171 results on '"Helena Tlaskalova-Hogenova"'
Search Results
2. Skin microbiota signature distinguishes IBD patients and reflects skin adverse events during anti-TNF therapy
- Author
-
Zuzana Reiss, Filip Rob, Martin Kolar, Dagmar Schierova, Jakub Kreisinger, Zuzana Jackova, Radka Roubalova, Stepan Coufal, Martin Mihula, Tomas Thon, Lukas Bajer, Michaela Novakova, Martin Vasatko, Klara Kostovcikova, Natalie Galanova, Milan Lukas, Miloslav Kverka, Jana Tresnak Hercogova, Helena Tlaskalova-Hogenova, and Zuzana Jiraskova Zakostelska
- Subjects
Microbiology (medical) ,Infectious Diseases ,Immunology ,Microbiology - Abstract
Crohn’s disease (CD) and ulcerative colitis (UC) are two forms of inflammatory bowel disease (IBD), where the role of gut but not skin dysbiosis is well recognized. Inhibitors of TNF have been successful in IBD treatment, but up to a quarter of patients suffer from unpredictable skin adverse events (SkAE). For this purpose, we analyzed temporal dynamics of skin microbiota and serum markers of inflammation and epithelial barrier integrity during anti-TNF therapy and SkAE manifestation in IBD patients. We observed that the skin microbiota signature of IBD patients differs markedly from healthy subjects. In particular, the skin microbiota of CD patients differs significantly from that of UC patients and healthy subjects, mainly in the retroauricular crease. In addition, we showed that anti-TNF-related SkAE are associated with specific shifts in skin microbiota profile and with a decrease in serum levels of L-FABP and I-FABP in IBD patients. For the first time, we showed that shifts in microbial composition in IBD patients are not limited to the gut and that skin microbiota and serum markers of the epithelium barrier may be suitable markers of SkAE during anti-TNF therapy.
- Published
- 2023
- Full Text
- View/download PDF
3. NMR- and MS-Based Untargeted Metabolomic Study of Stool and Serum Samples from Patients with Anorexia Nervosa
- Author
-
Martina Cermakova, Marek Vecka, Helena Tlaskalova-Hogenova, Radka Roubalová, Helena Pelantová, Blanka Šedivá, Hana Papežová, Petra Procházková, Petra Tomášová, Marek Kuzma, and Jiří Dvořák
- Subjects
Thyroid Hormones ,medicine.medical_specialty ,Magnetic Resonance Spectroscopy ,Thyrotropin ,Inflammation ,medicine.disease_cause ,Biochemistry ,anorexia nervosa ,Metabolomics ,Internal medicine ,medicine ,Humans ,Interleukin 6 ,mass spectrometry ,biology ,business.industry ,General Chemistry ,Metabolism ,metabolomics ,nuclear magnetic resonance ,Endocrinology ,Anorexia nervosa (differential diagnoses) ,Ketone bodies ,biology.protein ,medicine.symptom ,business ,Oxidative stress ,Hormone - Abstract
Anorexia nervosa (AN), a pathological restriction of food intake, leads to metabolic dysregulation. We conducted a metabolomics study to reveal changes caused by AN and the effect of hospital realimentation on metabolism. Both stool and serum from patients with AN and healthy controls were analyzed by NMR and MS. Statistical analysis revealed several altered biochemical and anthropometric parameters and 50 changed metabolites, including phospholipids, acylcarnitines, amino acids, derivatives of nicotinic acid, nucleotides, and energy metabolism intermediates. Biochemical and anthropometric parameters were correlated with metabolomic data. Metabolic changes in patients with AN described in our study imply serious system disruption defects, such as the development of inflammation and oxidative stress, changed free thyroxine (fT4) and thyroid-stimulating hormone (TSH) levels, a deficit of vitamins, muscle mass breakdown, and a decrease in ketone bodies as an important source of energy for the brain and heart. Furthermore, our data indicate only a very slight improvement after treatment. However, correlations of metabolomic results with body weight, interleukin 6, tumor necrosis factor α, fT4, and TSH might entail better prognoses and treatment effectiveness in patients with better system parameter status. Data sets are deposited in MassIVE: MSV000087713, DOI: 10.25345/C57R7X.
- Published
- 2021
4. Association between ustekinumab therapy and changes in specific anti-microbial response, serum biomarkers, and microbiota composition in patients with IBD: A pilot study
- Author
-
Filip Rob, Dagmar Schierova, Zuzana Stehlikova, Jakub Kreisinger, Radka Roubalova, Stepan Coufal, Martin Mihula, Zuzana Jackova, Miloslav Kverka, Tomas Thon, Klara Kostovcikova, Lukas Bajer, Pavel Drastich, Jana Tresnak Hercogova, Michaela Novakova, Martin Kolar, Martin Vasatko, Milan Lukas, Helena Tlaskalova-Hogenova, and Zuzana Jiraskova Zakostelska
- Subjects
Multidisciplinary - Abstract
Background Ustekinumab, is a new therapy for patients with IBD, especially for patients suffering from Crohn’s disease (CD) who did not respond to anti-TNF treatment. To shed light on the longitudinal effect of ustekinumab on the immune system, we investigated the effect on skin and gut microbiota composition, specific immune response to commensals, and various serum biomarkers. Methodology/Principal findings We recruited 11 patients with IBD who were monitored over 40 weeks of ustekinumab therapy and 39 healthy controls (HC). We found differences in the concentrations of serum levels of osteoprotegerin, TGF-β1, IL-33, and serum IgM antibodies against Lactobacillus plantarum between patients with IBD and HC. The levels of these biomarkers did not change in response to ustekinumab treatment or with disease improvement during the 40 weeks of observation. Additionally, we identified differences in stool abundance of uncultured Subdoligranulum, Faecalibacterium, and Bacteroides between patients with IBD and HC. Conclusion/Significance In this preliminary study, we provide a unique overview of the longitudinal monitoring of fecal and skin microbial profiles as well as various serum biomarkers and humoral and cellular response to gut commensals in a small cohort of patients with IBD on ustekinumab therapy.
- Published
- 2022
5. IBD Patients Show a Unique Skin Microbiota Composition and a Correlation Pattern Between Serological Biomarkers and Clinical Aspects During Skin Adverse Events in the Setting of Anti-TNFα Therapy
- Author
-
Zuzana Stehlikova, Filip Rob, Martin Kolar, Dagmar Schierova, Jakub Kreisinger, Zuzana Jackova, Radka Roubalova, Stepan Coufal, Martin Mihula, Tomas Thon, Lukas Bajer, Michaela Novakova, Martin Vasatko, Klara Kostovcikova, Natalie Galanova, Milan Lukas, Miloslav Kverka, Jana Hercogova, Helena Tlaskalova-Hogenova, and Zuzana Jiraskova Zakostelska
- Abstract
Background Crohn’s disease (CD) and ulcerative colitis (UC) are two forms of inflammatory bowel disease (IBD), where gut but not skin dysbiosis is well recognized. Tumor-necrosis factor alpha (TNFα) blockers have been successful in IBD treatment, but up to a quarter of patients suffer from skin adverse events (SkAE), for which we do not have a tool to predict them. Therefore, we examined the skin microbiota composition of IBD patients at several body sites by 16S bacterial profiling. Moreover, we quantified 22 serum markers of inflammation and epithelial barrier disruption in the setting of anti-TNFα therapy and SkAE manifestation by ELISA. Results The skin microbiota signature of IBD patients differed markedly from healthy subjects. In particular, skin microbiota of CD patients differed significantly from that of UC and HC at the retroauricular crease. CD patients showed high abundance of Corynebacterium (ASV86) and lower abundance of Actinomyces (ASV28), Cutibacterium (ASV212), Lawsonella (ASV87), and other taxa compared to HC. UC patients showed higher abundance of Delftia (ASV1039) and Pseudomonas (ASV1183), and lower abundance of Actinomyces (ASV28), Streptococcus (ASV607), or Haemophilus (ASV1150) than HC. Although microbiome changes were correlated at the ASV level in CD and UC patients compared to HC, we identified some bacterial ASV’s that exhibited changes typical of individual IBD forms. Manifestation of SkAE during anti-TNFα therapy was associated with a specific microbiota profile on the skin, and with a decrease in serum levels of L-FABP and I-FABP. Conclusions The overall proinflammatory effect of IBD might predispose for altered skin microbiota composition, hence promoting SkAE manifestation during anti-TNFα therapy. We identified specific serum markers associated with the incidence of SkAE during anti-TNFα therapy, however, further studies are needed to shed light on the impact of microbiota in this matter. Combining sequencing technology with search for serum biomarkers has opened up new opportunities in studying SkAE related to anti-TNFα therapy. Early detection of changes in microbiota-host equilibrium may have beneficial consequences in revealing the pathogenesis of SkAE in IBD.
- Published
- 2022
6. Multiparametric flow cytometry analysis of peripheral blood B cell trafficking differences among Epstein-Barr virus infected and uninfected subpopulations
- Author
-
Petr Hubacek, Petr Kosztyu, Milan Raska, Klara Kostovcikova, Josef Zadrazil, Karel Vondrak, Jiri Mestecky, Katerina Zachova, Helena Tlaskalova Hogenova, and Karel Matousovic
- Subjects
Adult ,Male ,Epstein-Barr Virus Infections ,igm ,plasma cell ,plasma blast ,medicine.medical_treatment ,Naive B cell ,Vesicular Transport Proteins ,lcsh:Medicine ,030204 cardiovascular system & hematology ,Plasma cell ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,B cell ,Aged ,Aged, 80 and over ,B-Lymphocytes ,epstein-barr virus ,medicine.diagnostic_test ,biology ,business.industry ,lcsh:R ,naïve b cells ,Middle Aged ,Flow Cytometry ,Epstein–Barr virus ,Molecular biology ,Isotype ,Healthy Volunteers ,memory b cells ,Blood ,medicine.anatomical_structure ,Cytokine ,030220 oncology & carcinogenesis ,biology.protein ,Female ,cell trafficking ,Antibody ,business ,iga - Abstract
Aims: Epstein-Barr virus (EBV) targets predominantly B cells and these cells could acquire new phenotype characteristics. Here we analyzed whether EBV-infected and -uninfected B cells from healthy subjects differ in proportion of dominant phenotypes, maturation stage, and homing receptors expression. Methods: EBV-infected and -uninfected cells were identified by flow cytometry using fluorophore-labeled EBV RNA-specific DNA probes combined with fluorophore-labeled antibody to surface lineage markers, integrins, chemokine receptors, and immunoglobulin isotypes, including intracellular ones. Results: Our results show that the trafficking characteristics of EBERpos B cells are distinct from EBERneg B cells with most dominant differences detected for α4β1 and α4β7 and CCR5 and CCR7. EBV-positive cells are predominantly memory IgM+ B cells or plasmablasts/plasma cells (PB/PC) positive for IgA or less for IgM. In comparison to uninfected B cells, less EBV-positive B cells express α4β7 and almost no cells express α4β1. EBV-positive B cells contained significantly higher proportion of CCR5+ and CCR7+ cells in comparison to EBV-negative cells. In vitro exposure of blood mononuclear cells to pro-inflammatory cytokine IL-6 reduces population of EBV-positive B cell. Conclusion: Although EBV-infected B cells represent only a minor subpopulation, their atypical functions could contribute in predisposed person to development abnormities such as some autoimmune diseases or tumors. Using multi-parameter flow cytometry we characterized differences in migration of EBV-positive and -negative B cells of various maturation stage and isotype of produced antibodies particularly different targeting to mucosal tissues of gastrointestinal and respiratory tracts.
- Published
- 2020
7. Secretory IgA N-glycans contribute to the protection against E. coli O55 infection of germ-free piglets
- Author
-
Jiri Dvorak, Marek Sinkora, Leona Raskova Kafkova, Milan Raska, Zuzana Jiraskova Zakostelska, Dagmar Srutkova, Jiri Mestecky, Renata Stepankova, Jozef Skarda, Luca Vannucci, Zdenek Novak, Helena Tlaskalova-Hogenova, Diana Brokesova, Michal Krupka, Stepan Coufal, Alena Fajstova, Ivo Uberall, Zuzana Stehlikova, Petra Hermanova, and Katerina Stepanova
- Subjects
0301 basic medicine ,Glycan ,Agglutination ,Glycosylation ,Secretory component ,Swine ,Immunology ,chemical and pharmacologic phenomena ,digestive system ,Article ,Microbiology ,03 medical and health sciences ,Immunoglobulin Fab Fragments ,0302 clinical medicine ,fluids and secretions ,stomatognathic system ,In vivo ,Polysaccharides ,Pregnancy ,Escherichia coli ,Immunology and Allergy ,Animals ,Germ-Free Life ,Secretory IgA ,Escherichia coli Infections ,Disease Resistance ,biology ,Chemistry ,food and beverages ,In vitro ,Agglutination (biology) ,Intestinal histology ,030104 developmental biology ,Animals, Newborn ,Immunoglobulin A, Secretory ,biology.protein ,Female ,030215 immunology ,Single-Chain Antibodies - Abstract
Mucosal surfaces are colonized by highly diverse commensal microbiota. Coating with secretory IgA (SIgA) promotes the survival of commensal bacteria while it inhibits the invasion by pathogens. Bacterial coating could be mediated by antigen-specific SIgA recognition, polyreactivity, and/or by the SIgA-associated glycans. In contrast to many in vitro studies, only a few reported the effect of SIgA glycans in vivo. Here, we used a germ-free antibody-free newborn piglets model to compare the protective effect of SIgA, SIgA with enzymatically removed N-glycans, Fab, and Fc containing the secretory component (Fc-SC) during oral necrotoxigenic E. coli O55 challenge. SIgA, Fab, and Fc-SC were protective, whereas removal of N-glycans from SIgA reduced SIgA-mediated protection as demonstrated by piglets' intestinal histology, clinical status, and survival. In vitro analyses indicated that deglycosylation of SIgA did not reduce agglutination of E. coli O55. These findings highlight the role of SIgA-associated N-glycans in protection. Further structural studies of SIgA-associated glycans would lead to the identification of those involved in the species-specific inhibition of attachment to corresponding epithelial cells.
