105 results on '"Xin M Luo"'
Search Results
2. SLE: Another Autoimmune Disorder Influenced by Microbes and Diet?
- Author
-
Qinghui eMu, Husen eZhang, and Xin M Luo
- Subjects
Diet ,Hygiene Hypothesis ,microbiota ,estrogen ,SLE ,Bacterial antigens ,Immunologic diseases. Allergy ,RC581-607 - 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.
- Published
- 2015
- Full Text
- View/download PDF
3. Paradoxical effects of all-trans-retinoic acid on lupus-like disease in the MRL/lpr mouse model.
- Author
-
Xiaofeng Liao, Jingjing Ren, Cheng-Hsin Wei, A Catharine Ross, Thomas E Cecere, Bernard S Jortner, S Ansar Ahmed, and Xin M Luo
- Subjects
Medicine ,Science - Abstract
Roles of all-trans-retinoic acid (tRA), a metabolite of vitamin A (VA), in both tolerogenic and immunogenic responses are documented. However, how tRA affects the development of systemic autoimmunity is poorly understood. Here we demonstrate that tRA have paradoxical effects on the development of autoimmune lupus in the MRL/lpr mouse model. We administered, orally, tRA or VA mixed with 10% of tRA (referred to as VARA) to female mice starting from 6 weeks of age. At this age, the mice do not exhibit overt clinical signs of lupus. However, the immunogenic environment preceding disease onset has been established as evidenced by an increase of total IgM/IgG in the plasma and expansion of lymphocytes and dendritic cells in secondary lymphoid organs. After 8 weeks of tRA, but not VARA treatment, significantly higher pathological scores in the skin, brain and lung were observed. These were accompanied by a marked increase in B-cell responses that included autoantibody production and enhanced expression of plasma cell-promoting cytokines. Paradoxically, the number of lymphocytes in the mesenteric lymph node decreased with tRA that led to significantly reduced lymphadenopathy. In addition, tRA differentially affected renal pathology, increasing leukocyte infiltration of renal tubulointerstitium while restoring the size of glomeruli in the kidney cortex. In contrast, minimal induction of inflammation with tRA in the absence of an immunogenic environment in the control mice was observed. Altogether, our results suggest that under a predisposed immunogenic environment in autoimmune lupus, tRA may decrease inflammation in some organs while generating more severe disease in others.
- Published
- 2015
- Full Text
- View/download PDF
4. The role of gut microbiota in different murine models of systemic lupus erythematosus
- Author
-
Ran Lu and Xin M. Luo
- Subjects
Lupus ,SLE ,gut microbiota ,mouse models ,leaky gut ,molecular mimicry ,Internal medicine ,RC31-1245 - Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by immune system dysfunction that can lead to serious health issues and mortality. Recent investigations highlight the role of gut microbiota alterations in modulating inflammation and disease severity in SLE. This review specifically summaries the variations in gut microbiota composition across various murine models of lupus. By focusing on these differences, we aim to elucidate the intricate relationship between gut microbiota dysbiosis and the development and progression of SLE in preclinical settings.
- Published
- 2024
- Full Text
- View/download PDF
5. IgA-mediated control of host-microbial interaction during weaning reaction influences gut inflammation
- Author
-
Wenjie Tang, Yusen Wei, Zhixiang Ni, Kangwei Hou, Xin M. Luo, and Haifeng Wang
- Subjects
Weaning reaction ,gut inflammation ,bacteroides uniformis ,CD138+ plasmacyte ,IgA-coated bacteria ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
ABSTRACTThe mechanisms of how host-microbe mutualistic relationships are established at weaning contingently upon B-cell surveillance remain inadequately elucidated. We found that CD138+ plasmacyte (PC)-mediated promotion of IgA response regulates the symbiosis between Bacteroides uniformis (B. uniformis) and the host during the weaning period. The IgA-skewed response of CD138+ PCs is essential for B. uniformis to occupy a defined gut luminal niche, thereby fostering stable colonization. Furthermore, B. uniformis within the natural gut niche was perturbed in the absence of IgA, resulting in exacerbated gut inflammation in IgA-deficient mice and weaned piglets. Thus, we propose that the priming and maintenance of intestinal IgA response from CD138+ PCs are required for host-microbial symbiosis, whereas the perturbation of which would enhance inflammation in weaning process.
- Published
- 2024
- Full Text
- View/download PDF
6. Recombination activating gene-2 regulates CpG-mediated interferon-α production in mouse bone marrow-derived plasmacytoid dendritic cells.
- Author
-
Xin M Luo and Margarida Y Y Lei
- Subjects
Medicine ,Science - Abstract
Using mice that lack recombination activating gene-2 (Rag2), we have found that bone marrow-derived plasmacytoid dendritic cells (pDCs) as main producers of interferon-α (IFNα) require Rag2 for normal development. This is a novel function for Rag2, whose classical role is to initiate B and T cell development. Here we showed that a population of common progenitor cells in the mouse bone marrow possessed the potential to become either B cells or pDCs upon appropriate stimulations, and the lack of Rag2 hindered the development of both types of progeny cells. A closer look at pDCs revealed that Rag2⁻/⁻ pDCs expressed a high level of Ly6C and were defective at producing IFNα in response to CpG, a ligand for toll-like receptor 9. This phenotype was not shared by Rag1⁻/⁻ pDCs. The induction of CCR7, CD40 and CD86 with CpG, however, was normal in Rag2⁻/⁻ pDCs. In addition, Rag2⁻/⁻ pDCs retained the function to promote antibody class switching and plasma cell formation through producing IL-6. Further analysis showed that interferon regulatory factor-8, a transcription factor important for both IFNα induction and pDC development, was dysregulated in pDCs lacking Rag2. These results indicate that the generation of interferon response in pDCs requires Rag2 and suggest the lymphoid origin of bone marrow-derived pDCs.
- Published
- 2012
- Full Text
- View/download PDF
7. Dimeric 2G12 as a potent protection against HIV-1.
- Author
-
Xin M Luo, Margarida Y Y Lei, Rana A Feidi, Anthony P West, Alejandro Benjamin Balazs, Pamela J Bjorkman, Lili Yang, and David Baltimore
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
We previously showed that broadly neutralizing anti-HIV-1 antibody 2G12 (human IgG1) naturally forms dimers that are more potent than monomeric 2G12 in in vitro neutralization of various strains of HIV-1. In this study, we have investigated the protective effects of monomeric versus dimeric 2G12 against HIV-1 infection in vivo using a humanized mouse model. Our results showed that passively transferred, purified 2G12 dimer is more potent than 2G12 monomer at preventing CD4 T cell loss and suppressing the increase of viral load following HIV-1 infection of humanized mice. Using humanized mice bearing IgG "backpack" tumors that provided 2G12 antibodies continuously, we found that a sustained dimer concentration of 5-25 µg/ml during the course of infection provides effective protection against HIV-1. Importantly, 2G12 dimer at this concentration does not favor mutations of the HIV-1 envelope that would cause the virus to completely escape 2G12 neutralization. We have therefore identified dimeric 2G12 as a potent prophylactic reagent against HIV-1 in vivo, which could be used as part of an antibody cocktail to prevent HIV-1 infection.
- Published
- 2010
- Full Text
- View/download PDF
8. RETRACTED: Tlr5 deficiency exacerbates lupus-like disease in the MRL/lpr mouse model
- Author
-
Razan M. Alajoleen, David N. Oakland, Rana Estaleen, Aida Shakeri, Ran Lu, Michael Appiah, Sha Sun, Jonathan Neumann, Shimako Kawauchi, Thomas E. Cecere, Ryan P. McMillan, Christopher M. Reilly, and Xin M. Luo
- Subjects
lupus ,TLR5 ,glomerulonephritis ,lymphoproliferation ,gut microbiota ,Immunologic diseases. Allergy ,RC581-607 - Abstract
IntroductionLeaky gut has been linked to autoimmune disorders including lupus. We previously reported upregulation of anti-flagellin antibodies in the blood of lupus patients and lupus-prone mice, which led to our hypothesis that a leaky gut drives lupus through bacterial flagellin-mediated activation of toll-like receptor 5 (TLR5).MethodsWe created MRL/lpr mice with global Tlr5 deletion through CRISPR/Cas9 and investigated lupus-like disease in these mice.ResultContrary to our hypothesis that the deletion of Tlr5 would attenuate lupus, our results showed exacerbation of lupus with Tlr5 deficiency in female MRL/lpr mice. Remarkably higher levels of proteinuria were observed in Tlr5-/- MRL/lpr mice suggesting aggravated glomerulonephritis. Histopathological analysis confirmed this result, and Tlr5 deletion significantly increased the deposition of IgG and complement C3 in the glomeruli. In addition, Tlr5 deficiency significantly increased renal infiltration of Th17 and activated cDC1 cells. Splenomegaly and lymphadenopathy were also aggravated in Tlr5-/- MRL/lpr mice suggesting impact on lymphoproliferation. In the spleen, significant decreased frequencies of regulatory lymphocytes and increased germinal centers were observed with Tlr5 deletion. Notably, Tlr5 deficiency did not change host metabolism or the existing leaky gut; however, it significantly reshaped the fecal microbiota.ConclusionGlobal deletion of Tlr5 exacerbates lupus-like disease in MRL/lpr mice. Future studies will elucidate the underlying mechanisms by which Tlr5 deficiency modulates host-microbiota interactions to exacerbate lupus.
