamina propria T cells in patients with UC. However, Fuss et al. (1996) showed that purified lamina propria CD4

+

T lymphocytes from UC patients produce equal amounts of the Th1 cytokine IFN-c but large amounts of the Th2 cy- tokine IL-5 upon stimulation with anti-CD2/CD28 antibodies com- pared to T cells from control patients. Interestingly, the production of another Th2 cytokine, IL-4, was reduced by UC lamina propria T cells compared to T cells from control patients, suggesting the po- tential existence of a modified Th2 cytokine profile in UC patients (Fuss et al. 1996). This cytokine profile may at least partially ac- count for the observed activation of B cells and the production of autoantibodies in UC patients, since Th2 cytokines are known to ac- tivate humoral immune responses. Taken together, the current data are consistent with a model in which co-stimulated T cells from CD patients produce a modified Th1 cytokine profile (high IFN-c, low IL-2), whereas T cells from UC patients exhibit a modified Th2 cytokine profile (high IL-5, low IL-4). Consistent with this concept, Fuss et al. recently showed that LP T cells from UC but not CD pa- tients produced large amounts of IL-13.

6.3 Cytokine Signaling in IBD T Cells

The data summarized above suggest that cytokines play a key role

in the pathogenesis of IBD in humans. This concept is underlined by

(5)

the therapeutic efficacy of several anti-cytokine strategies in IBD pa- tients. Regarding CD, recent studies have focused on understanding the molecular pathways leading to Th1 T-cell differentiation in this disease. The cytokine IL-12 heterodimer (p35/p40) produced by DCs or recently immigrated macrophages induces Th1 T-cell differentia- tion; this function requires the intracellular activation and nuclear translocation of the transcription factor Stat4 (signal transducer and activator of transcription 4) (Neurath et al. 2002a). However, the im- portance of the IL-12/Stat4 signaling pathway for CD pathogenesis has been highlighted by recent studies showing that IL-12 p35/p40 heterodimer secretion is increased in active CD, that T cells in this disease express large amounts of IL-12 receptor b2 chain, and that Stat4 is activated in mucosal T cells from active CD (Monteleone et al. 1997). Finally, IL-12 has been suggested to cause intestinal tissue injury, suggesting that it may play an active role in tissue destruc- tion in CD patients (Monteleone et al. 1999).

In addition to Stat4, the IFN-c inducible transcription factor Stat1 has been shown to be involved in Th1 T-cell differentiation. IFN-c signaling via Stat1 leads to activation of the transcription factor T- bet, a recently cloned member of the T-box protein family expressed in T lymphocytes (Szabo et al. 2000, 2002). Indeed, in CD T-bet is strongly upregulated in LP T cells, showing that these cells exhibit classical features of Th1 T cells (Neurath et al. 2002b). In contrast to CD, few data are available on cytokine signaling and T-cell differ- entiation in patients with UC. However, it appears that there is no increase in IL-12 production by LP cells in UC patients compared to control patients, while the expression of a potentially IL-12-antago- nizing cytokine, denoted EBI3, is increased (Christ et al. 1998).

6.4 Animal Models of IBD

Various animal models of chronic intestinal inflammation have been established recently which will likely provide new insights into the pathogenesis and potential treatment regimens of IBD (Elson et al.

1995; Wirtz and Neurath 2000). Although none of these models

truly represents human IBD, these models mimic key clinical, histo-

logical, and immunological findings in IBD patients. There have

(6)

been more than 70 models described for experimental colitis in ro- dents, and it is beyond the scope of the present review to discuss all of these models in detail. However, these models generally can be classified into four different groups. The first group of models con- sists of spontaneously occurring colitis forms such as the cotton top tamarin model and the C

3

H Bir mouse (Wirtz and Neurath 2000).

The second group consists of inducible models in normal healthy mice such as the DSS colitis model, the TNBS model, and the oxa- zolone colitis model (Neurath et al. 1995; Boirivant et al. 1998).

