Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Mol Immunol ; 156: 31-38, 2023 04.
Article En | MEDLINE | ID: mdl-36889184

Activation of PD-1 by anchoring it to Antigen Receptor (AR) components or associated co-receptors represents an attractive approach to treat autoimmune conditions. In this study, we provide evidence that CD48, a common lipid raft and Src kinase-associated coreceptor, induces significant Src kinase-dependent activation of PD-1 upon crosslinking, while CD71, a receptor excluded from these compartments, does not. Functionally, using bead-conjugated antibodies we demonstrate that CD48-dependent activation of PD-1 inhibits proliferation of AR-induced primary human T cells, and similarly, PD-1 activation using PD-1/CD48 bispecific antibodies inhibits IL-2, enhances IL-10 secretion, and reduces NFAT activation in primary human and Jurkat T cells, respectively. As a whole, CD48-dependent activation of PD-1 represents a novel mechanism to fine tune T cell activation, and by functionally anchoring PD-1 with receptors other than AR, this study provides a conceptual framework for rational development of novel therapies that activate inhibitory checkpoint receptors for treatment of immune-mediated diseases.


Lymphocyte Activation , Programmed Cell Death 1 Receptor , Humans , Jurkat Cells , src-Family Kinases , Apoptosis
2.
Am J Physiol Gastrointest Liver Physiol ; 321(5): G500-G512, 2021 11 01.
Article En | MEDLINE | ID: mdl-34494462

Mouse and human data implicate the NOD1 and NOD2 sensors of the intestinal microbiome and the associated signal transduction via the receptor interacting protein kinase 2 (RIPK2) as a potential key signaling node for the development of inflammatory bowel disease (IBD) and an attractive target for pharmacological intervention. The TRUC mouse model of IBD was strongly indicated for evaluating RIPK2 antagonism for its effect on intestinal inflammation based on previous knockout studies with NOD1, NOD2, and RIPK2. We identified and profiled the BI 706039 molecule as a potent and specific functional inhibitor of both human and mouse RIPK2 and with favorable pharmacokinetic properties. We dosed BI 706039 in the spontaneous TRUC mouse model from age 28 to 56 days. Oral, daily administration of BI 706039 caused dose-responsive and significant improvement in colonic histopathological inflammation, colon weight, and terminal levels of protein-normalized fecal lipocalin (all P values <0.001). These observations correlated with dose responsively increasing systemic levels of the BI 706039 compound, splenic molecular target engagement of RIPK2, and modulation of inflammatory genes in the colon. This demonstrates that a relatively low oral dose of a potent and selective RIPK2 inhibitor can modulate signaling in the intestinal immune system and significantly improve disease associated intestinal inflammation.NEW & NOTEWORTHY The RIPK2 kinase at the apex of microbiome immunosensing is an attractive target for pharmacological intervention. A low oral dose of a RIPK2 inhibitor leads to significantly improved intestinal inflammation in the murine TRUC model of colitis. A selective and potent inhibitor of the RIPK2 kinase may represent a new class of therapeutics that target microbiome-driven signaling for the treatment of IBD.


Colitis, Ulcerative/drug therapy , Colon/drug effects , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Animals , Biological Availability , Cells, Cultured , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Colon/enzymology , Colon/pathology , Crohn Disease/enzymology , Crohn Disease/pathology , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , Disease Models, Animal , Feces/chemistry , Humans , Inflammation Mediators/metabolism , Lipocalins/metabolism , Mice, Inbred BALB C , Mice, Knockout , Models, Biological , Monocytes/drug effects , Monocytes/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , T-Box Domain Proteins/genetics
3.
Immunity ; 43(4): 739-50, 2015 Oct 20.
Article En | MEDLINE | ID: mdl-26431947

Interleukin-23 (IL-23) and IL-17 are cytokines currently being targeted in clinical trials. Although inhibition of both of these cytokines is effective for treating psoriasis, IL-12 and IL-23 p40 inhibition attenuates Crohn's disease, whereas IL-17A or IL-17 receptor A (IL-17RA) inhibition exacerbates Crohn's disease. This dichotomy between IL-23 and IL-17 was effectively modeled in the multidrug resistance-1a-ablated (Abcb1a(-/-)) mouse model of colitis. IL-23 inhibition attenuated disease by decreasing colonic inflammation while enhancing regulatory T (Treg) cell accumulation. Exacerbation of colitis by IL-17A or IL-17RA inhibition was associated with severe weakening of the intestinal epithelial barrier, culminating in increased colonic inflammation and accelerated mortality. These data show that IL-17A acts on intestinal epithelium to promote barrier function and provide insight into mechanisms underlying exacerbation of Crohn's disease when IL-17A or IL-17RA is inhibited.


