Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nutrients ; 16(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999736

ABSTRACT

The etiology and mechanisms of autism and autism spectrum disorder (ASD) are not yet fully understood. There is currently no treatment for ASD for providing significant improvement in core symptoms. Recent studies suggest, however, that ASD is associated with gut dysbiosis, indicating that modulation of gut microbiota in children with ASD may thus reduce the manifestation of ASD symptoms. The aim of this pilot study (prospective randomized, double-blinded, placebo-controlled) was to evaluate efficacy of the biological response modifier Juvenil in modulating the microbiome of children with ASD and, in particular, whether Juvenil is able to alleviate the symptoms of ASD. In total, 20 children with ASD and 12 neurotypical children were included in our study. Supplementation of ASD children lasted for three months. To confirm Juvenil's impact on the gut microbiome, stool samples were collected from all children and the microbiome's composition was analyzed. This pilot study demonstrated that the gut microbiome of ASD children differed significantly from that of healthy controls and was converted by Juvenil supplementation toward a more neurotypical microbiome that positively modulated children's autism symptoms.


Subject(s)
Autism Spectrum Disorder , Dietary Supplements , Gastrointestinal Microbiome , Humans , Pilot Projects , Double-Blind Method , Male , Female , Autism Spectrum Disorder/microbiology , Child , Feces/microbiology , Child, Preschool , Prospective Studies , Autistic Disorder/microbiology , Dysbiosis/microbiology
2.
Front Microbiol ; 15: 1355872, 2024.
Article in English | MEDLINE | ID: mdl-38533334

ABSTRACT

Francisella tularensis secretes tubular outer membrane vesicles (OMVs) that contain a number of immunoreactive proteins as well as virulence factors. We have reported previously that isolated Francisella OMVs enter macrophages, cumulate inside, and induce a strong pro-inflammatory response. In the current article, we present that OMVs treatment of macrophages also enhances phagocytosis of the bacteria and suppresses their intracellular replication. On the other hand, the subsequent infection with Francisella is able to revert to some extent the strong pro-inflammatory effect induced by OMVs in macrophages. Being derived from the bacterial surface, isolated OMVs may be considered a "non-viable mixture of Francisella antigens" and as such, they present a promising protective material. Immunization of mice with OMVs isolated from a virulent F. tularensis subsp. holarctica strain FSC200 prolonged the survival time but did not fully protect against the infection with a lethal dose of the parent strain. However, the sera of the immunized animals revealed unambiguous cytokine and antibody responses and proved to recognize a set of well-known Francisella immunoreactive proteins. For these reasons, Francisella OMVs present an interesting material for future protective studies.

3.
Biology (Basel) ; 12(4)2023 Mar 25.
Article in English | MEDLINE | ID: mdl-37106700

ABSTRACT

Engagement of PRRs in recognition of PAMPs or DAMPs is one of the processes that initiates cellular stress. These sensors are involved in signaling pathways leading to induction of innate immune processes. Signaling initiated by PRRs is associated with the activation of MyD88-dependent signaling pathways and myddosome formation. MyD88 downstream signaling depends upon the context of signaling initiation, the cell (sub)type and the microenvironment of signal initiation. Recognition of PAMPs or DAMPs through PRRs activates the cellular autonomous defence mechanism, which orchestrates the cell responses to resolve specific insults at the single cell level. In general, stressed endoplasmic reticulum is directly linked with the induction of autophagy and initiation of mitochondrial stress. These processes are regulated by the release of Ca2+ from ER stores accepted by mitochondria, which respond through membrane depolarization and the production of reactive oxygen species generating signals leading to inflammasome activation. In parallel, signaling from PRRs initiates the accumulation of misfolded or inappropriately post-translationally modified proteins in the ER and triggers a group of conserved emergency rescue pathways known as unfolded protein response. The cell-autonomous effector mechanisms have evolutionarily ancient roots and were gradually specialized for the defence of specific cell (sub)types. All of these processes are common to the innate immune recognition of microbial pathogens and tumorigenesis as well. PRRs are active in both cases. Downstream are activated signaling pathways initiated by myddosomes, translated by the cellular autonomous defence mechanism, and finalized by inflammasomes.

