Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 923754, 2022.
Article in English | MEDLINE | ID: mdl-35967418

ABSTRACT

Commensal bacteria and the immune system have a close and strong relationship that maintains a balance to control inflammation. Alterations of the microbiota, known as dysbiosis, can direct reactivity to self-antigens not only in the intestinal mucosa but also at the systemic level. Our laboratory previously reported gut dysbiosis, particularly lower abundance of bacteria in the family Lactobacillaceae, in lupus-prone MRL/lpr mice, a model of systemic autoimmunity. Restoring the microbiota with a mix of 5 different Lactobacillus species (spp.), L. reuteri, L. oris, L. johnsonii, L. gasseri and L. rhamnosus, attenuated lupus-liked clinical signs, including splenomegaly and lymphadenopathy. However, our understanding of the mechanism was limited. In this study, we first investigated the effects of individual species. Surprisingly, none of the species individually recapitulated the benefits of the mix. Instead, Lactobacillus spp. acted synergistically to attenuate splenomegaly and renal lymphadenopathy through secreted factors and a CX3CR1-dependent mechanism. Interestingly, oral administration of MRS broth exerted the same benefits likely through increasing the relative abundance of endogenous Lactobacillus spp. Mechanistically, we found increased percentages of FOXP3-negative type 1 regulatory T cells with administration of the mix in both spleen and mesenteric lymph nodes. In addition, oral gavage of Lactobacillus spp. decreased the percentage of central memory T cells while increasing that of effector memory T cells in the lymphoid organs. Furthermore, a decreased percentage of double negative T cells was observed in the spleen with the mix. These results suggest that Lactobacillus spp. might act on T cells to attenuate splenomegaly and lymphadenopathy. Together, this study advances our understanding of how Lactobacillus spp. attenuate lupus in MRL/lpr mice. The synergistic action of these bacteria suggests that multiple probiotic bacteria in combination may dampen systemic autoimmunity and benefit lupus patients.


Subject(s)
Lactobacillus , Lymphadenopathy , Animals , Dysbiosis , Mice , Mice, Inbred MRL lpr , Splenomegaly
2.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33619092

ABSTRACT

Infants are prone to enteric infections due to an underdeveloped immune system. The maternal microbiota, through shaping the neonatal microbiota, helps establish a strong immune system in infants. We and others have observed the phenomenon of enhanced early neonatal immunoglobulin A (IgA) production in preweaning immunocompetent mice nursed by immunodeficient dams. Here, we show that this enhancement of IgA in neonates results from maternally derived microbiota. In addition, we have found that the neonatal IgA production can be induced by Lactobacillus reuteri, which is enriched in the milk of immunodeficient dams. Moreover, we show that while the production of neonatal IgA is dependent on neonatal T cells, the immunodeficient maternal microbiota-mediated enhancement of neonatal IgA has a T cell-independent component. Indeed, this enhancement may be dependent on type 3 innate lymphoid cells in the neonatal small intestinal lamina propria. Interestingly, maternal microbiota-induced neonatal IgA does not cross-react with common enteric pathogens. Future investigations will determine the functional consequences of having this extra IgA.


Subject(s)
Antibody Formation/immunology , Immunity, Maternally-Acquired , Immunoglobulin A/immunology , Immunomodulation , Microbiota/immunology , Animals , Animals, Newborn , Cross Reactions/immunology , Female , Host-Pathogen Interactions/immunology , Immunity, Innate , Intestinal Mucosa/immunology , Limosilactobacillus reuteri/immunology , Male , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
Front Immunol ; 11: 575179, 2020.
Article in English | MEDLINE | ID: mdl-33193366