The third and largest group comprises genetically modified animals such as transgenic mice or rats, mice in which certain genes have been inactivated by homologous recombination, and mice with tar- geted deletion of genes in certain cell populations only (conditional knockout mice). These models include rats carrying transgenes for HLA-B27 and b2-microglobulin, T-cell reconstituted Tge26mice transgenic for the human CD3e gene, mice carrying a dominant neg- ative N-cadherin mutant, and mice in which the genes for IL-2, IL- 10, Gai2, and the a- or b-chain of the T-cell receptor have been inac- tivated by homologous recombination (Kuhn et al. 1993; Mizoguchi et al. 1997, 1999). The fourth group consists of adoptive transfer models with either CD4

+

or CD8

+

T cells (Wirtz and Neurath 2000).

The most commonly used model in this group is an adoptive trans- fer of normal CD45RBhi or CD62L

+

T cells from healthy BALB/c donor mice into C.B.-17 SCID or RAG knockout mice (Powrie et al.

1994, 1996; Atreya et al. 2000) that results in colitis in the reconsti- tuted mice within 4±8 weeks (Table 1).

Table 1. Models of experimental colitis in rodents

Group 1 Group 2 Group 3 Group 4

Spontaneous Inducible Transfer Genetically

C

3

H Bir mouse DSS CD45Rbhigh IL-2 KO mouse

Cotton top tamarin TNBS CD62L

+

CD4

+

IL-10 KO mouse

Oxazolone hsp60 CD8

+

STAT4 transgenic

(7)

6.5 Lessons from the Animal Models

As discussed above, the IL-12/Stat-4 signaling pathway plays an im- portant role in Th1-mediated intestinal inflammation in humans.

This observation has been strongly supported by studies in mice with experimental colitis (Fig. 1). For instance, Th1-mediated mod- els of chronic intestinal inflammation are associated with increased production of IL-12, and neutralizing antibodies to IL-12 p40 pre- vent the development of Th1-mediated colitis, but they also suppress established Th1-mediated chronic intestinal inflammation (Neurath et al. 1995; Simpson et al. 1998). This effect is mediated by the prevention of Th1 T-cell development and the induction of Fas- mediated T-cell apoptosis in the inflamed gut (Fuss et al. 1999).

Furthermore, Stat4-deficient T cells fail to induce Th1-mediated co- litis in an adoptive transfer system in RAG knockout mice, whereas

Fig. 1. Model of Th1-mediated experimental colitis driven by bacterial anti- gens from the lumen and IL-12 production. (Modified from Neurath et al.

2002)

(8)

Stat4 transgenic mice (under control of the CMV promoter) develop Th1-mediated colitis upon immunization with DNP-KLH (Simpson et al. 1998; Wirtz et al. 1999). In addition to IL-12, however, the cy- tokine IL-23 (p19/p40) produced by APCs was shown to activate Stat4 in T lymphocytes (Oppmann et al. 2000), raising the possibili- ty that the observed Stat4 activation in colitis could be at least par- tially due to IL-23. Furthermore, the neutralizing antibodies to IL-12 used in the above studies on colitis activity appear to suppress the function of both IL-12 and IL-23, indicating that further studies are necessary to delineate the function of these cytokines individually.

In this context, high levels of IL-23 production by LP dendritic cells were recently described in the terminal ileum of mice (Oppmann et al. 2000), suggesting that this end-stage effector cytokine might be very important in driving memory T-cell activation in the small bowel.

Additional studies have been done on the role of T-bet in experi- mental colitis (Neurath et al. 2002b). These studies have shown that T-bet is an important factor for Th1 T-cell development and the reg- ulation of T-cell effector function. Specifically, in murine models of colitis it was found that T-bet-deficient T cells fail to induce Th1- mediated experimental colitis in an adoptive T-cell transfer system.

This observation cannot be attributed to the effects of T-bet on IFN- c production in CD4

+

T cells, since CD4

+

T cells from IFN-c knock- out mice are fully capable of inducing Th1-mediated colitis in an adoptive transfer system in RAG knockout mice. Instead these re- sults suggest a more general role of T-bet in Th1 T-cell differentia- tion via regulation of IL-12 receptor b2 chain expression. Finally, overexpression of T-bet in T cells was found to augment Th1- mediated colitis, suggesting that T-bet controls Th1-mediated muco- sal immune responses.