4.
Front Pharmacol ; 13: 663855, 2022.
Article in English | MEDLINE | ID: mdl-35847048

ABSTRACT

Purpose: Insulin-like growth factor-1 (IGF-1) stimulates epithelial regeneration but may also induce life-threatening hypoglycemia. In our study, we first assessed its safety. Subsequently, we examined the effect of IGF-1 administered in different dose regimens on gastrointestinal damage induced by high doses of gamma radiation. Material and methods: First, fasting C57BL/6 mice were injected subcutaneously with IGF-1 at a single dose of 0, 0.2, 1, and 2 mg/kg to determine the maximum tolerated dose (MTD). The glycemic effect of MTD (1 mg/kg) was additionally tested in non-fasting animals. Subsequently, a survival experiment was performed. Animals were irradiated (60Co; 14, 14.5, or 15 Gy; shielded head), and IGF-1 was administered subcutaneously at 1 mg/kg 1, 24, and 48 h after irradiation. Simultaneously, mice were irradiated (60Co; 12, 14, or 15 Gy; shielded head), and IGF-1 was administered subcutaneously under the same regimen. Jejunum and lung damage were assessed 84 h after irradiation. Finally, we evaluated the effect of six different IGF-1 dosage regimens administered subcutaneously on gastrointestinal damage and peripheral blood changes in mice 6 days after irradiation (60Co; 12 and 14 Gy; shielded head). The regimens differed in the number of doses (one to five doses) and the onset of administration (starting at 1 [five regimens] or 24 h [one regimen] after irradiation). Results: MTD was established at 1 mg/kg. MTD mitigated lethality induced by 14 Gy and reduced jejunum and lung damage caused by 12 and 14 Gy. However, different dosing regimens showed different efficacy, with three and four doses (administered 1, 24, and 48 h and 1, 24, 48, and 72 h after irradiation, respectively) being the most effective. The three-dose regimens supported intestinal regeneration even if the administration started at 24 h after irradiation, but its potency decreased. Conclusion: IGF-1 seems promising in the mitigation of high-dose irradiation damage. However, the selected dosage regimen affects its efficacy.

5.
Front Microbiol ; 12: 748706, 2021.
Article in English | MEDLINE | ID: mdl-34721352

ABSTRACT

Francisella tularensis is known to release unusually shaped tubular outer membrane vesicles (OMV) containing a number of previously identified virulence factors and immunomodulatory proteins. In this study, we present that OMV isolated from the F. tularensis subsp. holarctica strain FSC200 enter readily into primary bone marrow-derived macrophages (BMDM) and seem to reside in structures resembling late endosomes in the later intervals. The isolated OMV enter BMDM generally via macropinocytosis and clathrin-dependent endocytosis, with a minor role played by lipid raft-dependent endocytosis. OMVs proved to be non-toxic and had no negative impact on the viability of BMDM. Unlike the parent bacterium itself, isolated OMV induced massive and dose-dependent proinflammatory responses in BMDM. Using transmission electron microscopy, we also evaluated OMV release from the bacterial surface during several stages of the interaction of Francisella with BMDM. During adherence and the early phase of the uptake of bacteria, we observed numerous tubular OMV-like protrusions bulging from the bacteria in close proximity to the macrophage plasma membrane. This suggests a possible role of OMV in the entry of bacteria into host cells. On the contrary, the OMV release from the bacterial surface during its cytosolic phase was negligible. We propose that OMV play some role in the extracellular phase of the interaction of Francisella with the host and that they are involved in the entry mechanism of the bacteria into macrophages.

6.
Microorganisms ; 9(10)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34683457

ABSTRACT

Immune responses to intracellular pathogens depend largely upon the activation of T helper type 1-dependent mechanisms. The contribution of B cells to establishing protective immunity has long been underestimated. Francisella tularensis, including a number of subspecies, provides a suitable model for the study of immune responses against intracellular bacterial pathogens. We previously demonstrated that Francisella infects B cells and activates B-cell subtypes to produce a number of cytokines and express the activation markers. Recently, we documented the early production of natural antibodies as a consequence of Francisella infection in mice. Here, we summarize current knowledge on the innate and acquired humoral immune responses initiated by Francisella infection and their relationships with the immune defense systems.