ABSTRACT

Hypersensitivity reactions and immune dysregulation have been reported with the use of quaternary ammonium compound disinfectants (QACs). We hypothesized that QAC exposure would exacerbate autoimmunity associated with systemic lupus erythematosus (lupus). Surprisingly, however, we found that compared to QAC-free mice, ambient exposure of lupus-prone mice to QACs led to smaller spleens with no change in circulating autoantibodies or the severity of glomerulonephritis. This suggests that QACs may have immunosuppressive effects on lupus. Using a microfluidic device, we showed that ambient exposure to QACs reduced directional migration of bone marrow-derived neutrophils toward an inflammatory chemoattractant ex vivo. Consistent with this, we found decreased infiltration of neutrophils into the spleen. While bone marrow-derived neutrophils appeared to exhibit a pro-inflammatory profile, upregulated expression of PD-L1 was observed on neutrophils that infiltrated the spleen, which in turn interacted with PD-1 on T cells and modulated their fate. Specifically, QAC exposure hindered activation of splenic T cells and increased apoptosis of effector T-cell populations. Collectively, these results suggest that ambient QAC exposure decreases lupus-associated splenomegaly likely through neutrophil-mediated toning of T-cell activation and/or apoptosis. However, our findings also indicate that even ambient exposure could alter immune cell phenotypes, functions, and their fate. Further investigations on how QACs affect immunity under steady-state conditions are warranted.


Subject(s)
Disinfectants/pharmacology , Immunosuppressive Agents/pharmacology , Lupus Erythematosus, Systemic/drug therapy , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Quaternary Ammonium Compounds/pharmacology , Spleen/drug effects , Splenomegaly/prevention & control , T-Lymphocytes/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Disease Models, Animal , Female , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mice, Inbred MRL lpr , Neutrophils/immunology , Neutrophils/metabolism , Phenotype , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Splenomegaly/immunology , Splenomegaly/metabolism , Splenomegaly/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
Front Immunol ; 11: 593353, 2020.
Article in English | MEDLINE | ID: mdl-33240280

ABSTRACT

Autoimmune diseases, such as systemic lupus erythematosus, are characterized by excessive inflammation in response to self-antigens. Loss of appropriate immunoregulatory mechanisms contribute to disease exacerbation. We previously showed the suppressive effect of vancomycin treatment during the "active-disease" stage of lupus. In this study, we sought to understand the effect of the same treatment given before disease onset. To develop a model in which to test the regulatory role of the gut microbiota in modifying autoimmunity, we treated lupus-prone mice with vancomycin in the period before disease development (3-8 weeks of age). We found that administration of vancomycin to female MRL/lpr mice early, only during the pre-disease period but not from 3 to 15 weeks of age, led to disease exacerbation. Early vancomycin administration also reduced splenic regulatory B (Breg) cell numbers, as well as reduced circulating IL-10 and IL-35 in 8-week old mice. Further, we found that during the pre-disease period, administration of activated IL-10 producing Breg cells to mice treated with vancomycin suppressed lupus initiation, and that bacterial DNA from the gut microbiota was an inducer of Breg function. Oral gavage of bacterial DNA to mice treated with vancomycin increased Breg cells in the spleen and mesenteric lymph node at 8 weeks of age and reduced autoimmune disease severity at 15 weeks. This work suggests that a form of oral tolerance induced by bacterial DNA-mediated expansion of Breg cells suppress disease onset in the autoimmune-prone MRL/lpr mouse model. Future studies are warranted to further define the mechanism behind bacterial DNA promoting Breg cells.


Subject(s)
Autoimmunity , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , DNA, Bacterial/immunology , Gastrointestinal Microbiome/immunology , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Adoptive Transfer , Animals , Biomarkers , Disease Models, Animal , Disease Susceptibility , Female , Gastrointestinal Microbiome/drug effects , Immunomodulation , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/therapy , Mice , Mice, Inbred MRL lpr , Severity of Illness Index , Vancomycin/pharmacology
5.
Microbiome ; 7(1): 105, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31311609