- Published
- 2024
- Full Text
- View/download PDF
9. A double-edged sword: interactions of CX3CL1/CX3CR1 and gut microbiota in systemic lupus erythematosus
- Author
-
Rana A. Estaleen, Christopher M. Reilly, and Xin M. Luo
- Subjects
systemic lupus erythematosus ,lupus ,lupus nephritis ,CX3CR1 ,gut microbiota ,autoimmune disease ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Systemic lupus erythematosus (SLE) is a systemic chronic disease initiated by an abnormal immune response to self and can affect multiple organs. SLE is characterized by the production of autoantibodies and the deposition of immune complexes. In regard to the clinical observations assessed by rheumatologists, several chemokines and cytokines also contribute to disease progression. One such chemokine and adhesion molecule is CX3CL1 (otherwise known as fractalkine). CX3CL1 is involved in cell trafficking and inflammation through recognition by its receptor, CX3CR1. The CX3CL1 protein consists of a chemokine domain and a mucin-like stalk that allows it to function both as a chemoattractant and as an adhesion molecule. In inflammation and specifically lupus, the literature displays contradictory evidence for the functions of CX3CL1/CX3CR1 interactions. In addition, the gut microbiota has been shown to play an important role in the pathogenesis of SLE. This review highlights current studies that illustrate the interactions of the gut microbiota and CX3CR1 in SLE.
- Published
- 2024
- Full Text
- View/download PDF
10. Single-cell RNA sequencing analysis reveals the heterogeneity of IL-10 producing regulatory B cells in lupus-prone mice
- Author
-
Andrea R. Daamen, Razan M. Alajoleen, Amrie C. Grammer, Xin M. Luo, and Peter E. Lipsky
- Subjects
lupus ,Breg ,single-cell ,transcriptomics ,bioinformatics ,Immunologic diseases. Allergy ,RC581-607 - Abstract
IntroductionB cells can have both pathogenic and protective roles in autoimmune diseases, including systemic lupus erythematosus (SLE). Deficiencies in the number or immunosuppressive function of IL-10 producing regulatory B cells (Bregs) can cause exacerbated autoimmune inflammation. However, the exact role of Bregs in lupus pathogenesis has not been elucidated.MethodsWe carried out gene expression analysis by scRNA-seq to characterize differences in splenic Breg subsets and molecular profiles through stages of disease progression in lupus-prone mice. Transcriptome-based changes in Bregs from mice with active disease were confirmed by phenotypic analysis.ResultsWe found that a loss of marginal zone (MZ) lineage Bregs, an increase in plasmablast/plasma cell (PB-PC) lineage Bregs, and overall increases in inflammatory gene signatures were characteristic of active disease as compared to Bregs from the pre-disease stage. However, the frequencies of both MZ Bregs and PB-PCs expressing IL-10 were significantly decreased in active-disease mice.ConclusionOverall, we have identified changes to the repertoire and transcriptional landscape of Breg subsets associated with active disease that provide insights into the role of Bregs in lupus pathogenesis. These results could inform the design of Breg-targeted therapies and interventions to restore Breg suppressive function in autoimmunity.
- Published
- 2023
- Full Text
- View/download PDF
11. CX3CR1 modulates SLE-associated glomerulonephritis and cardiovascular disease in MRL/lpr mice
- Author
-
Xavier Cabana-Puig, Ran Lu, Shuo Geng, Jacquelyn S. Michaelis, Vanessa Oakes, Caitlin Armstrong, James C. Testerman, Xiaofeng Liao, Razan Alajoleen, Michael Appiah, Yao Zhang, Christopher M. Reilly, Liwu Li, and Xin M. Luo
- Subjects
Pharmacology ,Immunology - Published
- 2023
12. Hypovitaminosis A Drives the Progression of Tubulointerstitial Lupus Nephritis through Potentiating Predisease Cellular Autoreactivity
- Author
-
Leila Abdelhamid, Razan Alajoleen, Kathryn M. Kingsmore, Xavier Cabana-Puig, Ran Lu, Jing Zhu, James C. Testerman, Yaqi Li, A. Catharine Ross, Thomas E. Cecere, Christopher M. Reilly, Amrie C. Grammer, Peter E. Lipsky, and Xin M. Luo
- Subjects
Immunology ,Immunology and Allergy ,General Medicine - Abstract
Vitamin A (VA) deficiency (VAD) is observed in both humans and mice with lupus nephritis. However, whether VAD is a driving factor for accelerated progression of lupus nephritis is unclear. In this study, we investigated the effect of VAD on the progression of lupus nephritis in a lupus-prone mouse model, MRL/lpr. We initiated VAD either during gestation or after weaning to reveal a potential time-dependent effect. We found exacerbated lupus nephritis at ∼15 wk of age with both types of VAD that provoked tubulointerstitial nephritis leading to renal failure. This was concomitant with significantly higher mortality in all VAD mice. Importantly, restoration of VA levels after weaning reversed VAD-induced mortality. These results suggest VAD-driven acceleration of tubulointerstitial lupus nephritis. Mechanistically, at the earlier time point of 7 wk of age and before the onset of clinical lupus nephritis, continued VAD (from gestation until postweaning) enhanced plasma cell activation and augmented their autoantibody production, while also increasing the expansion of T lymphocytes that could promote plasma cell autoreactivity. Moreover, continued VAD increased the renal infiltration of plasmacytoid dendritic cells. VAD initiated after weaning, in contrast, showed modest effects on autoantibodies and renal plasmacytoid dendritic cells that were not statistically significant. Remarkably, analysis of gene expression in human kidney revealed that the retinoic acid pathway was decreased in the tubulointerstitial region of lupus nephritis, supporting our findings in MRL/lpr mice. Future studies will elucidate the underlying mechanisms of how VAD modulates cellular functions to exacerbate tubulointerstitial lupus nephritis.
- Published
- 2023
13. 1301 Lactobacillusspp. act in synergy to attenuate splenomegaly and lymphadenopathy in lupus- prone MRL/lprmice
- Author
-
Xavier Cabana-Puig, Qinghui Mu, Ran Lu, Brianna Swartwout, Leila Abdelhamid, Jing Zhu, Meeta Prakash, Thomas E Cecere, Zhuang Wang, Sabrina Callaway, Sha Sun, Christopher M Reilly, S Ansar Ahmed, and Xin M Luo
- Published
- 2022
14. Frontiers in Immunology
- Author
-
Xavier Cabana-Puig, Qinghui Mu, Ran Lu, Brianna Swartwout, Leila Abdelhamid, Jing Zhu, Meeta Prakash, Thomas E. Cecere, Zhuang Wang, Sabrina Callaway, Sha Sun, Christopher M. Reilly, S. Ansar Ahmed, and Xin M. Luo
- Subjects
type 1 regulatory T cells ,Mice, Inbred MRL lpr ,gut microbiota ,Inflammatory and immune system ,Immunology ,Lymphadenopathy ,lupus ,Autoimmune Disease ,double-negative T cells ,Lactobacillus ,Mice ,memory T cells ,Splenomegaly ,Animals ,Dysbiosis ,Immunology and Allergy - 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 familyLactobacillaceae, in lupus-prone MRL/lprmice, a model of systemic autoimmunity. Restoring the microbiota with a mix of 5 differentLactobacillusspecies (spp.),L. reuteri, L. oris, L. johnsonii, L. gasseriandL. 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,Lactobacillusspp. 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 endogenousLactobacillusspp. 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 ofLactobacillusspp. 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 thatLactobacillusspp. might act on T cells to attenuate splenomegaly and lymphadenopathy. Together, this study advances our understanding of howLactobacillusspp. attenuate lupus in MRL/lprmice. The synergistic action of these bacteria suggests that multiple probiotic bacteria in combination may dampen systemic autoimmunity and benefit lupus patients.
- Published
- 2022
- Full Text
- View/download PDF
15. AI-2/LuxS Quorum Sensing System Promotes Biofilm Formation of Lactobacillus rhamnosus GG and Enhances the Resistance to Enterotoxigenic Escherichia coli in Germ-Free Zebrafish
- Author
-
Zhaoxi Deng, Kangwei Hou, Teresa G. Valencak, Xin M. Luo, Jianxin Liu, and Haifeng Wang
- Subjects
Microbiology (medical) ,Inflammation ,General Immunology and Microbiology ,Ecology ,Physiology ,Lacticaseibacillus rhamnosus ,Quorum Sensing ,Cell Biology ,Gene Expression Regulation, Bacterial ,Carbon-Sulfur Lyases ,Infectious Diseases ,Bacterial Proteins ,Biofilms ,Genetics ,Animals ,Enterotoxigenic Escherichia coli ,Zebrafish - Abstract
Lactobacillus rhamnosus GG is a widely used probiotic to improve host intestinal health, promote growth, reduce diarrhea, and modulate immunity. In recent years, the bacterial quorum sensing system has attracted much attention; however, there has not been much research on the effect of the LuxS/AI-2 quorum sensing system of Lactobacillus on bacteriostasis, microbial ecology balance, and immune regulation in intestine.