- Published
- 2020
8. Urinary I-FABP, L-FABP, TFF-3, and SAA Can Diagnose and Predict the Disease Course in Necrotizing Enterocolitis at the Early Stage of Disease
- Author
-
Jiri Snajdauf, Barbora Frybova, Stepan Coufal, Alena Kokesova, Helena Tlaskalova-Hogenova, Miloslav Kverka, and Michal Rygl
- Subjects
Male ,medicine.medical_specialty ,Article Subject ,Immunology ,Fatty Acid-Binding Proteins ,Enteral administration ,Gastroenterology ,Veins ,Diagnosis, Differential ,Sepsis ,03 medical and health sciences ,0302 clinical medicine ,Enterocolitis, Necrotizing ,030225 pediatrics ,Internal medicine ,medicine ,Humans ,Immunology and Allergy ,Serum amyloid A ,Intestinal Mucosa ,Pneumatosis intestinalis ,Inflammation ,Enterocolitis ,Serum Amyloid A Protein ,business.industry ,Infant, Newborn ,General Medicine ,RC581-607 ,Prognosis ,medicine.disease ,digestive system diseases ,Early Diagnosis ,Gastrointestinal disease ,030220 oncology & carcinogenesis ,Necrotizing enterocolitis ,Disease Progression ,Female ,lipids (amino acids, peptides, and proteins) ,Immunologic diseases. Allergy ,Trefoil Factor-3 ,medicine.symptom ,business ,Biomarkers ,Research Article ,Abdominal surgery - Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease affecting mainly preterm newborns. It is characterized by unexpected onset and rapid progression with specific diagnostic signs as pneumatosis intestinalis or gas in the portal vein appearing later in the course of the disease. Therefore, we analyzed diagnostic and prognostic potential of the markers of early NEC pathogenesis, such as excessive inflammatory response (serum amyloid A (SAA)) and gut epithelium damage (intestinal and liver fatty acid-binding protein (I-FABP and L-FABP, respectively) and trefoil factor-3 (TFF-3)). We used ELISA to analyze these biomarkers in the urine of patients with suspected NEC, either spontaneous or surgery-related, or in infants without gut surgery (controls). Next, we compared their levels with the type of the disease (NEC or sepsis) and its severity. Already at the time of NEC suspicion, infants who developed NEC had significantly higher levels of all tested biomarkers than controls and higher levels of I-FABP and L-FABP than those who will later develop sepsis. Infants who will develop surgery-related NEC had higher levels of I-FABP and L-FABP than those who will develop sepsis already during the first 6 hours after the abdominal surgery. I-FABP was able to discriminate between infants who will develop NEC or sepsis and the SAA was able to discriminate between medical and surgical NEC. Moreover, the combination of TFF-3 with I-FABP and SAA could predict pneumatosis intestinalis, and the combination of I-FABP, L-FABP, and SAA could predict gas in the portal vein or long-term hospitalization and low SAA predicts early full enteral feeding. Thus, these biomarkers may be useful not only in the early, noninvasive diagnostics but also in the subsequent NEC management.
- Published
- 2020
9. Multiple Sclerosis and Microbiome
- Author
-
Jana Lizrova Preiningerova, Zuzana Jiraskova Zakostelska, Adhish Srinivasan, Veronika Ticha, Ivana Kovarova, Pavlina Kleinova, Helena Tlaskalova-Hogenova, and Eva Kubala Havrdova
- Subjects
Mice ,Multiple Sclerosis ,Microbiota ,Brain-Gut Axis ,Animals ,Humans ,digestive system ,Molecular Biology ,Biochemistry ,Gastrointestinal Microbiome - Abstract
The composition of microbiota and the gut-brain axis is increasingly considered a factor in the development of various pathological conditions. The etiology of multiple sclerosis (MS), a chronic autoimmune disease affecting the CNS, is complex and interactions within the gut-brain axis may be relevant in the development and the course of MS. In this article, we focus on the relationship between gut microbiota and the pathophysiology of MS. We review the contribution of germ-free mouse studies to our understanding of MS pathology and its implications for treatment strategies to modulate the microbiome in MS. This summary highlights the need for a better understanding of the role of the microbiota in patients’ responses to disease-modifying drugs in MS and disease activity overall.
- Published
- 2022
10. Severity of Experimental Autoimmune Uveitis Is Reduced by Pretreatment with Live Probiotic Escherichia coli Nissle 1917
- Author
-
Petra Svozilkova, Miloslav Kverka, Alena Fajstova, Jarmila Heissigerova, Tomas Hrncir, Stepan Coufal, Aneta Klimova, John V. Forrester, Otakar Dusek, Johan Slemin, and Helena Tlaskalova-Hogenova
- Subjects
0301 basic medicine ,experimental autoimmune uveitis ,Inflammation ,Gut flora ,law.invention ,03 medical and health sciences ,Probiotic ,0302 clinical medicine ,Immune system ,law ,medicine ,mucosal immune system ,Escherichia coli Nissle 1917 ,lcsh:QH301-705.5 ,Autoimmune disease ,biology ,business.industry ,General Medicine ,biology.organism_classification ,medicine.disease ,eye diseases ,macrophages ,030104 developmental biology ,medicine.anatomical_structure ,probiotics ,lcsh:Biology (General) ,Cervical lymph nodes ,030220 oncology & carcinogenesis ,Immunology ,Lymph ,medicine.symptom ,business ,Uveitis - Abstract
Non-infectious uveitis is considered an autoimmune disease responsible for a significant burden of blindness in developed countries and recent studies have linked its pathogenesis to dysregulation of the gut microbiota. We tested the immunomodulatory properties of two probiotics, Escherichia coli Nissle 1917 (EcN) and E. coli O83:K24:H31 (EcO), in a model of experimental autoimmune uveitis (EAU). To determine the importance of bacterial viability and treatment timing, mice were orally treated with live or autoclaved bacteria in both preventive and therapeutic schedules. Disease severity was assessed by ophthalmoscopy and histology, immune phenotypes in mesenteric and cervical lymph nodes were analyzed by flow cytometry and the gut immune environment was analyzed by RT-PCR and/or gut tissue culture. EcN, but not EcO, protected against EAU but only as a live organism and only when administered before or at the time of disease induction. Successful prevention of EAU was accompanied by a decrease in IRBP-specific T cell response in the lymph nodes draining the site of immunization as early as 7 days after the immunization and eye-draining cervical lymph nodes when the eye inflammation became apparent. Furthermore, EcN promoted an anti-inflammatory response in Peyer&rsquo, s patches, increased gut antimicrobial peptide expression and decreased production of inducible nitric oxide synthase in macrophages. In summary, we show here that EcN controls inflammation in EAU and suggest that probiotics may have a role in regulating the gut&ndash, eye axis.
- Published
- 2021
11. Fecal Microbiome Changes and Specific Anti-Bacterial Response in Patients with IBD during Anti-TNF Therapy
- Author
-
Stepan Coufal, Zuzana Jackova, Zuzana Stehlikova, Martin Mihula, Martin Modrak, Martin Vašátko, Jana Hercogová, Klara Kostovcikova, Miloslav Kverka, Dagmar Schierova, Lukas Bajer, Zuzana Jiraskova Zakostelska, Filip Rob, Pavel Drastich, Tomas Thon, Michaela Novakova, Radka Roubalová, Milan Lukáš, Martin Kolar, and Helena Tlaskalova-Hogenova
- Subjects
Adult ,Male ,QH301-705.5 ,microbiome ,Disease ,Gut flora ,Severity of Illness Index ,Inflammatory bowel disease ,digestive system ,Antibodies ,Article ,Feces ,mycobiome ,Immune system ,inflammatory bowel disease ,RNA, Ribosomal, 16S ,medicine ,Humans ,Microbiome ,Biology (General) ,Gastrointestinal tract ,biology ,business.industry ,Interleukin-17 ,Fungi ,Biodiversity ,General Medicine ,Inflammatory Bowel Diseases ,medicine.disease ,biology.organism_classification ,Ulcerative colitis ,Gastrointestinal Microbiome ,biological therapy ,Case-Control Studies ,Immunology ,Leukocytes, Mononuclear ,Female ,Tumor Necrosis Factor Inhibitors ,Metagenomics ,tumor necrosis factor-α ,business ,Dysbiosis - Abstract
Inflammatory bowel diseases (IBD) are chronic disorders of the gastrointestinal tract that have been linked to microbiome dysbiosis and immune system dysregulation. We investigated the longitudinal effect of anti-TNF therapy on gut microbiota composition and specific immune response to commensals in IBD patients. The study included 52 patients tracked over 38 weeks of therapy and 37 healthy controls (HC). To characterize the diversity and composition of the gut microbiota, we used amplicon sequencing of the V3V4 region of 16S rRNA for the bacterial community and of the ITS1 region for the fungal community. We measured total antibody levels as well as specific antibodies against assorted gut commensals by ELISA. We found diversity differences between HC, Crohn’s disease, and ulcerative colitis patients. The bacterial community of patients with IBD was more similar to HC at the study endpoint, suggesting a beneficial shift in the microbiome in response to treatment. We identified factors such as disease severity, localization, and surgical intervention that significantly contribute to the observed changes in the gut bacteriome. Furthermore, we revealed increased IgM levels against specific gut commensals after anti-TNF treatment. In summary, this study, with its longitudinal design, brings insights into the course of anti-TNF therapy in patients with IBD and correlates the bacterial diversity with disease severity in patients with ulcerative colitis (UC).
- Published
- 2021
- Full Text
- View/download PDF
12. Editorial: Employing Experimental Gnotobiotic Models to Decipher the Host-Microbiota Cross-Talk in Health and Disease
- Author
-
Martin Schwarzer, François Leulier, Irma Schabussova, Helena Tlaskalova-Hogenova, Institut de Génomique Fonctionnelle de Lyon (IGFL), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), and École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL)
- Subjects
[SDV]Life Sciences [q-bio] ,Immunology ,Biology ,Gut flora ,host-bacteria interactions ,Inflammatory bowel disease ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Species Specificity ,medicine ,Animals ,Germ-Free Life ,Humans ,Immunology and Allergy ,Microbiome ,Animal Husbandry ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,2. Zero hunger ,0303 health sciences ,Innate immune system ,Bacteria ,Microbiota ,Human microbiome ,RC581-607 ,medicine.disease ,biology.organism_classification ,Housing, Animal ,Bacterial Load ,gnotobiology ,Altered Schaedler flora ,Disease Models, Animal ,immune system ,Editorial ,germ-free ,Plant protein ,Host-Pathogen Interactions ,030211 gastroenterology & hepatology ,Immunologic diseases. Allergy - Abstract
Eukaryotic organisms have evolved in a world dominated by bacteria and archaea. Rather than face the daunting task of keeping their exposed surfaces germ-free (GF), they have developed close symbiotic relationships. Mucosal surfaces are associated with specific microbial communities that influence various aspects of host physiology and, most importantly, the development and fine-tuning of the immune system. In the last decade, we have witnessed a renewed interest in understanding the role of the microbiota for the homeostasis and disease. This has fostered the development of sophisticated multidisciplinary technologies that enable compositional and functional analysis of the microbiome (1). The most diverse and numerous microbial communities are found in the gastrointestinal tract. Alterations in the gut microbiome and/or disruptions of the cross-talk between host and microbiota has been linked to immune-mediated diseases such as allergies and autoimmune diseases such as inflammatory bowel disease, rheumatoid arthritis, multiple sclerosis and diabetes (2). The aforementioned complexity of the gut microbial ecosystem currently complicates the understanding of the microbiota-host cross-talk, with descriptive reports predominating over mechanistic studies. However, powerful tools for studying host-microbe interactions are germ-free (GF) and gnotobiotic animal models. Although historically mostly rodents and piglets (3) have been used, new vertebrate models, for example fish, and invertabrate models such as Drosophila have successfully been developed in recent years (4). The possibility to colonize GF animals with defined bacterial species or a consortium of bacteria has created a unique opportunity to evaluate the impact of the microbiota on host physiology and immune system (5). Colonization of GF mice with a defined bacterial consortium that mimics the complexity of a specific pathogen-free/specific opportunistic pathogen-free (SPF/SOPF) microbiota has increased the reproducibility of mouse experiments across different institutions and mouse facilities (6). In this regard, Eberl et al. show that one such consortium, Oligo-Mouse-Microbiota12 (Oligo-MM12), could be established in several different animal facilities with excellent reproducibility. Within two weeks after inoculation of the Oligo-MM12 consortium in GF mice similar stable microbial communities were found in all facilities. Furthermore, they showed that a second inoculation of the Oligo-MM12 strains after 72 h was even more effective at establishing a stable microbiota. Thus the reliable de novo generation of gnotobiotic rodents contributes greatly to experimental reproducibility, which has significantly benefitted biomedical research in recent years. Using the Oligo-MM12 consortium, Wyss et al. sought to explain the phenomenon behind elevated IgE levels in GF animals. Depending on the mouse facility, mouse model, and breeding conditions, GF mice exhibit elevated total serum IgE levels (6–9). Wyss et al. demonstrated that colonization of mice with a well-defined composition of bacterial species is required to inhibit elevated IgE levels. Features of bacterial consortia that successfully decrease the IgE include presence in early life, acetate production, and immunogenicity reflected in the induction of gut intestinal IgA. There is a growing interest in the development of microbial-based and microbial-targeted therapies (10). Preclinical studies with “humanized” mice (transfer of human microbiota to germ-free animals) are often necessary methodological requirement to analyze the potential therapeutic effects. This topic was discussed in detail by Rogala et al. GF mice colonized with single wild-type or genetically engineered microbial isolates are an invaluable tool to study the functions of individual bacterial genes and species. GF mice colonized with multiple defined isolates can be used to determine interactions between members of defined consortia. This is elegantly discussed and ways to improve studies of immune-microbial interactions using gnotobiotic mice are presented. On this theme, Bolsega et al. investigated whether different bacterial minimal consortia affect the outcome of murine norovirus-induced colitis in Il10-/- mice. By comparing two different minimal microbiota (Oligo-MM12 and Altered Schaedler Flora) colonized mice, they show that murine norovirus-triggered colitis depends on the composition of the microbiota. Lengfelder et al. characterized the colitogenic activity of Enterococcus faecalis as part of a simplified human microbial consortium based on seven enteric bacterial strains (SIHUMI). They showed that complex bacterial consortium interactions reprogram the gene expression profile and colitogenic activity of the opportunistic pathogen E. faecalis towards a protective function. Kostovcikova et al. examined colitis from a nutritional perspective and used a gnotobiotic approach to show that diet rich in animal protein exacerbates acute dextran sulfate sodium (DSS)-induced colitis, whereas diet rich in plant protein does not. They concluded that interactions between a dietary protein of animal origin and the gut microbiota increase sensitivity to intestinal inflammation by promoting the pro-inflammatory responses of monocytes. The gut microbiota can influence brain functions and behavior, including hypothalamic-pituitary-adrenocortical axis (HPA) activity. Using SPF and GF male BALB/c mice, Vagnerova et al. investigated the influence of the microbiota on the acute restraint stress (ARS) response in the pituitary, adrenal gland, and intestine, an organ of extra-adrenal glucocorticoid synthesis. They showed that GF animals have an exaggerated HPA response to stress. Finally, Murdoch and Rawls review the key insights provided by the gnotobiotic zebrafish model on the effects of microbiota on innate immunity. This included evidence that the perception of and response to the microbiota is evolutionarily conserved. They describe how to strengthen the zebrafish model system and provide new insights into the host-microbe interactions that would be difficult to study in mammalian models. Overall, we have assembled a set of eight research articles that bring new insights into host-microbiota interactions. These studies all highlight the advantages of gnotobiotic rodent and fish models, and demonstrate the modularity of defined bacterial minimal consortia that can be used to achieve greater standardization of biomedical research across different animal facilities.