ABSTRACT

BACKGROUND: Dysbiosis of gut microbiota exists in the pathogenesis of many autoimmune diseases, including systemic lupus erythematosus (lupus). Lupus patients who experienced pregnancy usually had more severe disease flares post-delivery. However, the possible role of gut microbiota in the link between pregnancy and exacerbation of lupus remains to be explored. RESULTS: In the classical lupus mouse model MRL/lpr, we compared the structures of gut microbiota in pregnant and lactating individuals vs. age-matched naïve mice. Consistent with studies on non-lupus mice, both pregnancy and lactation significantly changed the composition and diversity of gut microbiota. Strikingly, modulation of gut microbiota using the same strategy resulted in different disease outcomes in postpartum (abbreviated as "PP," meaning that the mice had undergone pregnancy and lactation) vs. control (naïve; i.e., without pregnancy or lactation) MRL/lpr females; while vancomycin treatment attenuated lupus in naïve mice, it did not do so, or even exacerbated lupus, in PP mice. Lactobacillus animalis flourished in the gut upon vancomycin treatment, and direct administration of L. animalis via oral gavage recapitulated the differential effects of vancomycin in PP vs. control mice. An enzyme called indoleamine 2,3-dioxygenase was significantly inhibited by L. animalis; however, this inhibition was only apparent in PP mice, which explained, at least partially, the lack of beneficial response to vancomycin in these mice. The differential production of immunosuppressive IL-10 and proinflammatory IFNγ in PP vs. control mice further explained why the disease phenotypes varied between the two types of mice bearing the same gut microbiota remodeling strategy. CONCLUSIONS: These results suggest that pregnancy and lactation interfere with the response of autoimmunity to modulation of gut microbiota. Further studies are necessary to better understand the complex relationship between pregnancy and lupus.


Subject(s)
Autoimmunity , Gastrointestinal Microbiome/immunology , Lactation , Lupus Erythematosus, Systemic/microbiology , Pregnancy, Animal , Animals , Anti-Bacterial Agents/administration & dosage , Female , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/immunology , Interleukin-10/immunology , Lactobacillus/immunology , Lactobacillus/metabolism , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred MRL lpr , Pregnancy , Vancomycin/administration & dosage
6.
Front Microbiol ; 9: 757, 2018.
Article in English | MEDLINE | ID: mdl-29725324

ABSTRACT

Lactobacillus reuteri (L. reuteri) is a well-studied probiotic bacterium that can colonize a large number of mammals. In humans, L. reuteri is found in different body sites, including the gastrointestinal tract, urinary tract, skin, and breast milk. The abundance of L. reuteri varies among different individuals. Several beneficial effects of L. reuteri have been noted. First, L. reuteri can produce antimicrobial molecules, such as organic acids, ethanol, and reuterin. Due to its antimicrobial activity, L. reuteri is able to inhibit the colonization of pathogenic microbes and remodel the commensal microbiota composition in the host. Second, L. reuteri can benefit the host immune system. For instance, some L. reuteri strains can reduce the production of pro-inflammatory cytokines while promoting regulatory T cell development and function. Third, bearing the ability to strengthen the intestinal barrier, the colonization of L. reuteri may decrease the microbial translocation from the gut lumen to the tissues. Microbial translocation across the intestinal epithelium has been hypothesized as an initiator of inflammation. Therefore, inflammatory diseases, including those located in the gut as well as in remote tissues, may be ameliorated by increasing the colonization of L. reuteri. Notably, the decrease in the abundance of L. reuteri in humans in the past decades is correlated with an increase in the incidences of inflammatory diseases over the same period of time. Direct supplementation or prebiotic modulation of L. reuteri may be an attractive preventive and/or therapeutic avenue against inflammatory diseases.

7.
Inflamm Bowel Dis ; 24(2): 361-379, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29361089

ABSTRACT

One of the best characterized mouse models of the inflammatory bowel diseases (IBD; Crohn's disease, ulcerative colitis) is the CD4+CD45RBhigh T cell transfer model of chronic colitis. Following our relocation to Texas Tech University Health Sciences Center (TTUHSC), we observed a dramatic reduction in the incidence of moderate-to-severe colitis from a 16-year historical average of 90% at Louisiana State University Health Sciences Center (LSUHSC) to <30% at TTUHSC. We hypothesized that differences in the commensal microbiota at the 2 institutions may account for the differences in susceptibility to T cell-induced colitis. Using bioinformatic analyses of 16S rRNA amplicon sequence data, we quantified and compared the major microbial populations in feces from healthy and colitic mice housed at the 2 institutions. We found that the bacterial composition differed greatly between mice housed at LSUHSC vs TTUHSC. We identified several genera strongly associated with, and signficantly overrepresented in high responding RAG-/- mice housed at LSUHSC. In addition, we found that colonization of healthy TTUHSC RAG-/- mice with feces obtained from healthy or colitic RAG-/- mice housed at LSUHSC transferred susceptibility to T cell-induced colitis such that the recipients developed chronic colitis with incidence and severity similar to mice generated at LSUHSC. Finally, we found that the treatment of mice with preexisting colitis with antibiotics remarkably attenuated disease. Taken together, our data demonstrate that specific microbial communities determine disease susceptibility and that manipulation of the intestinal microbiota alters the induction and/or perpetuation of chronic colitis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Colitis/immunology , Colitis/microbiology , Colon/pathology , Gastrointestinal Microbiome/drug effects , Adoptive Transfer , Animals , Bacteria/classification , Colon/drug effects , Disease Models, Animal , Feces/microbiology , Gastrointestinal Microbiome/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Ribosomal, 16S/genetics , T-Lymphocytes/immunology
8.
Appl Environ Microbiol ; 84(4)2018 02 15.
Article in English | MEDLINE | ID: mdl-29196292