- Published
- 2022
16. Analyses of Proteinuria, Renal Infiltration of Leukocytes, and Renal Deposition of Proteins in Lupus-prone MRL/lpr Mice
- Author
-
Xin M. Luo and Xavier Cabana-Puig
- Subjects
Inflammation ,Mice, Inbred MRL lpr ,General Immunology and Microbiology ,General Chemical Engineering ,General Neuroscience ,Kidney ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Proteinuria ,Leukocytes ,Animals ,Humans ,Lupus Erythematosus, Systemic ,Female - Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder with no known cure and is characterized by persistent inflammation in many organs, including the kidneys. Under such circumstances, the kidney loses its ability to clean waste from the blood and regulate salt and fluid concentrations, eventually leading to renal failure. Women, particularly those of childbearing age, are diagnosed nine times more often than men. Kidney disease is the leading cause of mortality in SLE patients. The present protocol describes a quick and simple method to measure excreted protein levels in collected urine, tracking lupus progression over time. In addition, an approach to isolate kidney mononuclear cells is provided based on size and density selection to investigate renal infiltration of leukocytes. Furthermore, an immunohistochemical method has been developed to characterize protein deposition in the glomeruli and leukocyte infiltration in the tubulointerstitial space. Together, these methods can help investigate the progression of chronic inflammation associated with the kidneys of lupus-prone MRL/lpr mice.
- Published
- 2022
17. Frontiers in Immunology
- Author
-
Anna Christovich and Xin M. Luo
- Subjects
gut microbiota ,type 1 diabetes ,Immunology ,leaky gut ,Autoimmunity ,multiple sclerosis ,Autoimmune Diseases ,Gastrointestinal Microbiome ,Mice ,systemic lupus erythematosus ,Immunology and Allergy ,Animals ,Dysbiosis ,Humans ,Lupus Erythematosus, Systemic - Abstract
With the rising prevalence of autoimmune diseases, the role of the environment, specifically the gut microbiota, in disease development has grown to be a major area of study. Recent advances show a relationship and possible cause and effect between the gut microbiota and the initiation or exacerbation of autoimmune diseases. Furthermore, microbial dysbiosis and leaky gut are frequent phenomena in both human autoimmune diseases and the murine autoimmunity models. This review will focus on literature in recent years concerning the gut microbiota and leaky gut in relation to the autoimmune diseases, including systemic lupus erythematosus, type 1 diabetes, and multiple sclerosis. Published version
- Published
- 2022
18. Analysis of Fecal Microbiota Dynamics in Lupus-Prone Mice Using a Simple, Cost-Effective DNA Isolation Method
- Author
-
Xin M. Luo, Christopher M. Reilly, and Xavier Cabana Puig
- Subjects
Mice, Inbred MRL lpr ,General Immunology and Microbiology ,Cost-Benefit Analysis ,Microbiota ,General Chemical Engineering ,General Neuroscience ,DNA ,General Biochemistry, Genetics and Molecular Biology ,Autoimmune Diseases ,Feces ,Mice ,RNA, Ribosomal, 16S ,Animals ,Female - Abstract
Gut microbiota has an important role in educating the immune system. This relationship is extremely important for understanding autoimmune diseases that are not only driven by genetic factors, but also environmental factors that can trigger the onset and/or worsen the disease course. A previously published study on the dynamics of the gut microbiota in lupus-prone MRL/lpr female mice showed how changes of the gut microbiota can alter disease progression. Here, a protocol is described for extracting representative samples from the gut microbiota for studies of autoimmunity. Microbiota samples are collected from the anus and processed, from which the DNA is extracted using a phenol-chloroform method and purified by alcohol precipitation. After PCR is performed, purified amplicons are sequenced using a Next Generation Sequencing platform at Argonne National Laboratory. Finally, the 16S ribosomal RNA gene sequencing data is analyzed. As an example, data obtained from gut microbiota comparisons of MRL/lpr mice with or without CX3CR1 are shown. Results showed significant differences in genera containing pathogenic bacteria such as those in the phylum Proteobacteria, as well as the genus Bifidobacterium, which is considered part of the healthy commensal microbiota. In summary, this simple, cost-effective DNA isolation method is reliable and can help the investigation of gut microbiota changes associated with autoimmune diseases.
- Published
- 2022
19. Inflamed synovial fluid induces a homeostatic response in bone marrow mononuclear cells in vitro: Implications for joint therapy
- Author
-
Linda A. Dahlgren, Theodore S. Kalbfleisch, Bruno C. Menarim, Xin M. Luo, Caitlin Mason, Kiersten H. Gillis, Stephen R. Werre, James N. MacLeod, Christopher R. Byron, and Andrea Oliver
- Subjects
Male ,0301 basic medicine ,Interleukin-1beta ,Inflammation ,Cartilage metabolism ,Biochemistry ,Peripheral blood mononuclear cell ,Regulatory macrophages ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Synovial Fluid ,Genetics ,medicine ,Animals ,Synovial fluid ,Macrophage ,Horses ,Insulin-Like Growth Factor I ,Molecular Biology ,Cells, Cultured ,Cell Proliferation ,Synovitis ,business.industry ,Macrophages ,Flow Cytometry ,Interleukin-10 ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Leukocytes, Mononuclear ,Cytokines ,Female ,Bone marrow ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Synovial inflammation is a central feature of osteoarthritis (OA), elicited when local regulatory macrophages (M2-like) become overwhelmed, activating an inflammatory response (M1-like). Bone marrow mononuclear cells (BMNC) are a source of naive macrophages capable of reducing joint inflammation and producing molecules essential for cartilage metabolism. This study investigated the response of BMNC to normal (SF) and inflamed synovial fluid (ISF). Equine BMNC cultured in autologous SF or ISF (n = 8 horses) developed into macrophage-rich cultures with phenotypes similar to cells native to normal SF and became more confluent in ISF (~100%) than SF (~25%). BMNC cultured in SF or ISF were neither M1- nor M2-like, but exhibited aspects of both phenotypes and a regulatory immune response, characterized by increasing counts of IL-10+ macrophages, decreasing IL-1β concentrations and progressively increasing IL-10 and IGF-1 concentrations. Changes were more marked in ISF and suggest that homeostatic mechanisms were preserved over time and were potentially favored by progressive cell proliferation. Collectively, our data suggest that intra-articular BMNC could increase synovial macrophage counts, potentiating the macrophage- and IL-10-associated mechanisms of joint homeostasis lost during the progression of OA, preserving the production of cytokines involved in tissue repair (PGE2 , IL-10) generally impaired by frequently used corticosteroids.
- Published
- 2020
20. Diet and Hygiene in Modulating Autoimmunity During the Pandemic Era
- Author
-
Leila Abdelhamid and Xin M. Luo
- Subjects
SARS-CoV-2 ,Incidence ,autoimmunity ,Immunology ,COVID-19 ,Hygiene ,Review ,RC581-607 ,immunomodulation ,Severity of Illness Index ,immune homeostasis ,Autoimmune Diseases ,Diet ,Immune Tolerance ,Animals ,Humans ,Immunology and Allergy ,Immunologic diseases. Allergy ,Pandemics - Abstract
The immune system is an efficiently toned machinery that discriminates between friends and foes for achieving both host defense and homeostasis. Deviation of immune recognition from foreign to self and/or long-lasting inflammatory responses results in the breakdown of tolerance. Meanwhile, educating the immune system and developing immunological memory are crucial for mounting defensive immune responses while protecting against autoimmunity. Still to elucidate is how diverse environmental factors could shape autoimmunity. The emergence of a world pandemic such as SARS-CoV-2 (COVID-19) not only threatens the more vulnerable individuals including those with autoimmune conditions but also promotes an unprecedented shift in people’s dietary approaches while urging for extraordinary hygiene measures that likely contribute to the development or exacerbation of autoimmunity. Thus, there is an urgent need to understand how environmental factors modulate systemic autoimmunity to better mitigate the incidence and or severity of COVID-19 among the more vulnerable populations. Here, we discuss the effects of diet (macronutrients and micronutrients) and hygiene (the use of disinfectants) on autoimmunity with a focus on systemic lupus erythematosus.