- Published
- 2021
13. Altered Serum Immunological and Biochemical Parameters and Microbiota Composition in Patients With AN During Realimentation
- Author
-
Radka Roubalova, Petra Prochazkova, Jiri Dvorak, Martin Hill, Hana Papezova, Jakub Kreisinger, Josef Bulant, Alena Lambertova, Petra Holanova, Martin Bilej, and Helena Tlaskalova-Hogenova
- Subjects
0301 basic medicine ,Endocrinology, Diabetes and Metabolism ,Gut flora ,medicine.disease_cause ,anorexia nervosa ,Serology ,Autoimmunity ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Enterobacteriaceae ,microbiota ,Medicine ,TX341-641 ,Nutrition ,Original Research ,Nutrition and Dietetics ,alpha-melanocyte stimulating hormone ,biology ,Nutrition. Foods and food supply ,business.industry ,Autoantibody ,biology.organism_classification ,realimentation ,immune system ,030104 developmental biology ,Anorexia nervosa (differential diagnoses) ,Immunology ,business ,autoantibody ,030217 neurology & neurosurgery ,Food Science ,Hormone - Abstract
Anorexia nervosa (AN) is a life-threatening psychiatric disorder with not well-described pathogenesis. Besides the genetic and sociological factors, autoimmunity is also considered to take part in AN pathogenesis. We evaluated general serological factors showing the physiological state of 59 patients with AN at hospital admission and their discharge. We detected the altered levels of some general biochemical and immunological parameters. We also detected decreased levels of appetite-regulating alpha-melanocyte stimulating hormone (α-MSH) in patients at hospital admission. Moreover, elevated anti-α-MSH IgM levels and decreased anti-α-MSH IgA levels were observed in patients with AN. Therefore, we analyzed the gut microbiota composition with special focus on α-MSH antigen-mimetic containing microbes from the Enterobacteriaceae family. We correlated gut bacterial composition with anti-α-MSH Ig levels and detected decreasing IgG levels with increasing alpha diversity. The upregulation of pro-inflammatory cytokines IL-6, IL-17, and TNF-α were detected in patients with AN both prior and after hospitalization. We also evaluated the treatment outcome and improvement was observed in the majority of patients with AN. We provide new data about various serum biochemical parameters and their changes during the patients' hospitalization, with emphasis on the immune system, and its possible participation in AN pathogenesis.
- Published
- 2021
14. The role of gut microbiota in intestinal and liver diseases
- Author
-
Helena Tlaskalova-Hogenova, Miloslav Kverka, Tomas Hrncir, and Lucia Hrncirova
- Subjects
040301 veterinary sciences ,Gut flora ,Inflammatory bowel disease ,Coeliac disease ,0403 veterinary science ,Pathogenesis ,Mice ,03 medical and health sciences ,Immune system ,medicine ,Animals ,Germ-Free Life ,030304 developmental biology ,0303 health sciences ,General Veterinary ,biology ,business.industry ,Liver Diseases ,Cancer ,04 agricultural and veterinary sciences ,medicine.disease ,biology.organism_classification ,Gastrointestinal Microbiome ,Disease Models, Animal ,Intestinal Diseases ,Immunology ,Dysbiosis ,Animal Science and Zoology ,business - Abstract
The world-wide incidence of many immune-mediated and metabolic diseases, including those of the intestines and liver, is steadily increasing. Gut microbiota plays a central role in the pathogenesis of these diseases as it mediates environmental changes to the intestinal immune system. Various environmental factors including diet, food additives and medication also trigger the compositional and functional alterations of microbiota, that is, dysbiosis, and this dysbiosis is closely associated with many chronic inflammatory diseases. However, the causal relationship remains unclear for the majority of these diseases. In this review, we discuss essential epidemiological data, known pathogenetic factors including those of genetic and environmental nature, while mainly focusing on the role of gut microbiota in the development of selected intestinal and liver diseases. Using specific examples, we also briefly describe some of the most widely-used animal models including gnotobiotic models and their contribution to the research of pathogenetic mechanisms of the host–microbiota relationship.
- Published
- 2018
15. Current Aspects of the Role of Autoantibodies Directed Against Appetite-Regulating Hormones and the Gut Microbiome in Eating Disorders
- Author
-
Kvido Smitka, Petra Prochazkova, Radka Roubalova, Jiri Dvorak, Hana Papezova, Martin Hill, Jaroslav Pokorny, Otomar Kittnar, Martin Bilej, and Helena Tlaskalova-Hogenova
- Subjects
anorexia nervosa and bulimia ,alpha-MSH ,ghrelin ,digestive, oral, and skin physiology ,gut and blood-brain barrier permeability ,caseinolytic peptidase B ,RC648-665 ,Diseases of the endocrine glands. Clinical endocrinology ,fecal microbial transplantation - Abstract
The equilibrium and reciprocal actions among appetite-stimulating (orexigenic) and appetite-suppressing (anorexigenic) signals synthesized in the gut, brain, microbiome and adipose tissue (AT), seems to play a pivotal role in the regulation of food intake and feeding behavior, anxiety, and depression. A dysregulation of mechanisms controlling the energy balance may result in eating disorders such as anorexia nervosa (AN) and bulimia nervosa (BN). AN is a psychiatric disease defined by chronic self-induced extreme dietary restriction leading to an extremely low body weight and adiposity. BN is defined as out-of-control binge eating, which is compensated by self-induced vomiting, fasting, or excessive exercise. Certain gut microbiota-related compounds, like bacterial chaperone protein Escherichia coli caseinolytic protease B (ClpB) and food-derived antigens were recently described to trigger the production of autoantibodies cross-reacting with appetite-regulating hormones and neurotransmitters. Gut microbiome may be a potential manipulator for AT and energy homeostasis. Thus, the regulation of appetite, emotion, mood, and nutritional status is also under the control of neuroimmunoendocrine mechanisms by secretion of autoantibodies directed against neuropeptides, neuroactive metabolites, and peptides. In AN and BN, altered cholinergic, dopaminergic, adrenergic, and serotonergic relays may lead to abnormal AT, gut, and brain hormone secretion. The present review summarizes updated knowledge regarding the gut dysbiosis, gut-barrier permeability, short-chain fatty acids (SCFA), fecal microbial transplantation (FMT), blood-brain barrier permeability, and autoantibodies within the ghrelin and melanocortin systems in eating disorders. We expect that the new knowledge may be used for the development of a novel preventive and therapeutic approach for treatment of AN and BN.
- Published
- 2021
16. Monoassociation of Preterm Germ-Free Piglets with Bifidobacterium animalis Subsp. lactis BB-12 and Its Impact on Infection with Salmonella Typhimurium
- Author
-
Zbynek Stranak, Alla Splichalova, Sharon M. Donovan, Helena Tlaskalova-Hogenova, Zdislava Splichalova, and Igor Splichal
- Subjects
0301 basic medicine ,Salmonella ,inflammatory cytokines ,030106 microbiology ,Medicine (miscellaneous) ,Ileum ,Spleen ,Biology ,medicine.disease_cause ,Occludin ,digestive system ,preterm host ,General Biochemistry, Genetics and Molecular Biology ,Article ,Microbiology ,Proinflammatory cytokine ,law.invention ,03 medical and health sciences ,Probiotic ,law ,medicine ,Mesenteric lymph nodes ,lcsh:QH301-705.5 ,Bifidobacterium animalis subsp. lactis BB-12 ,Salmonella Typhimurium ,biology.organism_classification ,Bifidobacterium animalis ,intestinal barrier ,immunocompromised ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,bacteria - Abstract
Preterm germ-free piglets were monoassociated with probiotic Bifidobacterium animalis subsp. lactis BB-12 (BB12) to verify its safety and to investigate possible protection against subsequent infection with Salmonella Typhimurium strain LT2 (LT2). Clinical signs of salmonellosis, bacterial colonization in the intestine, bacterial translocation to mesenteric lymph nodes (MLN), blood, liver, spleen, and lungs, histopathological changes in the ileum, claudin-1 and occludin mRNA expression in the ileum and colon, intestinal and plasma concentrations of IL-8, TNF-α, and IL-10 were evaluated. Both BB12 and LT2 colonized the intestine of the monoassociated piglets. BB12 did not translocate in the BB12-monoassociated piglets. BB12 was detected in some cases in the MLN of piglets, consequently infected with LT2, but reduced LT2 counts in the ileum and liver of these piglets. LT2 damaged the luminal structure of the ileum, but a previous association with BB12 mildly alleviated these changes. LT2 infection upregulated claudin-1 mRNA in the ileum and colon and downregulated occludin mRNA in the colon. Infection with LT2 increased levels of IL-8, TNF-α, and IL-10 in the intestine and plasma, and BB12 mildly downregulated them compared to LT2 alone. Despite reductions in bacterial translocation and inflammatory cytokines, clinical signs of LT2 infection were not significantly affected by the probiotic BB12. Thus, we hypothesize that multistrain bacterial colonization of preterm gnotobiotic piglets may be needed to enhance the protective effect against the infection with S. Typhimurium LT2.
- Published
- 2021
17. Contribution of Infectious Agents to the Development of Celiac Disease
- Author
-
Adéla Szczepanková, Daniel Sánchez, Věra Hábová, Helena Tlaskalova-Hogenova, and Iva Hoffmanová
- Subjects
0301 basic medicine ,Microbiology (medical) ,Disease ,Review ,parasites ,Microbiology ,immune response ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,gluten-free diet ,Virology ,microbiota ,Medicine ,Ingestion ,infections ,Mucosal immunity ,Pathological ,lcsh:QH301-705.5 ,chemistry.chemical_classification ,biology ,business.industry ,nutritional and metabolic diseases ,Gluten ,digestive system diseases ,030104 developmental biology ,chemistry ,lcsh:Biology (General) ,Immunology ,biology.protein ,030211 gastroenterology & hepatology ,business ,Gliadin ,celiac disease - Abstract
The ingestion of wheat gliadin (alcohol-soluble proteins, an integral part of wheat gluten) and related proteins induce, in genetically predisposed individuals, celiac disease (CD), which is characterized by immune-mediated impairment of the small intestinal mucosa. The lifelong omission of gluten and related grain proteins, i.e., a gluten-free diet (GFD), is at present the only therapy for CD. Although a GFD usually reduces CD symptoms, it does not entirely restore the small intestinal mucosa to a fully healthy state. Recently, the participation of microbial components in pathogenetic mechanisms of celiac disease was suggested. The present review provides information on infectious diseases associated with CD and the putative role of infections in CD development. Moreover, the involvement of the microbiota as a factor contributing to pathological changes in the intestine is discussed. Attention is paid to the mechanisms by which microbes and their components affect mucosal immunity, including tolerance to food antigens. Modulation of microbiota composition and function and the potential beneficial effects of probiotics in celiac disease are discussed.
- Published
- 2021
18. The intestinal microbiota and metabolites in patients with anorexia nervosa
- Author
-
Marek Kuzma, Martin Bilej, Alena Lambertova, Jakub Kreisinger, Petra Procházková, Jiri Dvorak, Radka Roubalová, Josef Bulant, Helena Tlaskalova-Hogenova, Helena Pelantová, Kvido Smitka, Martina Cermakova, Hana Papezova, Petra Tomášová, Martin Hill, and Petra Holanova
- Subjects
0301 basic medicine ,Anorexia Nervosa ,Physiology ,microbiome ,RC799-869 ,Gut flora ,Body Mass Index ,Feces ,0302 clinical medicine ,Brain-Gut Axis ,Longitudinal Studies ,Neurotransmitter Agents ,biology ,Gastroenterology ,dysbiosis ,Diseases of the digestive system. Gastroenterology ,renourishment ,Infectious Diseases ,Female ,030211 gastroenterology & hepatology ,Research Article ,Research Paper ,Ruminococcaceae ,neurotransmitter ,Adult ,Microbiology (medical) ,Microbiology ,ede-q ,Young Adult ,03 medical and health sciences ,mycobiome ,scfa ,medicine ,Humans ,Microbiome ,bacteriome ,Alistipes ,gut-brain-microbiota axis ,Bacteria ,Clostridiales ,Fungi ,Bacteriome ,Fatty Acids, Volatile ,biology.organism_classification ,medicine.disease ,Archaea ,Gastrointestinal Microbiome ,bmi ,030104 developmental biology ,Metagenome ,Bacteroides ,Dysbiosis - Abstract
Brain-gut microbiota interactions are intensively studied in connection with various neurological and psychiatric diseases. While anorexia nervosa (AN) pathophysiology is not entirely clear, it is presumably linked to microbiome dysbiosis. We aimed to elucidate the gut microbiota contribution in AN disease pathophysiology. We analyzed the composition and diversity of the gut microbiome of patients with AN (bacteriome and mycobiome) from stool samples before and after renourishment, and compared them to healthy controls. Further, levels of assorted neurotransmitters and short-chain fatty acids (SCFA) were analyzed in stool samples by MS and NMR, respectively. Biochemical, anthropometric, and psychometric profiles were assessed. The bacterial alpha-diversity parameter analyses revealed only increased Chao 1 index in patients with AN before the realimentation, reflecting their interindividual variation. Subsequently, core microbiota depletion signs were observed in patients with AN. Overrepresented OTUs (operation taxonomic units) in patients with AN taxonomically belonged to Alistipes, Clostridiales, Christensenellaceae, and Ruminococcaceae. Underrepresented OTUs in patients with AN were Faecalibacterium, Agathobacter, Bacteroides, Blautia, and Lachnospira. Patients exhibited greater interindividual variation in the gut bacteriome, as well as in metagenome content compared to controls, suggesting altered bacteriome functions. Patients had decreased levels of serotonin, GABA, dopamine, butyrate, and acetate in their stool samples compared to controls. Mycobiome analysis did not reveal significant differences in alpha diversity and fungal profile composition between patients with AN and healthy controls, nor any correlation of the fungal composition with the bacterial profile. Our results show the changed profile of the gut microbiome and its metabolites in patients with severe AN. Although therapeutic partial renourishment led to increased body mass index and improved psychometric parameters, SCFA, and neurotransmitter profiles, as well as microbial community compositions, did not change substantially during the hospitalization period, which can be potentially caused by only partial weight recovery.
