Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Immunohorizons ; 7(12): 872-885, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38147032

ABSTRACT

Our bodies are home to individual-specific microbial ecosystems that have recently been found to be modified by cancer immunotherapies. The interaction between the gut microbiome and islet autoimmunity leading to type I diabetes (T1D) is well described and highlights the microbiome contribution during the onset and T1D development in animals and humans. As cancer immunotherapies induce gut microbiome perturbations and immune-mediated adverse events in susceptible patients, we hypothesized that NOD mice can be used as a predictive tool to investigate the effects of anti-PD-1 treatment on the onset and severity of T1D, and how microbiota influences immunopathology. In this longitudinal study, we showed that anti-PD-1 accelerated T1D onset, increased glutamic acid decarboxylase-reactive T cell frequency in spleen, and precipitated destruction of ß cells, triggering high glucose levels and pancreatic islet reduction. Anti-PD-1 treatment also resulted in temporal microbiota changes and lower diversity characteristic of T1D. Finally, we identified known insulin-resistance regulating bacteria that were negatively correlated with glucose levels, indicating that anti-PD-1 treatment impacts the early gut microbiota composition. Moreover, an increase of mucin-degrading Akkermansia muciniphila points to alterations of barrier function and immune system activation. These results highlight the ability of microbiota to readily respond to therapy-triggered pathophysiological changes as rescuers (Bacteroides acidifaciens and Parabacteroides goldsteinii) or potential exacerbators (A. muciniphila). Microbiome-modulating interventions may thus be promising mitigation strategies for immunotherapies with high risk of immune-mediated adverse events.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Immune Checkpoint Inhibitors , Animals , Humans , Mice , Glucose , Longitudinal Studies , Mice, Inbred NOD , Neoplasms , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/pharmacology
2.
Int J Cancer ; 145(3): 678-685, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30653264

ABSTRACT

Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths worldwide and the need for novel biomarkers and therapeutic strategies to improve diagnosis and surveillance is obvious. This study aims to identify ß6 -integrin (ITGB6) as a novel serum tumor marker for diagnosis, prognosis, and surveillance of CRC. ITGB6 serum levels were validated in retro- and prospective CRC patient cohorts. ITGB6 serum levels were analyzed by ELISA. Using an initial cohort of 60 CRC patients, we found that ITGB6 is present in the serum of CRC, but not in non-CRC control patients. A cut-off of ≥2 ng/mL ITGB6 reveals 100% specificity for the presence of metastatic CRC. In an enlarged study cohort of 269 CRC patients, ITGB6 predicted the onset of metastatic disease and was associated with poor prognosis. Those data were confirmed in an independent, prospective cohort consisting of 40 CRC patients. To investigate whether ITGB6 can also be used for tumor surveillance, serum ITGB6-levels were assessed in 26 CRC patients, pre- and post-surgery, as well as during follow-up visits. After complete tumor resection, ITGB6 serum levels declined completely. During follow-up, a new rise in ITGB6 serum levels indicated tumor recurrence or the onset of new metastasis as confirmed by CT scan. ITGB6 was more accurate for prognosis of advanced CRC and for tumor surveillance as the established marker carcinoembryonic antigen (CEA). Our findings identify ITGB6 as a novel serum marker for diagnosis, prognosis, and surveillance of advanced CRC. This might essentially contribute to an optimized patient care.


Subject(s)
Colorectal Neoplasms/blood , Integrin beta Chains/blood , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Case-Control Studies , Colorectal Neoplasms/genetics , Humans , Integrin beta Chains/biosynthesis , Integrin beta Chains/genetics , Prognosis , Proof of Concept Study , Proportional Hazards Models , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reproducibility of Results
3.
Int J Antimicrob Agents ; 50(3): 342-351, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28689869