7.
Pathogens ; 10(3)2021 Mar 22.
Article in English | MEDLINE | ID: mdl-33809954

ABSTRACT

SARS-CoV-2 infection induces the production of autoantibodies, which is significantly associated with complications during hospitalization and a more severe prognosis in COVID-19 patients. Such a response of the patient's immune system may reflect (1) the dysregulation of the immune response or (2) it may be an attempt to regulate itself in situations where the non-infectious self poses a greater threat than the infectious non-self. Of significance may be the primary virus-host cell interaction where the surface-bound ACE2 ectoenzyme plays a critical role. Here, we present a brief analysis of recent findings concerning the immune recognition of SARS-CoV-2, which, we believe, favors the second possibility as the underlying reason for the production of autoantibodies during COVID-19.

8.
J Proteome Res ; 20(3): 1716-1732, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33543941

ABSTRACT

Release of outer membrane vesicles (OMV) is an important phenomenon in Gram-negative bacteria playing multiple roles in their lifestyle, including in relation to virulence and host-pathogen interaction. Francisella tularensis, unlike other bacteria, releases unusually shaped, tubular OMV. We present a proteomic comparison of OMV and membrane fractions from two F. tularensis strains: moderately virulent subsp. holarctica strain FSC200 and highly virulent subsp. tularensis strain SchuS4. Proteomic comparison studies routinely evaluate samples from the same proteome, but sometimes we must compare samples from closely related organisms. This raises quantification issues. We propose a novel approach to cross-species proteomic comparison based on an intersection protein database from the individual single-species databases. This is less prone to quantification errors arising from differences in the sequences. Consecutively comparing subproteomes of OMV and membranes of the two strains allows distinguishing differences in relative protein amounts caused by global expression changes from those caused by preferential protein packing to OMV or membranes. Among the proteins most differently packed into OMV between the two strains, we detected proteins involved in biosynthesis and metabolism of bacterial envelope components like O-antigen, lipid A, phospholipids, and fatty acids, as well as some major structural outer membrane proteins. The data are available via ProteomeXchange with identifier PXD022406.


Subject(s)
Francisella tularensis , Tularemia , Bacterial Outer Membrane , Francisella , Humans , Proteome/genetics , Proteomics , Virulence
9.
Sci Rep ; 11(1): 1541, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33452341

ABSTRACT

There remains to this day a great gap in understanding as to the role of B cells and their products-antibodies and cytokines-in mediating the protective response to Francisella tularensis, a Gram-negative coccobacillus belonging to the group of facultative intracellular bacterial pathogens. We previously have demonstrated that Francisella interacts directly with peritoneal B-1a cells. Here, we demonstrate that, as early as 12 h postinfection, germ-free mice infected with Francisella tularensis produce infection-induced antibody clones reacting with Francisella tularensis proteins having orthologs or analogs in eukaryotic cells. Production of some individual clones was limited in time and was influenced by virulence of the Francisella strain used. The phylogenetically stabilized defense mechanism can utilize these early infection-induced antibodies both to recognize components of the invading pathogens and to eliminate molecular residues of infection-damaged self cells.


Subject(s)
B-Lymphocytes/metabolism , Tularemia/immunology , Animals , Antibody Formation , B-Lymphocytes/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Francisella tularensis/pathogenicity , Mice , Mice, Inbred BALB C , Tularemia/microbiology , Virulence
10.
Front Pharmacol ; 11: 1199, 2020.
Article in English | MEDLINE | ID: mdl-32903478

ABSTRACT

PURPOSE: Therapeutic thorax irradiation as an intervention in lung cancer has its limitations due to toxic effects leading to pneumonitis and/or pulmonary fibrosis. It has already been confirmed that hyaluronic acid (HA), an extracellular matrix glycosaminoglycan, is involved in inflammation disorders and wound healing in lung tissue. We examined the effects after gamma irradiation of hyaluronic acid nanoparticles (HANPs) applied into lung prior to that irradiation in a dose causing radiation-induced pulmonary injuries (RIPI). MATERIALS AND METHODS: Biocompatible HANPs were first used for viability assay conducted on the J774.2 cell line. For in vivo experiments, HANPs were administered intratracheally to C57Bl/6 mice 30 min before thoracic irradiation by 17 Gy. Molecular, cellular, and histopathological parameters were measured in lung and peripheral blood at days 113, 155, and 190, corresponding to periods of significant morphological and/or biochemical alterations of RIPI. RESULTS: Modification of linear hyaluronic acid molecule into nanoparticles structure significantly affected the physiological properties and caused long-term stability against ionizing radiation. The HANPs treatments had significant effects on the expression of the cytokines and particularly on the pro-fibrotic signaling pathway in the lung tissue. The radiation fibrosis phase was altered significantly in comparison with a solely irradiated group. CONCLUSIONS: The present study provides evidence that application of HANPs caused significant changes in molecular and cellular patterns associated with RIPI. These findings suggest that HANPs could diminish detrimental radiation-induced processes in lung tissue, thereby potentially decreasing the extracellular matrix degradation leading to lung fibrosis.