- Published
- 2021
19. Diet Rich in Simple Sugars Promotes Pro-Inflammatory Response via Gut Microbiota Alteration and TLR4 Signaling
- Author
-
Marek Kuzma, Blanka Šedivá, Martin Kostovčík, Alena Fajstova, Stepan Coufal, Miloslav Kverka, Helena Tlaskalova-Hogenova, Martina Cermakova, Tomas Hudcovic, Tomas Hrncir, Helena Pelantová, Jana Malkova, Klara Kostovcikova, and Natalie Galanova
- Subjects
endocrine system ,medicine.medical_specialty ,T-Lymphocytes ,microbiome ,Spleen ,Gut flora ,Severity of Illness Index ,inflammatory bowel diseases ,Permeability ,Article ,Feces ,Immune system ,neutrophils ,Internal medicine ,medicine ,Animals ,Microbiome ,Colitis ,Receptor ,Immunity, Mucosal ,lcsh:QH301-705.5 ,mucosal barrier ,Acute colitis ,metabolites ,Inflammation ,Mice, Inbred BALB C ,biology ,Dextran Sulfate ,Monosaccharides ,General Medicine ,biology.organism_classification ,medicine.disease ,Diet ,Gastrointestinal Microbiome ,DNA-Binding Proteins ,Intestines ,Toll-Like Receptor 4 ,medicine.anatomical_structure ,Endocrinology ,Gene Expression Regulation ,lcsh:Biology (General) ,high-sugar diet ,Chronic Disease ,TLR4 ,Female ,Signal Transduction - Abstract
Diet is a strong modifier of microbiome and mucosal microenvironment in the gut. Recently, components of western-type diets have been associated with metabolic and immune diseases. Here, we studied how high-sugar diet (HSD) consumption influences gut mucosal barrier and immune response under steady state conditions and in a mouse model of acute colitis. We found that HSD significantly increased gut permeability, spleen weight, and neutrophil levels in spleens of healthy mice. Subsequent dextran sodium sulfate administration led to severe colitis. In colon, HSD significantly promoted neutrophil infiltration and increased the levels of IL-6, IL-1&beta, and TNF-&alpha, Moreover, HSD-fed mice had significantly higher abundance of pathobionts, such as Escherichia coli and Candida, in fecal samples. Although germ-free mice colonized with microbiota of conventionally reared mice that consumed different diets had equally severe colitis, mice colonized with HSD microbiota showed markedly increased infiltration of neutrophils to the gut. The induction of colitis in Toll-like receptor 4 (TLR4)-deficient HSD-fed mice led to significantly milder colitis than in wild-type mice. In conclusion, our results suggested a significant role of HSD in disruption of barrier integrity and balanced mucosal and systemic immune response. In addition, these processes seemed to be highly influenced by resident potentially pathogenic microbiota or metabolites via the TLR4 signaling pathway.
- Published
- 2020
- Full Text
- View/download PDF
20. Severity of Experimental Autoimmune Uveitis Is Reduced by Pretreatment with Live Probiotic
- Author
-
Otakar, Dusek, Alena, Fajstova, Aneta, Klimova, Petra, Svozilkova, Tomas, Hrncir, Miloslav, Kverka, Stepan, Coufal, Johan, Slemin, Helena, Tlaskalova-Hogenova, John V, Forrester, and Jarmila, Heissigerova
- Subjects
Inflammation ,experimental autoimmune uveitis ,Probiotics ,eye diseases ,Article ,Escherichia coli Nissle 1917 ,Autoimmune Diseases ,macrophages ,Mice, Inbred C57BL ,Uveitis ,Disease Models, Animal ,Mice ,Escherichia coli ,Animals ,Female ,mucosal immune system ,Intestinal Mucosa - Abstract
Non-infectious uveitis is considered an autoimmune disease responsible for a significant burden of blindness in developed countries and recent studies have linked its pathogenesis to dysregulation of the gut microbiota. We tested the immunomodulatory properties of two probiotics, Escherichia coli Nissle 1917 (EcN) and E. coli O83:K24:H31 (EcO), in a model of experimental autoimmune uveitis (EAU). To determine the importance of bacterial viability and treatment timing, mice were orally treated with live or autoclaved bacteria in both preventive and therapeutic schedules. Disease severity was assessed by ophthalmoscopy and histology, immune phenotypes in mesenteric and cervical lymph nodes were analyzed by flow cytometry and the gut immune environment was analyzed by RT-PCR and/or gut tissue culture. EcN, but not EcO, protected against EAU but only as a live organism and only when administered before or at the time of disease induction. Successful prevention of EAU was accompanied by a decrease in IRBP-specific T cell response in the lymph nodes draining the site of immunization as early as 7 days after the immunization and eye-draining cervical lymph nodes when the eye inflammation became apparent. Furthermore, EcN promoted an anti-inflammatory response in Peyer’s patches, increased gut antimicrobial peptide expression and decreased production of inducible nitric oxide synthase in macrophages. In summary, we show here that EcN controls inflammation in EAU and suggest that probiotics may have a role in regulating the gut–eye axis.
- Published
- 2020
21. Immunomodulatory Components of Human Colostrum and Milk
- Author
-
Miloslav Kverka, Jiří Hrdý, and Helena Tlaskalova-Hogenova
- Subjects
Allergy ,Lacteal ,medicine.medical_treatment ,Inflammation ,Biology ,medicine.disease ,Immune system ,medicine.anatomical_structure ,Cytokine ,Immunity ,Immunology ,medicine ,Colostrum ,medicine.symptom ,Breast feeding - Abstract
Human milk is a unique and complex secretion differing from lacteal secretions of other species. Besides nutrition, it provides protection during the newborn's adaption to the extrauterine environment and reduces the morbidity and mortality caused by both infectious and noninfectious diseases. Its components act directly against infectious agents, but they also accelerate the newborn's immune system development, increasing its capacity for defense and reducing the risk of allergy and other immune-related diseases. Cytokines show the most refined immunomodulatory effects, but oligosaccharides, hormones, and other components affect the newborn's immunity as well. Furthermore, milk components substantially affect the microbial colonization of infant mucosa, which substantially influences the development of all parts of the immune system. All these components act primarily locally, on the mucosal membranes, preventing the penetration of microbes and other antigenic components into the circulation thus ensuring effective defense without the damaging inflammation. Human lacteal secretions contain a number of live cells. Although there are no major differences in the cytokine production between allergic and healthy mothers, they are able to respond to multiple stimuli. By increasing happiness, boosting protective immunity, and decreasing the risk of breast cancer, breastfeeding may have multiple benefits for the mother as well.
- Published
- 2020
22. Probiotic bacteria Escherichia coli Nissle decrease the severity of intraocular inflammation in experimental autoimmune uveitis
- Author
-
Jarmila Heissigerova, Michaela Brichova, Petra Svozilkova, Miloslav Kverka, Tomáš Kučera, Tomas Hrncir, Aneta Klimova, Helena Tlaskalova-Hogenova, Otakar Dusek, Stepan Coufal, and Alena Fajstova
- Subjects
Intraocular inflammation ,Ophthalmology ,business.industry ,medicine ,Probiotic bacteria ,Autoimmune uveitis ,General Medicine ,medicine.disease_cause ,business ,Escherichia coli ,Microbiology - Published
- 2019
23. Occurrence of Serum Antibodies Against Wheat Alpha-Amylase Inhibitor 0.19 in Celiac Disease
- Author
-
S Štěpánová Honzová, Iva Hoffmanová, Petr Halada, Ludmila Tučková, Daniel Sánchez, M Hospodková, Věra Hábová, and Helena Tlaskalova-Hogenova
- Subjects
Adult ,Male ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Antigenicity ,Physiology ,Alpha amylase inhibitor ,Gastroenterology ,law.invention ,Cohort Studies ,Pathogenesis ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Intestinal mucosa ,law ,Internal medicine ,Humans ,Medicine ,Young adult ,Aged ,Plant Proteins ,biology ,business.industry ,General Medicine ,Middle Aged ,respiratory system ,Antibodies, Anti-Idiotypic ,Immunoglobulin A ,respiratory tract diseases ,Celiac Disease ,030104 developmental biology ,Immunoglobulin G ,Recombinant DNA ,biology.protein ,Female ,030211 gastroenterology & hepatology ,Antibody ,business - Abstract
The alcohol-soluble fraction of wheat gluten (gliadins) induces in genetically susceptible individuals immunologically mediated celiac disease (CLD). However, gliadins and related cereal proteins are not unique foodstuff targets of CLD patients´ immune system. Non-gluten wheat alpha-amylase inhibitor 0.19 (AAI 0.19) has been found to be capable of activating human monocyte-derived dendritic cells and inducing pro-inflammatory status in intestinal mucosa of patients with celiac disease (CLD). The possible contribution of this reactivity in incomplete remission of CLD patients on a gluten-free diet (GFD) is matter of contention. In an attempt to characterize the antigenicity of AAI 0.19 in patients with active CLD, patients on a GFD and healthy controls we developed ELISA employing wheat recombinant AAI 0.19. Using this test we revealed a significant (P
- Published
- 2018
24. Oral Microbiota Composition and Antimicrobial Antibody Response in Patients with Recurrent Aphthous Stomatitis
- Author
-
Lydie IzakovičováHollá, Zuzana Jiraskova Zakostelska, Klara Kostovcikova, Stepan Coufal, Petra Borilova Linhartova, Miloslav Kverka, Marketa Libanska, Jirina Bartova, Radka Roubalová, Vojtech Tlaskal, Petra Procházková, Natalie Galanova, Dagmar Schierova, Helena Tlaskalova-Hogenova, Zuzana Stehlikova, Jitka Petanová, Radka Cermakova, Jiri Dvorak, Antonín Fassmann, and Jakub Kreisinger
- Subjects
0301 basic medicine ,Microbiology (medical) ,microbiome ,Recurrent aphthous stomatitis ,medicine.disease_cause ,Microbiology ,Article ,03 medical and health sciences ,mycobiome ,0302 clinical medicine ,Virology ,Medicine ,Labial Mucosa ,Oral mucosa ,Leptotrichia ,biology ,oral mucosa ,business.industry ,Streptococcus ,030206 dentistry ,medicine.disease ,biology.organism_classification ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Fusobacterium ,Malassezia ,business ,Dysbiosis - Abstract
Recurrent aphthous stomatitis (RAS) is the most common disease of the oral mucosa, and it has been recently associated with bacterial and fungal dysbiosis. To study this link further, we investigated microbial shifts during RAS manifestation at an ulcer site, in its surroundings, and at an unaffected site, compared with healed mucosa in RAS patients and healthy controls. We sampled microbes from five distinct sites in the oral cavity. The one site with the most pronounced differences in microbial alpha and beta diversity between RAS patients and healthy controls was the lower labial mucosa. Detailed analysis of this particular oral site revealed strict association of the genus Selenomonas with healed mucosa of RAS patients, whereas the class Clostridia and genera Lachnoanaerobaculum, Cardiobacterium, Leptotrichia, and Fusobacterium were associated with the presence of an active ulcer. Furthermore, active ulcers were dominated by Malassezia, which were negatively correlated with Streptococcus and Haemophilus and positively correlated with Porphyromonas species. In addition, RAS patients showed increased serum levels of IgG against Mogibacterium timidum compared with healthy controls. Our study demonstrates that the composition of bacteria and fungi colonizing healthy oral mucosa is changed in active RAS ulcers, and that this alteration persists to some extent even after the ulcer is healed.
- Published
- 2019
25. The Pros and Cons of Using Oat in a Gluten-Free Diet for Celiac Patients
- Author
-
Adéla Szczepanková, Iva Hoffmanová, Helena Tlaskalova-Hogenova, and Daniel Sánchez
- Subjects
0301 basic medicine ,medicine.medical_specialty ,animal structures ,Avena ,Clinical Decision-Making ,Food Contamination ,lcsh:TX341-641 ,Recommended Dietary Allowances ,Gastroenterology ,Risk Assessment ,Gliadin ,03 medical and health sciences ,Diet, Gluten-Free ,0302 clinical medicine ,gluten-free diet ,gluten-free oat ,Internal medicine ,amylase/trypsin inhibitors ,medicine ,otorhinolaryngologic diseases ,Humans ,Health risk ,oat ,chemistry.chemical_classification ,Discussion ,030109 nutrition & dietetics ,Nutrition and Dietetics ,biology ,business.industry ,High protein ,Patient Selection ,food and beverages ,nutritional and metabolic diseases ,digestive system diseases ,chemistry ,biology.protein ,030211 gastroenterology & hepatology ,Gluten free ,business ,Edible Grain ,Nutritive Value ,lcsh:Nutrition. Foods and food supply ,celiac disease ,Food Science ,Polyunsaturated fatty acid - Abstract
A therapeutic gluten-free diet often has nutritional limitations. Nutritional qualities such as high protein content, the presence of biologically active and beneficial substances (fiber, beta-glucans, polyunsaturated fatty acids, essential amino acids, antioxidants, vitamins, and minerals), and tolerance by the majority of celiac patients make oat popular for use in gluten-free diet. The health risk of long-time consumption of oat by celiac patients is a matter of debate. The introduction of oat into the diet is only recommended for celiac patients in remission. Furthermore, not every variety of oat is also appropriate for a gluten-free diet. The risk of sensitization and an adverse immunologically mediated reaction is a real threat in some celiac patients. Several unsolved issues still exist which include the following: (1) determination of the susceptibility markers for the subgroup of celiac patients who are at risk because they do not tolerate dietary oat, (2) identification of suitable varieties of oat and estimating the safe dose of oat for the diet, and (3) optimization of methods for detecting the gliadin contamination in raw oat used in a gluten-free diet.
