Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 12: 642802, 2021.
Article in English | MEDLINE | ID: mdl-33936060

ABSTRACT

Protection against Staphylococcus aureus is determined by the polarization of the anti-bacterial immune effector mechanisms. Virulence factors of S. aureus can modulate these and induce differently polarized immune responses in a single individual. We proposed that this may be due to intrinsic properties of the bacterial proteins. To test this idea, we selected two virulence factors, the serine protease-like protein B (SplB) and the glycerophosphoryl diester phosphodiesterase (GlpQ). In humans naturally exposed to S. aureus, SplB induces a type 2-biased adaptive immune response, whereas GlpQ elicits type 1/type 3 immunity. We injected the recombinant bacterial antigens into the peritoneum of S. aureus-naïve C57BL/6N mice and analyzed the immune response. This was skewed by SplB toward a Th2 profile including specific IgE, whereas GlpQ was weakly immunogenic. To elucidate the influence of adjuvants on the proteins' polarization potential, we studied Montanide ISA 71 VG and Imject™Alum, which promote a Th1 and Th2 response, respectively. Alum strongly increased antibody production to the Th2-polarizing protein SplB, but did not affect the response to GlpQ. Montanide enhanced the antibody production to both S. aureus virulence factors. Montanide also augmented the inflammation in general, whereas Alum had little effect on the cellular immune response. The adjuvants did not override the polarization potential of the S. aureus proteins on the adaptive immune response.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Phosphoric Diester Hydrolases/immunology , Staphylococcus aureus/immunology , Virulence Factors/immunology , Animals , Antibodies, Bacterial/blood , Antigen-Presenting Cells/immunology , Cytokines/biosynthesis , Female , Immunization , Mice , Mice, Inbred C57BL , Th1 Cells/immunology , Th2 Cells/immunology
2.
Pancreatology ; 20(8): 1637-1647, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33097430

ABSTRACT

BACKGROUND: Acute pancreatitis is a gastrointestinal disorder of high incidence resulting in life threatening complications in up to 20% of patients. Its severe form is characterized by an extensive and systemic immune response. We investigated the role of the adaptive immune response in two experimental models of pancreatitis. METHODS: In C57BI/6-mice mild pancreatitis was induced by 8-hourly injections of caerulein and severe pancreatitis by additional, partial pancreatic duct ligation. T-cell-activation was determined by flow-cytometry of CD25/CD69, T-cell-differentiation by nuclear staining of the transcription-factors Tbet, Gata3 and Foxp3. In vivo CD4+ T-cells were depleted using anti-CD4 antibody. Disease severity was determined by histology, serum amylase and lipase activities, lung MPO and serum cytokine levels (IL-6, TNFα, IL-10). RESULTS: In both models T-cells were activated. Th1-differentiation (Tbet) was absent during pancreatitis but we detected a pronounced Th2/Treg (Gata3/Foxp3) response which paralleled disease severity in both models. The complete depletion of CD4+ T-cells via anti-CD4 antibody, surprisingly, reduced disease severity significantly, as well as granulocyte infiltration and pro- and anti-inflammatory cytokine levels. Co-incubation of acini and T-cells did not lead to T-cell-activation by acinar cells but to acinar damage by T-cells. During pancreatitis no significant T-cell-infiltration into the pancreas was observed. CONCLUSION: T cells orchestrate the early local as well as the systemic immune responses in pancreatitis and are directly involved in organ damage. The Th2 response appears to increase disease severity, rather than conferring an immunological protection.


Subject(s)
Adaptive Immunity , Cell Differentiation , Pancreatitis , T-Lymphocytes, Regulatory , Th2 Cells , Animals , Cytokines , Disease Models, Animal , Lymphocyte Activation , Mice , Pancreatitis/immunology
3.
Gastroenterology ; 158(1): 253-269.e14, 2020 01.
Article in English | MEDLINE | ID: mdl-31593700