11.
Microb Pathog ; 140: 103937, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31862393

ABSTRACT

Mycobacterium tuberculosis is the main etiological agent of tuberculosis. The Bacillus Calmette-Guérin (BCG) microbes that are primarily used as a vaccine against tuberculosis also constitute the dominant infection model for studying the interaction of mycobacteria with the host cell types. The majority of interaction experiments have been conducted using macrophages and monocytes as prototype phagocyte cell types. Here, we report that M. bovis BCG infects mouse primary B cells as well as human B cell line. The complement receptors, along with B cell receptors, are engaged in the process of bacterial entry into the host B cells. Once inside the B cells, the intracellular trafficking of BCG follows the complete endocytic pathway of the ingested particles, which is in contrast to the events taking place during ingestion of BCG by macrophages. In vivo infection of mice with M. bovis BCG activated peritoneal as well as splenic B cells to produce proinflammatory cytokines. This paper further supports the evidence that B cells are involved in a host's early interactions with intracellular bacterial pathogens and participate in the induction of innate defense responses.


Subject(s)
B-Lymphocytes , Cytokines/metabolism , Mycobacterium bovis/immunology , Tuberculosis/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/microbiology , BCG Vaccine , Humans , Immunity, Innate , Mice , Primary Cell Culture , Tuberculosis/microbiology
12.
Article in English | MEDLINE | ID: mdl-31334134

ABSTRACT

Primary interaction of an intracellular bacterium with its host cell is initiated by activation of multiple signaling pathways in response to bacterium recognition itself or as cellular responses to stress induced by the bacterium. The leading molecules in these processes are cell surface membrane receptors as well as cytosolic pattern recognition receptors recognizing pathogen-associated molecular patterns or damage-associated molecular patterns induced by the invading bacterium. In this review, we demonstrate possible sequences of events leading to recognition of Francisella tularensis, present findings on known mechanisms for manipulating cell responses to protect Francisella from being killed, and discuss newly published data from the perspective of early stages of host-pathogen interaction.


Subject(s)
Francisella tularensis/immunology , Host-Pathogen Interactions/immunology , Immunity, Innate , Pathogen-Associated Molecular Pattern Molecules/immunology , Receptors, Pattern Recognition/immunology , Tularemia/immunology , Alarmins/genetics , Alarmins/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Humans , Macrophages/immunology , Macrophages/microbiology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Phagocytosis/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Pattern Recognition/genetics , Signal Transduction , Tularemia/genetics , Tularemia/microbiology
13.
Microb Pathog ; 123: 314-322, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30055244

ABSTRACT

Bacteria that are highly virulent, expressing high infectivity, and able to survive nebulization, pose great risk to the human population. One of these is Francisella tularensis, the etiological agent of tularemia. F. tularensis is a subject of intense scientific interest due to the fact that vaccines for its immunoprophylaxis in humans are not yet routinely available. One of the substantial obstacles in developing such vaccines is our insufficient knowledge of processes that initiate and regulate the expression of effective protective immunity against intracellular bacteria. Here, we present data documenting the different pattern of cellular behavior occurring in an environment unaffected by microbiota using the model of germ-free mice mono-associated with F. tularensis subsp. holarctica strain LVS in comparison with a classic specific-pathogen-free murine model during early stages of infection.


