Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Lancet Neurol ; 21(6): 528-536, 2022 06.
Article in English | MEDLINE | ID: mdl-35568047

ABSTRACT

BACKGROUND: Perinatal arterial ischaemic stroke (PAIS) is an important cause of neurodevelopmental disabilities. In this first-in-human study, we aimed to assess the feasibility and safety of intranasally delivered bone marrow-derived allogeneic mesenchymal stromal cells (MSCs) to treat PAIS in neonates. METHODS: In this open-label intervention study in collaboration with all neonatal intensive care units in the Netherlands, we included neonates born at full term (≥36 weeks of gestation) with MRI-confirmed PAIS in the middle cerebral artery region. All eligible patients were transferred to the neonatal intensive care unit of the Wilhelmina Children's Hospital. Neonates received one dose of 45-50 × 106 bone-marrow derived MSCs intranasally within 7 days of presenting signs of PAIS. The primary endpoints were acute and subacute safety outcomes, including vital signs, blood markers, and the occurrence of toxicity, adverse events, and serious adverse events. The occurrence of unexpected cerebral abnormalities by a repeat MRI at 3 months of age was a secondary endpoint. As part of standard clinical follow-up at Wilhelmina Children's Hospital, we assessed corticospinal tract development on MRI and performed motor assessments at 4 months of age. This study is registered with ClinicalTrials.gov, NCT03356821. FINDINGS: Between Feb 11, 2020, and April 29, 2021, ten neonates were enrolled in the study. Intranasal administration of MSCs was well tolerated in all ten neonates. No serious adverse events were observed. One adverse event was seen: a mild transient fever of 38°C without the need for clinical intervention. Blood inflammation markers (C-reactive protein, procalcitonin, and leukocyte count) were not significantly different pre-administration versus post-administration and, although thrombocyte levels increased (p=0·011), all were within the physiological range. Follow-up MRI scans did not show unexpected structural cerebral abnormalities. All ten patients had initial pre-Wallerian changes in the corticospinal tracts, but only four (40%) patients showed asymmetrical corticospinal tracts at follow-up MRI. Abnormal early motor assessment was found in three (30%) infants. INTERPRETATION: This first-in-human study demonstrates that intranasal bone marrow-derived MSC administration in neonates after PAIS is feasible and no serious adverse events were observed in patients followed up until 3 months of age. Future large-scale placebo-controlled studies are needed to determine the therapeutic effect of intranasal MSCs for PAIS. FUNDING: Netherlands Organization for Health Research and Development (ZonMw).


Subject(s)
Brain Ischemia , Ischemic Stroke , Mesenchymal Stem Cells , Stroke , Child , Feasibility Studies , Humans , Infant , Infant, Newborn , Netherlands , Research , Stroke/diagnostic imaging , Stroke/therapy , Treatment Outcome
2.
Front Immunol ; 11: 559152, 2020.
Article in English | MEDLINE | ID: mdl-33101274

ABSTRACT

Hematopoietic cell transplantation (HCT) is a last resort, potentially curative treatment option for pediatric patients with refractory acute myeloid leukemia (AML). Cord blood transplantation (CBT) results in less relapses and less graft-versus-host disease when compared to other sources. Nevertheless, still more than half of the children die from relapses. We therefore designed a strategy to prevent relapses by inducing anti-AML immunity after CBT, using a CB-derived dendritic cell (CBDC) vaccine generated from CD34+ CB cells from the same graft. We here describe the optimization and validation of good manufacturing practice (GMP)-grade production of the CBDC vaccine. We show the feasibility of expanding low amounts of CD34+ cells in a closed bag system to sufficient DCs per patient for at least three rounds of vaccinations. The CBDCs showed upregulated costimulatory molecules after maturation and showed enhanced CCR7-dependent migration toward CCL19 in a trans-well migrations assay. CBDCs expressed Wilms' tumor 1 (WT1) protein after electroporation with WT1-mRNA, but were not as potent as CBDCs loaded with synthetic long peptides (peptivator). The WT1-peptivator loaded CBDCs were able to stimulate T-cells both in a mixed lymphocyte reaction as well as in an antigen-specific (autologous) setting. The autologous stimulated T-cells lysed not only the WT1+ cell line, but most importantly, also primary pediatric AML cells. Altogether, we provide a GMP-protocol of a highly mature CBDC vaccine, loaded with WT1 peptivator and able to stimulate autologous T-cells in an antigen-specific manner. Finally, these T-cells lysed primary pediatric AML demonstrating the competence of the CBDC vaccine strategy.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fetal Blood/cytology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , WT1 Proteins/genetics , Antigen Presentation , Biomarkers , Cancer Vaccines/therapeutic use , Combined Modality Therapy , Cord Blood Stem Cell Transplantation , Cytotoxicity, Immunologic , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Prognosis , Recurrence , T-Cell Antigen Receptor Specificity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome , WT1 Proteins/immunology
4.
PLoS One ; 14(10): e0221924, 2019.
Article in English | MEDLINE | ID: mdl-31581194