- Published
- 2019
26. Microbiota, Microbial Metabolites, and Barrier Function in A Patient with Anorexia Nervosa after Fecal Microbiota Transplantation
- Author
-
Petra Procházková, Marek Kuzma, Martin Bilej, Radka Roubalová, Josef Bulant, Pavel Hrabák, Hana Papezova, Blanka Šedivá, Jiri Dvorak, Martina Cermakova, Eva Meisnerova, Petra Tomášová, Helena Tlaskalova-Hogenova, and Alena Lambertova
- Subjects
0301 basic medicine ,Microbiology (medical) ,microbial metabolites ,short-chain fatty acids ,microbiome ,Case Report ,Microbial dysbiosis ,Microbiology ,small intestinal bacterial overgrowth syndrome ,03 medical and health sciences ,0302 clinical medicine ,Beneficial bacteria ,Virology ,Small intestinal bacterial overgrowth ,Firmicutes/Bacteroides ,medicine ,Akkermansia ,Microbiome ,lcsh:QH301-705.5 ,Barrier function ,biology ,fecal microbiota transplantation (FMT) ,digestive, oral, and skin physiology ,Fecal bacteriotherapy ,biology.organism_classification ,medicine.disease ,030104 developmental biology ,lcsh:Biology (General) ,Anorexia nervosa (differential diagnoses) ,030211 gastroenterology & hepatology - Abstract
The change in the gut microbiome and microbial metabolites in a patient suffering from severe and enduring anorexia nervosa (AN) and diagnosed with small intestinal bacterial overgrowth syndrome (SIBO) was investigated. Microbial gut dysbiosis is associated with both AN and SIBO, and therefore gut microbiome changes by serial fecal microbiota transplantation (FMT) is a possible therapeutic modality. This study assessed the effects of FMT on gut barrier function, microbiota composition, and the levels of bacterial metabolic products. The patient treatment with FMT led to the improvement of gut barrier function, which was altered prior to FMT. Very low bacterial alpha diversity, a lack of beneficial bacteria, together with a great abundance of fungal species were observed in the patient stool sample before FMT. After FMT, both bacterial species richness and gut microbiome evenness increased in the patient, while the fungal alpha diversity decreased. The total short-chain fatty acids (SCFAs) levels (molecules presenting an important source of energy for epithelial gut cells) gradually increased after FMT. Contrarily, one of the most abundant intestinal neurotransmitters, serotonin, tended to decrease throughout the observation period. Overall, gut microbial dysbiosis improvement after FMT was considered. However, there were no signs of patient clinical improvement. The need for an in-depth analysis of the donor´s stool and correct selection pre-FMT is evident.
- Published
- 2019
27. Microbiota, immunity and immunologically-mediated diseases
- Author
-
Jitka Petanová, Zuzana Jiraskova Zakostelska, Miloslav Kverka, and Helena Tlaskalova Hogenova
- Subjects
0301 basic medicine ,Modern medicine ,Oral cavity ,digestive system ,03 medical and health sciences ,Human health ,0302 clinical medicine ,Immune system ,Immunity ,Internal Medicine ,medicine ,Humans ,Microbiome ,business.industry ,Microbiota ,Probiotics ,medicine.disease ,Commensalism ,Gastrointestinal Microbiome ,stomatognathic diseases ,030104 developmental biology ,Immune System Diseases ,030220 oncology & carcinogenesis ,Immunology ,Dysbiosis ,Cardiology and Cardiovascular Medicine ,business - Abstract
Each individual is colonized by broad spectrum of microbes. Recent surge of interest in microbiota across all fields of medicine was motivated by an increasing body of knowledge on how commensals influence human health. This is most notable in the gut, where most microbes reside, but microbes colonizing other niches, such as oral cavity or skin, may influence health as well. Microbiota fundamentally influences the immune system development and its perturbation, i.e. dysbiosis, is associated with many inflammatory, autoimmune and neoplastic diseases. Microbiota forms a symbiotic relationship with the host - maintaining balanced and efficient immune response and protects from colonization by pathogens. Modern medicine may benefit greatly by adopting these ideas for therapeutic or prophylactic purposes. These may include manipulation with microbiota by diet, changes in lifestyle or directly by probiotics or fecal microbiota transfer.
- Published
- 2019
28. Dysbiosis of Skin Microbiota in Psoriatic Patients: Co-occurrence of Fungal and Bacterial Communities
- Author
-
Zuzana Stehlikova, Martin Kostovcik, Klara Kostovcikova, Miloslav Kverka, Katerina Juzlova, Filip Rob, Jana Hercogova, Petr Bohac, Yishay Pinto, Atara Uzan, Omry Koren, Helena Tlaskalova-Hogenova, and Zuzana Jiraskova Zakostelska
- Subjects
Microbiology (medical) ,skin ,Mycobiota ,lcsh:QR1-502 ,medicine.disease_cause ,Microbiology ,lcsh:Microbiology ,03 medical and health sciences ,Lactobacillus ,Psoriasis ,microbiota ,medicine ,Original Research ,030304 developmental biology ,0303 health sciences ,integumentary system ,biology ,030306 microbiology ,psoriasis ,sequencing ,biology.organism_classification ,medicine.disease ,Kocuria ,Elbow Skin ,Malassezia sympodialis ,mycobiota ,Staphylococcus ,Dysbiosis - Abstract
Psoriasis is a chronic inflammatory skin disease, whose pathogenesis involves dysregulated interplay among immune cells, keratinocytes and environmental triggers, including microbiota. Bacterial and fungal dysbiosis has been recently associated with several chronic immune-mediated diseases including psoriasis. In this comprehensive study, we investigated how different sampling sites and methods reflect the uncovered skin microbiota composition. After establishing the most suitable approach, we further examined correlations between bacteria and fungi on the psoriatic skin. We compared microbiota composition determined in the same sample by sequencing two distinct hypervariable regions of the 16S rRNA gene. We showed that using the V3V4 region led to higher species richness and evenness than using the V1V2 region. In particular, genera, such as Staphylococcus and Micrococcus were more abundant when using the V3V4 region, while Planococcaceae, on the other hand, were detected only by the V1V2 region. We performed a detailed analysis of skin microbiota composition of psoriatic lesions, unaffected psoriatic skin, and healthy control skin from the back and elbow. Only a few discriminative features were uncovered, mostly specific for the sampling site or method (swab, scraping, or biopsy). Swabs from psoriatic lesions on the back and the elbow were associated with increased abundance of Brevibacterium and Kocuria palustris and Gordonia, respectively. In the same samples from psoriatic lesions, we found a significantly higher abundance of the fungus Malassezia restricta on the back, while Malassezia sympodialis dominated the elbow mycobiota. In psoriatic elbow skin, we found significant correlation between occurrence of Kocuria, Lactobacillus, and Streptococcus with Saccharomyces, which was not observed in healthy skin. For the first time, we showed here a psoriasis-specific correlation between fungal and bacterial species, suggesting a link between competition for niche occupancy and psoriasis. However, it still remains to be elucidated whether observed microbial shift and specific inter-kingdom relationship pattern are of primary etiological significance or secondary to the disease.
- Published
- 2019
29. Diagnostic pitfalls of celiac disease
- Author
-
Iva Hoffmanová, Helena Tlaskalova-Hogenova, and Daniel Sánchez
- Subjects
Gynecology ,010407 polymers ,medicine.medical_specialty ,Glutens ,business.industry ,Duodenum ,01 natural sciences ,0104 chemical sciences ,Celiac Disease ,Internal Medicine ,medicine ,Humans ,Intestinal Mucosa ,Cardiology and Cardiovascular Medicine ,business ,Autoantibodies - Abstract
Celiac disease is a very common autoimmune disorder caused by the ingestion of dietary gluten products in genetically susceptible persons. Its global prevalence is estimated around 1 %. However, the most cases are not diagnosed. Clinical presentation is widely variable with the involvement of various human systems. Besides a clinical picture (that is often non characteristic), the diagnosis is based on positivity of serological testing (tissue transglutaminase autoantibodies) and histological evaluation of small intestinal mucosa. The article presents a rational diagnostic approach to celiac disease. Key words: celiac disease - Marsh classification - tissue transglutaminase autoantibodies - villous atrophy.
- Published
- 2019
30. Crucial Role of Microbiota in Experimental Psoriasis Revealed by a Gnotobiotic Mouse Model
- Author
-
Zuzana Stehlikova, Klara Kostovcikova, Miloslav Kverka, Pavel Rossmann, Jiri Dvorak, Iva Novosadova, Martin Kostovcik, Stepan Coufal, Dagmar Srutkova, Petra Prochazkova, Tomas Hudcovic, Hana Kozakova, Renata Stepankova, Filip Rob, Katerina Juzlova, Jana Hercogova, Helena Tlaskalova-Hogenova, and Zuzana Jiraskova Zakostelska
- Subjects
Microbiology (medical) ,skin ,medicine.drug_class ,Segmented filamentous bacteria ,Antibiotics ,lcsh:QR1-502 ,Inflammation ,Biology ,Microbiology ,antibiotics ,lcsh:Microbiology ,Pathogenesis ,03 medical and health sciences ,Immune system ,Psoriasis ,medicine ,microbiota ,intestine ,Original Research ,030304 developmental biology ,Bifidobacterium ,0303 health sciences ,030306 microbiology ,animal model ,psoriasis ,medicine.disease ,biology.organism_classification ,imiquimod ,germ-free ,Immunology ,medicine.symptom ,Lactobacillus plantarum - Abstract
Psoriatic patients have altered microbiota, both in the intestine and on the skin. It is not clear, however, whether this is a cause or consequence of the disease. In this study, using an experimental mouse model of psoriasis induced by imiquimod (IMQ), we show that oral treatment with a broad spectrum of antibiotics (MIX) or metronidazole (MET) alone mitigates the severity of skin inflammation through downregulation of Th17 immune response in conventional mice. Since some antibiotics, including MET, can influence immune system reactivity, we also evaluated the effect of MIX in the same model under germ-free (GF) conditions. GF mice treated with MET did not show milder signs of imiquimod-induced skin inflammation (IISI) which supports the conclusion that the therapeutic effect is mediated by changes in microbiota composition. Moreover, compared to controls, mice treated with MIX had a significantly higher abundance of the genus Lactobacillus in the intestine and on the skin. Mice treated with MET had a significantly higher abundance of the genera Bifidobacterium and Enterococcus both on the skin and in the intestine and of Parabacteroides distasonis in the intestine. Additionally, GF mice and mice monocolonized with either Lactobacillus plantarum or segmented filamentous bacteria (SFB) were more resistant to IISI than conventional mice. Interestingly, compared to GF mice, IMQ induced a higher degree of systemic Th17 activation in mice monocolonized with SFB but not with L. plantarum. The present findings provide evidence that intestinal and skin microbiota directly regulates IISI and emphasizes the importance of microbiota in the pathogenesis of psoriasis.
- Published
- 2019
31. Unique Gene Expression Signatures in the Intestinal Mucosa and Organoids Derived from Germ-Free and Monoassociated Mice
- Author
-
Monika Stastna, Klara Kostovcikova, Nikol Baloghova, Hynek Strnad, Jolana Tureckova, Lucie Janeckova, Helena Tlaskalova-Hogenova, Katerina Galuskova, Jiri Svec, Michal Kolar, Eva Sloncova, Jitka Stancikova, Vladimir Korinek, and Tomas Hudcovic
- Subjects
Colon ,Adipose tissue ,Biology ,Catalysis ,Article ,Inorganic Chemistry ,lcsh:Chemistry ,Intestinal mucosa ,ANGPTL4 ,Gene expression ,Organoid ,medicine ,Escherichia coli ,Animals ,Germ-Free Life ,Physical and Theoretical Chemistry ,Intestinal Mucosa ,Onecut2 ,Molecular Biology ,lcsh:QH301-705.5 ,Immunity, Mucosal ,Spectroscopy ,Enricher tool ,Mice, Inbred BALB C ,Gene Expression Profiling ,Microbiota ,Organic Chemistry ,Lipid metabolism ,General Medicine ,Molecular biology ,Small intestine ,Computer Science Applications ,Gene expression profiling ,Organoids ,medicine.anatomical_structure ,lcsh:Biology (General) ,lcsh:QD1-999 ,Gene Expression Regulation ,expression profiling ,Immune System ,monoassociation ,Biomarkers - Abstract
Commensal microbiota contribute to gut homeostasis by inducing transcription of mucosal genes. Analysis of the impact of various microbiota on intestinal tissue provides an important insight into the function of this organ. We used cDNA microarrays to determine the gene expression signature of mucosa isolated from the small intestine and colon of germ-free (GF) mice and animals monoassociated with two E. coli strains. The results were compared to the expression data obtained in conventionally reared (CR) mice. In addition, we analyzed gene expression in colon organoids derived from CR, GF, and monoassociated animals. The analysis revealed that the complete absence of intestinal microbiota mainly affected the mucosal immune system, which was not restored upon monoassociation. The most important expression changes observed in the colon mucosa indicated alterations in adipose tissue and lipid metabolism. In the comparison of differentially expressed genes in the mucosa or organoids obtained from GF and CR mice, only six genes were common for both types of samples. The results show that the increased expression of the angiopoietin-like 4 (Angptl4) gene encoding a secreted regulator of lipid metabolism indicates the GF status.