Subject(s)
Francisella tularensis/immunology , Francisella tularensis/pathogenicity , Host-Pathogen Interactions/immunology , Tularemia/immunology , Animals , Bacterial Vaccines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Germ-Free Life/immunology , Immunity, Innate , Mice , Mice, Inbred BALB C , Microbiota , Peritoneum/microbiology , Peritoneum/pathology , Specific Pathogen-Free Organisms/immunology , Spleen/microbiology , Spleen/pathology , Tularemia/microbiology , Tularemia/pathology
14.
Res Microbiol ; 169(3): 127-134, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29407045

ABSTRACT

Bacterial biofilms pose a serious medical problem due to their significant resistance to antimicrobials, and staphylococci are recognized as the most frequent cause of biofilm-associated infections. The hop plant (Humulus lupulus L.) contains substances that have been determined to act as anti-infective agents against bacteria, mainly in planktonic form. Therefore, we decided to investigate the antibiofilm properties of H. lupulus L.-derived compounds (humulone, lupulone and xanthohumol) against a selected group of Staphylococcus spp., including methicillin-susceptible and resistant strains. All tested hop compounds were shown to possess antimicrobial properties against all tested staphylococci, both planktonic and biofilm-dwelling, with no significant difference between resistant and susceptible strains. All compounds lowered the number of bacterial cells released from the biofilm, with the strongest effect seen for lupulone, followed by xanthohumol. Moreover, lupulone and xanthohumol were not only able to penetrate the biofilm and reduce the number of bacteria within it, but their higher concentrations (∼60 µg/mL for xanthohumol and ∼125 µg/mL for lupulone) reduced the number of surviving bacterial cells to zero.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Cyclohexenes/pharmacology , Flavonoids/pharmacology , Propiophenones/pharmacology , Staphylococcus/drug effects , Staphylococcus/growth & development , Terpenes/pharmacology , Cell Line , Cell Survival/drug effects , Humans , Humulus/chemistry , Microbial Sensitivity Tests , Plant Extracts/pharmacology , Staphylococcus/genetics
15.
Article in English | MEDLINE | ID: mdl-29085810

ABSTRACT

The intracellular bacterial pathogen Francisella tularensis causes serious infectious disease in humans and animals. Moreover, F. tularensis, a highly infectious pathogen, poses a major concern for the public as a bacterium classified under Category A of bioterrorism agents. Unfortunately, research has so far failed to develop effective vaccines, due in part to the fact that the pathogenesis of intracellular bacteria is not fully understood and in part to gaps in our understanding of innate immune recognition processes leading to the induction of adaptive immune response. Recent evidence supports the concept that immune response to external stimuli in the form of bacteria is guided by the primary interaction of the bacterium with the host cell. Based on data from different Francisella models, we present here the basic paradigms of the emerging innate immune recognition concept. According to this concept, the type of cell and its receptor(s) that initially interact with the target constitute the first signaling window; the signals produced in the course of primary interaction of the target with a reacting cell act in a paracrine manner; and the innate immune recognition process as a whole consists in a series of signaling windows modulating adaptive immune response. Finally, the host, in the strict sense, is the interacting cell.


Subject(s)
Francisella tularensis/immunology , Host-Pathogen Interactions/immunology , Immunity, Innate , Tularemia/immunology , Adaptive Immunity , Animals , Disease Models, Animal , Humans , Immune System , Mice , Paracrine Communication/immunology
16.
Radiat Res ; 186(3): 264-74, 2016 09.
Article in English | MEDLINE | ID: mdl-27538113

ABSTRACT

We examined the effect of epidermal growth factor (EGF) treatment in mice that received bone marrow transplantation (BMT) after 11 Gy whole-body irradiation. C57Bl/6 mice were divided into three treatment groups: 0 Gy; 11 Gy ((60)Co, single dose, 0.51 Gy/min) with BMT (5 × 10(6) bone marrow cells isolated from green fluorescent protein syngeneic mice, 3-4 h postirradiation); and 11 Gy with BMT and EGF (2 mg/kg applied subcutaneously 1, 3 and 5 days postirradiation). Survival data were collected. Bone marrow, peripheral blood count and cytokines, gastrointestine and liver parameters and migration of green fluorescent protein-positive cells were evaluated at 63 days postirradiation. Epidermal growth factor increased survival of irradiated animals that received BMT from 10.7 to 85.7% at 180 days postirradiation. In the BMT group, we found changes in differential bone marrow and blood count, plasma cytokine levels, gastrointestinal tissues and liver at 63 days postirradiation. These alterations were completely or in some parameters at least partially restored by epidermal growth factor. These findings indicate that epidermal growth factor, administered 1, 3 and 5 days postirradiation in combination with bone marrow transplantation, significantly improves long-term prognosis.