ABSTRACT

BACKGROUND: The Pregnane X Receptor (PXR) is a principal signal transducer in mucosal responses to xenobiotic stress. It is well-recognized that inflammatory bowel disease is accompanied by xenobiotic stress, but the importance of the PXR in limiting inflammatory responses in inflammatory bowel disease remains obscure at best. METHODS: We stimulate a total of 106 colonic biopsies from 19 Crohn's disease patients with active disease, 36 colonic biopsies from 8 control patients, colonic organoids and various cell culture models (either proficient or genetically deficient with respect to PXR) in vitro with the PXR ligand rifampicin or vehicle. Effects on NF-κB activity are assessed by measuring interleukin-8 (IL-8) and interleukin-1ß (IL-1ß) mRNA levels by qPCR and in cell culture models by NF-κB reporter-driven luciferase activity and Western blot for signal transduction elements. RESULTS: We observe a strict inverse correlation between colonic epithelial PXR levels and NF-κB target gene expression in colonic biopsies from Crohn's disease patients. PXR, activated by rifampicin, is rate-limiting for mucosal NF-κB activation in IBD. The correlation between colonic epithelial PXR levels and NF-κB target gene expression was also observed in intestinal organoids system. Furthermore, in preclinical in vitro models of intestinal inflammation, including intestinal organoids, genetic inactivation of PXR unleashes NF-κB-dependent signal transduction whereas conversely NF-κB signaling reduces levels of PXR expression. CONCLUSIONS: Our data indicate that the PXR is a major and clinically relevant antagonist of NF-κB activity in the intestinal epithelial compartment during inflammatory bowel disease.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , NF-kappa B/metabolism , Pregnane X Receptor/metabolism , Biopsy , Cell Line, Tumor , Cytokines/metabolism , Humans , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Rifampin/pharmacology , Signal Transduction/drug effects
5.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(8): 1157-1167, 2019 08.
Article in English | MEDLINE | ID: mdl-31051284

ABSTRACT

BACKGROUND: Natural killer T (NKT) cells in adipose tissue (AT) contribute to whole body energy homeostasis. RESULTS: Inhibition of the glucosylceramide synthesis in adipocytes impairs iNKT cell activity. CONCLUSION: Glucosylceramide biosynthesis pathway is important for endogenous lipid antigen activation of iNKT cells in adipocytes. SIGNIFICANCE: Unraveling adipocyte-iNKT cell communication may help to fight obesity-induced AT dysfunction. Overproduction and/or accumulation of ceramide and ceramide metabolites, including glucosylceramides, can lead to insulin resistance. However, glucosylceramides also fulfill important physiological functions. They are presented by antigen presenting cells (APC) as endogenous lipid antigens via CD1d to activate a unique lymphocyte subspecies, the CD1d-restricted invariant (i) natural killer T (NKT) cells. Recently, adipocytes have emerged as lipid APC that can activate adipose tissue-resident iNKT cells and thereby contribute to whole body energy homeostasis. Here we investigate the role of the glucosylceramide biosynthesis pathway in the activation of iNKT cells by adipocytes. UDP-glucose ceramide glucosyltransferase (Ugcg), the first rate limiting step in the glucosylceramide biosynthesis pathway, was inhibited via chemical compounds and shRNA knockdown in vivo and in vitro. ß-1,4-Galactosyltransferase (B4Galt) 5 and 6, enzymes that convert glucosylceramides into potentially inactive lactosylceramides, were subjected to shRNA knock down. Subsequently, (pre)adipocyte cell lines were tested in co-culture experiments with iNKT cells (IFNγ and IL4 secretion). Inhibition of Ugcg activity shows that it regulates presentation of a considerable fraction of lipid self-antigens in adipocytes. Furthermore, reduced expression levels of either B4Galt5 or -6, indicate that B4Galt5 is dominant in the production of cellular lactosylceramides, but that inhibition of either enzyme results in increased iNKT cell activation. Additionally, in vivo inhibition of Ugcg by the aminosugar AMP-DNM results in decreased iNKT cell effector function in adipose tissue. Inhibition of endogenous glucosylceramide production results in decreased iNKT cells activity and cytokine production, underscoring the role of this biosynthetic pathway in lipid self-antigen presentation by adipocytes.