A potential role for EBI3 in colitis is supported by the recent ob-

servation that EBI3-deficient mice show reduced Th2 cytokine re-

sponses and are protected from oxazolone-induced colitis, a colitis

model mediated by iNK T cells producing IL-4 and IL-13. Further-

more, experimental colitis models support the notion that Th2 cyto-

kines may play a key pathogenic role in regulating colitis activity in

vivo. In fact, transfer of Th2 cells has recently been shown to cause

colitis in an antigen-specific colitis model. Furthermore, antibodies

(9)

to IL-4 attenuate T cell-mediated colitis induced by the hapten re- agent oxazolone (Boirivant et al. 1998). In addition, inactivation of the IL-4 but not the IFN-c gene has been shown to suppress colitis activity in TCR knockout animals that normally display some fea- tures of human UC. Furthermore, removal of the cecal tip early in life prevents colitis in TCR knockout mice (Mizoguchi et al. 1996), suggesting that the immune system in this region initiates rather than perpetuates such colitis. This is reminiscent of the observed protective effect of early appendectomy on the development of coli- tis in UC patients.

There is some recent indication that the role of individual cyto- kines may vary during the course of experimental colitis. For in- stance, administration of recombinant IL-4 prevents Th1-mediated colitis in an adoptive transfer model but can augment established Th1-mediated colitis, suggesting that the effects of the Th2-type cytokine IL-4 may be dependent on the stage of the disease in T cell-mediated colitis. Consistently, Spencer and coworkers recently showed that colitis in young IL-10-deficient mice is mediated by IL- 12-driven Th1 T cells, whereas later stages of disease appear to be mediated by the Th2-type cytokines IL-4 and IL-13 (Spencer et al.

2002). These findings support the notion that cytokine production and cytokine function may differ during different phases of colitis in vivo.

The above findings on transcriptional polarization of T cells not only give valuable new insights into the immunopathogenesis of IBD but also provide a rationale for selective targeting of signaling cascades in IBD.

6.6 Treatment of Experimental Colitis with Recombinant Cytokines or Anti-cytokine Antibodies

Administration of neutralizing antibodies to cytokines with assumed

pathogenic roles is a commonly used approach for treatment of ex-

perimental colitis. For instance, IFN-c and TNF were shown to be

involved in the pathogenesis of colitis in CD45Rbhi CD4

+

T cell re-

stored SCID mice, as disease was prevented by administration of an-

tibodies to IFN-c and significantly reduced in severity by treatment

(10)

with antibodies to TNF (Powrie et al. 1994). In this experimental model, IFN-c may either act directly by causing damage to colonic epithelial cells or may act primarily by activating macrophages with subsequent production of inflammatory mediators, such as reactive oxygen and nitrogen intermediates, IL-1, IL-6, IL-8, IL-12, and TNF. Furthermore, it was shown that IL-12 is essential for disease induction in TNBS-induced colitis, as treatment with antibodies to IL-12 prevented disease induction. Furthermore, administration of anti-gp39 (CD40L) antibodies during the induction phase of Th1 re- sponses in TNBS-colitis was associated with decreased IL-12 pro- duction, and prevented IFN-c production by LP T cells and disease activity (Stçber et al. 1996). Conversely, injection of rIL-12 hetero- dimer reversed the effect of anti-gp39 treatment and resulted in se- vere disease activity. However, when anti-gp39 was given after dis- ease was established, no effect on disease activity was observed, suggesting that CD40/CD40L interactions are crucial for the in vivo priming of TH1 T cells via the stimulation of IL-12 secretion by APCs in the early phase of colitis. In contrast to anti-gp39, antibod- ies to IL-12 were also highly effective in treating established chronic colitis in the murine TNBS model.