- Published
- 2022
21. Phenotypic Drift in Lupus-Prone MRL/lpr Mice: Potential Roles of MicroRNAs and Gut Microbiota
- Author
-
Xavier Cabana-Puig, Jacob M. Bond, Zhuang Wang, Rujuan Dai, Ran Lu, Amy Lin, Vanessa Oakes, Amy Rizzo, Brianna Swartwout, Leila Abdelhamid, Jiangdi Mao, Meeta Prakash, Constanza Sangmeister, Nathaniel Cheung, Catharine Cowan, Christopher M. Reilly, Sha Sun, S. Ansar Ahmed, and Xin M. Luo
- Subjects
Male ,Mice, Inbred MRL lpr ,Immunology ,General Medicine ,Kidney ,Lupus Nephritis ,Gastrointestinal Microbiome ,Disease Models, Animal ,Mice ,MicroRNAs ,RNA, Ribosomal, 16S ,Animals ,Immunology and Allergy ,Female ,Spleen - Abstract
MRL/lpr mice have been extensively used as a murine model of lupus. Disease progression in MRL/lpr mice can differ among animal facilities, suggesting a role for environmental factors.We noted a phenotypic drift of our in-house colony, which was the progeny of mice obtained from The Jackson Laboratory (JAX; stocking number 000485), that involved attenuated glomerulonephritis, increased splenomegaly, and reduced lymphadenopathy. To validate our in-house mice as a model of lupus, we compared these mice with those newly obtained from JAX, which were confirmed to be genetically identical to our in-house mice. Surprisingly, the new JAX mice exhibited a similar phenotypic drift, most notably the attenuation of glomerulonephritis. Interestingly, our in-house colony differed from JAX mice in body weight and kidney size (both sexes), as well as in splenic size, germinal center formation, and level of anti-dsDNA auto-IgG in the circulation (male only). In addition, we noted differential expression of microRNA (miR)-21 and miR-183 that might explain the splenic differences in males. Furthermore, the composition of gut microbiota was different between in-house and new JAX mice at early time points, which might explain some of the renal differences (e.g., kidney size). However, we could not identify the reason for attenuated glomerulonephritis, a shared phenotypic drift between the two colonies. It is likely that this was due to certain changes of environmental factors present in both JAX and our facilities. Taken together, these results suggest a significant phenotypic drift in MRL/lpr mice in both colonies that may require strain recovery from cryopreservation. This work was supported by internal funding from Virginia-Maryland College of Veterinary Medicine and National Institutes of Health Grants AR067418 and AR073240 (X.M.L.). Published version
- Published
- 2022
22. Intermittent Fasting Reshapes the Gut Microbiota and Metabolome and Reduces Weight Gain More Effectively Than Melatonin in Mice
- Author
-
Jingliang Liu, Yifan Zhong, Yanfei Ma, Haifeng Wang, Xin M. Luo, and Jianxin Liu
- Subjects
medicine.medical_specialty ,media_common.quotation_subject ,Endocrinology, Diabetes and Metabolism ,melatonin ,Gut flora ,liver ,Melatonin ,chemistry.chemical_compound ,Adipocyte ,Internal medicine ,Intermittent fasting ,medicine ,Metabolome ,TX341-641 ,metabolites ,media_common ,Nutrition ,Original Research ,Nutrition and Dietetics ,Triglyceride ,biology ,gut microbiota ,Nutrition. Foods and food supply ,intermittent fasting ,Appetite ,intestinal morphology ,biology.organism_classification ,Endocrinology ,chemistry ,medicine.symptom ,Weight gain ,medicine.drug ,Food Science - Abstract
Background: Intermittent fasting (IF) can reduce energy intake and body weight (BW). Melatonin has many known functions, which include reducing appetite and preventing excessive weight gain.Objective: This study aimed to investigate the effects of IF on body fat and the gut microbiota and metabolome as well as a potential interaction with melatonin.Methods: Male C57BL/6J mice (23.0 ± 0.9 g, 6 wk old) were randomly assigned into four groups (12 mice/group): control (C), intermittent fasting (F), melatonin (M), and intermittent fasting plus melatonin (MF). The C and M groups mice were provided with ad libitum access to food and water, while the F and MF groups underwent alternative-day feed deprivation (15 cycles total). Melatonin was administered in the drinking water of the M and MF groups. Blood, epididymal fat, liver tissue, and intestinal tissue and contents were collected for lab measurements, histology, and microbiota and metabolome analysis. Main effects and interactions were tested by 2-factor ANOVA.Results: IF significantly reduced BW gain and serum glucose, total cholesterol (TC) and triglyceride (TG) levels. Adipocyte size significantly decreased with IF, then the number of adipocytes per square millimeter significantly increased (P < 0.05). Compared to the C group, the M and MF groups had significantly higher serum melatonin levels (17 and 21%, respectively), although melatonin monotherapy had no effect on serum parameters and adipocytes. There was no interaction between IF and melatonin on BW gain and serum parameters except for on adipocyte area and number per square millimeter, Bacteroidetes and Akkermansia bacterial abundance, and the levels of the intestinal metabolites alanine, valine and isoleucine. IF changed the intestinal microbiota structure, with the F and MF groups clearly separating from the C and M groups. Metabolomic analysis showed that there was obvious separation between all four groups.Conclusions: IF, but neither melatonin nor the interaction between IF and melatonin, could alter intestinal microbiota and metabolism and prevent obesity by reducing BW gain, serum glucose, TC, and TG, and adipocyte size in mice.
- Published
- 2021
- Full Text
- View/download PDF
23. Lactobacillus reuteri ZJ617 Culture Supernatant Attenuates Acute Liver Injury Induced in Mice by Lipopolysaccharide
- Author
-
Yanjun Cui, Renlong Tang, Haifeng Wang, Jiangdi Mao, Sirui Qi, Wenming Zhang, Chong Wang, Jianxin Liu, and Xin M. Luo
- Subjects
Limosilactobacillus reuteri ,Lipopolysaccharides ,Male ,0301 basic medicine ,medicine.medical_specialty ,Lipopolysaccharide ,MAP Kinase Signaling System ,medicine.medical_treatment ,Intraperitoneal injection ,Medicine (miscellaneous) ,Aspartate transaminase ,Apoptosis ,Protective Agents ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Autophagy ,medicine ,Animals ,Nutrition and Dietetics ,biology ,business.industry ,Probiotics ,NF-kappa B ,biology.organism_classification ,Lactobacillus reuteri ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Liver ,Alanine transaminase ,chemistry ,Culture Media, Conditioned ,030220 oncology & carcinogenesis ,biology.protein ,TLR4 ,Tumor necrosis factor alpha ,Chemical and Drug Induced Liver Injury ,Inflammation Mediators ,business ,Signal Transduction - Abstract
BACKGROUND Lactobacillus rhamnosus GG culture supernatant (LGGs) promotes intestinal integrity and ameliorates acute liver injury induced by alcohol in mice. OBJECTIVES The aim of this study was to investigate the protective effects and molecular mechanisms of Lactobacillus reuteri ZJ617 culture supernatant (ZJ617s) on acute liver injury induced by lipopolysaccharide (LPS) in mice. METHODS Male C57BL/6 mice (20 ± 2 g, 8 wk old) were randomly divided into 4 groups (6 mice/group): oral inoculation with phosphate-buffered saline (control), intraperitoneal injection of LPS (10 mg/kg body weight) (LPS), oral inoculation with ZJ617s 2 wk before intraperitoneal injection of LPS (ZJ617s + LPS), or oral inoculation with LGGs 2 wk before intraperitoneal injection of LPS (LGGs + LPS). Systemic inflammation, intestinal integrity, biomarkers of hepatic function, autophagy, and apoptosis signals in the liver were determined. RESULTS Twenty-four hours after LPS injection, the activities of serum alanine transaminase and aspartate transaminase were 32.2% and 30.3% lower in the ZJ617s + LPS group compared with the LPS group, respectively (P
- Published
- 2019
24. Autologous bone marrow mononuclear cells modulate joint homeostasis in an equine in vivo model of synovitis
- Author
-
Andrea Oliver, Sarah H. Barrett, Bruno C. Menarim, Stephen R. Werre, Linda A. Dahlgren, Christopher R. Byron, Ying Ngo, Kiersten H. Gillis, Xin M. Luo, and Caitlin Mason
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,business.industry ,Osteoarthritis ,medicine.disease ,Autologous bone ,Biochemistry ,Peripheral blood mononuclear cell ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,In vivo ,Synovitis ,Genetics ,medicine ,Macrophage ,Bone marrow ,business ,Molecular Biology ,030217 neurology & neurosurgery ,Homeostasis ,Biotechnology - Abstract
Osteoarthritis (OA) is characterized by macrophage-driven synovitis. Macrophages promote synovial health but become inflammatory when their regulatory functions are overwhelmed. Bone marrow mononuc...
- Published
- 2019
25. Regulation of neonatal IgA production by the maternal microbiota
- Author
-
Grace Lee, Qinghui Mu, Kristin Eden, Irving C. Allen, Michael R. Edwards, Dylan K. McDaniel, Christopher M. Reilly, Xavier Cabana-Puig, Jing Zhu, Jiangdi Mao, Brianna K. Swartwout, Rebecca M. Brock, Leila Abdelhamid, and Xin M. Luo
- Subjects
Limosilactobacillus reuteri ,Male ,Immunoglobulin A ,T-Lymphocytes ,Lactobacillus reuteri ,T cells ,Cross Reactions ,Biology ,digestive system ,Immunomodulation ,Mice ,fluids and secretions ,Immunology and Inflammation ,Immune system ,Animals ,Intestinal Mucosa ,Multidisciplinary ,maternal microbiota ,type 3 innate lymphoid cells ,Microbiota ,Innate lymphoid cell ,Biological Sciences ,biology.organism_classification ,neonatal IgA ,Immunity, Innate ,stomatognathic diseases ,Animals, Newborn ,Antibody Formation ,Host-Pathogen Interactions ,Immunology ,biology.protein ,Small Intestinal Lamina Propria ,Female ,Immunity, Maternally-Acquired - Abstract
Significance Infants are born without an established gut microbiota, which develops rapidly after birth and is shaped by the maternal microbiota. However, how the maternal microbiota, through shaping the neonatal microbiota, would affect the establishment of a strong immune system in neonates remains unclear. Here, we show mechanistically how the maternal microbiota regulates the de novo production of neonatal IgA., 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.