- Published
- 2019
32. Diet Rich in Animal Protein Promotes Pro-inflammatory Macrophage Response and Exacerbates Colitis in Mice
- Author
-
Klara Kostovcikova, Stepan Coufal, Natalie Galanova, Alena Fajstova, Tomas Hudcovic, Martin Kostovcik, Petra Prochazkova, Zuzana Jiraskova Zakostelska, Martina Cermakova, Blanka Sediva, Marek Kuzma, Helena Tlaskalova-Hogenova, and Miloslav Kverka
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,Colon ,Immunology ,macrophage ,Gut flora ,Adaptive Immunity ,Inflammatory bowel disease ,T-Lymphocytes, Regulatory ,digestive system ,Monocytes ,microbiota, colitis ,Microbiology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,dietary protein ,RAG2 ,medicine ,microbiota ,Immunology and Allergy ,Macrophage ,Animals ,Colitis ,Original Research ,Inflammation ,Mice, Knockout ,Mice, Inbred BALB C ,biology ,Chemistry ,Macrophages ,biology.organism_classification ,medicine.disease ,Acquired immune system ,Ulcerative colitis ,Diet ,Gastrointestinal Microbiome ,DNA-Binding Proteins ,Intestines ,Disease Models, Animal ,030104 developmental biology ,Plant protein ,germ-free ,Th17 Cells ,Female ,Dietary Proteins ,lcsh:RC581-607 ,030215 immunology - Abstract
Diet is a major factor determining gut microbiota composition and perturbances in this complex ecosystem are associated with the inflammatory bowel disease (IBD). Here, we used gnotobiotic approach to analyze, how interaction between diet rich in proteins and gut microbiota influences the sensitivity to intestinal inflammation in murine model of ulcerative colitis. We found that diet rich in animal protein (aHPD) exacerbates acute dextran sulfate sodium (DSS)-induced colitis while diet rich in plant protein (pHPD) does not. The deleterious effect of aHPD was also apparent in chronic DSS colitis and was associated with distinct changes in gut bacteria and fungi. Therefore, we induced acute DSS-colitis in germ-free mice and transferred gut microbiota from aCD or aHPD fed mice to find that this effect requires presence of microbes and aHPD at the same time. The aHPD did not change the number of regulatory T cells or Th17 cells and still worsened the colitis in immuno-deficient RAG2 knock-out mice suggesting that this effect was not dependent on adaptive immunity. The pro-inflammatory effect of aHPD was, however, abrogated when splenic macrophages were depleted with clodronate liposomes. This treatment prevented aHPD induced increase in colonic Ly-6Chigh pro-inflammatory monocytes, but the ratio of resident Ly-6C-/low macrophages was not changed. These data show that the interactions between dietary protein of animal origin and gut microbiota increase sensitivity to intestinal inflammation by promoting pro-inflammatory response of monocytes.
- Published
- 2019
33. Gut Microbiota and NAFLD: Pathogenetic Mechanisms, Microbiota Signatures, and Therapeutic Interventions
- Author
-
Klara Kostovcikova, Jan Krejsek, Pavlína Králíčková, Vladimira Machova, Helena Tlaskalova-Hogenova, Tomas Hrncir, Miloslav Kverka, Robert Hromadka, Eva Trckova, and Lucia Hrncirova
- Subjects
gut microbiota dysbiosis ,0301 basic medicine ,Microbiology (medical) ,QH301-705.5 ,Review ,Disease ,Gut flora ,Chronic liver disease ,Bioinformatics ,digestive system ,Microbiology ,loss of diversity ,liver steatosis ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Virology ,Medicine ,Biology (General) ,faecal microbiota transplantation ,Intestinal permeability ,biology ,intestinal permeability ,business.industry ,cirrhosis ,Fatty liver ,nutritional and metabolic diseases ,hepatocellular carcinoma ,medicine.disease ,biology.organism_classification ,digestive system diseases ,030104 developmental biology ,030211 gastroenterology & hepatology ,Metabolic syndrome ,business ,Dysbiosis - Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease. Its worldwide prevalence is rapidly increasing and is currently estimated at 24%. NAFLD is highly associated with many features of the metabolic syndrome, including obesity, insulin resistance, hyperlipidaemia, and hypertension. The pathogenesis of NAFLD is complex and not fully understood, but there is increasing evidence that the gut microbiota is strongly implicated in the development of NAFLD. In this review, we discuss the major factors that induce dysbiosis of the gut microbiota and disrupt intestinal permeability, as well as possible mechanisms leading to the development of NAFLD. We also discuss the most consistent NAFLD-associated gut microbiota signatures and immunological mechanisms involved in maintaining the gut barrier and liver tolerance to gut-derived factors. Gut-derived factors, including microbial, dietary, and host-derived factors involved in NAFLD pathogenesis, are discussed in detail. Finally, we review currently available diagnostic and prognostic methods, summarise latest knowledge on promising microbiota-based biomarkers, and discuss therapeutic strategies to manipulate the microbiota, including faecal microbiota transplantation, probiotics and prebiotics, deletions of individual strains with bacteriophages, and blocking the production of harmful metabolites.
- Published
- 2021
34. Anti-calreticulin antibodies and calreticulin in sera of patients diagnosed with dilated or hypertrophic cardiomyopathy
- Author
-
Karol Curila, Daniel Sánchez, Iva Hoffmanová, Pavel Gregor, Věra Hábová, Helena Tlaskalova-Hogenova, and Ludmila Tučková
- Subjects
Adult ,Cardiomyopathy, Dilated ,Male ,0301 basic medicine ,genetic structures ,Immunology ,Autoimmunity ,Enzyme-Linked Immunosorbent Assay ,030204 cardiovascular system & hematology ,medicine.disease_cause ,Autoantigens ,law.invention ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,law ,medicine ,Humans ,Immunology and Allergy ,In patient ,cardiovascular diseases ,Aged ,Autoantibodies ,biology ,Hypertrophic cardiomyopathy ,Dilated cardiomyopathy ,Cardiomyopathy, Hypertrophic ,Middle Aged ,equipment and supplies ,medicine.disease ,Immunoglobulin A ,030104 developmental biology ,Pepscan ,Case-Control Studies ,Immunoglobulin G ,cardiovascular system ,biology.protein ,Recombinant DNA ,Female ,Antibody ,Calreticulin ,Biomarkers ,circulatory and respiratory physiology - Abstract
Distinct cellular level of the Ca2+-binding chaperone calreticulin (CRT) is essential for correct embryonal cardiac development and postnatal function. However, CRT is also a potential autoantigen eliciting formation of antibodies (Ab), whose role is not yet clarified. Immunization with CRT leads to cardiac injury, while overexpression of CRT in cardiomyocytes induces dilated cardiomyopathy (DCM) in animals. Hence, we analysed levels of anti-CRT Ab and calreticulin in the sera of patients with idiopatic DCM and hypertrophic cardiomyopathy (HCM). ELISA and immunoblot using human recombinant CRT and Pepscan with synthetic, overlapping decapeptides of CRT were used to detect anti-CRT Ab. Serum CRT concentration was tested by ELISA. Significantly increased levels of anti-CRT Ab of isotypes IgA (p
- Published
- 2016
35. Effect of Lactobacillus casei on the Pharmacokinetics of Amiodarone in Male Wistar Rats
- Author
-
Zuzana Matuskova, Pavel Anzenbacher, Helena Tlaskalova-Hogenova, Rostislav Vecera, Michal Siller, Jan Strojil, and Eva Anzenbacherova
- Subjects
Male ,Lactobacillus casei ,Clinical chemistry ,medicine.medical_treatment ,Metabolite ,Administration, Oral ,Amiodarone ,Pharmacology ,030226 pharmacology & pharmacy ,law.invention ,03 medical and health sciences ,Probiotic ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,law ,medicine ,Animals ,Pharmacology (medical) ,Saline ,biology ,business.industry ,Probiotics ,Amiodarone Hydrochloride ,biology.organism_classification ,Rats ,Lacticaseibacillus casei ,chemistry ,030220 oncology & carcinogenesis ,business ,medicine.drug - Abstract
The probiotic bacterium Escherichia coli strain Nissle 1917 has previously been shown to alter the pharmacokinetics of amiodarone. The aim of this study was to determine whether the probiotic bacterium Lactobacillus casei produces similar alterations in amiodarone disposition. A suspension of live probiotic bacteria L. casei strain DN-114 001 (1.5 × 109 CFU/dose; probiotic pre-treated group) or a saline solution (control group) was administered directly into the stomach of male Wistar rats (N = 30 in each group) by oral gavage daily for 7 consecutive days. On the eighth day, all rats (N = 60) were given a single oral dose of an amiodarone hydrochloride suspension (model drug; 50 mg/kg). The concentrations of amiodarone and of its main metabolite N-desethylamiodarone were determined in rat plasma by high-performance liquid chromatography. Comparison of the pharmacokinetics of amiodarone in the control group and probiotic pre-treated group revealed that the peak plasma concentration of amiodarone was delayed by >2 h in the probiotic pre-treated group. The plasma level of N-desethylamiodarone was unchanged in the probiotic pre-medicated group and its pharmacokinetic parameters were not altered. The slower absorption of amiodarone in the probiotic pre-treated rats compared to the control ones and the unchanged pharmacokinetics of its main metabolite suggest that the probiotic strain of L. casei DN-114 001 has probably no clinical consequences as the difference was not statistically significant.
- Published
- 2016
36. Anorexia nervosa: Gut microbiota-immune-brain interactions
- Author
-
Petra Procházková, Martin Bilej, Helena Tlaskalova-Hogenova, Radka Roubalová, Kvido Smitka, and Hana Papežová
- Subjects
0301 basic medicine ,Anorexia Nervosa ,030209 endocrinology & metabolism ,Disease ,Gut flora ,Critical Care and Intensive Care Medicine ,digestive system ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Humans ,030109 nutrition & dietetics ,Nutrition and Dietetics ,biology ,business.industry ,Brain ,medicine.disease ,biology.organism_classification ,Anorexia nervosa (differential diagnoses) ,Immunology ,business ,Neurohormones ,Dysbiosis ,Psychoneuroimmunology - Abstract
Anorexia nervosa is a psychiatric disorder defined by an extremely low body weight, a devastating fear of weight gain, and body image disturbance, however the etiopathogenesis remains unclear. The objective of the article is to provide a comprehensive review on the potential role of gut microbiota in pathogenesis of anorexia nervosa. Recent advances in sequencing techniques used for microbial detection revealed that this disease is associated with disruption of the composition of normal gut microbiota (dysbiosis), manifested by low microbial diversity and taxonomic differences as compared to healthy individuals. Microorganisms present in the gut represent a part of the so called "microbiota-gut-brain" axis that affect the central nervous system and thus human behavior via the production of various neuroactive compounds. In addition, cells of the immune system are equipped with receptors for these neuroactive substances. Microbiota of the intestinal system also represent a very important antigenic source. These antigens can mimic some host neuropeptides and neurohormones and thus trigger the production of autoantibodies which cross-react with these compounds. The levels and affinities of these antibodies are thought to be associated with neuropsychiatric conditions including anxiety, depression, and eating and sleep disorders. The study of microbiota function in diseases could bring new insights to the pathogenetic mechanisms.
- Published
- 2018
37. Oral Bacterial and Fungal Microbiome Impacts Colorectal Carcinogenesis
- Author
-
Zuzana Jiraskova Zakostelska, Helena Tlaskalova-Hogenova, and Klara Klimesova
- Subjects
0301 basic medicine ,Microbiology (medical) ,lcsh:QR1-502 ,microbiome ,Review ,Microbiology ,biofilm ,lcsh:Microbiology ,03 medical and health sciences ,mycobiome ,0302 clinical medicine ,Immune system ,medicine ,oral diseases ,Microbiome ,Pathogen ,Barrier function ,biology ,Obligate ,Biofilm ,dysbiosis ,Fusobacterium ,medicine.disease ,biology.organism_classification ,stomatognathic diseases ,030104 developmental biology ,030220 oncology & carcinogenesis ,pathobiont ,Dysbiosis - Abstract
Host’s physiology is significantly influenced by microbiota colonizing the epithelial surfaces. Complex microbial communities contribute to proper mucosal barrier function, immune response, and prevention of pathogen invasion and have many other crucial functions. The oral cavity and large intestine are distant parts of the digestive tract, both heavily colonized by commensal microbiota. Nevertheless, they feature different proportions of major bacterial and fungal phyla, mostly due to distinct epithelial layers organization and different oxygen levels. A few obligate anaerobic strains inhabiting the oral cavity are involved in the pathogenesis of oral diseases. Interestingly, these microbiota components are also enriched in gut inflammatory and tumor tissue. An altered microbiota composition – dysbiosis – and formation of polymicrobial biofilms seem to play important roles in the development of oral diseases and colorectal cancer. In this review, we describe the differences in composition of commensal microbiota in the oral cavity and large intestine and the mechanisms by which microbiota affect the inflammatory and carcinogenic response of the host.
- Published
- 2018
38. Intestinal Microbiota: Facts and Fiction
- Author
-
Helena Tlaskalova-Hogenova and Miloslav Kverka
- Subjects
0301 basic medicine ,biology ,Gastrointestinal Microbiome ,Gastroenterology ,Inflammatory Bowel Diseases ,Zoology ,General Medicine ,Gut flora ,medicine.disease ,biology.organism_classification ,Complex ecosystem ,Microbiology ,03 medical and health sciences ,030104 developmental biology ,Immune system ,Immune System Diseases ,medicine ,Humans ,Dysbiosis ,Bacteria ,Archaea - Abstract
In humans, the gut microbiota forms a complex ecosystem consisting of a vast number of bacteria, Archaea, fungi and viruses. It represents a major stimulus to the development of the immune system and many other physiological functions, so that it may shape the individual's susceptibility to infectious and immune-mediated diseases. The emergence of new ‘-omics' methods recently revolutionized the way we study the host-microbe interactions, but they also raised new questions and issues. In this review, we discuss the impact of these new data on the current and future therapies for chronic inflammatory diseases. We also outline the major conceptual, technical and interpretational issues that recently led to some misleading conclusions and discuss in brief the current research directions in the field.