ABSTRACT

Gut microbiota dysbiosis has been observed in a number of autoimmune diseases. However, the role of the gut microbiota in systemic lupus erythematosus (SLE), a prototypical autoimmune disease characterized by persistent inflammation in multiple organs of the body, remains elusive. Here we report the dynamics of the gut microbiota in a murine lupus model, NZB/W F1, as well as intestinal dysbiosis in a small group of SLE patients with active disease. The composition of the gut microbiota changed markedly before and after the onset of lupus disease in NZB/W F1 mice, with greater diversity and increased representation of several bacterial species as lupus progressed from the predisease stage to the diseased stage. However, we did not control for age and the cage effect. Using dexamethasone as an intervention to treat SLE-like signs, we also found that a greater abundance of a group of lactobacilli (for which a species assignment could not be made) in the gut microbiota might be correlated with more severe disease in NZB/W F1 mice. Results of the human study suggest that, compared to control subjects without immune-mediated diseases, SLE patients with active lupus disease possessed an altered gut microbiota that differed in several particular bacterial species (within the genera Odoribacter and Blautia and an unnamed genus in the family Rikenellaceae) and was less diverse, with increased representation of Gram-negative bacteria. The Firmicutes/Bacteroidetes ratios did not differ between the SLE microbiota and the non-SLE microbiota in our human cohort.IMPORTANCE SLE is a complex autoimmune disease with no known cure. Dysbiosis of the gut microbiota has been reported for both mice and humans with SLE. In this emerging field, however, more studies are required to delineate the roles of the gut microbiota in different lupus-prone mouse models and people with diverse manifestations of SLE. Here, we report changes in the gut microbiota in NZB/W F1 lupus-prone mice and a group of SLE patients with active disease.


Subject(s)
Dysbiosis/microbiology , Gastrointestinal Microbiome , Lupus Erythematosus, Systemic/microbiology , Adult , Aged , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Bacteroidetes/isolation & purification , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Disease Models, Animal , Female , Firmicutes/isolation & purification , Gram-Negative Bacteria/classification , Gram-Negative Bacteria/isolation & purification , Humans , Lactobacillus/isolation & purification , Lupus Erythematosus, Systemic/drug therapy , Male , Mice , Middle Aged , Young Adult
9.
Sci Rep ; 7(1): 13675, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29057975

ABSTRACT

Gut microbiota and the immune system interact to maintain tissue homeostasis, but whether this interaction is involved in the pathogenesis of systemic lupus erythematosus (SLE) is unclear. Here we report that oral antibiotics given during active disease removed harmful bacteria from the gut microbiota and attenuated SLE-like disease in lupus-prone mice. Using MRL/lpr mice, we showed that antibiotics given after disease onset ameliorated systemic autoimmunity and kidney histopathology. They decreased IL-17-producing cells and increased the level of circulating IL-10. In addition, antibiotics removed Lachnospiraceae and increased the relative abundance of Lactobacillus spp., two groups of bacteria previously shown to be associated with deteriorated or improved symptoms in MRL/lpr mice, respectively. Moreover, we showed that the attenuated disease phenotype could be recapitulated with a single antibiotic vancomycin, which reshaped the gut microbiota and changed microbial functional pathways in a time-dependent manner. Furthermore, vancomycin treatment increased the barrier function of the intestinal epithelium, thus preventing the translocation of lipopolysaccharide, a cell wall component of Gram-negative Proteobacteria and known inducer of lupus in mice, into the circulation. These results suggest that mixed antibiotics or a single antibiotic vancomycin ameliorate SLE-like disease in MRL/lpr mice by changing the composition of gut microbiota.