ABSTRACT

BACKGROUND & AIMS: Pancreatitis starts with primarily sterile local inflammation that induces systemic inflammatory response syndrome, followed by compensatory anti-inflammatory response syndrome (CARS). We investigated the mechanisms of these processes in mice and human serum. METHODS: We induced severe acute pancreatitis by partial duct ligation with caerulein stimulation or intraperitoneal injection of l-arginine in mice with deletion of interleukin (IL)12B, NLRP3, or IL18 and in mice given MCC950, a small molecule inhibitor of the NLRP3-inflammasome. Pancreata were collected from mice and analyzed by histology, and cytokine levels were measured in serum samples. We measured activation of adaptive immune responses in mice with pancreatitis by flow cytometry analysis of T cells (CD25 and CD69) isolated from the spleen. Differentiation of T-helper (Th1) cells, Th2 cells, and T-regulatory cells was determined by nuclear staining for TBET, GATA3, and FOXP3. We performed transcriptome analysis of mouse lymph nodes and bone marrow-derived macrophages after incubation with acini. We measured levels of cytokines in serum samples from patients with mild and severe acute pancreatitis. RESULTS: Activation of the adaptive immune response in mice was initiated by macrophage-derived, caspase 1-processed cytokines and required activation of NLRP3 (confirmed in serum samples from patients with pancreatitis). Spleen cells from mice with pancreatitis had increases in Th2 cells but not in Th1 cells. Bone marrow-derived macrophages secreted IL1B and IL18, but not IL12, after co-incubation with pancreatic acini. T-cell activation and severity of acute pancreatitis did not differ significantly between IL12B-deficient and control mice. In contrast, NLRP3- or IL18-deficient mice had reduced activation of T cells and no increase in Th2 cell-mediated responses compared with control mice. The systemic type 2 immune response was mediated by macrophage-derived cytokines of the IL1 family. Specifically, IL18 induced a Th2 cell-mediated response in the absence of IL12. MCC950 significantly reduced neutrophil infiltration, T-cell activation, and disease severity in mice. CONCLUSIONS: In mice with severe pancreatitis, we found systemic inflammatory response syndrome and compensatory anti-inflammatory response syndrome developed in parallel. Infiltrating macrophages promote inflammation and simultaneously induce a Th2 cell-mediated response via IL18. Inhibition of NLRP3 reduces systemic inflammatory response syndrome and compensatory anti-inflammatory response syndrome and might be used to treat patients with severe pancreatitis.


Subject(s)
Furans/administration & dosage , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pancreatitis/immunology , Sulfonamides/administration & dosage , Systemic Inflammatory Response Syndrome/immunology , Acinar Cells , Adaptive Immunity , Animals , Arginine/toxicity , Cells, Cultured , Ceruletide/toxicity , Cytokines/blood , Cytokines/immunology , Disease Models, Animal , Heterocyclic Compounds, 4 or More Rings , Humans , Indenes , Injections, Intraperitoneal , Interleukin-18/immunology , Interleukin-18/metabolism , Macrophages/immunology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pancreas/cytology , Pancreas/immunology , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/drug therapy , Primary Cell Culture , Sulfones , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/drug therapy , Th2 Cells/immunology , Th2 Cells/metabolism
4.
Sci Rep ; 9(1): 16774, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31727956

ABSTRACT

Acute pancreatitis is characterized by an early intracellular protease activation and invasion of leukocytes into the pancreas. Cathepsins constitute a large group of lysosomal enzymes, that have been shown to modulate trypsinogen activation and neutrophil infiltration. Cathepsin G (CTSG) is a neutrophil serine protease of the chymotrypsin C family known to degrade extracellular matrix components and to have regulatory functions in inflammatory disorders. The aim of this study was to investigate the role of CTSG in pancreatitis. Isolated acinar cells were exposed to recombinant CTSG and supramaximal cholezystokinin stimulation. In CTSG-/- mice and corresponding controls acute experimental pancreatitis was induced by serial caerulein injections. Severity was assessed by histology, serum enzyme levels and zymogen activation. Neutrophil infiltration was quantified by chloro-acetate ersterase staining and myeloperoxidase measurement. CTSG was expessed in inflammatory cells but not in pancreatic acinar cells. CTSG had no effect on intra-acinar-cell trypsinogen activation. In CTSG-/- mice a transient decrease of neutrophil infiltration into the pancreas and lungs was found during acute pancreatitis while the disease severity remained largely unchanged. CTSG is involved in pancreatic neutrophil infiltration during pancreatitis, albeit to a lesser degree than the related neutrophil (PMN) elastase. Its absence therefore leaves pancreatitis severity essentially unaffected.