Subject(s)
Adipocytes/metabolism , Glucosylceramides/biosynthesis , Natural Killer T-Cells/metabolism , Adipocytes/cytology , Antigen Presentation , Cell Communication , Cell Line , Coculture Techniques , Cytokines/metabolism , Glucosylceramides/metabolism , Humans , Insulin Resistance , Lipids/immunology , Lymphocyte Activation , Natural Killer T-Cells/cytology
6.
Cancers (Basel) ; 11(2)2019 Feb 05.
Article in English | MEDLINE | ID: mdl-30764500

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells which instruct both the innate and adaptive immune systems. Once mature, they have the capacity to activate and prime naïve T cells for recognition and eradication of pathogens and tumor cells. These characteristics make them excellent candidates for vaccination strategies. Most DC vaccines have been generated from ex vivo culture of monocytes (mo). The use of mo-DCs as vaccines to induce adaptive immunity against cancer has resulted in clinical responses but, overall, treatment success is limited. The application of primary DCs or DCs generated from CD34⁺ stem cells have been suggested to improve clinical efficacy. Cord blood (CB) is a particularly rich source of CD34⁺ stem cells for the generation of DCs, but the dynamics and plasticity of the specific DC lineage development are poorly understood. Using flow sorting of DC progenitors from CB cultures and subsequent RNA sequencing, we found that CB-derived DCs (CB-DCs) exclusively originate from CD115⁺-expressing progenitors. Gene set enrichment analysis displayed an enriched conventional DC profile within the CD115-derived DCs compared with CB mo-DCs. Functional assays demonstrated that these DCs matured and migrated upon good manufacturing practice (GMP)-grade stimulation and possessed a high capacity to activate tumor-antigen-specific T cells. In this study, we developed a culture protocol to generate conventional DCs from CB-derived stem cells in sufficient numbers for vaccination strategies. The discovery of a committed DC precursor in CB-derived stem cell cultures further enables utilization of conventional DC-based vaccines to provide powerful antitumor activity and long-term memory immunity.

7.
PLoS One ; 12(8): e0182308, 2017.
Article in English | MEDLINE | ID: mdl-28767696

ABSTRACT

BACKGROUND: SH2 domain containing inositol-5-phosphatase (SHIP1) is an important modulator of innate and adaptive immunity. In mice, loss of SHIP1 provokes severe ileitis resembling Crohn's disease (CD), as a result of deregulated immune responses, altered cytokine production and intestinal fibrosis. Recently, SHIP1 activity was shown to be correlated to the presence of a CD-associated single nucleotide polymorphism in ATG16L1. Here, we studied SHIP1 activity and expression in an adult cohort of CD patients. METHODS: SHIP1 activity, protein and mRNA expression in peripheral blood mononuclear cells from CD patients in clinical remission were determined by Malachite green assay, Western blotting and qRT-PCR respectively. Genomic DNA was genotyped for ATG16L1 rs2241880. RESULTS: SHIP1 protein levels are profoundly diminished in a subset of patients; however, SHIP1 activity and expression are not correlated to ATG16L1 SNP status in this adult cohort. CONCLUSIONS: Aberrant SHIP1 activity can contribute to disease in a subset of adult CD patients, and warrants further investigation.


Subject(s)
Autophagy-Related Proteins/genetics , Crohn Disease/genetics , Down-Regulation , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Polymorphism, Single Nucleotide , Adult , Cell Line , Cohort Studies , Crohn Disease/metabolism , Female , Gene Expression Regulation , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Young Adult
8.
Clin Exp Gastroenterol ; 10: 83-90, 2017.
Article in English | MEDLINE | ID: mdl-28496351