Subject(s)
Anti-Bacterial Agents/pharmacology , Lupus Erythematosus, Systemic/drug therapy , Animals , Disease Models, Animal , Female , Gastrointestinal Microbiome/drug effects , Interleukins/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Kidney/drug effects , Kidney/metabolism , Lipopolysaccharides/blood , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mice , Spleen/drug effects , Spleen/metabolism , Spleen/pathology
10.
Microbiome ; 5(1): 73, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28697806

ABSTRACT

BACKGROUND: Systemic lupus erythematosus, characterized by persistent inflammation, is a complex autoimmune disorder with no known cure. Immunosuppressants used in treatment put patients at a higher risk of infections. New knowledge of disease modulators, such as symbiotic bacteria, can enable fine-tuning of parts of the immune system, rather than suppressing it altogether. RESULTS: Dysbiosis of gut microbiota promotes autoimmune disorders that damage extraintestinal organs. Here we report a role of gut microbiota in the pathogenesis of renal dysfunction in lupus. Using a classical model of lupus nephritis, MRL/lpr, we found a marked depletion of Lactobacillales in the gut microbiota. Increasing Lactobacillales in the gut improved renal function of these mice and prolonged their survival. We used a mixture of 5 Lactobacillus strains (Lactobacillus oris, Lactobacillus rhamnosus, Lactobacillus reuteri, Lactobacillus johnsonii, and Lactobacillus gasseri), but L. reuteri and an uncultured Lactobacillus sp. accounted for most of the observed effects. Further studies revealed that MRL/lpr mice possessed a "leaky" gut, which was reversed by increased Lactobacillus colonization. Lactobacillus treatment contributed to an anti-inflammatory environment by decreasing IL-6 and increasing IL-10 production in the gut. In the circulation, Lactobacillus treatment increased IL-10 and decreased IgG2a that is considered to be a major immune deposit in the kidney of MRL/lpr mice. Inside the kidney, Lactobacillus treatment also skewed the Treg-Th17 balance towards a Treg phenotype. These beneficial effects were present in female and castrated male mice, but not in intact males, suggesting that the gut microbiota controls lupus nephritis in a sex hormone-dependent manner. CONCLUSIONS: This work demonstrates essential mechanisms on how changes of the gut microbiota regulate lupus-associated immune responses in mice. Future studies are warranted to determine if these results can be replicated in human subjects.


Subject(s)
Gastrointestinal Microbiome , Kidney/physiopathology , Lactobacillus/physiology , Lupus Nephritis/microbiology , Lupus Nephritis/therapy , Animals , Disease Models, Animal , Female , Immunoglobulin G/blood , Interleukin-10/biosynthesis , Interleukin-10/blood , Interleukin-6/biosynthesis , Kidney/immunology , Kidney/pathology , Lactobacillus/classification , Lactobacillus/growth & development , Lactobacillus/isolation & purification , Lupus Nephritis/immunology , Lupus Nephritis/physiopathology , Male , Mice , Mice, Inbred MRL lpr , Orchiectomy , Sex Factors , T-Lymphocytes, Regulatory
11.
Int Immunol ; 29(6): 263-276, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28637300

ABSTRACT

The course and severity of lupus in spontaneous murine lupus models varies among laboratories, which may be due to variations in diet, housing and/or local environmental conditions. In this study, we investigated the influence of common rodent diets while keeping other factors constant. Female lupus-prone MRL/lpr (MRL/MpJ-Faslpr/J) mice were subjected to the same housing conditions and given one of the three diets: Teklad 7013 containing isoflavone-rich soy and alfalfa, Harlan 2018 isoflavone-rich soy-based diet or Research Diets Inc. D11112226 (RD) purified-ingredients diet containing casein and no phytoestrogens. While the total caloric intake was similar among all three treatment groups, mice fed on the 2018 diet developed higher levels of proteinuria and mice fed on either 7013 or 2018 developed higher levels of glomerular immune complex deposition. Remarkably, mice fed the RD diet had markedly decreased proteinuria with diminished C3, total IgG, IgG1 and IgG3 immune complex deposition, along with reduced CD11b+ cellular infiltration into the glomeruli. The type of diet intake also influenced cytokine production, fecal microbiota (increased Lachnospiraceae in mice fed on 2018), altered microRNAs (miRNAs; higher levels of lupus-associated miR-148a and miR-183 in mice fed on 7013 and/or 2018) and altered DNA methylation. This is the first study to comprehensively compare the cellular, molecular and epigenetic effects of these commercial diets in murine lupus.