- Published
- 2021
26. Glyceraldehyde-3-Phosphate Dehydrogenase Increases the Adhesion of Lactobacillus reuteri to Host Mucin to Enhance Probiotic Effects
- Author
-
Dongyan Fu, Junli Zhu, Jianxin Liu, Zhaoxi Deng, Xin M. Luo, Wenming Zhang, Tian Dai, and Haifeng Wang
- Subjects
0301 basic medicine ,Membrane permeability ,030106 microbiology ,Ileum ,Catalysis ,Bacterial cell structure ,law.invention ,Microbiology ,lcsh:Chemistry ,Inorganic Chemistry ,03 medical and health sciences ,Probiotic ,mucin ,In vivo ,law ,medicine ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,intestine ,Spectroscopy ,biology ,Chemistry ,GAPDH ,Organic Chemistry ,Mucin ,food and beverages ,General Medicine ,biology.organism_classification ,Computer Science Applications ,Lactobacillus reuteri ,adhesion ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,lcsh:QD1-999 ,Bacteria - Abstract
The ability to adhere to the intestinal mucus layer is an important property of probiotic bacteria. Lactobacillus reuteri strains ZJ615 and ZJ617 show low and high adhesion, respectively, to intestinal epithelial cells. In this study, we quantified bacterial cell wall-associated glyceraldehyde-3-phosphate dehydrogenases (cw-GAPDH) and bacterial cell membrane permeability in both strains using immunoblotting and flow cytometry, respectively. Highly adhesive L. reuteri ZJ617 possessed significantly more cw-GAPDH, higher cell membrane permeability, and significantly higher adhesive ability toward mucin compared with low-adhesive L. reuteri ZJ615. In vitro adhesion studies and analysis of interaction kinetics using the Octet, the system revealed significantly decreased interaction between L. reuteri and mucin when mucin was oxidized when bacterial surface proteins were removed when bacteria were heat-inactivated at 80 °, C for 30 min, and when the interaction was blocked with an anti-GAPDH antibody. SWISS-MODEL analysis suggested intensive interactions between mucin glycans (GalNAc&alpha, 1-O-Ser, GalNAc&alpha, Ser, and Gal&beta, 3GalNAc) and GAPDH. Furthermore, in vivo studies revealed significantly higher numbers of bacteria adhering to the jejunum, ileum, and colon of piglets orally inoculated with L. reuteri ZJ617 compared with those inoculated with L. reuteri ZJ615, this led to a significantly decreased rate of diarrhea in piglets inoculated with L. reuteri ZJ617. In conclusion, there are strong correlations among the abundance of cw-GAPDH in L. reuteri, the ability of the bacterium to adhere to the host, and the health benefits of this probiotic.
- Published
- 2020
- Full Text
- View/download PDF
27. Importance of microRNAs and gut microbiota in the characterization of a phenotypic drift in lupus-prone MRL/lpr mice
- Author
-
Xavier Cabana Puig, Jacob M. Bond, Zhuang Wang, Rujuan Dai, Ran Lu, Amy Lin, Vanessa oakes, Amy Rizzo, Brianna Swarwout, Leila Abdelhamid, Jiangdi Mao, Meeta Prakash, Constanza Sangmeister, Nathaniel Cheung, Catharine R Cowan, Christopher M Reilly, Sha Sun, S Ansar Ahmed, and Xin M Luo
- Subjects
Immunology ,Immunology and Allergy - Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease. The cause of SLE is not only genetic; in fact, environmental factors may play a more important role in disease development. The MRL/lpr mouse model lupus-like symptoms due to multiple SLE susceptible loci in the MRL background, and offers an accelerated model compared to the MRL parent strain due to the Faslpr mutation. Recently, our laboratory witnessed a loss of disease phenotype in our in-house colony of MRL/lpr mice. We thus compared mice newly obtained from The Jackson Laboratory (JAX; Stock Number 000485) to our in-house mice. The single-nucleotide polymorphism (SNP) analysis showed no genetic drift, suggesting that environmental factors could be triggering a phenotypic drift. Surprisingly, the newly purchased JAX mice also had attenuation of glomerulonephritis. Even though JAX mice manifested similar attenuation of lupus nephritis, our in-house colony showed differences in organ weights. Furthermore, males showed a significantly higher level of anti-double stranded DNA auto-IgG consistent with germinal center maturation. In addition, in-house males had significantly higher levels of microRNA-21 and microRNA-183 explaining spleen size difference. Moreover, the composition of gut microbiota was different between in-house and new JAX mice at early age, with many groups of bacteria differing at later time points, which might explain some of the phenotypic differences. These results suggest that microRNAs and gut microbiota might be responsible for the phenotypic differences of MRL/lpr mice in JAX and our colonies as they were genetically identical. On the other hand, the attenuation of nephritis in both groups requires further investigation. Supported by R01AR073240
- Published
- 2022
28. Glyceraldehyde-3-Phosphate Dehydrogenase Increases the Adhesion of
- Author
-
Zhaoxi, Deng, Tian, Dai, Wenming, Zhang, Junli, Zhu, Xin M, Luo, Dongyan, Fu, Jianxin, Liu, and Haifeng, Wang
- Subjects
Limosilactobacillus reuteri ,Swine ,GAPDH ,Probiotics ,Lactobacillus reuteri ,Mucins ,food and beverages ,Glyceraldehyde-3-Phosphate Dehydrogenases ,Epithelial Cells ,Bacterial Adhesion ,Article ,adhesion ,mucin ,Animals ,Intestinal Mucosa ,intestine - Abstract
The ability to adhere to the intestinal mucus layer is an important property of probiotic bacteria. Lactobacillus reuteri strains ZJ615 and ZJ617 show low and high adhesion, respectively, to intestinal epithelial cells. In this study, we quantified bacterial cell wall-associated glyceraldehyde-3-phosphate dehydrogenases (cw-GAPDH) and bacterial cell membrane permeability in both strains using immunoblotting and flow cytometry, respectively. Highly adhesive L. reuteri ZJ617 possessed significantly more cw-GAPDH, higher cell membrane permeability, and significantly higher adhesive ability toward mucin compared with low-adhesive L. reuteri ZJ615. In vitro adhesion studies and analysis of interaction kinetics using the Octet, the system revealed significantly decreased interaction between L. reuteri and mucin when mucin was oxidized when bacterial surface proteins were removed when bacteria were heat-inactivated at 80 °C for 30 min, and when the interaction was blocked with an anti-GAPDH antibody. SWISS-MODEL analysis suggested intensive interactions between mucin glycans (GalNAcα1-O-Ser, GalNAcαSer, and Galβ3GalNAc) and GAPDH. Furthermore, in vivo studies revealed significantly higher numbers of bacteria adhering to the jejunum, ileum, and colon of piglets orally inoculated with L. reuteri ZJ617 compared with those inoculated with L. reuteri ZJ615; this led to a significantly decreased rate of diarrhea in piglets inoculated with L. reuteri ZJ617. In conclusion, there are strong correlations among the abundance of cw-GAPDH in L. reuteri, the ability of the bacterium to adhere to the host, and the health benefits of this probiotic.
- Published
- 2020
29. Frontiers in Immunology
- Author
-
Jing Zhu, Amrie C. Grammer, Thomas E. Cecere, Brianna K. Swartwout, Xavier Cabana Puig, Meeta Prakash, Joe Grieco, Christopher M. Reilly, Michael R. Edwards, Jiangdi Mao, Xin M. Luo, Peter E. Lipsky, Christopher E Puglisi, Qinghui Mu, Prathyusha Bachali, Virginia Tech Carilion School of Medicine, and Biomedical Sciences and Pathobiology
- Subjects
lcsh:Immunologic diseases. Allergy ,DNA, Bacterial ,0301 basic medicine ,Mice, Inbred MRL lpr ,immunoregulation ,Regulatory B cells ,Immunology ,Inflammation ,Spleen ,Gut flora ,medicine.disease_cause ,Severity of Illness Index ,Autoimmunity ,Immunomodulation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,systemic lupus erythematosus ,Vancomycin ,Animals ,Lupus Erythematosus, Systemic ,Immunology and Allergy ,Medicine ,Original Research ,Autoimmune disease ,B-Lymphocytes, Regulatory ,Systemic lupus erythematosus ,gut microbiota ,biology ,business.industry ,autoimmunity ,medicine.disease ,biology.organism_classification ,Adoptive Transfer ,Gastrointestinal Microbiome ,bacterial DNA ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Female ,Disease Susceptibility ,medicine.symptom ,lcsh:RC581-607 ,business ,Biomarkers ,030215 immunology ,medicine.drug - 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. NIAMSUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USANIH National Institute of Arthritis & Musculoskeletal & Skin Diseases (NIAMS) [AR073240, AR067418] NIAMS AR073240, AR067418 (XL).
- Published
- 2020
30. Olive Extract Ameliorates Oxidative Stress and Inflammation, and Protects Intestinal Villus and Microbiota in Piglets Induced by Lipopolysaccharides
- Author
-
Jianxin Liu, Xin M. Luo, Yu Zhang, Zhaoxi Deng, Mao-Long He, and Haifeng Wang
- Subjects
Olive extract ,medicine.anatomical_structure ,Chemistry ,Intestinal villus ,medicine ,Inflammation ,medicine.symptom ,Pharmacology ,medicine.disease_cause ,Oxidative stress - Abstract
Background: The olive extract contains compounds with antioxidant and anti-inflammatory properties. This study was designed to investigate whether olive cake extract, enriched with maslinic acid and hydroxytyrosol, alleviates the lipopolysaccharides (LPS)-induced oxidative stress, inflammation and intestinal villus damage in piglets.Methods: Thirty weaned piglets (6.9±0.9 kg) were assigned to five groups using a randomized complete block design. Piglets were fed a basal diet before intraperitoneal (i.p.) injection of physiological saline (C); fed a basal diet alone (CL) or fed a basal diet plus olive extract (OL), antibiotics (AL), or olive extract and antibiotics (OAL) before i.p. injection of LPS. The feeding lasted for 2 weeks. Piglets were euthanized 4h after LPS injection. Systemic anti-oxidant and inflammation levels were measured and villus morphology in the intestine was examined.Results: Compared with those in the C group, piglets in the CL group had significantly lower GSH-Px, SOD, ALB levels and higher MDA, NO, LDH, ALT and AST levels in the serum (PPPPPPLactobacillus and Clostridium at genus level.Conclusion: Dietary supplementation with olive extract maslinic acid and hydroxytyrosol improved anti-oxidative and anti-inflammatory capacity, intestinal structure morphology, and increased the abundance of beneficial intestinal bacteria in weaned piglets challenged by LPS.