ABSTRACT

Inflammatory bowel disease (IBD) may develop due to an immunogenic response to commensal gut microbiota triggered by environmental factors in the genetically susceptible host. Isotretinoin, applied in the treatment of severe acne, has been variably associated with IBD, but prior treatment with antibiotics, also associated with IBD development, confounds confirmation of this association. This study investigated the effects of doxycycline, metronidazole (frequently used in the treatment of acne and IBD, respectively) and isotretinoin on murine gut (faecal) microbiota after 2 weeks of treatment and after a 4-week recovery period. Faecal microbiota composition was assessed by 16S rRNA gene sequencing on the GS-FLX 454 platform with primers directed against the variable regions V1-V2. Doxycycline had a modest effect on bacterial richness and evenness, but had pronounced persistent and significant effects on the abundance of certain operational taxonomic units compared with the control group. In contrast, metronidazole induced a pronounced reduction in diversity after treatment, but these effects did not persist after the recovery period. This study demonstrates differential effects of antibiotics on the gut microbiota with doxycycline, unlike metronidazole, mediating long-term changes in the murine gut microbiota. Isotretinoin had no significant effect on the faecal microbiota.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Dermatologic Agents/administration & dosage , Doxycycline/administration & dosage , Gastrointestinal Microbiome/drug effects , Isotretinoin/administration & dosage , Metronidazole/administration & dosage , Animals , Cluster Analysis , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Female , Mice, Inbred BALB C , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
4.
Sci Rep ; 6: 37082, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27853192

ABSTRACT

Inflammatory bowel disease (IBD) may develop due to an inflammatory response to commensal gut microbiota triggered by environmental factors in a genetically susceptible host. Isotretinoin (acne therapy) has been inconsistently associated with IBD onset and flares but prior treatment with antibiotics, also associated with IBD development, complicates the confirmation of this association. Here we studied in mice whether doxycycline, metronidazole or isotretinoin induce epigenetic modifications, and consequently change T-cell mRNA expression and/or function directly after treatment and after a 4 week recovery period. Isotretinoin induced IL-10 signaling in Tregs and naive T-cells directly after treatment and reduced effector T-cell proliferation alone and in co-culture with Tregs. Metronidazole activated processes associated with anti-inflammatory pathways in both T-cell subsets directly after the treatment period whereas doxycycline induced an immediate pro-inflammatory expression profile that resolved after the recovery period. Long-term changes indicated an inhibition of proliferation by doxycycline and induction of beneficial immune and metabolic pathways by metronidazole. Persistent alterations in microRNA and mRNA expression profiles after the recovery period indicate that all three medications may induce long-term epigenetic modifications in both T-cell subsets. Yet, our data do not support the induction of a long-term pro-inflammatory phenotype in murine Tregs and naive T-cells.


Subject(s)
Doxycycline/pharmacology , Epigenesis, Genetic/drug effects , Isotretinoin/pharmacology , Metronidazole/pharmacology , MicroRNAs/biosynthesis , RNA, Messenger/biosynthesis , T-Lymphocytes, Regulatory/metabolism , Animals , Female , Gene Expression Profiling , Mice , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/cytology
5.
Inflamm Bowel Dis ; 19(11): 2356-65, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23899542