Subject(s)
DNA Methylation , Diet/adverse effects , Glomerulonephritis/etiology , Lupus Erythematosus, Systemic/etiology , MicroRNAs/genetics , Microbiota/immunology , Proteinuria/etiology , Animals , Antigen-Antibody Complex/metabolism , Caseins/administration & dosage , Commerce , Disease Models, Animal , Energy Intake , Female , Glomerulonephritis/genetics , Humans , Isoflavones/administration & dosage , Lupus Erythematosus, Systemic/genetics , Mice , Mice, Inbred MRL lpr , Rodentia , Soy Foods/adverse effects
12.
Front Immunol ; 8: 598, 2017.
Article in English | MEDLINE | ID: mdl-28588585

ABSTRACT

The intestinal epithelial lining, together with factors secreted from it, forms a barrier that separates the host from the environment. In pathologic conditions, the permeability of the epithelial lining may be compromised allowing the passage of toxins, antigens, and bacteria in the lumen to enter the blood stream creating a "leaky gut." In individuals with a genetic predisposition, a leaky gut may allow environmental factors to enter the body and trigger the initiation and development of autoimmune disease. Growing evidence shows that the gut microbiota is important in supporting the epithelial barrier and therefore plays a key role in the regulation of environmental factors that enter the body. Several recent reports have shown that probiotics can reverse the leaky gut by enhancing the production of tight junction proteins; however, additional and longer term studies are still required. Conversely, pathogenic bacteria that can facilitate a leaky gut and induce autoimmune symptoms can be ameliorated with the use of antibiotic treatment. Therefore, it is hypothesized that modulating the gut microbiota can serve as a potential method for regulating intestinal permeability and may help to alter the course of autoimmune diseases in susceptible individuals.

13.
Front Immunol ; 6: 608, 2015.
Article in English | MEDLINE | ID: mdl-26648937

ABSTRACT

Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease. Despite years of study, the etiology of SLE is still unclear. Both genetic and environmental factors have been implicated in the disease mechanisms. In the past decade, a growing body of evidence has indicated an important role of gut microbes in the development of autoimmune diseases, including type 1 diabetes, rheumatoid arthritis, and multiple sclerosis. However, such knowledge on SLE is little, though we have already known that environmental factors can trigger the development of lupus. Several recent studies have suggested that alterations of the gut microbial composition may be correlated with SLE disease manifestations, while the exact roles of either symbiotic or pathogenic microbes in this disease remain to be explored. Elucidation of the roles of gut microbes - as well as the roles of diet that can modulate the composition of gut microbes - in SLE will shed light on how this autoimmune disorder develops, and provide opportunities for improved biomarkers of the disease and the potential to probe new therapies. In this review, we aim to compile the available evidence on the contributions of diet and gut microbes to SLE occurrence and pathogenesis.

14.
Virology ; 435(2): 301-7, 2013 Jan 20.
Article in English | MEDLINE | ID: mdl-23063406

ABSTRACT

Twelve avian-origin H3N2 influenza strains were isolated from dogs with signs of respiratory disease in northern China during 2009-2010. Phylogenetic analysis showed that eight gene segments of all the isolates had a close relationship with those of avian-origin H3N2 canine influenza viruses (CIVs) from South Korea and southern China. Genetic analysis indicated that these isolates had a PERQTR/G HA cleavage motif, which differed from the PEKQTR/G motif of canine viruses before 2007. Noteworthy, one of our isolates had an additional basic amino acid at position -3 of the HA cleavage site, with a sequence of PERRTR/G which might facilitate the HA cleavage. An insertion of two amino acids at positions 74-75 in the neuraminidase stalk were found in all H3N2 CIVs isolated since 2009. Our findings show the continued evolution of avian-origin H3N2 CIVs and emphasize the necessity of continued surveillance of influenza virus in dogs.