- Published
- 2020
31. Quorum Sensing, Biofilm, and Intestinal Mucosal Barrier: Involvement the Role of Probiotic
- Author
-
Haifeng Wang, Jianxin Liu, Xin M. Luo, and Zhaoxi Deng
- Subjects
0301 basic medicine ,Microbiology (medical) ,Microorganism ,030106 microbiology ,Immunology ,lcsh:QR1-502 ,Review ,Biology ,Microbiology ,lcsh:Microbiology ,biofilm ,law.invention ,03 medical and health sciences ,Probiotic ,Immune system ,Cellular and Infection Microbiology ,law ,Intestinal Mucosa ,intestine ,mucosal barrier ,Barrier function ,Bacteria ,Probiotics ,Biofilm ,Quorum Sensing ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Quorum sensing ,030104 developmental biology ,Infectious Diseases ,Biofilms ,probiotic ,Symbiotic bacteria - Abstract
The intestine is a particularly dynamic environment in which the host constantly interacts with trillions of symbiotic bacteria called the microbiota. Using quorum sensing (QS) communication, bacteria can coordinate their social behavior and influence host cell activities in a non-invasive manner. Nowadays, a large amount of research has greatly spurred the understanding of how bacterial QS communication regulates bacterial cooperative behaviors due to coexistence and host-microbe interactions. In this review, we discuss bacterial QS in the gut and its role in biofilm formation. As a biological barrier, the mucosal immune system can effectively prevent pathogenic microorganisms and other immunogenic components from entering the internal environment of the host. We focus on the relationship between biofilm and intestinal mucosal immunity, and how probiotic bacteria may regulate them. This review is to provide a theoretical basis for the development of new techniques including probiotics targeting the intestinal barrier function, thereby improving gut health.
- Published
- 2020
32. The Specific Immune Response
- Author
-
S. Ansar Ahmed, Gerhardt G. Schurig, and Xin M. Luo
- Subjects
business.industry ,Immunology ,Medicine ,business ,Acquired immune system - Published
- 2020
33. Contributors
- Author
-
S. Ansar Ahmed, Steven P. Brinsko, Autumn P. Davidson, Susan L. Ewart, Deborah S. Greco, Steven R. Heidemann, Thomas H. Herdt, Bradley G. Klein, Xin M. Luo, Brian K. Petroff, Juan E. Romano, John H. Rossmeisl, Gerhardt G. Schurig, George H. Stabenfeldt, Robert B. Stephenson, Jill W. Verlander, and Sharon G. Witonsky
- Published
- 2020
34. Retinoic Acid Exerts Disease Stage-Dependent Effects on Pristane-Induced Lupus
- Author
-
Stephen R. Werre, Sha Sun, Xin M. Luo, Tanya LeRoith, Yaqi Li, Brianna K. Swartwout, Thomas E. Cecere, Christopher M. Reilly, Haifeng Wang, Song Li, Xavier Cabana-Puig, A. Catharine Ross, Jiyoung Lee, and Leila Abdelhamid
- Subjects
0301 basic medicine ,Chemokine ,Retinoic acid ,medicine.disease_cause ,pristane-induced ,Autoimmunity ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Glomerulonephritis ,retinoic acid ,Immunology and Allergy ,Lupus Erythematosus, Systemic ,RNA-Seq ,Vitamin A ,reproductive and urinary physiology ,Original Research ,Kidney ,Mice, Inbred BALB C ,Systemic lupus erythematosus ,biology ,Chemistry ,Drug Synergism ,Lupus Nephritis ,Specific Pathogen-Free Organisms ,medicine.anatomical_structure ,embryonic structures ,Disease Progression ,Female ,Immunosuppressive Agents ,lcsh:Immunologic diseases. Allergy ,kidney ,Immunology ,Tretinoin ,Real-Time Polymerase Chain Reaction ,Drug Administration Schedule ,Proinflammatory cytokine ,03 medical and health sciences ,Downregulation and upregulation ,medicine ,Animals ,stage-dependent ,gut microbiota ,Terpenes ,Contraindications, Drug ,Dendritic Cells ,lupus ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Gastrointestinal Microbiome ,Disease Models, Animal ,030104 developmental biology ,Gene Expression Regulation ,Bacterial Translocation ,Cancer research ,biology.protein ,Dysbiosis ,RNA ,lcsh:RC581-607 ,Spleen ,030215 immunology - Abstract
We previously showed that all-trans-retinoic acid (tRA), an active metabolite of vitamin A, exacerbated pre-existing autoimmunity in lupus; however, its effects before the development of autoimmunity are unknown. Here, using a pristane-induced model, we show that tRA exerts differential effects when given at the initiation vs. continuation phase of lupus. Unlike tRA treatment during active disease, pre-pristane treatment with tRA aggravated glomerulonephritis through increasing renal expression of pro-fibrotic protein laminin β1, activating bone marrow conventional dendritic cells (cDCs), and upregulating the interaction of ICAM-1 and LFA-1 in the spleen, indicating an active process of leukocyte activation and trafficking. Transcriptomic analysis revealed that prior to lupus induction, tRA significantly upregulated the expression of genes associated with cDC activation and migration. Post-pristane tRA treatment, on the other hand, did not significantly alter the severity of glomerulonephritis; rather, it exerted immunosuppressive functions of decreasing circulatory and renal deposition of autoantibodies as well as suppressing the renal expression of proinflammatory cytokines and chemokines. Together, these findings suggest that tRA differentially modulate lupus-associated kidney inflammation depending on the time of administration. Interestingly, both pre- and post-pristane treatments with tRA reversed pristane-induced leaky gut and modulated the gut microbiota in a similar fashion, suggesting a gut microbiota-independent mechanism by which tRA affects the initiation vs. continuation phase of lupus.
- Published
- 2019
35. Lactobacillus rhamnosus GG Attenuates Lipopolysaccharide-Induced Inflammation and Barrier Dysfunction by Regulating MAPK/NF-κB Signaling and Modulating Metabolome in the Piglet Intestine
- Author
-
Tao Zhu, Yanfei Ma, Xin M. Luo, Siri Qi, Jianxin Liu, Yanjun Cui, Jiangdi Mao, Haifeng Wang, and Xiaoxiao Dou
- Subjects
Lipopolysaccharides ,Male ,medicine.medical_specialty ,Lipopolysaccharide ,Gastrointestinal Diseases ,MAP Kinase Signaling System ,medicine.medical_treatment ,Intraperitoneal injection ,Sus scrofa ,Medicine (miscellaneous) ,Stimulation ,Inflammation ,Ileum ,Occludin ,p38 Mitogen-Activated Protein Kinases ,chemistry.chemical_compound ,Internal medicine ,medicine ,Animals ,Nutrition and Dietetics ,Tight Junction Proteins ,Chemistry ,Kinase ,Lacticaseibacillus rhamnosus ,Probiotics ,Transcription Factor RelA ,Gastroenteritis ,Up-Regulation ,medicine.anatomical_structure ,Endocrinology ,Metabolome ,Tumor necrosis factor alpha ,Female ,medicine.symptom ,Signal Transduction - Abstract
BACKGROUND Probiotic Lactobacillius rhamnosus GG (LGG) shows beneficial immunomodulation on cultured cell lines in vitro and in mouse models. OBJECTIVE The aim was to investigate the effects of LGG on intestinal injury and the underlying mechanisms by elucidating inflammatory signaling pathways and metabolomic response to LPS stimulation in the piglet intestine. METHODS Piglets (Duroc × Landrace × Large White, including males and female; 8.6 ± 1.1 kg) aged 28 d were assigned to 3 groups (n = 6/group): oral inoculation with PBS for 2 wk before intraperitoneal injection of physiological saline [control (CON)] or LPS (25 μg/kg body weight; LPS) or oral inoculation with LGG for 2 wk before intraperitoneal injection of LPS (LGG+LPS). Piglets were killed 4 h after LPS injection. Systemic inflammation, intestinal integrity, inflammation signals, and metabolomic characteristics in the intestine were determined. RESULTS Compared with CON, LPS stimulation significantly decreased ileal zonula occludens 1 (ZO-1; 44%), claudin-3 (44%), and occludin (41%) expression; increased serum diamineoxidase (73%), D-xylose (19%), TNF-α (43%), and IL-6 (55%) concentrations; induced p38 mitogen-activated protein kinase (p38 MAPK; 85%), extracellular signal-regulated kinase (ERK; 96%), and NF-κB p65 phosphorylation (37%) (P
- Published
- 2019
36. Lactobacillus rhamnosus GG components, SLP, gDNA and CpG, exert protective effects on mouse macrophages upon lipopolysaccharide challenge
- Author
-
J.X. Liu, Xin M. Luo, Haixia Wang, Yanjun Cui, and S.R. Qi
- Subjects
0106 biological sciences ,MAPK/ERK pathway ,DNA, Bacterial ,Lipopolysaccharides ,Lipopolysaccharide ,MAP Kinase Signaling System ,medicine.medical_treatment ,Oligonucleotides ,01 natural sciences ,Applied Microbiology and Biotechnology ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Lactobacillus rhamnosus ,010608 biotechnology ,medicine ,Animals ,RNA, Messenger ,Protein kinase A ,Receptor ,Extracellular Signal-Regulated MAP Kinases ,0303 health sciences ,Base Composition ,biology ,030306 microbiology ,Kinase ,Chemistry ,Interleukin-6 ,Lacticaseibacillus rhamnosus ,Tumor Necrosis Factor-alpha ,Macrophages ,Probiotics ,Toll-Like Receptors ,NF-kappa B ,Epithelial Cells ,biology.organism_classification ,Molecular biology ,Real-time polymerase chain reaction ,Cytokine ,RAW 264.7 Cells ,Cytokines - Abstract