ABSTRACT

BACKGROUND: Although it is well recognized that fatigue is an important problem in many of the quiescent inflammatory bowel disease (IBD) patients, it is unknown whether the immune status is different in fatigued versus non-fatigued patients. In this study, we contrasted various characteristics of the immune system in fatigued against non-fatigued patients with IBD in clinical remission. PATIENTS AND METHODS: Patients with IBD in clinical remission were phenotyped according to the Montreal classification, and the checklist individual strength-fatigue (CIS-fatigue) was used to assess fatigue (CIS-fatigue ≥ 35). Flow cytometry on peripheral blood samples was used to investigate differences in leukocyte subsets. The expression of various cytokines was determined in stimulated whole blood and serum samples using enzyme-linked immunosorbent assay. Differences between fatigued and non-fatigued patients with IBD were assessed. RESULTS: In total, 55 patients were included in the fatigue group (FG) and 29 patients in the non-fatigue group (NFG). No differences in demographic and clinical characteristics were observed between the groups. Flow cytometry data showed a significantly lower percentage of monocytes (p = 0.011) and a higher percentage of memory T-cells (p = 0.005) and neutrophils (p = 0.033) in the FG compared with the NFG. Whole blood stimulation showed increased TNF-α (p = 0.022) and IFN-γ (p = 0.047) in the FG. The median serum level was significantly higher for IL-12 (p < 0.001) and IL-10 (p = 0.005) and lower for IL-6 (p = 0.002) in the FG compared with NFG. CONCLUSION: Significant differences in immune profile between fatigued and non-fatigued patients with IBD in clinical remission were found, which point out to a chronically active and Th1-skewed immune system in patients with fatigue. Whether these immune differences are directly involved in the fatigue complaints via immune-to-brain communication pathways remains to be determined. As such, further exploration of the underlying immune effects associated with fatigue is warranted to determine potential treatment options.

9.
Oncoimmunology ; 4(11): e1023973, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26451309

ABSTRACT

The poor survival rates of refractory/relapsed acute myeloid leukemia (AML) patients after haematopoietic cell transplantation (HCT) requires the development of additional immune therapeutic strategies. As the elicitation of tumor-antigen specific cytotoxic T lymphocytes (CTLs) is associated with reduced relapses and enhanced survival, enhanced priming of these CTLs using an anti-AML vaccine may result in long-term immunity against AML. Cord blood (CB), as allogeneic HCT source, may provide a unique setting for such post-HCT vaccination, considering its enhanced graft-versus-leukemia (GvL) effects and population of highly responsive naïve T cells. It is our goal to develop a powerful and safe immune therapeutic strategy composed of CB-HCT followed by vaccination with CB CD34+-derived dendritic cells (DCs) presenting the oncoprotein Wilms Tumor-1 (WT1), which is expressed in AML-blasts in the majority of patients. Here, we describe the optimization of a clinically applicable DC culture protocol. This two-step protocol consisting of an expansion phase followed by the differentiation toward DCs, enables us to generate sufficient cord blood-derived DCs (CBDCs) in the clinical setting. At the end of the culture, the CBDCs exhibit a mature surface phenotype, are able to migrate, express tumor antigen (WT1) after electroporation with mRNA encoding the full-length WT1 protein, and stimulate WT1-specific T cells.

10.
Front Immunol ; 5: 218, 2014.
Article in English | MEDLINE | ID: mdl-24904573

ABSTRACT

Hematopoietic cell transplantation (HCT) is a last treatment resort and only potentially curative treatment option for several hematological malignancies resistant to chemotherapy. The induction of profound immune regulation after allogeneic HCT is imperative to prevent graft-versus-host reactions and, at the same time, allow protective immune responses against pathogens and against tumor cells. Dendritic cells (DCs) are highly specialized antigen-presenting cells that are essential in regulating this balance and are of major interest as a tool to modulate immune responses in the complex and challenging phase of immune reconstitution early after allo-HCT. This review focuses on the use of DC vaccination to prevent cancer relapses early after allo-HCT. It describes the role of host and donor-DCs, various vaccination strategies, different DC subsets, antigen loading, DC maturation/activation, and injection sites and dose. At last, clinical trials using DC vaccination post-allo-HCT and the future perspectives of DC vaccination in combination with other cancer immunotherapies are discussed.

11.
J Biol Chem ; 289(32): 22128-39, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24966328

ABSTRACT

Obesity-induced adipose tissue (AT) dysfunction results in a chronic low-grade inflammation that predisposes to the development of insulin resistance and type 2 diabetes. During the development of obesity, the AT-resident immune cell profile alters to create a pro-inflammatory state. Very recently, CD1d-restricted invariant (i) natural killer T (NKT) cells, a unique subset of lymphocytes that are reactive to so called lipid antigens, were implicated in AT homeostasis. Interestingly, recent data also suggest that human and mouse adipocytes can present such lipid antigens to iNKT cells in a CD1d-dependent fashion, but little is known about the lipid antigen presentation machinery in adipocytes. Here we show that CD1d, as well as the lipid antigen loading machinery genes pro-saposin (Psap), Niemann Pick type C2 (Npc2), α-galactosidase (Gla), are up-regulated in early adipogenesis, and are transcriptionally controlled by CCAAT/enhancer-binding protein (C/EBP)-ß and -δ. Moreover, adipocyte-induced Th1 and Th2 cytokine release by iNKT cells also occurred in the absence of exogenous ligands, suggesting the display of endogenous lipid antigen-D1d complexes by 3T3-L1 adipocytes. Furthermore, we identified microsomal triglyceride transfer protein, which we show is also under the transcriptional regulation of C/EBPß and -δ, as a novel player in the presentation of endogenous lipid antigens by adipocytes. Overall, our findings indicate that adipocytes can function as non-professional lipid antigen presenting cells, which may present an important aspect of adipocyte-immune cell communication in the regulation of whole body energy metabolism and immune homeostasis.