Subject(s)
Acinar Cells/cytology , Cathepsin G/genetics , Ceruletide/adverse effects , Neutrophils/metabolism , Pancreatitis/immunology , Acinar Cells/drug effects , Acinar Cells/immunology , Animals , Cells, Cultured , Disease Models, Animal , Gene Knockout Techniques , Granulocytes/metabolism , Male , Mice , Neutrophil Infiltration , Pancreatitis/chemically induced , Pancreatitis/genetics , Trypsinogen/metabolism
5.
Gastroenterology ; 154(3): 704-718.e10, 2018 02.
Article in English | MEDLINE | ID: mdl-29079517

ABSTRACT

BACKGROUND & AIMS: Acute pancreatitis is characterized by premature intracellular activation of digestive proteases within pancreatic acini and a consecutive systemic inflammatory response. We investigated how these processes interact during severe pancreatitis in mice. METHODS: Pancreatitis was induced in C57Bl/6 wild-type (control), cathepsin B (CTSB)-knockout, and cathepsin L-knockout mice by partial pancreatic duct ligation with supramaximal caerulein injection, or by repetitive supramaximal caerulein injections alone. Immune cells that infiltrated the pancreas were characterized by immunofluorescence detection of Ly6g, CD206, and CD68. Macrophages were isolated from bone marrow and incubated with bovine trypsinogen or isolated acinar cells; the macrophages were then transferred into pancreatitis control or cathepsin-knockout mice. Activities of proteases and nuclear factor (NF)-κB were determined using fluorogenic substrates and trypsin activity was blocked by nafamostat. Cytokine levels were measured using a cytometric bead array. We performed immunohistochemical analyses to detect trypsinogen, CD206, and CD68 in human chronic pancreatitis (n = 13) and acute necrotizing pancreatitis (n = 15) specimens. RESULTS: Macrophages were the predominant immune cell population that migrated into the pancreas during induction of pancreatitis in control mice. CD68-positive macrophages were found to phagocytose acinar cell components, including zymogen-containing vesicles, in pancreata from mice with pancreatitis, as well as human necrotic pancreatic tissues. Trypsinogen became activated in macrophages cultured with purified trypsinogen or co-cultured with pancreatic acini and in pancreata of mice with pancreatitis; trypsinogen activation required macrophage endocytosis and expression and activity of CTSB, and was sensitive to pH. Activation of trypsinogen in macrophages resulted in translocation of NF-kB and production of inflammatory cytokines; mice without trypsinogen activation (CTSB-knockout mice) in macrophages developed less severe pancreatitis compared with control mice. Transfer of macrophage from control mice to CTSB-knockout mice increased the severity of pancreatitis. Inhibition of trypsin activity in macrophages prevented translocation of NF-κB and production of inflammatory cytokines. CONCLUSIONS: Studying pancreatitis in mice, we found activation of digestive proteases to occur not only in acinar cells but also in macrophages that infiltrate pancreatic tissue. Activation of the proteases in macrophage occurs during endocytosis of zymogen-containing vesicles, and depends on pH and CTSB. This process involves macrophage activation via NF-κB-translocation, and contributes to systemic inflammation and severity of pancreatitis.


Subject(s)
Cathepsin B/metabolism , Endocytosis , Macrophages/enzymology , Pancreas/enzymology , Pancreatitis, Acute Necrotizing/enzymology , Trypsinogen/metabolism , Adoptive Transfer , Animals , Cathepsin B/deficiency , Cathepsin B/genetics , Cathepsin L/deficiency , Cathepsin L/genetics , Cells, Cultured , Ceruletide , Coculture Techniques , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation , Genetic Predisposition to Disease , Humans , Hydrogen-Ion Concentration , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/pathology , Macrophages/transplantation , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Necrosis , Pancreas/immunology , Pancreas/pathology , Pancreatectomy , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/immunology , Pancreatitis, Acute Necrotizing/pathology , Phagocytosis , Phenotype , Severity of Illness Index , Time Factors
6.
Gut ; 65(11): 1838-1849, 2016 11.
Article in English | MEDLINE | ID: mdl-27196585