The aim of this study was to determine whether Lactobacillus rhamnosus GG (LGG) components (surface layer protein, SLP; genomic DNA, gDNA; unmethylated cytosine-phosphate-guanine-containing oligodeoxynucleotide, CpG-ODN), alone or in combination, could affect immunomodulation, and evaluate the signalling mechanism in mouse macrophage RAW264.7 cells challenged with lipopolysaccharide (LPS). LGG components were used to treat cells before LPS stimulation. Cytokine and Toll-like receptor (TLR) expression were assessed using real-time quantitative PCR (RT-qPCR). Mitogen-activated protein kinase (MAPK), extracellular regulated protein kinase (ERK) and nuclear factor-kappa B (NF-κB) signalling pathways were evaluated using immunoblots and immunofluorescence. SLP or SLP + gDNA pre-treatment significantly reduced the LPS-induced mRNA expression of tumour necrosis factor alpha (TNF-α). Pre-treatment with LGG single components (SLP, gDNA or CpG) or their combinations (SLP + gDNA or SLP + CpG) significantly decreased the LPS-induced interleukin-6 (IL-6) mRNA level (P 0·05). Pre-treatment with SLP or gDNA, alone or in combination, significantly suppressed LPS-induced TLR2 and TLR4 mRNA levels (P 0·05). SLP pre-treatment also significantly decreased the LPS-induced expression of TLR9 (P 0·05). Pre-treatment with LGG single components or combinations significantly suppressed the LPS-induced phosphorylation levels of ERK (P 0·05). In conclusion, pre-incubation with LGG components, singly or in combination, generally inhibited the activation of TLR, MAPK and NF-κB signalling pathways in LPS-stimulated cells, leading to attenuated inflammatory cytokine TNF-α and IL-6 production. These results indicate that nonviable probiotic LGG components exert an anti-inflammation effect on epithelial cells. SIGNIFICANCE AND IMPACT OF THE STUDY: Lactobacillus rhamnosus GG (LGG) is widely used as probiotics. However, its main components are not well known for affecting immunomodulation. This study investigated the effects of pre-treatments with different components such as surface layer protein, genomic DNA and unmethylated cytosine-phosphate-guanine-containing oligodeoxynucleotides, alone or in combination on immunomodulation, and evaluated the signalling mechanism in mouse macrophage RAW264.7 cells challenged with lipopolysaccharide. Pre-incubation with components alone or in combination generally inhibited the activation of Toll-like receptor, mitogen-activated protein kinases, extracellular regulated protein kinases and nuclear factor-kappa B signalling pathways in lipopolysaccharide-stimulated cells, which generally leads to attenuated inflammatory cytokine interleukin-6 and tumour necrosis factor alpha production. These results indicate that nonviable probiotic LGG components exert an anti-inflammation effect on epithelial cells.
- Published
- 2019
37. Pregnancy and lactation interfere with the response of autoimmunity to modulation of gut microbiota
- Author
-
Jiangdi Mao, Haifeng Wang, Xavier Cabana-Puig, Christopher M. Reilly, Thomas E. Cecere, Qinghui Mu, Brianna K. Swartwout, Xin M. Luo, and Leila Abdelhamid
- Subjects
Mice, Inbred MRL lpr ,Autoimmunity ,Gut flora ,medicine.disease_cause ,IDO ,Pathogenesis ,Mice ,Pregnancy ,immune system diseases ,Lactation ,Lupus Erythematosus, Systemic ,skin and connective tissue diseases ,0303 health sciences ,Systemic lupus erythematosus ,biology ,Anti-Bacterial Agents ,Interleukin-10 ,3. Good health ,Treg ,medicine.anatomical_structure ,Leaky gut ,lcsh:QR100-130 ,Female ,Microbiology (medical) ,Lupus ,Gut microbiota ,digestive system ,Microbiology ,lcsh:Microbial ecology ,Proinflammatory cytokine ,Interferon-gamma ,03 medical and health sciences ,Vancomycin ,medicine ,Animals ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,030304 developmental biology ,030306 microbiology ,Research ,Lactobacillus animalis ,biology.organism_classification ,medicine.disease ,Gastrointestinal Microbiome ,Lactobacillus ,Immunology ,Pregnancy, Animal ,Dysbiosis ,IFNγ - 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. Electronic supplementary material The online version of this article (10.1186/s40168-019-0720-8) contains supplementary material, which is available to authorized users.
- Published
- 2019
38. Implications of Probiotics on the Maternal-Neonatal Interface: Gut Microbiota, Immunomodulation, and Autoimmunity
- Author
-
Brianna K. Swartwout and Xin M. Luo
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,Immunology ,Disease ,Review ,Gut flora ,medicine.disease_cause ,Autoimmunity ,Immunomodulation ,neonatal ,03 medical and health sciences ,Immune system ,Medicine ,Animals ,Humans ,Immunology and Allergy ,Autoimmune disease ,Pregnancy ,biology ,gut microbiota ,business.industry ,Probiotics ,autoimmunity ,Immune dysregulation ,medicine.disease ,biology.organism_classification ,Mother-Child Relations ,3. Good health ,Gastrointestinal Microbiome ,maternal ,030104 developmental biology ,probiotics ,Immune System ,business ,lcsh:RC581-607 ,Dysbiosis - Abstract
Probiotics are being investigated for the treatment of autoimmune disease by re-balancing dysbiosis induced changes in the immune system. Pregnancy is a health concern surrounding autoimmune disease, both for the mother and her child. Probiotics for maternity are emerging on the market and have gained significant momentum in the literature. Thus far, evidence supports that probiotics alter the structure of the normal microbiota and the microbiota changes significantly during pregnancy. The interaction between probiotics-induced changes and normal changes during pregnancy is poorly understood. Furthermore, there is emerging evidence that the maternal gut microbiota influences the microbiota of offspring, leading to questions on how maternal probiotics may influence the health of neonates. Underpinning the development and balance of the immune system, the microbiota, especially that of the gut, is significantly important, and dysbiosis is an agent of immune dysregulation and autoimmunity. However, few studies exist on the implications of maternal probiotics for the outcome of pregnancy in autoimmune disease. Is it helpful or harmful for mother with autoimmune disease to take probiotics, and would this be protective or pathogenic for her child? Controversy surrounds whether probiotics administered maternally or during infancy are healthful for allergic disease, and their use for autoimmunity is relatively unexplored. This review aims to discuss the use of maternal probiotics in health and autoimmune disease and to investigate their immunomodulatory properties. Preparation of this publication was supported by the National Institute of Arthritis and Musculoskeletal and Skin Diseases of the National Institutes of Health under Award Number 1R01AR073240 and 1R15AR067418. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
- Published
- 2018
- Full Text
- View/download PDF
39. Roles of CX3CR1 and probiotic lactobacilli on the development of autoimmunity in MRL/lpr mice
- Author
-
Xavier Cabana Puig, Leila Abdelhamid, Brianna Swartwout, Jing Zhu, Xiaofeng Liao, Christopher M Reilly, and Xin M Luo
- Subjects
Immunology ,Immunology and Allergy - Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease with no known cure. Gut microbiota and the immune system have a close relationship that controls inflammation. Alterations in the gut microbiota, known as dysbiosis, can trigger SLE in both human and mouse. We previously published differences between the gut microbiota in lupus-prone MRL/Mp-Faslpr (lpr) mice compared to healthy controls, with lower Lactobacillaceae abundance in lpr mice. Restoring the gut microbiota with a mix of 5 different strains of this family significantly attenuated lupus-liked disease in lpr mice. In this study, we sought to understand the potential benefit of this mix in lpr mice with Cx3cr1 deficiency (KO). KO mice on C57BL/6 background display increased translocation of commensal bacteria into the mesenteric lymph node (MLN) and increased susceptibility to intestinal inflammation. However, whether CX3CR1+ macrophages play a role in controlling systemic inflammation was unclear. After backcrossing the KO allele to lpr mice, we found that KO lpr mice exhibited exacerbated splenomegaly and lymphadenopathy, as well as increased proteinuria. Interestingly, treating the KO lpr mice with the 5 Lactobacillus strains attenuated the enlargement of MLN and abrogated the increase of proteinuria. Furthermore, the effects of the probiotic bacteria were accompanied by downregulation of serum endotoxin, suggesting the reversal of a leaky gut that may explain their benefits on KO lpr mice. Together, our results show an important role of CX3CR1+ macrophages in controlling systemic inflammation in lupus, and that probiotic Lactobacillus spp. may be an effective treatment against SLE through preventing bacterial translocation.