Subject(s)
Influenza A Virus, H3N2 Subtype/genetics , Influenza in Birds/transmission , Orthomyxoviridae Infections/veterinary , Animals , Birds/virology , China/epidemiology , Dog Diseases/epidemiology , Dog Diseases/physiopathology , Dog Diseases/virology , Dogs , Evolution, Molecular , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza in Birds/virology , Molecular Sequence Data , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/virology , Phylogeny , Sequence Analysis, DNA
15.
J Hazard Mater ; 237-238: 1-9, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-22959477

ABSTRACT

Highly dispersed Co(0.5)Zn(0.5)Fe(2)O(4)/polypyrrole (CZFO/PPy) nanocomposites with enhanced electromagnetic properties and large surface area were rapidly and controllably prepared using microfluidic reactors. A novel magnetically controllable microdevice using the new adsorbent in a highly dispersed form was assembled and used for fluoride adsorption. Compared with traditional adsorption methods, the device displayed high adsorption efficiency and capacity. The adsorbents were regenerated with no significant loss in defluoridation ability, which indicates that the device is a realistic and highly efficient alternative way of removing fluoride pollution at low cost.


Subject(s)
Fluorides/chemistry , Nanocomposites/chemistry , Water Pollutants, Chemical/chemistry , Water Purification/instrumentation , Adsorption , Cobalt/chemistry , Cost-Benefit Analysis , Equipment Design , Iron/chemistry , Magnetic Phenomena , Polymers/chemistry , Pyrroles/chemistry , Water Purification/methods , Zinc/chemistry
16.
J Colloid Interface Sci ; 365(1): 308-13, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21974921

ABSTRACT

Herein we report the direct fabrication of TiO(2) subwavelength structures with 1-dimensional TiO(2) nanorods on glass substrate through solvothermal process to form self-cleaning antireflection coatings. TiO(2) precursor solutions with different solvent constituents create TiO(2) nanorods with much different morphologies grown on glass substrates. Apiculate TiO(2) nanorods with vertical orientation are grown on the glass substrate which is solvothermally treated in the precursor solution containing ethylene glycol. This glass substrate exhibit the highest transmittance of 70-85% in the range of 520-800 nm and negligible absorption in visible light region (400-800 nm). Furthermore, the TiO(2) nanorod arrays show high hydrophobicity and photocatalytic degradation ability which offer the glass substrate self-cleaning properties for both hydrophilic and oily contaminants.


Subject(s)
Ethylene Glycol/chemistry , Glass/chemistry , Hydrophobic and Hydrophilic Interactions , Nanotubes/chemistry , Titanium/chemistry , Catalysis , Photochemistry , Surface Properties
17.
J Hazard Mater ; 188(1-3): 363-8, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21345584

ABSTRACT

High-density arrays of vertically oriented TiO(2) nanorods with uniform distribution on Ti foil have been formed through template-free oxidation of Ti in hydrogen peroxide solutions. Subsequent thermal treatment was applied for growing mixed crystal structures to pursue higher performance. Morphology characterization using field emission scanning electron microscopy (FESEM) shows a nanorod diameter in the range of 20-50 nm with a length of 1.5 µm. X-ray diffraction (XRD) measurement demonstrates the crystallization of the TiO(2) nanorods prior to thermal treatment and the formation of anatase and rutile mixed phase after thermal treatment. The mixed crystal TiO(2) nanorods show a much higher performance than pure anatase in photoelectrochemical experiments. Steady-state photocurrent resulted from photocatalytic oxidation of organic compounds by TiO(2) nanorods is employed as response signal in determination of the organics to yield a linear range of 0-1.1mM for glucose. For other organics, an excellent linear relationship between the net steady-state photocurrent and the concentration of electrons transferred in exhaustive oxidation for these organics is obtained, which empowers the mixed crystal TiO(2) nanorods to serve as versatile material in organics-sensing application.


Subject(s)
Glucose/analysis , Nanotubes/chemistry , Organic Chemicals/analysis , Titanium/chemistry , Crystallization , Electrochemical Techniques , Hydrogen Peroxide , Oxidation-Reduction , Photochemistry/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...