Subject(s)
Adipocytes/immunology , Adipocytes/metabolism , Antigen Presentation , Antigens, CD1d/metabolism , Carrier Proteins/metabolism , Lipids/immunology , 3T3-L1 Cells , Adipogenesis/genetics , Adipogenesis/immunology , Animals , Antigens, CD1d/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-delta/genetics , CCAAT-Enhancer-Binding Protein-delta/metabolism , Carrier Proteins/genetics , Cell Communication/immunology , Energy Metabolism , Gene Expression , Gene Knockdown Techniques , Humans , Mice , Natural Killer T-Cells/immunology , Transcription, Genetic
12.
Sci Transl Med ; 6(233): 233ra53, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24760188

ABSTRACT

In inflammatory bowel disease (IBD), large areas of apparently healthy mucosa lie adjacent to ulcerated intestine. Knowledge of the mechanisms that maintain remission in an otherwise inflamed intestine could provide important clues to the pathogenesis of this disease and provide rationale for clinical treatment strategies. We used kinome profiling to generate comprehensive descriptions of signal transduction pathways in inflamed and noninflamed colonic mucosa in a cohort of IBD patients, and compared the results to non-IBD controls. We observed that p21Rac1 guanosine triphosphatase (GTPase) signaling was strongly suppressed in noninflamed colonic mucosa in IBD. This suppression was due to both reduced guanine nucleotide exchange factor activity and increased intrinsic GTPase activity. Pharmacological p21Rac1 inhibition correlated with clinical improvement in IBD, and mechanistically unrelated pharmacological p21Rac1 inhibitors increased innate immune functions such as phagocytosis, bacterial killing, and interleukin-8 production in healthy controls and patients. Thus, suppression of p21Rac activity assists innate immunity in bactericidal activity and may induce remission in IBD.


Subject(s)
Crohn Disease/immunology , Crohn Disease/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Immunity, Innate , Signal Transduction , rac1 GTP-Binding Protein/metabolism , Animals , Biopsy , Crohn Disease/pathology , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Down-Regulation , Enzyme Inhibitors/pharmacology , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Inflammation/pathology , Intestinal Mucosa/pathology , Protein Kinases/metabolism , Remission Induction , Thioguanine/pharmacology , rac1 GTP-Binding Protein/antagonists & inhibitors
13.
Gut ; 63(7): 1081-91, 2014 Jul.
Article in English | MEDLINE | ID: mdl-23964099

ABSTRACT

OBJECTIVE: Although genome wide association studies have partly uncovered the genetic basis of Crohn's disease (CD), it remains a challenge to link genetic polymorphisms to functional intestinal phenotypes. Paneth cells are specialised antimicrobial epithelial cells localised to the small-intestinal crypt-base. Here, we investigate whether genomic variations in ATG16L1 affect Paneth cell function. DESIGN: Genomic variation of ATG16L1 (T300A, rs2241880) was determined in DNA from 78 patients with CD and 12 healthy controls. Paraffin-embedded ileal biopsies from patients with genotype AA (n=17), GA (n=38) and patients with the GG allele (n=23) were stained for GRP78, phospho-EIF2α, lysozyme, cleaved-caspase 3, phosphohistone H3, phospho-IκB, p65, phospho-p38MAPK and PHLDA1. Microbial composition of biopsies was assessed by PCR. Disease phenotype was scored. RESULTS: In patients with quiescent disease but with an ATG16L1 risk allele, the endoplasmic reticulum (ER) stress markers GRP78 and pEIF2α were highly expressed in Paneth cells. Other CD risk gene variations did not correlate with Paneth cell ER stress. Functionally, patients with ER-stressed Paneth cells showed no changes in intestinal epithelial cells proliferation or apoptosis, Paneth cell or stem cell numbers, p65, phospho-IκB and phospho-p38 staining. However, a significantly increased presence of adherent-invasive Escherichia coli was observed in biopsies from patients with ER-stressed Paneth cells. Phenotypically, patients with GRP78 positive Paneth cells have relatively less colonic disease over ileal disease (-21%, p=0.04), more fistulas (+21%, p=0.05) and an increased need for intestinal surgery (+38%, p=0.002). CONCLUSIONS: The ATG16L1 T300A polymorphism defines a specific subtype of patients with CD, characterised by Paneth cell ER stress even during quiescent disease. Paneth cell ER stress correlates with bacterial persistence, and is thus likely to modulate antimicrobial functionality of this cell type in patients with CD.