There is now overwhelming evidence that regulatory T-cell popu-

lations may prevent development of colitis by production of cyto-

kines whose functions antagonize those of Th1 T cells. Among the

regulatory subsets identified in the gut are Tr1 cells producing IL-

10, Foxp3-expressing CD4

+

CD25

+

T cells, and TGF-b-producing

Th3 cells. Thus, a second approach for treatment of intestinal in-

flammation consists of administration of cells producing anti-inflam-

matory cytokines or recombinant cytokines with assumed protective

roles. For instance, administration of rIL-10 inhibited development

of colitis in SCID mice reconstituted with CD45RBhi T cells. In ad-

dition to IL-10, TGF-b may also play a pivotal role in the regulation

of inflammatory reactions of the intestine. TGF-b is a pluripotent cy-

tokine known to be involved in immune and inflammatory cell pro-

cesses. Of particular relevance is the fact that TGF-b is a potent in-

hibitor of T- and B-cell proliferation and down-regulates macro-

phage cytokine production. The importance of this inhibitory func-

tion is underscored by the finding that TGF-b1 null mice show mul-

tiorgan inflammation, excessive lymphocytic infiltrations, and early

(11)

death. TGF-b has also been shown to be induced upon oral exposure of various antigens and to be an important mediator of oral toler- ance. Experimental studies showed that protection from CD45Rbhi CD4

+

T cell-induced colitis in SCID mice by co-transfer of CD45RBlo CD4

+

T cell population could be reversed by adding an- tibodies to TGF-b, suggesting a key negative role for this cytokine in disease induction (Powrie et al. 1996; Powrie and Maloy 2003).

These data on the functional importance of TGF-b are further sup- ported by an oral tolerance model in mice with TNBS-colitis, in- duced by oral administration of haptenized colonic proteins (HCPs) prior to rectal administration of TNBS that resulted in suppression of TNBS-induced disease. This suppression (oral tolerance) appears to be due to the generation of mucosal T cells producing TGF-b.

These data suggested that regulation of IL-12-driven Th1-responses and TGF-b levels may have relevance for treatment of human in- flammatory bowel diseases.

6.7 Therapeutic Modulation of Cytokines in IBD

The above data have encouraged clinical studies to determine

whether selective regulation of cytokine levels could be an addi-

tional therapeutic option to treat patients with IBD and whether such

regulation could be better than treatment with immunosuppressive

drugs. Thus, several recent studies have attempted to treat patients

with IBD by administration of recombinant cytokines with assumed

protective functions or by administration of antibodies to cytokines

with assumed pathogenic roles. For instance, systemic administration

of antibodies to TNF-a to patients with CD resulted in reduction of

disease activity or even remission of disease in about 65% of the pa-

tients (Targan et al. 1997). Furthermore, many additional clinical

therapies based on modulation of cytokine function are now in phase

II trials (e.g., anti-IFN-c, anti-IL-12, anti-IL6R antibodies) that are

based on initial work in experimental models of colitis.

(12)

References

Atreya R, Mudter J, Finotto S, et al (2000) Blockade of IL-6trans-signaling suppresses T cell resistance against apoptosis in chronic intestinal inflam- mation: evidence in Crohn's disease and experimental colitis in vivo. Nat Med 6:583±588

Boirivant M, Fuss IJ, Chu A, Strober W (1998) Oxazolone colitis: a murine model of T helper cell type 2 colitis treatable with antibodies to interleu- kin-4. J Exp Med 188:1929±1939

Christ AD, Stevens AC, Koeppen H, Walsh S, Omata F, Devergne O, Bir- kenbach M, Blumberg RS (1998) An interleukin 12-related cytokine is up-regulated in ulcerative colitis but not in Crohn's disease. Gastroenter- ology 115:307±313

Duchmann R, Zeitz M (1998) Crohn's disease. In: Ogra P, Strober W (eds) Handbook of mucosal immunology. Academic Press, New York

Elson CO (2002) Genes, microbes, and T cells ± new therapeutic targets in Crohn's disease. New Engl J Med 346:614±616