- Published
- 2017
39. The effect of probiotic Escherichia coli strain Nissle 1917 lipopolysaccharide on the 5-aminosalicylic acid transepithelial transport across Caco-2 cell monolayers
- Author
-
Zbyněk Svoboda, Zdeněk Zídek, Věra Štětinová, Libuse Smetanova, Jaroslav Květina, Dagmar Kholova, and Helena Tlaskalova-Hogenova
- Subjects
Lipopolysaccharides ,Aminosalicylic acid ,Lipopolysaccharide ,Physiology ,Metabolite ,Intracellular Space ,Biophysics ,medicine.disease_cause ,High-performance liquid chromatography ,Permeability ,law.invention ,Microbiology ,Probiotic ,chemistry.chemical_compound ,law ,Escherichia coli ,medicine ,Humans ,Mesalamine ,Probiotics ,Anti-Inflammatory Agents, Non-Steroidal ,Biological Transport ,Epithelial Cells ,General Medicine ,In vitro ,chemistry ,Caco-2 ,Culture Media, Conditioned ,Caco-2 Cells - Abstract
The object of this study was to investigate the effect of probiotic Escherichia coli strain Nissle 1917 (EcN) (i) EcN lipopolysaccharide (EcN LPS) and (ii) bacteria-free supernatant of EcN suspension (EcN supernatant) on in vitro transepithelial transport of mesalazine (5-aminosalicylic acid, 5-ASA), the most commonly prescribed anti-inflammatory drug in inflammatory bowel disease (IBD). Effect of co-administered EcN LPS (100 µg/ml) or EcN supernatant (50 µg/ml) on the 5-ASA transport (300 µmol/l) was studied using the Caco-2 monolayer (a human colon carcinoma cell line) as a model of human intestinal absorption. Permeability characteristics for absorptive and secretory transport of parent drug and its intracellularly-formed metabolite were determined. The quantification of 5-ASA and its main metabolite N-acetyl-5-amino-salicylic acid (N-Ac-5-ASA) was performed by high performance liquid chromatography. Obtained results suggest that neither EcN LPS nor EcN supernatant had effect on the total 5-ASA transport (secretory flux greater than absorptive flux) and on the transport of intracellularly formed N-Ac-5-ASA (preferentially transported in the secretory direction). The percent cumulative transport of the total 5-ASA alone or in combination with EcN LPS or EcN supernatant did not exceed 1%.
- Published
- 2014
40. Inflammatory Bowel Disease Types Differ in Markers of Inflammation, Gut Barrier and in Specific Anti-Bacterial Response
- Author
-
Zuzana Stehlikova, Helena Tlaskalova-Hogenova, Klara Kostovcikova, Lukas Bajer, Dagmar Schierova, Pavel Drastich, Zuzana Jiraskova Zakostelska, Zuzana Jackova, Zuzana Gajdarova, Natalie Galanova, Miloslav Kverka, and Stepan Coufal
- Subjects
Adult ,Male ,0301 basic medicine ,Cholangitis, Sclerosing ,T cells ,Inflammation ,digestive system ,Inflammatory bowel disease ,Article ,Primary sclerosing cholangitis ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Crohn Disease ,Osteoprotegerin ,inflammatory bowel disease ,microbiota ,medicine ,Humans ,antibodies ,lcsh:QH301-705.5 ,gut barrier ,business.industry ,biomarkers ,General Medicine ,Middle Aged ,medicine.disease ,Ulcerative colitis ,digestive system diseases ,030104 developmental biology ,lcsh:Biology (General) ,Immunology ,Dysbiosis ,Colitis, Ulcerative ,Female ,030211 gastroenterology & hepatology ,medicine.symptom ,business - Abstract
Crohn&rsquo, s disease (CD), ulcerative colitis (UC) and inflammatory bowel disease (IBD) associated with primary sclerosing cholangitis (PSC-IBD), share three major pathogenetic mechanisms of inflammatory bowel disease (IBD)-gut dysbiosis, gut barrier failure and immune system dysregulation. While clinical differences among them are well known, the underlying mechanisms are less explored. To gain an insight into the IBD pathogenesis and to find a specific biomarker pattern for each of them, we used protein array, ELISA and flow cytometry to analyze serum biomarkers and specific anti-microbial B and T cell responses to the gut commensals. We found that decrease in matrix metalloproteinase (MMP)-9 and increase in MMP-14 are the strongest factors discriminating IBD patients from healthy subjects and that PSC-IBD patients have higher levels of Mannan-binding lectin, tissue inhibitor of metalloproteinases 1 (TIMP-1), CD14 and osteoprotegerin than patients with UC. Moreover, we found that low transforming growth factor-&beta, 1 (TGF-&beta, 1) is associated with disease relapse and low osteoprotegerin with anti-tumor necrosis factor-alpha (TNF-&alpha, ) therapy. Patients with CD have significantly decreased antibody and increased T cell response mainly to genera Eubacterium, Faecalibacterium and Bacteroides. These results stress the importance of the gut barrier function and immune response to commensal bacteria and point at the specific differences in pathogenesis of PSC-IBD, UC and CD.
- Published
- 2019
41. Segmented filamentous bacteria in a defined bacterial cocktail induce intestinal inflammation in SCID mice reconstituted with CD45RBhigh CD4+ T cells
- Author
-
Simon Read, Magda Strus, Tomas Hrncir, Olga Kofronova, Tomas Hudcovic, Paul W. Bland, Helena Tlaskalova-Hogenova, Fiona Powrie, Pioter Heczko, Oldrich Benada, Holm H. Uhlig, Z Reháková, Hana Kozakova, and Renata Stepankova
- Subjects
CD4-Positive T-Lymphocytes ,Adoptive cell transfer ,Segmented filamentous bacteria ,Spleen ,Mice, SCID ,Biology ,Inflammatory bowel disease ,Microbiology ,Mice ,Fusobacterium mortiferum ,Intestinal mucosa ,medicine ,Immunology and Allergy ,Animals ,Colitis ,Intestinal Mucosa ,In Situ Hybridization, Fluorescence ,Severe combined immunodeficiency ,Mice, Inbred BALB C ,Hyperplasia ,Gastroenterology ,Hypertrophy ,medicine.disease ,Flow Cytometry ,Adoptive Transfer ,Disease Models, Animal ,medicine.anatomical_structure ,Microscopy, Electron, Scanning ,Leukocyte Common Antigens - Abstract
Background: The aim was to analyze the influence of intestinal microbiota on the development of intestinal inflammation. We used the model of chronic inflammation that develops spontaneously in the colon of conventional severe combined immunodeficiency (SCID) mice restored with the CD45 RBhigh subset of CD4+T cells isolated from the spleen of normal BALB/c mice. Methods: A CD4+CD45RBhigh subpopulation of T cells was purified from the spleen of conventional BALB/c mice by magnetic separation (MACS) and transferred into immunodeficient SCID mice. Germ-free (GF) SCID mice or SCID mice monoassociated with Enterococcus faecalis, SFB (segmented filamentous bacteria), Fusobacterium mortiferum, Bacteroides distasonis, and in combination Fusobacterium mortiferum + SFB or Bacteroides distasonis + SFB were used as recipients. SCID mice were colonized by a defined cocktail of specific pathogen-free (SPF) bacteria. Mice were evaluated 8–12 weeks after the cell transfer for clinical and morphological signs of inflammatory bowel disease (IBD). Results: After the transfer of the CD4+CD45RBhigh T-cell subpopulation to SCID mice severe colitis was present in conventional animals and in mice colonized with a cocktail of SPF microflora plus SFB. Altered intestinal barrier in the terminal ileum of mice with severe colitis was documented by immunohistology using antibodies to ZO-1 (zona occludens). Conclusions: Only SFB bacteria together with a defined SPF mixture were effective in triggering intestinal inflammation in the model of IBD in reconstituted SCID mice, while no colitis was detected in GF mice or in mice colonized either with SPF microflora or monoassociated only with SFB or colonized by Bacteroides distasonis + SFB or Fusobacterium mortiferum + SFB. (Inflamm Bowel Dis 2007)
- Published
- 2016
42. Differential activity of IL-12 and IL-23 in mucosal and systemic innate immune pathology
- Author
-
Brent S. McKenzie, Daniel J. Cua, Barbara Joyce-Shaikh, Nicolas Robinson, Fiona Powrie, Holm H. Uhlig, Sophie Hue, Renata Stepankova, Helena Tlaskalova-Hogenova, Claire Thompson, and Sofia Buonocore
- Subjects
Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,HUMDISEASE ,Biology ,Systemic inflammation ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Interleukin 23 ,medicine ,Immunology and Allergy ,030304 developmental biology ,0303 health sciences ,Innate immune system ,Innate lymphoid cell ,3. Good health ,Cytokine ,Infectious Diseases ,Mucosal immunology ,CELLIMMUNO ,medicine.symptom ,030215 immunology - Abstract
The CD40-CD154 pathway is important in the pathogenesis of inflammatory bowel disease. Here we show that injection of an agonistic CD40 mAb to T and B cell-deficient mice was sufficient to induce a pathogenic systemic and intestinal innate inflammatory response that was functionally dependent on tumor necrosis factor-alpha and interferon-gamma as well as interleukin-12 p40 and interleukin-23 p40 secretion. CD40-induced colitis, but not wasting disease or serum proinflammatory cytokine production, depended on interleukin-23 p19 secretion, whereas interleukin-12 p35 secretion controlled wasting disease and serum cytokine production but not mucosal immunopathology. Intestinal inflammation was associated with IL-23 (p19) mRNA-producing intestinal dendritic cells and IL-17A mRNA within the intestine. Our experiments identified IL-23 as an effector cytokine within the innate intestinal immune system. The differential role of IL-23 in local but not systemic inflammation suggests that it may make a more specific target for the treatment of IBD.
- Published
- 2016
43. The Microbiota Determines Susceptibility to Experimental Autoimmune Uveoretinitis
- Author
-
Helena Tlaskalova-Hogenova, Tomas Hrncir, Jarmila Heissigerova, Petra Seidler Stangova, Petra Svozilkova, John V. Forrester, Aneta Klimova, Renata Stepankova, and Miloslav Kverka
- Subjects
0301 basic medicine ,Male ,Pathology ,Adaptive Immunity ,Eye ,Lymphocyte Activation ,Autoantigens ,T-Lymphocytes, Regulatory ,Mice ,0302 clinical medicine ,Immunophenotyping ,Immunology and Allergy ,Interferon gamma ,medicine.diagnostic_test ,Microbiota ,Interleukin-17 ,General Medicine ,Acquired immune system ,Flow Cytometry ,Anti-Bacterial Agents ,Female ,Interleukin 17 ,Lymph ,medicine.symptom ,medicine.drug ,Research Article ,lcsh:Immunologic diseases. Allergy ,medicine.medical_specialty ,Article Subject ,Immunology ,Inflammation ,Biology ,Retina ,Flow cytometry ,Autoimmune Diseases ,Uveitis ,03 medical and health sciences ,Interferon-gamma ,Immune system ,medicine ,Animals ,Germ-Free Life ,Eye Proteins ,Macrophages ,Retinitis ,Bacterial Load ,eye diseases ,Mice, Inbred C57BL ,Retinol-Binding Proteins ,Disease Models, Animal ,030104 developmental biology ,sense organs ,lcsh:RC581-607 ,030215 immunology - Abstract
The microbiota is a crucial modulator of the immune system. Here, we evaluated how its absence or reduction modifies the inflammatory response in the murine model of experimental autoimmune uveoretinitis (EAU). We induced EAU in germ-free (GF) or conventionally housed (CV) mice and in CV mice treated with a combination of broad-spectrum antibiotics either from the day of EAU induction or from one week prior to induction of disease. The severity of the inflammation was assessed by fundus biomicroscopy or by histology, including immunohistology. The immunophenotyping of T cells in local and distant lymph nodes was performed by flow cytometry. We found that GF mice and mice where the microbiota was reduced one week before EAU induction were protected from severe autoimmune inflammation. GF mice had lower numbers of infiltrating macrophages and significantly less T cell infiltration in the retina than CV mice with EAU. GF mice also had reduced numbers of IFN-γand IL-17-producing T cells and increased numbers of regulatory T cells in the eye-draining lymph nodes. These data suggest that the presence of microbiota during autoantigen recognition regulates the inflammatory response by influencing the adaptive immune response.
- Published
- 2016
44. Intestinal Microbiota Promotes Psoriasis-Like Skin Inflammation by Enhancing Th17 Response
- Author
-
Jana Malkova, Klara Klimesova, Renata Stepankova, Zuzana Stehlikova, Helena Tlaskalova-Hogenova, Martin Kostovčík, Jana Hercogová, Michaela Hornova, Iva Novosádová, Kateřina Jůzlová, Miloslav Kverka, Tomas Hudcovic, Pavel Rossmann, and Zuzana Jiraskova Zakostelska
- Subjects
0301 basic medicine ,Gene Expression ,lcsh:Medicine ,Imiquimod ,Gut flora ,Pathology and Laboratory Medicine ,Lymphocyte Activation ,030207 dermatology & venereal diseases ,White Blood Cells ,Mice ,0302 clinical medicine ,Antibiotics ,Animal Cells ,Lactobacillales ,Medicine and Health Sciences ,lcsh:Science ,Immune Response ,Skin ,Clostridiales ,Mice, Inbred BALB C ,Multidisciplinary ,Antimicrobials ,T Cells ,Interleukin-17 ,Drugs ,Receptors, Antigen, T-Cell, gamma-delta ,Animal Models ,Genomics ,Nuclear Receptor Subfamily 1, Group F, Member 3 ,Anti-Bacterial Agents ,Actinobacteria ,medicine.anatomical_structure ,Medical Microbiology ,Aminoquinolines ,Female ,Interleukin 17 ,medicine.symptom ,Cellular Types ,medicine.drug ,Research Article ,T cell ,Inflammatory Diseases ,Immune Cells ,Immunology ,Inflammation ,Mouse Models ,Microbial Genomics ,Biology ,Research and Analysis Methods ,Microbiology ,Autoimmune Diseases ,03 medical and health sciences ,Immune system ,Model Organisms ,Signs and Symptoms ,Species Specificity ,Diagnostic Medicine ,Psoriasis ,Microbial Control ,medicine ,Genetics ,Animals ,Germ-Free Life ,Humans ,Microbiome ,Pharmacology ,Blood Cells ,Bacteria ,Gut Bacteria ,lcsh:R ,Organisms ,Biology and Life Sciences ,Cell Biology ,medicine.disease ,biology.organism_classification ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Th17 Cells ,Clinical Immunology ,lcsh:Q ,Clinical Medicine - Abstract
Psoriasis is a chronic inflammatory skin disease in which Th17 cells play a crucial role. Since indigenous gut microbiota influences the development and reactivity of immune cells, we analyzed the link among microbiota, T cells and the formation of psoriatic lesions in the imiquimod-induced murine model of psoriasis. To explore the role of microbiota, we induced skin inflammation in germ-free (GF), broad-spectrum antibiotic (ATB)-treated or conventional (CV) BALB/c and C57BL/6 mice. We found that both mice reared in GF conditions for several generations and CV mice treated with ATB were more resistant to imiquimod-induced skin inflammation than CV mice. The ATB treatment dramatically changed the diversity of gut bacteria, which remained stable after subsequent imiquimod application; ATB treatment resulted in a substantial increase in the order Lactobacillales and a significant decrease in Coriobacteriales and Clostridiales. Moreover, as compared to CV mice, imiquimod induced a lower degree of local and systemic Th17 activation in both GF and ATB-treated mice. These findings suggest that gut microbiota control imiquimod-induced skin inflammation by altering the T cell response.