Subject(s)
Bone Marrow Transplantation , EGF Family of Proteins/pharmacology , Radiation Injuries/drug therapy , Radiation Injuries/therapy , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Bone Marrow/drug effects , Bone Marrow/immunology , Bone Marrow/radiation effects , Cell Count , Cytokines/blood , Dose-Response Relationship, Radiation , Female , Intestines/drug effects , Intestines/pathology , Intestines/radiation effects , Mice , Mitosis/drug effects , Mitosis/radiation effects , Organ Size/drug effects , Organ Size/radiation effects , Radiation Injuries/blood , Radiation Injuries/pathology , Safety , Spleen/drug effects , Spleen/pathology , Spleen/radiation effects , Time Factors , Whole-Body Irradiation/adverse effects
17.
Cell Mol Life Sci ; 73(20): 3961-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27544211

ABSTRACT

This brief review is dedicated to the legacy of Prof. Jaroslav Sterzl and his colleagues, who laid the foundation for gnotobiology in the former Czechoslovakia 55 years. Prof. Sterzl became one of the founders of modern Czechoslovak immunology, which was characterized by work on a wide range of problems needing to be solved. While examining the mechanisms of innate immunity, he focused his studies on the induction of antibody production by immunocompetent cells involved in adaptive immune transmission while using the model of pig fetuses and germ-free piglets and characterizing immunoglobulins in the sera of these piglets. Although not fully appreciated to this day, his experimental proof of the hypothesis focused on the common precursor of cell-forming antibodies of different isotypes was later confirmed in experiments at the gene level. Prof. Sterzl's work represented a true milestone in the development of not solely Czechoslovak but also European and global immunology. He collaborated closely with the World Health Organization for many years, serving there as leader of the Reference Laboratory for Factors of Innate Immunity.


Subject(s)
Germ-Free Life , Host-Pathogen Interactions , Animals , Disease Models, Animal , Gastrointestinal Microbiome , Mice
18.
PLoS One ; 10(7): e0132571, 2015.
Article in English | MEDLINE | ID: mdl-26161475

ABSTRACT

Francisella tularensis, the etiological agent of tularemia, is an intracellular pathogen that dominantly infects and proliferates inside phagocytic cells but can be seen also in non-phagocytic cells, including B cells. Although protective immunity is known to be almost exclusively associated with the type 1 pathway of cellular immunity, a significant role of B cells in immune responses already has been demonstrated. Whether their role is associated with antibody-dependent or antibody-independent B cell functions is not yet fully understood. The character of early events during B cell-pathogen interaction may determine the type of B cell response regulating the induction of adaptive immunity. We used fluorescence microscopy and flow cytometry to identify the basic requirements for the entry of F. tularensis into B cells within in vivo and in vitro infection models. Here, we present data showing that Francisella tularensis subsp. holarctica strain LVS significantly infects individual subsets of murine peritoneal B cells early after infection. Depending on a given B cell subset, uptake of Francisella into B cells is mediated by B cell receptors (BCRs) with or without complement receptor CR1/2. However, F. tularensis strain FSC200 ΔiglC and ΔftdsbA deletion mutants are defective in the ability to enter B cells. Once internalized into B cells, F. tularensis LVS intracellular trafficking occurs along the endosomal pathway, albeit without significant multiplication. The results strongly suggest that BCRs alone within the B-1a subset can ensure the internalization process while the BCRs on B-1b and B-2 cells need co-signaling from the co receptor containing CR1/2 to initiate F. tularensis engulfment. In this case, fluidity of the surface cell membrane is a prerequisite for the bacteria's internalization. The results substantially underline the functional heterogeneity of B cell subsets in relation to F. tularensis.