Elson CO, Sartor RB, Tennyson GS, Riddell RH (1995) Experimental mod- els of inflammatory bowel disease. Gastroenterology 109:1344±1367 Emmrich J, Seyfarth M, Fleig W, Emmrich F (1991) Treatment of inflam-

matory bowel disease with anti-CD4 monoclonal antibody. Lancet 338:

570±571

Fuss I, Neurath MF, Boirivant M, Klein JS, Motte CD, Strong SA, Fiocchi C, Strober W (1996) Disparate CD4+ lamina propria (LP) lymphocyte secretion profiles in inflammatory bowel disease. J Immunol 157:1261±

Fuss IJ, Marth T, Neurath MF, Pearlstein GR, Jain A, Strober W (1999) 1270 Anti-interleukin 12 treatment regulates apoptosis of Th1 T cells in ex- perimental colitis in mice. Gastroenterology 117:1078±1088

Hove TT, Montfrans CV, Peppelenbosch MP, Deventer SJv (2002) Inflixi- mab treatment induces apoptosis of lamina propria T lymphocytes in Crohn's disease. Gut 50:206±211

Kuhn R, Lohler J, Rennick D, Rajewsky K, Mueller W (1993) Interleukin- 10 deficient mice develop chronic enterocolitis. Cell 75:263±274 Mizoguchi A, Mizoguchi E, Chiba C, Bhan AK (1996) Role of appendix in

the development of inflammatory bowel disease in TCR-a mutant mice. J Exp Med 184:707±715

Mizoguchi A, Mizoguchi E, Smith RN, Preffer FI, Bhan AK (1997) The suppressive role of B cells in chronic colitis of T cell receptor a mutant mice. J Exp Med 186:1749±1756

Mizoguchi A, Mizoguchi E, Bhan AK (1999) The critical role for interleu- kin-4 but not interferon-c in the pathogenesis of colitis in T-cell receptor a mutant mice. Gastroenterology 116:320±326

Monteleone G, Biancone L, Marasco R, Morrone G, Marasco O, Luzza F,

Pallone F (1997) Interleukin-12 is expressed and actively released by

(13)

Crohn's disease intestinal lamina propria mononuclear cells. Gastroenter- ology 112:1169±1178

Monteleone G, MacDonald TT, Wathen NC, Pallone F, Pender SL (1999) Enhancing Lamina propria Th1 cell responses with interleukin 12 pro- duces severe tissue injury. Gastroenterology 117:1069±1077

Neurath MF, Fuss I, Kelsall BL, Stuber E, Strober W (1995) Antibodies to IL-12 abrogate established experimental colitis in mice. J Exp Med 182:1280±1289

Neurath MF, Duchmann R, Bçschenfelde KHMz (1996) Zytokine bei chro- nisch entzçndlichen Darmerkrankungen. DMW 121:735±741

Neurath MF, Finotto S, Strober W (2001) Immunology of inflammatory bowel disease. Chap. 87: Clinical Immunology, 2nd edn. In press Neurath MF, Finotto S, Glimcher LH (2002a) The role of Th1/Th2 polariza-

tion in mucosal immunity. Nat Med 8:567±573

Neurath MF, Weigmann B, Finotto S, et al (2002b) The transcription factor T-b regulates mucosal T cell activation in experimental colitis and Crohn's disease. J Exp Med 195:1129±1143

Oppmann B, Lesley R, Blom B et al (2000) Novel p19 protein engages IL- 12p40 to form a cytokine, IL-23, with biological activities similar to as well as distinct from IL-12. Immunity 13(5):715±725

Podolsky DK (1991) Inflammatory bowel disease. New Engl J Med 325:

928±937

Powrie F, Maloy KJ (2003) Regulating the regulators. Science 299:1030±

Powrie F, Leach MW, Mauze S, Menon S, Caddle LB, Coffman RL (1994) 1031 Inhibition of Th1 responses prevents inflammatory bowel disease in scid mice reconstituted with CD45RBhi CD4+ T cells. Immunity 2:553±562 Powrie F, Carlino J, Leach MW, Mauze S, Coffman RL (1996) A critical

role for transforming growth factor-b but not interleukin-4 in the suppres- sion of T helper type 1-mediated colitis by CD45Rb(low) CD4+ T cells.