- Published
- 2016
45. Two faces of microbiota in inflammatory and autoimmune diseases: triggers and drugs
- Author
-
Miloslav Kverka and Helena Tlaskalova-Hogenova
- Subjects
Microbiology (medical) ,Allergy ,Inflammation ,Biology ,medicine.disease_cause ,Inflammatory bowel disease ,Autoimmune Diseases ,Pathology and Forensic Medicine ,Autoimmunity ,law.invention ,Probiotic ,Immune system ,law ,Hypersensitivity ,medicine ,Humans ,Immunology and Allergy ,Microbiome ,Intestinal Mucosa ,Periodontal Diseases ,Probiotics ,General Medicine ,Inflammatory Bowel Diseases ,medicine.disease ,Gastrointestinal Tract ,Celiac Disease ,Host-Pathogen Interactions ,Immunology ,Metagenome ,medicine.symptom - Abstract
The prevalence of chronic autoimmune and inflammatory diseases, such as inflammatory bowel disease, allergies, or rheumatic diseases, is steadily increasing in developed countries. This increase is probably accelerated by environmental factors, such as decrease in infectious burden or changes in food processing. These lifestyle changes then strongly influence the strongest stimulus for the immune system - commensal microbiota. Despite the differences in the affected organ, the immune-mediated diseases have one or more factors in common - microbe either as a trigger or as a protector, mucosal barrier dysfunction, and dysregulation of the immune system. The core questions, which microbes are involved and how these diseases can be cured or even prevented still remain unsolved. Powered by the recent progress in technology, by new insights into the function of immune system, by advances in microbiome research, and extended use of gnotobiological techniques, these mechanisms are now being unravelled and new therapeutic possibilities are emerging. To secure their niche, the microbes devised many ingenious ways, how to dampen the inflammation. Nonpathogenic microorganisms or microbial components isolated from probiotic, commensal or even pathogenic microbes could be, therefore, used to interfere with the pathogenetic mechanisms of immune-mediated diseases.
- Published
- 2012
46. Increased expression of chemokine receptors CCR1 and CCR3 in nasal polyps: molecular basis for recruitment of the granulocyte infiltrate
- Author
-
D. Kovář, M. Navara, David P. Funda, R. Holý, Petra Fundová, and Helena Tlaskalova-Hogenova
- Subjects
CCR1 ,Chemokine ,Stromal cell ,Receptors, CCR3 ,CCR3 ,Receptors, CCR1 ,Mucous membrane of nose ,General Medicine ,Biology ,medicine.disease ,Microbiology ,Eosinophils ,Pathogenesis ,Nasal Mucosa ,Chemokine receptor ,Nasal Polyps ,Immunology ,medicine ,biology.protein ,Humans ,Nasal polyps ,Granulocytes - Abstract
Inflammatory processes play an important role in the development of nasal polyps (NP), but the etiology and, to a high degree also, the pathogenesis of NP are not fully understood. The role of several cytokines and chemokines such as eotaxins, IL-4, IL-5, IL-6, IL-8, and RANTES has been reported in NP. Herewith, we investigated the expression and pattern of distribution of chemokine receptors CCR1 and CCR3 in nasal polyps. Immunohistochemical detection was carried out in frozen sections of biopsies from 22 NP and 18 nasal mucosa specimens in both the epithelial and stromal compartments. Fluorescence microscopy and computerized image analysis revealed a statistically significant increased number of CCR1 (45.2 ± 2.8 vs. 15.1 ± 1.9, p
- Published
- 2012
47. Colostrum of Healthy Mothers Contains Broad Spectrum of Secretory IgA Autoantibodies
- Author
-
Miloslav Kverka, Jaroslava Pribylova, Klara Klimesova, Helena Tlaskalova-Hogenova, Ingrid Kocourkova, Klara Krausova, and Pavel Rossmann
- Subjects
Adult ,Immunoglobulin A ,Adolescent ,animal diseases ,Immunology ,Mothers ,Enzyme-Linked Immunosorbent Assay ,Autoantigens ,Broad spectrum ,fluids and secretions ,Immune system ,Antibody Specificity ,Pregnancy ,Animals ,Humans ,Lactation ,Immunology and Allergy ,Secretory IgA ,Autoantibodies ,biology ,Colostrum ,Autoantibody ,Infant ,Proteins ,food and beverages ,Haplorhini ,Immunohistochemistry ,Rats ,Breast Feeding ,Immunoglobulin A, Secretory ,biology.protein ,Female ,Antibody ,Breast feeding ,Protein Binding - Abstract
Human colostrum and milk provide a newborn with immunomodulatory components, ensuring protection and proper development of the immune system. Secretory IgA antibodies in colostrum represent the first line of defence against harmful substances, but their potential spectra of reactivity with autoantigens remains unclear. Here, we characterised the repertoire of natural sectretory IgA autoantibodies in colostrum of healthy mothers.The human colostrum samples from 39 healthy mothers were analyzed for autoantibodies by indirect immunofluorescence, dot blots, immunoblots and ELISA.We found that there is high diversity in reactivities of colostral IgA antibodies to autoantigens among individual samples. Using tissue sections and biochips commonly used for autoimmunity testing, we found that most samples reacted with monkey ovary (79.3%), monkey pancreatic tissue (78.6%), human HEp-2 cells (69%) and monkey adrenal gland (69.0%), fewer samples reacted with monkey liver tissue (47.2%), rat stomach (42.9%), monkey testicular tissue (41.4%), monkey salivary gland (39.3%), rat kidney (32.1%) and monkey cerebellar tissue (17.9%). At the protein level, we detected reactivity of IgA with 21 out of 25 (auto) antigens. The majority of the samples reacted with the pyruvate dehydrogenase complex, E3 ubiquitin ligase, cytosolic liver antigen, promyelocytic leukemia protein and nuclear pore glycoprotein-210. Using ELISA, we found reactivity of colostral IgA antibodies against examined extractable nuclear antigens, double stranded DNA, phospholipids and neutrophil cytoplasm.The broad spectrum of polyreactive natural autoantibodies present in human colostrum may contribute to proper development of mucosal immune system of the breastfed infant.
- Published
- 2012
48. Spontaneous in vitro IL-6 production in various intestinal segments in patients with inflammatory bowel disease
- Author
-
P. Drastich, Helena Tlaskalova-Hogenova, J. Spicak, L. Frolova-Brizova, and P. Zanvit
- Subjects
Adult ,Male ,medicine.medical_specialty ,Rectum ,Enzyme-Linked Immunosorbent Assay ,Inflammation ,Polymerase Chain Reaction ,Microbiology ,Inflammatory bowel disease ,Gastroenterology ,Cecum ,Crohn Disease ,Internal medicine ,Biopsy ,medicine ,Humans ,RNA, Messenger ,Intestinal Mucosa ,Colitis ,Interleukin 6 ,Cells, Cultured ,biology ,medicine.diagnostic_test ,Interleukin-6 ,business.industry ,General Medicine ,Middle Aged ,medicine.disease ,Ulcerative colitis ,medicine.anatomical_structure ,biology.protein ,Colitis, Ulcerative ,Female ,medicine.symptom ,business - Abstract
Interleukin-6 (IL-6) plays an important role in regulation of intestinal inflammatory processes in inflammatory bowel disease (IBD). The levels of IL-6 in media from cultured biopsy samples were determined by ELISA in 14 Crohn’s disease (CD) patients, 17 patients with ulcerative colitis (UC), and 24 healthy controls in terminal ileum, cecum, and rectum. Results were confirmed by measuring mRNA expression in selected patients. In CD patients, there were increased levels of IL-6 (expressed in picograms per milligram of biopsy tissue mass) in terminal ileum compared with controls (median, 617 vs. 90.4; p
- Published
- 2011
49. Negative regulation of Toll-like receptor signaling plays an essential role in homeostasis of the intestine
- Author
-
Jeanette Wilmanski, Huamei Forsman, Koichi Kobayashi, Helena Tlaskalova-Hogenova, Liming Hao, Tomas Hrncir, and Amlan Biswas
- Subjects
Immunology ,Caspase 1 ,Biology ,Article ,Mice ,Immune system ,medicine ,Animals ,Homeostasis ,Immunology and Allergy ,Colitis ,Toll-like receptor ,Toll-Like Receptors ,medicine.disease ,Interleukin-10 ,Intestines ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,Disease Models, Animal ,Interleukin 10 ,Interleukin-1 Receptor-Associated Kinases ,Myeloid Differentiation Factor 88 ,TLR4 ,Signal transduction ,Signal Transduction - Abstract
A healthy intestinal tract is characterized by controlled homeostasis due to the balanced interaction between commensal bacteria and the host mucosal immune system. Human and animal model studies have supported the hypothesis that breakdown of this homeostasis may underlie the pathogenesis of inflammatory bowel diseases (IBDs). However it is not well understood how intestinal microflora stimulate the intestinal mucosal immune system and how such activation is regulated. Using a spontaneous, commensal bacteria-dependent colitis model in IL-10-deficient mice, we investigated the role of Toll-like receptors (TLRs) and their negative regulation in intestinal homeostasis. In addition to IL-10−/−MyD88−/− mice, IL-10−/−TLR4−/− mice exhibited reduced colitis compared to IL-10−/− mice, indicating that TLR4 signaling plays an important role in inducing colitis. Interestingly, the expression of IRAK-M, a negative regulator of TLR signaling, is dependent on intestinal commensal flora, as IRAK-M expression was reduced in mice re-derived into a germ-free environment, and introduction of commensal bacteria into germ-free mice induced IRAK-M expression. IL-10−/−IRAK-M−/− mice exhibited exacerbated colitis with increased inflammatory cytokine gene expression. Therefore, this study indicates that intestinal microflora stimulate the colitogenic immune system through TLRs and negative regulation of TLR signaling is essential in maintaining intestinal homeostasis.
- Published
- 2010
50. Neonatal colonization of mice with Lactobacillus plantarum producing the aeroallergen Bet v 1 biases towards Th1 and T-regulatory responses upon systemic sensitization
- Author
-
Martin Schwarzer, Andreas Repa, Bruno Pot, Irma Schabussova, Tomas Hrncir, Ursula Wiedermann, Renata Stepankova, Tomas Hudcovic, Hana Kozakova, Helena Tlaskalova-Hogenova, Catherine Daniel, and Arnold Pollak
- Subjects
0303 health sciences ,Allergy ,030306 microbiology ,Immunology ,FOXP3 ,chemical and pharmacologic phenomena ,Aeroallergen ,Spleen ,Biology ,medicine.disease_cause ,Immunoglobulin E ,medicine.disease ,3. Good health ,Microbiology ,03 medical and health sciences ,Allergen ,medicine.anatomical_structure ,Immune system ,medicine ,biology.protein ,Immunology and Allergy ,Sensitization ,030304 developmental biology - Abstract
To cite this article: Schwarzer M, Repa A, Daniel C, Schabussova I, Hrncir T, Pot B, Stepankova R, Hudcovic T, Pollak A, Tlaskalova-Hogenova H, Wiedermann U, Kozakova H. Neonatal colonization of mice with Lactobacillus plantarum producing the aeroallergen Bet v 1 biases towards Th1 and T-regulatory responses upon systemic sensitization. Allergy 2011; 66: 368–375. Abstract Background: The use of recombinant lactic acid bacteria (LAB) as vehicles for mucosal delivery of recombinant allergens is an attractive concept for antigen-defined allergy prevention/treatment. Interventions with LAB are of increasing interest early in life when immune programming is initiated. Here, we investigated the effect of neonatal colonization with a recombinant LAB producing the major birch pollen allergen Bet v 1 in a murine model of type I allergy. Methods: We constructed a recombinant Lactobacillus (L.) plantarum NCIMB8826 strain constitutively producing Bet v 1 to be used for natural mother-to-offspring mono-colonization of germ-free BALB/c mice. Allergen-specific immunomodulatory effects of the colonization on humoral and cellular immune responses were investigated prior and after sensitization to Bet v 1. Results: Mono-colonization with the Bet v 1 producing L. plantarum induced a Th1-biased immune response at the cellular level, evident in IFN-γ production of splenocytes upon stimulation with Bet v 1. After sensitization with Bet v 1 these mice displayed suppressed IL-4 and IL-5 production in spleen and mesenteric lymph node cell cultures as well as decreased allergen-specific antibody responses (IgG1, IgG2a, and IgE) in sera. This suppression was associated with a significant up-regulation of the regulatory marker Foxp3 at the mRNA level in the spleen cells. Conclusion: Intervention at birth with a live recombinant L. plantarum producing a clinically relevant allergen reduces experimental allergy and might therefore become an effective strategy for early intervention against the onset of allergic diseases.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.