Subject(s)
B-Lymphocytes/metabolism , Francisella tularensis/physiology , Receptors, Antigen, B-Cell/metabolism , Receptors, Complement/metabolism , Tularemia/microbiology , Animals , B-Lymphocytes/microbiology , Biological Transport , Cell Line , Female , Genes, Bacterial , Host-Pathogen Interactions , Membrane Microdomains/metabolism , Mice, Inbred BALB C , Microbial Viability , Receptors, IgG/metabolism , Sequence Deletion
19.
Int J Radiat Biol ; 91(9): 703-14, 2015.
Article in English | MEDLINE | ID: mdl-25994811

ABSTRACT

PURPOSE: We examined the effect of epidermal growth factor (EGF) and bone marrow transplantation (BMT) on gastrointestinal damage after high-dose irradiation of mice. MATERIAL AND METHODS: C57Black/6 mice were used. Two survival experiments were performed (12 and 13 Gy; (60)Co, 0.59-0.57 Gy/min). To evaluate BMT and EGF action, five groups were established - 0 Gy, 13 Gy, 13 Gy + EGF (at 2 mg/kg, first dose 24 h after irradiation and then every 48 h), 13 Gy + BMT (5 × 10(6) cells from green fluorescent protein [GFP] syngenic mice, 4 h after irradiation), and 13 Gy + BMT + EGF. Survival data, blood cell counts, gastrointestine and liver parameters and GFP positive cell migration were measured. RESULTS: BMT and EGF (three doses, at 2 mg/kg, administered 1, 3 and 5 days after irradiation) significantly increased survival (13 Gy). In blood, progressive cytopenia was observed with BMT, EGF or their combination having no improving effect early after irradiation. In gastrointestinal system, BMT, EGF and their combination attenuated radiation-induced atrophy and increased regeneration during first week after irradiation with the combination being most effective. Signs of systemic inflammatory reaction were observed 30 days after irradiation. CONCLUSIONS: Our data indicate that BMT together with EGF is a promising strategy in the treatment of high-dose whole-body irradiation damage.


Subject(s)
Bone Marrow Transplantation , Epidermal Growth Factor/therapeutic use , Gastrointestinal Tract/injuries , Gastrointestinal Tract/radiation effects , Radiation Injuries, Experimental/therapy , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Combined Modality Therapy , Epidermal Growth Factor/administration & dosage , Female , Gastrointestinal Tract/pathology , Inflammation/pathology , Lithostathine/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitosis/drug effects , Mitosis/radiation effects , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/pathology , Whole-Body Irradiation/adverse effects
20.
PLoS One ; 9(10): e109898, 2014.
Article in English | MEDLINE | ID: mdl-25296161

ABSTRACT

Our laboratory's investigations into mechanisms of protective immunity against Francisella tularensis Live Vaccine Strain (LVS) have uncovered mediators important in host defense against primary infection, as well as those correlated with successful vaccination. One such potential correlate was IL-12p40, a pleiotropic cytokine that promotes Th1 T cell function as part of IL-12p70. LVS-infected IL-12p40 deficient knockout (KO) mice maintain a chronic infection, but IL-12p35 KO mice clear LVS infection; thus the role that IL-12p40 plays in immunity to LVS is independent of the IL-12p70 heterodimer. IL-12p40 can also partner with IL-23p19 to create the heterodimeric cytokine IL-23. Here, we directly tested the role of IL-23 in LVS resistance, and found IL-23 to be largely dispensable for immunity to LVS following intradermal or intranasal infection. IL-23p19 KO splenocytes were fully competent in controlling intramacrophage LVS replication in an in vitro overlay assay. Further, antibody responses in IL-23p19 KO mice were similar to those of normal wild type mice after LVS infection. IL-23p19 KO mice or normal wild type mice that survived primary LVS infection survived maximal doses of LVS secondary challenge. Thus p40 has a novel role in clearance of LVS infection that is unrelated to either IL-12 or IL-23.


Subject(s)
Bacterial Vaccines , Coinfection/metabolism , Francisella tularensis/physiology , Interleukin-23 Subunit p19/deficiency , Interleukin-23 Subunit p19/genetics , Tularemia/metabolism , Animals , Coinfection/genetics , Coinfection/immunology , Female , Francisella tularensis/immunology , Interleukin-23 Subunit p19/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tularemia/genetics , Tularemia/immunology , Vaccines, Attenuated
SELECTION OF CITATIONS
SEARCH DETAIL
...