ABSTRACT

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies and is projected to be the second leading cause of cancer-related death by 2030. Despite extensive knowledge and insights into biological properties and genetic aberrations of PDAC, therapeutic options remain temporary and ineffective. One plausible explanation for the futile response to therapy is an insufficient and non-specific delivery of anticancer drugs to the tumour site. DESIGN: Superparamagnetic iron oxide nanoparticles (SPIONs) coupled with siRNA directed against the cell cycle-specific serine-threonine-kinase, Polo-like kinase-1 (siPLK1-StAv-SPIONs), could serve a dual purpose for delivery of siPLK1 to the tumour and for non-invasive assessment of efficiency of delivery in vivo by imaging the tumour response. siPLK1-StAv-SPIONs were designed and synthesised as theranostics to function via a membrane translocation peptide with added advantage of driving endosomal escape for mediating transportation to the cytoplasm (myristoylated polyarginine peptides) as well as a tumour-selective peptide (EPPT1) to increase intracellular delivery and tumour specificity, respectively. RESULTS: A syngeneic orthotopic as well as an endogenous cancer model was treated biweekly with siPLK1-StAv-SPIONs and tumour growth was monitored by small animal MRI. In vitro and in vivo experiments using a syngeneic orthotopic PDAC model as well as the endogenous LSL-KrasG12D, LSL-Trp53R172H, Pdx-1-Cre model revealed significant accumulation of siPLK1-StAv-SPIONs in PDAC, resulting in efficient PLK1 silencing. Tumour-specific silencing of PLK1 halted tumour growth, marked by a decrease in tumour cell proliferation and an increase in apoptosis. CONCLUSIONS: Our data suggest siPLK1-StAv-SPIONs with dual specificity residues for tumour targeting and membrane translocation to represent an exciting opportunity for targeted therapy in patients with PDAC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal , Cell Cycle Proteins , Magnetite Nanoparticles/therapeutic use , Pancreatic Neoplasms , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , RNA, Small Interfering , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Drug Delivery Systems/methods , Drug Monitoring/methods , Gene Silencing , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Polo-Like Kinase 1
7.
Gut ; 63(3): 494-505, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23564336

ABSTRACT

BACKGROUND: Autoimmune pancreatitis (AIP) in humans invariably responds to steroid treatment, but little is known about the underlying pathogenesis and the benefits of alternative treatments. OBJECTIVE: To study the pathogenesis, and the efficacy of alternative immunosuppressant agents in the MRL/Mp mouse model of AIP. DESIGN: MRL/Mp mice were pretreated for 4 weeks with polyinosinic:polycytidylic acid to induce AIP. Pancreatic sections of mice genetically deleted for CTLA-4 were analysed. Blockage of CTLA-4 was achieved by intraperitoneal antibody treatment with 2 µg/g anti-mouse-CD152. Subsequent therapeutic studies were performed for a period of 4 weeks using cyclosporine A (40 µg/g), rapamycin (1 µg/g) or azathioprine (15 µg/g). RESULTS: Blockage of CTLA-4 in MRL/Mp mice suppressed regulatory T cell (Treg) function and raised the effector T cell (Teff) response with subsequent histomorphological organ destruction, indicating that AIP is a T cell-driven disease. Using an established histopathological score, we found that dexamethasone, cyclosporine A and rapamycin, but less so azathioprine, reduced pancreatic damage. However, the beneficial effects of cyclosporine A and rapamycin were achieved via different mechanisms: cyclosporine A inhibited Teff activation and proliferation whereas rapamycin led to selective expansion of Tregs which subsequently suppressed the Teff response. CONCLUSIONS: The calcineurin inhibitor cyclosporine A and the mammalian target of rapamycin (mTOR) inhibitor, rapamycin, improve the course of AIP in MRL/Mp mice via different mechanisms. These findings further support the concept of autoreactive T cells as key players in the pathogenesis of AIP and suggest that cyclosporine A and rapamycin should be considered for treatment of AIP in humans.