Colitis/immunology , Interleukin-17/physiology , Interleukin-23/physiology , Receptors, Interleukin-17/physiology , ATP Binding Cassette Transporter, Subfamily B/deficiency , Animals , Colitis/drug therapy , Colitis/etiology , Colitis/microbiology , Disease Models, Animal , Disease Progression , Epithelium/physiopathology , Female , Forkhead Transcription Factors/analysis , Gene Expression Regulation/immunology , Helicobacter Infections/complications , Helicobacter Infections/immunology , Immunization, Passive , Immunoglobulin G/therapeutic use , Interleukin-12 Subunit p40/antagonists & inhibitors , Interleukin-17/immunology , Interleukin-23/immunology , Interleukin-23 Subunit p19/antagonists & inhibitors , Interleukin-23 Subunit p19/immunology , Intestinal Mucosa/physiopathology , Mice , Mice, Knockout , Permeability , Receptors, Interleukin-17/antagonists & inhibitors , Receptors, Interleukin-17/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Transcriptome
4.
Int Immunopharmacol ; 28(1): 354-8, 2015 Sep.
Article En | MEDLINE | ID: mdl-26130567

Staphylococcus aureus is a common nosocomial infection and its resistance to penicillin and methicillin antibiotics is a growing clinical problem. We previously described the development of a humanized anti-Staphylococcus enterotoxin B (SEB) antibody derived from the mouse antibody made by the 20B1 hybridoma. This antibody was humanized and characterized kinetically by surface plasmon resonance demonstrating that the humanized clones retained binding to SEB. Clones were then functionally characterized in an in vitro assay demonstrating that the murine 20B1, chimeric and humanized antibodies potently inhibited SEB-induced murine splenocyte proliferation assay. Here, we describe a human cell-based screening assay, optimized by varying multiple experimental parameters that resulted in an assay that was used to demonstrate full and potent neutralization by the parental, chimeric and humanized antibodies. The replacement of fetal bovine serum (FBS) with normal human serum (NHS) was found to be a crucial factor in the performance of the human cell based screening assay that enabled the calculation of mAb efficacy and potency. In addition, we found that anti-SEB antibodies showed similar efficacy and potency with a triple mutant Fc region (designed to be effector function null) or a wild-type Fc region, which is in contrast to previously described studies.


Antibodies, Monoclonal, Humanized/immunology , Enterotoxins/immunology , Animals , Biological Assay , Cell Proliferation , Cells, Cultured , Humans , Leukocytes/immunology , Male , Mice , Middle Aged , Receptors, IgG/immunology , Serum
5.
J Leukoc Biol ; 97(6): 1011-22, 2015 Jun.
Article En | MEDLINE | ID: mdl-25637591

The regulation of T cell and DC retention and lymphatic egress within and from the intestine is critical for intestinal immunosurveillance; however, the cellular processes that orchestrate this balance during IBD remain poorly defined. With the use of a mouse model of TNF-driven Crohn's-like ileitis (TNF(Δ) (ARE)), we examined the role of CCR7 in the control of intestinal T cell and DC retention/egress during experimental CD. We observed that the frequency of CCR7-expressing TH1/TH17 effector lymphocytes increased during active disease in TNF(Δ) (ARE) mice and that ΔARE/CCR7(-/-) mice developed exacerbated ileitis and multiorgan inflammation, with a marked polarization and ileal retention of TH1 effector CD4(+) T cells. Furthermore, adoptive transfer of ΔARE/CCR7(-/-) effector CD4(+) into lymphopenic hosts resulted in ileo-colitis, whereas those transferred with ΔARE/CCR7(+/+) CD4(+) T cells developed ileitis. ΔARE/CCR7(-/-) mice had an acellular draining MLN, decreased CD103(+) DC, and decreased expression of RALDH enzymes and of CD4(+)CD25(+)FoxP3(+) Tregs. Lastly, a mAb against CCR7 exacerbated ileitis in TNF(Δ) (ARE) mice, phenocopying the effects of congenital CCR7 deficiency. Our data underscore a critical role for the lymphoid chemokine receptor CCR7 in orchestrating immune cell traffic and TH1 versus TH17 bias during chronic murine ileitis.