J Exp Med 183:2669±2674

Schunk K, Kern A, Oberholzer K, Kalden P, Orth T, Mayer I, Wanitschke R (2000) Hydro-MRI in Crohn's disease: Appraisal of disease activity. In- vest Radiol 35:431±437

Simpson SJ, Shah S, Comiskey M, de-Jong VP, Wang B, Mizoguchi E, Bhan AK, Terhorst C (1998) T cell-mediated pathology in two models of experimental colitis depends predominantly on the interleukin 12/Signal transducer and activator of transcription (Stat)-4 pathway, but is not con- ditional on interferon c expression by T cells. J Exp Med 187:1225±1234 Spencer DM, Veldman GM, Banerjee S, Willis J, Levine AD (2002) Distinct

inflammatory mechanisms mediate early versus late colitis in mice. Gas- troenterology 122:94±105

Strober W, Neurath MF (1995) Immunological diseases of the gastrointesti- nal tract. In: Rich RR (ed) Clinical immunology, Chap. 94. Mosby, St.

Louis, pp 1401±1428

(14)

Strober W, Kelsall B, Fuss I, Marth T, Ludviksson B, Ehrhardt R, Neurath MF (1997) Reciprocal IFN-c and TGF-b responses regulate the occur- rence of mucosal inflammation. Immunol Today 18:61±64

Stçber E, Strober W, Neurath MF (1996) Blocking the CD40L-CD40 inter- action in vivo specifically prevents the priming of Th1-T cells through the inhibition of IL-12 secretion. J Exp Med 183:693±698

Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH (2000) A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell 100:655±669

Szabo SJ, Sullivan BM, Stemmann C, Satoskar AR, Sleckman BP, Glimcher LH (2002) T-bet is essential for Th1 lineage commitment and IFN-c pro- duction in CD4 but not CD8 T Cell. Sci 295:338±342

Targan SR, Hanauer SB, Deventer SJV, Mayer L, Present DH, Braakman T, Woody KLD, Schaible TF, Rutgeerts PJ (1997) A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor a for Crohn's disease. New Engl J Med 337:1029±1035

Wirtz S, Neurath MF (2000) Animal models of intestinal inflammation: new insights into the molecular pathogenesis and immunotherapy of inflam- matory bowel disease. Int J Colorectal Dis 15:144±160

Wirtz S, Finotto S, Kanzler S, Lohse AW, Blessing M, Lehr HA, Galle PR,

Neurath MF (1999) Cutting Edge: Chronic intestinal inflammation in

STAT-4 transgenic mice: characterization of disease and adoptive transfer

by TNF- plus IFN-c producing CD4+ T cells that respond to bacterial

antigens. J Immunol 162:1884±1888

Riferimenti

Documenti correlati

Results with probiotics in Crohn’s disease are con- flicting. In a small pilot study, treatment with cap- sules containing E. coli Nissle 1917 was compared to the administration

The diagnosis of Crohn’s disease should include assessment of the presence, severity, and extent of disease, inflammatory lesion activity, and the pres- ence of

Inflammatory bowel diseases (IBD), which appear mainly in the form of ulcerative colitis (UC) and Crohn’s disease (CD), represent complex disorders as documented by the wide

It is now apparent that the major hepatobiliary diseases seen in association with both UC and Crohn's disease (CD) - namely pericholangitis, primary sclerosing cholangitis

previsto ai sensi del II comma dell’art. 42 in data 28.02.2005, esecutiva a tutti gli effetti, il Comune ha adottato il Programma Integrato di Riqualificazione Urbanistica

automobile starter interrupters. These are devices that are installed in cars by creditors, allowing them to remotely disable the car if a debtor has breached the terms of

al trattamento d zati  e  non  di  com di cui al punto 4 l re     . al trattamento d i e ad attività di p

al trattamento d zati  e  non  di  com di cui al punto 4 l re     . al trattamento d i e ad attività di p