Subject(s)
Autoimmune Diseases/drug therapy , Cyclosporine/therapeutic use , Immunosuppressive Agents/therapeutic use , Pancreas/immunology , Pancreatitis, Chronic/drug therapy , Sirolimus/therapeutic use , T-Lymphocyte Subsets/metabolism , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Azathioprine/therapeutic use , Biomarkers/metabolism , CTLA-4 Antigen/antagonists & inhibitors , Cell Proliferation/drug effects , Cyclosporine/pharmacology , Dexamethasone/therapeutic use , Drug Administration Schedule , Female , Flow Cytometry , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred Strains , Pancreas/drug effects , Pancreas/pathology , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/immunology , Pancreatitis, Chronic/pathology , Poly I-C , Random Allocation , Sirolimus/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Treatment Outcome
8.
J Gastroenterol Hepatol ; 27 Suppl 2: 47-51, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22320916

ABSTRACT

The incidence of acute pancreatitis per 100,000 of population ranges from 5 to 80. Patients suffering from hemorrhagic-necrotizing pancreatitis die in 10-24% of cases. 80% of all cases of acute pancreatitis are etiologically linked to gallstone disease immoderate alcohol consumption. As of today no specific causal treatment for acute pancreatitis exists. Elevated C-reactive protein levels above 130,mg/L can also predict a severe course of acute pancreatitis. The essential medical treatment for acute pancreatitis is the correction of hypovolemia. Prophylactic antibiotics should be restricted to patients with necrotizing pancreatitis, infected necrosis or other infectious complications. However, as premature intracellular protease activation is known to be the primary event in acute pancreatitis. Severe acute pancreatitis is characterized by an early inflammatory immune response syndrome (SIRS) and a subsequent compensatory anti-inflammatory response syndrome (CARS) contributing to severity as much as protease activation does. CARS suppresses the immune system and facilitates nosocomial infections including infected pancreatic necrosis, one of the most feared complications of the disease. A number of attempts have been made to suppress the early systemic inflammatory response but even if these mechanisms have been found to be beneficial in animal models they failed in daily clinical practice.


Subject(s)
Inflammation Mediators/metabolism , Pancreas/immunology , Pancreatitis/immunology , Pancreatitis/therapy , Acute Disease , Animals , Biomarkers/metabolism , Disease Progression , Humans , Pancreas/drug effects , Pancreas/pathology , Pancreatitis/diagnosis , Pancreatitis/pathology , Pancreatitis, Acute Necrotizing/immunology , Pancreatitis, Acute Necrotizing/therapy , Severity of Illness Index , Treatment Outcome
9.
Blood ; 117(16): 4349-57, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21224471

ABSTRACT

Emergency mobilization of neutrophil granulocytes (neutrophils) from the bone marrow (BM) is a key event of early cellular immunity. The hematopoietic cytokine granulocyte-colony stimulating factor (G-CSF) stimulates this process, but it is unknown how individual neutrophils respond in situ. We show by intravital 2-photon microscopy that a systemic dose of human clinical-grade G-CSF rapidly induces the motility and entry of neutrophils into blood vessels within the tibial BM of mice. Simultaneously, the neutrophil-attracting chemokine KC (Cxcl1) spikes in the blood. In mice lacking the KC receptor Cxcr2, G-CSF fails to mobilize neutrophils and antibody blockade of Cxcr2 inhibits the mobilization and induction of neutrophil motility in the BM. KC is expressed by megakaryocytes and endothelial cells in situ and is released in vitro by megakaryocytes isolated directly from BM. This production of KC is strongly increased by thrombopoietin (TPO). Systemic G-CSF rapidly induces the increased production of TPO in BM. Accordingly, a single injection of TPO mobilizes neutrophils with kinetics similar to G-CSF, and mice lacking the TPO receptor show impaired neutrophil mobilization after short-term G-CSF administration. Thus, a network of signaling molecules, chemokines, and cells controls neutrophil release from the BM, and their mobilization involves rapidly induced Cxcr2-mediated motility controlled by TPO as a pacemaker.


Subject(s)
Bone Marrow Cells/cytology , Granulocyte Colony-Stimulating Factor/immunology , Neutrophils/cytology , Receptors, Interleukin-8B/immunology , Thrombopoietin/immunology , Animals , Bone Marrow/immunology , Bone Marrow Cells/immunology , Bone and Bones/cytology , Cell Line , Cell Movement , Cells, Cultured , Humans , Megakaryocytes/cytology , Megakaryocytes/immunology , Mice , Mice, Inbred C57BL , Neutrophils/immunology
SELECTION OF CITATIONS
SEARCH DETAIL