Ileitis/immunology , Ileum/immunology , Receptors, CCR7/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Adoptive Transfer , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , Cell Movement/drug effects , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/pathology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Models, Animal , Gene Expression Regulation , Humans , Ileitis/genetics , Ileitis/pathology , Ileum/pathology , Isoenzymes/genetics , Isoenzymes/immunology , Mice , Mice, Transgenic , Receptors, CCR7/antagonists & inhibitors , Receptors, CCR7/deficiency , Receptors, CCR7/genetics , Signal Transduction , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology , Th1 Cells/drug effects , Th1 Cells/pathology , Th1 Cells/transplantation , Th17 Cells/drug effects , Th17 Cells/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
6.
J Infect Dis ; 210(6): 973-81, 2014 Sep 15.
Article En | MEDLINE | ID: mdl-24803533

BACKGROUND: Staphylococcal enterotoxin B (SEB), a potential biological warfare agent, is a potent superantigen that contributes to the virulence of methicillin-resistant Staphylococcus aureus (MRSA), which is a major health threat in the United States. Efforts to develop toxin-neutralizing antibodies as adjunctive therapies are justified, given the high mortality and frequent failure of therapy despite available antibiotics. METHODS: Murine SEB-specific mAb 20B1 was humanized, and treatment benefits of Hu-1.6/1.1 and Hu-1.4/1.1 variants were investigated in mice in an SEB intoxication model, as well as in sepsis and deep-tissue infection models. RESULTS: Hu-1.6/1.1 and Hu-1.4/1.1 protected mice against SEB-induced lethal shock. Hu-1.6/1.1 also enhanced survival of mice that developed fatal sepsis after challenge with a SEB-producing MRSA strain. Combined treatment of Hu-1.6/1.1 with vancomycin further increased survival and altered cytokine responses, compared with monotherapy with either monoclonal antibody or vancomycin alone. Efficacy was also demonstrated in the deep-tissue infection model, where Hu-1.4/1.1 bound to SEB in vivo and decreased abscess formation, as well as proinflammatory cytokine levels. CONCLUSIONS: SEB-neutralizing mAb 20B1 was successfully humanized. The mAb affects outcome by modulating the proinflammatory host response in both the sepsis and the intoxication models, which justifies further development.


Anti-Bacterial Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Enterotoxins/immunology , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections/prevention & control , Vancomycin/therapeutic use , Abscess/immunology , Abscess/prevention & control , Animals , Anti-Bacterial Agents/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/immunology , Binding Sites, Antibody/immunology , Cytokines/blood , Enterotoxins/genetics , Methicillin-Resistant Staphylococcus aureus/immunology , Mice , Mice, Inbred BALB C , Staphylococcal Infections/immunology , Vancomycin/administration & dosage
7.
J Infect Dis ; 208(12): 2058-66, 2013 Dec 15.
Article En | MEDLINE | ID: mdl-23922375

BACKGROUND: Methicillin-resistant Staphylococcus aureus (MRSA) has become a major health threat in the United States. Staphylococcal enterotoxin B (SEB) is a potent superantigen that contributes to its virulence. High mortality and frequent failure of therapy despite available antibiotics have stimulated research efforts to develop adjunctive therapies. METHODS: Treatment benefits of SEB-specific monoclonal antibody (mAb) 20B1 were investigated in mice in sepsis, superficial skin, and deep-tissue infection models. RESULTS: Mice challenged with a SEB-producing MRSA strain developed fatal sepsis, extensive tissue skin infection, and abscess-forming deep-seeded thigh muscle infection. Animals preimmunized against SEB or treated passively with mAb 20B1 exhibited enhanced survival in the sepsis model, whereas decrease of bacterial burden was observed in the superficial skin and deep-tissue models. mAb 20B1 bound to SEB in the infected tissue and decreased abscess formation and proinflammatory cytokine levels, lymphocyte proliferation, and neutrophil recruitment. CONCLUSIONS: mAb 20B1, an SEB-neutralizing mAb, is effective against MRSA infection. mAb 20B1 protects against lethal sepsis and reduces skin tissue invasion and deep-abscess formation. The mAb penetrates well into the abscess and binds to SEB. It affects the outcome of S. aureus infection by modulating the host's proinflammatory immune response.