ABSTRACT

BACKGROUND: In vitro and in vivo data have shown that retinoid treatment promotes an anti-inflammatory milieu with few adverse effects toward the gastrointestinal tract. The in vivo studies reported here further evaluate retinoid effects in 2 mouse models of inflammatory bowel disease. METHOD: Chronic dextran sulfate sodium colitis was induced in age- and weight-matched C57Bl/6 mice by 4 cycles of dextran sulfate sodium administration (6-8 animals/group). At cycle 4, animals were administered 13-cis-retinoic acid (isotretinoin, 30 mg/kg) or vehicle (oral gavage) or 4-oxo-13-cis-retinoic acid (15 mg/kg, intraperitoneal) daily. T-cell transfer colitis was induced in CB17 SCID mice by transfer of naive CD4CD62L T cells and treated by transfer of regulatory CD4CD25 T cells (4-6 animals/group); isolated from BALB/c mice after treatment with isotretinoin or vehicle, as above, for 2 weeks. Assessments included endoscopic and histological scores, myeloperoxidase activity, serum cytokines, and plasma isotretinoin levels. RESULTS: Retinoid-treated animals with colitis showed comparable changes in myeloperoxidase activity, and endoscopic and histological scores, versus untreated animals with colitis. Modest and comparable changes were seen in body weight and colon length in animals injected with naive T cells from isotretinoin-treated donors versus those injected with T cells from vehicle-treated donors. Retinoid treatment was consistently associated with lower interleukin-12 levels, which, after the transfer of naive T cells from isotretinoin-treated donors, supported isotretinoin-mediated predisposition of naive T cells toward reduced proinflammatory cytokine expression. Colitis had no effect on isotretinoin exposure. CONCLUSIONS: Retinoids attenuate the proinflammatory cytokine response in vivo, with only modest effects on body weight and parameters of gastrointestinal morphology.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Colitis/drug therapy , Disease Models, Animal , Gastrointestinal Tract/drug effects , Isotretinoin/pharmacology , T-Lymphocytes, Regulatory/drug effects , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Colitis/chemically induced , Colitis/immunology , Cytokines/metabolism , Dermatologic Agents/pharmacology , Dextran Sulfate/toxicity , Female , Flow Cytometry , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Peroxidase/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
6.
Arch Toxicol ; 87(4): 633-43, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23135549

ABSTRACT

Cadmium (Cd) affects the expression of estrogen receptor (ER) and aryl hydrocarbon receptor (AhR)-associated genes in rat uterus and elicits estrogen-like activity in vitro. The small intestine is highly exposed to dietary Cd which may mimic or antagonize estrogen action in this tissue. We investigated the effects of Cd and 17-alpha-ethinylestradiol (EE2) on AhR-associated gene expression after oral exposure of ovariectomized female Wistar rats, and metallothionein (Mt1a) expression as a typical metal-response marker. Mt1a in the small intestine was strongly induced by co-treatment with CdCl2 at 2 mg/kg b.wt (Cd 2) and 0.1 mg/kg b.wt EE2 than by the single compound (3-day gavage). The Cd 2-induced down-regulation of Cyp1a1, Gsta2, and Nqo1 mRNA was not antagonized by pure anti-estrogen (2.5 mg/kg b.wt ZK191703 s.c., ZK). Interestingly, the EE2-induced down-regulation of Cyp1a1, Gsta2, and Nqo1 mRNA was antagonized by Cd 2 in vivo and in colon cancer cell lines (HT-29 and CaCo-2, treated 5 days with Cd 1 µM and/or E2 0.01 µM) with low or no ER-beta expression. Dose dependency was studied after Cd exposure with drinking water (5 and 50 ppm CdCl2 equivalent to 0.4 and 4 mg/kg b.wt; Cd 0.4, Cd 4) for 28 days and EE2 as reference. Intestinal Mt1a expression was dose dependently induced, while AhR target genes were down-regulated by Cd 0.4 similar to EE2 and more pronounced than by Cd 4. We propose that Cd modulates intestinal AhR-associated gene expression similar to estrogens, but (contrary to its effects in uterus) via ER-independent and/or ER-beta-mediated mechanisms. Our new data suggest interference of Cd with estrogen and AhR signaling in the small intestine.


Subject(s)
Cadmium Chloride/toxicity , Estrogens/pharmacology , Ethinyl Estradiol/pharmacology , Gene Expression Regulation/drug effects , Intestine, Small/drug effects , Receptors, Aryl Hydrocarbon/genetics , Animals , Caco-2 Cells , Cell Survival/drug effects , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Dose-Response Relationship, Drug , Drug Therapy, Combination , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Female , Fluorocarbons , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Intestine, Small/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Metallothionein/genetics , Metallothionein/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Organ Size/drug effects , Ovariectomy , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Aryl Hydrocarbon/metabolism , Uterus/drug effects , Uterus/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...