Subject(s)
Carrier Proteins/genetics , Crohn Disease/genetics , Endoplasmic Reticulum Stress/genetics , Genetic Predisposition to Disease , Paneth Cells/metabolism , Polymorphism, Single Nucleotide , Autophagy-Related Proteins , Biopsy , Case-Control Studies , Cells, Cultured , Crohn Disease/metabolism , Crohn Disease/microbiology , Crohn Disease/pathology , DNA, Bacterial/analysis , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/physiology , Escherichia coli/genetics , Escherichia coli/isolation & purification , Genetic Markers , Genotyping Techniques , Humans , Ileum/metabolism , Ileum/microbiology , Ileum/pathology , Microbiota , Multivariate Analysis , Paneth Cells/microbiology , Paneth Cells/pathology , Phenotype , Risk Factors
14.
Int J Biochem Cell Biol ; 45(4): 798-806, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23291352

ABSTRACT

The epithelial layer of our intestines must meet two opposing requirements. On one hand it must allow for efficient uptake of nutrients and fluids, on the other hand it is a vital defence barrier between the milieu interior and the milieu exterior. In contrast to the lung that by virtue of cilia movement is kept virtually sterile, the gut epithelium is confronted by a stupendous microbiological load and a substantial xenobiotic challenge. The efficiency by which our intestinal epithelium manages to deal with the challenge of efficient nutrient absorption while simultaneously fulfilling its barrier function is testimony to what the forces of evolution can accomplish. Importantly, our understanding as to how our gut epithelial compartment manages this balancing act is now rapidly emerging, answering one of the oldest questions in cell biology. Importantly, when aberrations in this balance occur, for instance as a consequence genetic polymorphisms, increased propensity to develop chronic inflammation and inflammatory bowel disease is the result. Thus the knowledge on intestinal cell biology and biochemistry is not only of academic interest but may also aid design of novel avenues for the rational treatment of mucosal disease.


Subject(s)
Cell Biology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Animals , Environment , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Metagenome
15.
Inflamm Bowel Dis ; 19(1): 132-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22535619

ABSTRACT

BACKGROUND: Most biomarkers predicting mucosal relapse of ulcerative colitis (UC) patients in clinical remission represent low levels of mucosal inflammation. Since SOCS3 expression may increase the vulnerability of intestinal epithelial cells (IECs) to various insults, we investigated whether its expression predicts mucosal relapse in UC patients in clinical remission without any signs of mucosal inflammation. METHODS: UC patients (n = 32) in clinical, endoscopic, and histological remission were followed up for 9 years. IEC expression of SOCS3, p-STAT3, and p-STAT1 were assessed with biopsies from the baseline colonoscopy, last colonoscopy before relapse, and colonoscopy at relapse. Clinical data, endoscopy, and histology reports were collected from patient charts. RESULTS: Twenty-six (81%) patients had histological relapse, 19 (59%) developed an endoscopic relapse, and 17 (53%) had a clinical relapse during follow-up. SOCS3 expression at first colonoscopy during remission correlated with shorter time to histological, endoscopic, and clinical relapse. SOCS3 expression was increased at the last colonoscopy before relapse, approaching relapse levels, whereas p-STAT3 expression was low during the entire remission. A positive correlation between IEC SOCS3 and its inducer p-STAT1 was shown. CONCLUSIONS: SOCS3 IEC expression during remission may be useful in predicting mucosal relapse in patients without any signs of mucosal inflammation. These data strengthen our hypothesis that SOCS3 contributes to enhanced vulnerability of IEC during remission. Thus, SOCS3 levels during remission may function as a therapeutic target for clinical monitoring and early induction of mucosal healing.