Antibodies, Monoclonal/pharmacology , Enterotoxins/immunology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/immunology , Abscess/microbiology , Abscess/pathology , Animals , Antibodies, Monoclonal/immunology , Enterotoxins/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Interleukins/blood , Interleukins/immunology , Mice , Mice, Inbred BALB C , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcal Skin Infections , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Superantigens/immunology , Superantigens/metabolism , Survival Analysis , T-Lymphocytes/immunology , Thigh/microbiology , Thigh/pathology , Virulence
9.
Autoimmunity ; 42(3): 171-82, 2009 Mar.
Article En | MEDLINE | ID: mdl-19301198

IP-10 secretion is induced by pro-inflammatory cytokines and mediates the migration of CXCR3+ cells. Its elevation in clinical samples has been associated with multiple inflammatory diseases and its antagonism has been reported to be effective in several animal models of inflammatory disease. We generated a mouse anti-mouse IP-10 monoclonal antibody (mAb; Clone 20A9) that specifically bound murine IP-10 with high affinity and inhibited in vitro IP-10 induced BaF3/mCXCR3 cell migration with an IC(50) of approximately 4 nM. The 20A9 mAb was completely absorbed in vivo and had dose proportional pharmacokinetic exposure with a serum half life of 2.4-6 days. The 20A9 mAb inhibited IP-10 mediated T-cell recruitment to the airways, indicating that it is effective in vivo. However, administration of the 20A9 mAb had no significant effect on disease in mouse models of delayed type hypersensitivity, collagen induced arthritis, cardiac allograft transplantation tolerance, EAE or CD4+ CD45RBHi T-cell transfer-induced IBD. These data suggest that the 20A9 mAb can antagonize IP-10 mediated chemotaxis in vitro and in vivo and that this is insufficient to cause a therapeutic benefit in multiple mouse models of inflammatory disease.


Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cell Movement/drug effects , Chemokine CXCL10/antagonists & inhibitors , Chemokine CXCL10/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Arthritis, Experimental/pathology , Arthritis, Experimental/therapy , Bronchoalveolar Lavage Fluid/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Cell Movement/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Graft Rejection/prevention & control , Heart Transplantation/immunology , Inflammation/pathology , Inflammation/therapy , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Inbred Strains , Mice, SCID , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/drug effects , Treatment Outcome
10.
Curr Protoc Pharmacol ; Chapter 5: Unit5.58, 2009 Dec.
Article En | MEDLINE | ID: mdl-22294404

Animal models of experimentally induced inflammatory bowel disease (IBD) are useful for understanding more about the mechanistic basis of disease, identifying new targets for therapeutic intervention, and testing novel therapeutic agents. This unit provides detailed protocols for four of the most commonly used mouse models of experimentally induced intestinal inflammation: chemical induction of colitis by dextran sodium sulfate (DSS), hapten-induced colitis via 2,4,6-trinitrobenzene sulfonic acid (TNBS), Helicobacter-induced colitis in mdr1a(-/-) mice, and the CD4(+) CD45RB(hi) SCID transfer colitis model.


Colitis/etiology , Disease Models, Animal , Adoptive Transfer/methods , Animals , CD4-Positive T-Lymphocytes/transplantation , Colitis/drug therapy , Dextran Sulfate/toxicity , Female , Helicobacter Infections , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Specimen Handling/methods , Trinitrobenzenesulfonic Acid/toxicity
11.
J Immunol ; 181(8): 5278-88, 2008 Oct 15.
Article En | MEDLINE | ID: mdl-18832683

The B7 costimulatory molecules govern many aspects of T cell immune responses by interacting with CD28 for costimulation, but also with CTLA-4 for immune suppression. Although blockade of CTLA-4 with Ab in humans undergoing cancer immune therapy has led to some cases of inflammatory bowel disease, spontaneous animal models of colitis that depend upon modulation of B7 interactions have not been previously described. In this study, we demonstrate that mice expressing a soluble B7-2 Ig Fc chimeric protein spontaneously develop colitis that is dependent on CD28-mediated costimulation of CD4(+) T cells. We show that the chimeric protein has mixed agonistic/antagonist properties, and that it acts in part by blocking the cell intrinsic effects on T cell activation of engagement of CTLA-4. Disease occurred in transgenic mice that lack expression of the endogenous B7 molecules (B7 double knock-out mice), because of the relatively weak costimulatory delivered by the chimeric protein. Surprisingly, colitis was more severe in this context, which was associated with the decreased number of Foxp3(+) regulatory T cells in transgenic B7 double knock-out mice. This model provides an important tool for examining how B7 molecules and their effects on CTLA-4 modulate T cell function and the development of inflammatory diseases.