Subject(s)
Biomarkers/analysis , Colitis, Ulcerative/complications , Intestinal Mucosa/pathology , Mucositis/diagnosis , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Adult , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/therapy , Colonoscopy , Endoscopy, Gastrointestinal , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Intestinal Mucosa/metabolism , Male , Middle Aged , Mucositis/etiology , Mucositis/metabolism , Phosphorylation , Prognosis , Recurrence , Remission Induction , Suppressor of Cytokine Signaling 3 Protein , Young Adult
16.
Mol Med ; 18: 1412-9, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23154639

ABSTRACT

Ulcerative colitis (UC) is a chronic disease associated with long periods of quiescent disease followed by fulminant exacerbation. Imminent relapse in UC is associated with high mucosal expression of suppressor of cytokine signaling 3 (SOCS3); hence, knowledge of the mechanisms driving mucosal SOCS3 expression may provide important clues as to rational therapy. Thus, here we aim to characterize the molecular forces driving SOCS3 expression in the mucosal compartment, focusing on druggable pathways. The colon epithelial cell line Caco-2 was stimulated with interferon (IFN)-γ, interleukin (IL)-4 or prostaglandin E(2) (PGE(2)) to allow correlations between SOCS3 expression with signal transducer and activator of transcription 1 (STAT1), STAT6 and adenosine 3',5'-cyclic monophosphate (cAMP) signaling, respectively. The physiological relevance of the findings obtained was assessed by immunohistochemical staining for the activated forms of STAT1, STAT6, protein kinase A (PKA)-Cγ and cAMP response element-binding protein (CREB) in biopsies from inactive UC patients and controls. Stimulation with IFN-γ, IL-4 or PGE(2) induced activation of STAT1, STAT6 and cAMP, respectively, in colonic cells, without any signs of concomitant STAT3 activation. Forced activation of all these signaling pathways was sufficient for SOCS3 expression. Biopsies from patients with inactive UC showed significant increase of phosphorylated STAT1 (p-STAT1) (p < 0.0001), p-STAT6 (p = 0.0001), p-PKA-Cγ (p = 0.0003) and p-CREB (p = 0.0025) expression compared with controls. STAT3-independent SOCS3 induction in inactive UC involves multiple proinflammatory signaling pathways and contradicts the usefulness of pathway-specific antiinflammatory drugs for preventing relapse. Our findings suggest that broad-spectrum antiinflammatory drugs are essential to counteract increases in SOCS3 expression and exacerbation of disease. Our results highlight the multifactorial nature of the factors that cause exacerbation in UC.


Subject(s)
Colitis, Ulcerative/metabolism , Cyclic AMP/metabolism , STAT1 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , Adult , Caco-2 Cells , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/pathology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Cytokines/pharmacology , Enterocytes/drug effects , Enterocytes/enzymology , Enterocytes/pathology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein
17.
Eur J Immunol ; 42(12): 3150-66, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22936024

ABSTRACT

Dendritic cells (DCs) and monocyte-derived macrophages (MΦs) are key components of intestinal immunity. However, the lack of surface markers differentiating MΦs from DCs has hampered understanding of their respective functions. Here, we demonstrate that, using CD64 expression, MΦs can be distinguished from DCs in the intestine of both mice and humans. On that basis, we revisit the phenotype of intestinal DCs in the absence of contaminating MΦs and we delineate a developmental pathway in the healthy intestine that leads from newly extravasated Ly-6C(hi) monocytes to intestinal MΦs. We determine how inflammation impacts this pathway and show that T cell-mediated colitis is associated with massive recruitment of monocytes to the intestine and the mesenteric lymph node (MLN). There, these monocytes differentiate into inflammatory MΦs endowed with phagocytic activity and the ability to produce inducible nitric oxide synthase. In the MLNs, inflammatory MΦs are located in the T-cell zone and trigger the induction of proinflammatory T cells. Finally, T cell-mediated colitis develops irrespective of intestinal DC migration, an unexpected finding supporting an important role for MLN-resident inflammatory MΦs in the etiology of T cell-mediated colitis.


Subject(s)
Colitis/immunology , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Lymph Nodes/immunology , Macrophages/immunology , Mesentery/immunology , Receptors, IgG/immunology , Th1 Cells/immunology , Animals , Antigens, Ly/immunology , Cell Differentiation/immunology , Colitis/pathology , Dendritic Cells/pathology , Humans , Immunity, Mucosal , Intestinal Mucosa/pathology , Lymph Nodes/pathology , Macrophages/pathology , Mesentery/pathology , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Th1 Cells/pathology
18.
Carcinogenesis ; 33(10): 1889-96, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22739025