B7-1 Antigen/immunology , Colitis/immunology , Inflammatory Bowel Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , B7-1 Antigen/genetics , CD28 Antigens/genetics , CD28 Antigens/immunology , CTLA-4 Antigen , Colitis/genetics , Disease Models, Animal , Humans , Inflammatory Bowel Diseases/genetics , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
12.
Curr Opin Drug Discov Devel ; 9(2): 207-17, 2006 Mar.
Article En | MEDLINE | ID: mdl-16566291

The past decade has seen an abundance of new mouse models that mimic the human inflammatory bowel diseases (IBDs), Crohn's disease and ulcerative colitis. These mouse models of IBD have provided great insight into the potential mechanisms that drive homeostatic dysregulation in the intestine, which manifests as mucosal inflammation. Within this review, the different animal models that have been employed to gain a greater understanding of the pathogenesis of IBD are discussed and some of the new biological drugs that have emerged as potential therapeutics as a result of these mouse modeling studies are reviewed.


Inflammatory Bowel Diseases/drug therapy , Animals , Cytokines/biosynthesis , Disease Models, Animal , Humans , Inflammatory Bowel Diseases/pathology , Lymphocyte Activation/immunology , Lymphocyte Activation/physiology , Mice
13.
Mol Imaging Biol ; 5(4): 271-7, 2003.
Article En | MEDLINE | ID: mdl-14499142

Treatment of inflammatory bowel disease (IBD) generally relies on long-term use of anti-inflammatory and immunosuppressive agents. The adverse effects of those drugs make it important to prescribe the minimal regimen that is effective. An objective method for noninvasively quantifying severity of bowel inflammation would thus be valuable in guiding inflammatory bowel disease therapy. Using positron emission tomography (PET), we show that white blood cells (WBCs) labeled with 2-deoxy-2-[18F]fluoro-D-glucose (FDG) can serve as a quantitative marker for identifying the presence and severity of intestinal inflammation. In both murine and human subjects, PET images of FDG-labeled WBCs demonstrated little tracer uptake in healthy gastrointestinal and urinary tracts, where physiologic distribution of FDG images of glucose metabolism often compromises abdominopelvic PET imaging of intestinal pathology. Intestinal foci of FDG-labeled WBCs were confirmed to represent inflamed bowel through histopathologic or colonoscopic analysis, and intensity of foci measured in PET images correlated well with histopathologic measures of degree of inflammation. FDG-labeled WBC's, in conjunction with PET, can be used to provide quantitative assessment of bowel inflammation noninvasively, accurately, and rapidly.


Colon/diagnostic imaging , Inflammatory Bowel Diseases/diagnostic imaging , Leukocytes/metabolism , Tomography, Emission-Computed , Animals , Colon/pathology , Crohn Disease/diagnostic imaging , Crohn Disease/pathology , Kinetics , Mice , Radiography , Time Factors
14.
Am J Physiol Gastrointest Liver Physiol ; 282(4): G690-701, 2002 Apr.
Article En | MEDLINE | ID: mdl-11897629

There is an acute need for effective therapy for inflammatory bowel disease (IBD), particularly at the level of repair of the damaged epithelium. We evaluated the efficacy of recombinant human keratinocyte growth factor (rHuKGF) in both the dextran sodium sulfate (DSS) and the CD4(+)CD45RB(Hi) T cell transfer models of IBD. Disease was induced either by the ad libitum administration to normal mice of 4% DSS in the drinking water or by the injection of 4 x 10(5) CD4(+)CD45RB(Hi) T cells into immunodeficient scid/scid mice. rHuKGF was administered by subcutaneous injection at doses of 1.0 or 3.0 mg/kg in both preventative and therapeutic regimens during both studies. rHuKGF significantly improved survival and body weight loss in the DSS model in both preventative and therapeutic dosing regimens. It also improved diarrhea, hematochezia, and hematological parameters, as well as large intestine histopathology. In the T cell transfer model, rHuKGF improved body weight loss, diarrhea, and levels of serum amyloid A, as well as large intestine histopathology. In both models of IBD, the colonic levels of intestinal trefoil factor (ITF) were elevated by the disease state and further elevated by treatment with rHuKGF. These data suggest that rHuKGF may prove useful in the clinical management of IBD and its effects are likely mediated by its ability to locally increase the levels of ITF.


CD4-Positive T-Lymphocytes/transplantation , Dextran Sulfate , Fibroblast Growth Factors/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Leukocyte Common Antigens/analysis , Animals , CD4-Positive T-Lymphocytes/immunology , Diarrhea , Disease Models, Animal , Female , Fibroblast Growth Factor 7 , Humans , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/physiopathology , Intestinal Mucosa/pathology , Leukocyte Count , Male , Mice , Mice, SCID , Neutrophils , Recombinant Proteins/therapeutic use , Serum Amyloid A Protein/analysis , Weight Loss
...