ABSTRACT

Ulcerative colitis (UC) is associated with a high risk of developing colorectal cancer (CRC). The mechanisms by which chronic inflammatory responses in the colon may promote CRC remain only partially understood, but may involve reduced negative regulation of interleukin (IL)-6 signaling towards signal transducer and activator of transcription 3 activation through the loss of SOCS3 expression, unleashing the full carcinogenic potential of this transcription factor. Thus, we analyzed SOCS3 expression in the colon of healthy controls, as well as in a cohort of UC patients with varying degrees of dysplasia. We observe that the loss of epithelial SOCS3 expression delimits the areas subject to dysplasia in UC, suggesting an important tumour-suppressive role of SOCS3 downregulation, early in the transformation process. Importantly, methylation of the SOCS3 promotor appears to constitute an important regulatory mechanism for colonic SOCS3 expression as SOCS3 methylation status in CRC cells correlates with a disability to upregulate SOCS3 upon IL-6 stimulation, whereas forced demethylation using 5-aza-2'-deoxycytidine restores SOCS3 expression and inhibits IL-6-induced p-signal transducer and activator of transcription 3 activation and proliferation. Expression of the DNA methyltransferase gene DMNT1 is prominent in dysplastic cells and correlates with low or absent SOCS3 expression. Thus, induction of DNMT1 expression in the chronically inflamed colon may release IL-6 signaling towards signal transducer and activator of transcription 3 from inhibition through SOCS3 increasing the propensity to malignant transformation. Hence, DNMT1 emerges as a rational target in preventive strategies aimed at counteracting UC-CRC.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Colitis, Ulcerative/metabolism , Colorectal Neoplasms/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Interleukin-6/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Colitis, Ulcerative/pathology , Colon/metabolism , Colorectal Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Gene Expression Regulation, Neoplastic , Humans , Intestinal Mucosa/metabolism , Male , Suppressor of Cytokine Signaling 3 Protein
19.
Cytokine Growth Factor Rev ; 23(3): 127-38, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22591635

ABSTRACT

Inflammatory bowel disease (IBD) has unclear pathogenesis and it is related to the increasing risk of developing colorectal cancer (CRC). Recent studies have uncovered the molecular mechanism of intracellular signaling pathways of inflammatory cytokines such as tumor necrosis factor (TNF)-α, interferon (IFN)-γ and interleukin (IL)-6. The major transcription factors including STAT3 have been shown to play a major role in transmitting inflammatory cytokine signals to the nucleus. The suppressors of cytokine signaling (SOCS) 3 protein is the key physiological regulators of cytokine-mediated STAT3 signaling. As such it influences the development of inflammatory and malignant disorders like this associated with IBD. Here we review the complex function of SOCS3 in innate and adaptive immunity, different cell types (macrophages, neutrophils, dendritic cells, B cells, T cells and intestinal epithelial cells) and the role of SOCS3 on the pathogenesis of inflammatory bowel disease (IBD) and IBD-related cancer. Finally, we explore how this knowledge may open novel avenues for the rational treatment of IBD and IBD-related cancer.


Subject(s)
Colorectal Neoplasms/etiology , Inflammatory Bowel Diseases/immunology , Suppressor of Cytokine Signaling Proteins/physiology , Adaptive Immunity/physiology , B-Lymphocytes/immunology , Dendritic Cells/immunology , Humans , Immunity, Innate/physiology , Inflammation/complications , Inflammatory Bowel Diseases/complications , Macrophages/immunology , Neutrophils/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Suppressor of Cytokine Signaling 3 Protein , T-Lymphocytes/immunology
20.
J Crohns Colitis ; 6(3): 373-85, 2012 04.
Article in English | MEDLINE | ID: mdl-22405177

ABSTRACT

The second scientific workshop of the European Crohn's and Colitis Organization (ECCO) focused on the relevance of intestinal healing for the disease course of inflammatory bowel disease (IBD). The objective was to better understand basic mechanisms, markers for disease prediction, detection and monitoring of intestinal healing, impact of intestinal healing on the disease course of IBD as well as therapeutic strategies. The results of this workshop are presented in four separate manuscripts. This section describes basic mechanisms of intestinal healing, identifies open questions in the field and provides a framework for future studies.


Subject(s)
Colitis, Ulcerative/physiopathology , Crohn Disease/physiopathology , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Wound Healing/drug effects , Wound Healing/physiology , Adrenal Cortex Hormones/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibodies, Monoclonal/pharmacology , Azathioprine/pharmacology , Butyric Acid/pharmacology , Colitis, Ulcerative/genetics , Crohn Disease/genetics , Cyclosporine/pharmacology , Defensins/physiology , Extracellular Matrix/physiology , Humans , Immunosuppressive Agents/pharmacology , Infliximab , Leptin/physiology , Matrix Metalloproteinases/physiology , Mesalamine/pharmacology , Paneth Cells/physiology , Probiotics/pharmacology , Reactive Nitrogen Species/physiology , Reactive Oxygen Species/metabolism , Sulfasalazine/pharmacology , Tumor Necrosis Factor-alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...