- Published
- 2021
40. Vitamin A Deficiency Deteriorates Lupus Nephritis
- Author
-
Leila Abdelhamid, Xavier Cabana-Puig, Brianna Swartwout, Jing Zhu, Yaqi Li, A. Catharine Ross, Thomas E. Cecere, Phillip Ruiz, Christopher M. Reilly, and Xin M Luo
- Subjects
Immunology ,Immunology and Allergy - Abstract
Vitamin A deficiency (VAD) is observed in both human and mice with lupus nephritis (LN). However, whether VAD is a driving factor for LN progression is unclear. Here, we investigated the effect of VAD on the progression of LN in a lupus-prone mouse model, MRL/lpr. In these mice, while LN is not apparent before 8 weeks of age, the inflammation of lymphoid tissues suggesting initiation of lupus is evident at weaning. Thus, we initiated VAD either before or after weaning to reveal a potential time-dependent effect. At around 15 weeks of age with both types of VAD, we found exacerbated LN that was characterized by deteriorated kidney inflammation, impaired renal epithelial integrity, and dramatic squamous metaplasia of the renal pelvic urothelium. Both types of VAD provoked severe neutrophilic tubulointerstitial nephritis and accelerated renal failure. This was concomitant with significantly higher mortality in all VAD mice. Looking at an earlier time point of 7 weeks of age and before the onset of clinical LN, both types of VAD increased splenomegaly, lymphadenopathy, and circulating autoantibodies; three additional hallmarks of lupus, as well as renal infiltration of conventional and plasmacytoid dendritic cells. These results suggest VAD-driven deterioration of LN regardless of the time of VAD initiation. Our findings raise significant public health concerns that concurrent lupus and VAD may lead to more severe SLE. Future studies will elucidate the underlying mechanisms of how VAD modulates myeloid cells to accelerate the initiation of LN.
- Published
- 2021
41. Frontiers in Immunology
- Author
-
Leila Abdelhamid, Xavier Cabana-Puig, Qinghui Mu, Maryam Moarefian, Brianna Swartwout, Kristin Eden, Prerna Das, Ryan P. Seguin, Libin Xu, Sarah Lowen, Mital Lavani, Terry C. Hrubec, Caroline N. Jones, Xin M. Luo, Biomedical Sciences and Pathobiology, Mechanical Engineering, and Biological Sciences
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,Mice, Inbred MRL lpr ,Neutrophils ,T-Lymphocytes ,T cell ,Immunology ,quaternary ammonium compound (QAC) ,T cells ,Apoptosis ,Spleen ,medicine.disease_cause ,Autoimmunity ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunity ,medicine ,Animals ,Lupus Erythematosus, Systemic ,Immunology and Allergy ,Cells, Cultured ,Original Research ,lupus (SLE) ,splenomegaly ,Systemic lupus erythematosus ,Chemistry ,autoimmunity ,neutrophil ,Immune dysregulation ,medicine.disease ,Quaternary Ammonium Compounds ,Disease Models, Animal ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,Neutrophil Infiltration ,Female ,lcsh:RC581-607 ,Immunosuppressive Agents ,Ex vivo ,Disinfectants ,030215 immunology - 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. National Institute of Arthritis and Musculoskeletal and Skin DiseasesUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USANIH National Institute of Arthritis & Musculoskeletal & Skin Diseases (NIAMS); National Institute of Environmental Health SciencesUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USANIH National Institute of Environmental Health Sciences (NIEHS) [T32-ES007032]; National Institute of General Medical Sciences of the National Institutes of HealthUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USANIH National Institute of General Medical Sciences (NIGMS) [R35GM133610] Funding was received from National Institute of Arthritis and Musculoskeletal and Skin Diseases. We also would like to acknowledge the following funding sources. Environmental Pathology and Toxicology Training Grant funded by the National Institute of Environmental Health Sciences (T32-ES007032) for supporting RP. Seguin. The National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM133610 for supporting M Moarefian.
- Published
- 2021
42. Activation of B10 cells via gut microbiota in the pathogenesis of systemic lupus erythematosus
- Author
-
Meeta B Prakash, Xin M Luo, Qinghui Mu, and Michael R Edwards
- Subjects
Immunology ,Immunology and Allergy - Abstract
Systemic Lupus Erythematosus(SLE), a disease of various genetic and environmental origins, is often characterized by immune complexes causing inflammation and damage in various organ systems, such as the brain, kidneys, lung, joint, and skin. Current treatment for SLE include steroidal and nonsteroidal anti-inflammatory drugs, antimalarial agents, and immunosuppressive medications. These treatments primarily reduce symptoms, and patients may develop side effects with long-term care. The gut microbiome has an important role in various metabolic and immunological diseases. A previous study found that increased Lactobacillus colonization restored the mucosal barrier, and increased abundance in these bacteria attenuated SLE symptoms through reversing a ‘leaky gut’. This allows for potential therapies of SLE with probiotics. In preliminary trials, vancomycin treatment during active disease killed gram-positive bacteria, but spared Lactobacillus, and attenuated symptoms. Surprisingly, significantly decreased amounts of CD10+CD19+ B cells that produce IL-10 (B10) were found when vancomycin was given before the onset of lupus. Vancomycin treatment before disease onset significantly removed bacterial DNA from the gut and circulation, causing exacerbation of clinical symptoms. Oral gavage of activated B10 cells in vancomycin treated mice rescued the negative effects of vancomycin treated mice at the pre-disease stage. This work suggests that a form of oral tolerance induced by bacterial DNA-mediated expansion of B10 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 B10 cells.
- Published
- 2021
43. Deficiency of maternal antibodies modulates the development of lupus-like disease in BXSB offspring
- Author
-
Jing Zhu, Nina Bowman, Samantha Naughton, Xin M Luo, and Caroline McPhee Leeth
- Subjects
Immunology ,Immunology and Allergy - Abstract
Maternal factors play an essential role in shaping the immune system of the newborn, yet it is unknown whether maternal factors could modulate the development of systemic lupus erythematosus (SLE) in the offspring. Activation-induced cytidine deaminase (AID) is an enzyme required for somatic hypermutation and class switch recombination. Given that IgG and IgA isotypes account for the vast majority of passive immunity in rodents, our previously established AID-deficient BXSB mice provide a model in which no IgG or IgA antibodies are transferred to the offspring. In this study, we compared genotypically identical mice born to either AID-sufficient dams or AID-deficient dams and evaluated the effects of maternal antibodies in disease progression. Offspring from knockout dams developed disease at a faster rate, as shown by more severe nephritis and elevated pathogenic autoantibodies compared to their counterparts born to wild type dams. In addition, the splenic structure was intensively disrupted with disorganized follicles and fewer discernible germinal centers. A more activated T cell phenotype was also characterized in maternal antibody-deficient mice. When immune competent pups were cross-fostered onto AID deficient dams, these mice exhibited more severe disease phenotypes, indicating the protective antibody effect contributes to the modulation of SLE progression in post-natal period. Overall, these findings highlight the importance of maternal antibodies in programming the immune system and altering the SLE development in the offspring.
- Published
- 2021
44. All-Trans-Retinoic Acid Augments the Histopathological Outcome of Neuroinflammation and Neurodegeneration in Lupus-Prone MRL/lpr Mice
- Author
-
Joshua B Sparks, Jingjing Ren, Michelle H. Theus, Xiaofeng Liao, and Xin M. Luo
- Subjects
0301 basic medicine ,Mice, Inbred MRL lpr ,Histology ,Central nervous system ,Retinoic acid ,Tretinoin ,Inflammation ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Lupus Erythematosus, Systemic ,Meningitis ,Vitamin A ,Neuroinflammation ,Autoantibodies ,business.industry ,Neurodegeneration ,Autoantibody ,Brain ,Complement C3 ,Vitamins ,Articles ,medicine.disease ,Astrogliosis ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Immunoglobulin G ,Immunology ,Female ,Anatomy ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Recently, we demonstrated that treatment with all- trans-retinoic acid (tRA) induced a paradoxical effect on immune activation during the development of autoimmune lupus. Here, we further describe its negative effects on mediating neuroinflammation and neurodegeneration. Female MRL/lpr mice were orally administered tRA or VARA (retinol mixed with 10% tRA) from 6 to 14 weeks of age. Both treatments had a significant effect on brain weight, which correlated with histopathological evidence of focal astrogliosis, meningitis, and ventriculitis. Infiltration of CD138- and Iba1-positve immune cells was observed in the third ventricle and meninges of treated mice that co-labeled with ICAM-1, indicating their inflammatory nature. Increased numbers of circulating plasma cells, autoantibodies, and total IgG were also apparent. IgG and C3 complement deposition in these brain regions were also prominent as was focal astrogliosis surrounding the ventricular lining and meninges. Using Fluoro-Jade staining, we further demonstrate that neuroinflammation was accompanied by neurodegeneration in the cortex of treated mice compared with vehicle controls. These findings indicate that vitamin A exposure exacerbates the immunogenic environment of the brain during the onset of systemic autoimmune disease. Vitamin A may therefore compromise the immuno-privileged nature of the central nervous system under a predisposed immunogenic environment.
- Published
- 2016
45. Specific HDAC6 inhibition by ACY-738 reduces SLE pathogenesis in NZB/W mice
- Author
-
Nicole L. Regna, Abdul Gafoor Puthiyaveetil, Sarah E. Hammond, Cristen B. Chafin, Christopher M. Reilly, Xin M. Luo, David L. Caudell, Miranda D. Vieson, and Matthew Jarpe
- Subjects
0301 basic medicine ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,T cells ,Fluorescent Antibody Technique ,Spleen ,Enzyme-Linked Immunosorbent Assay ,Biology ,Histone Deacetylase 6 ,Hydroxamic Acids ,Real-Time Polymerase Chain Reaction ,Histone Deacetylases ,Pathogenesis ,03 medical and health sciences ,Mice ,Systemic lupus erythematosus ,Immune system ,HDAC ,Internal medicine ,medicine ,Animals ,Humans ,Lupus Erythematosus, Systemic ,Immunology and Allergy ,Enzyme Inhibitors ,B cell ,B cells ,Mice, Inbred NZB ,Lymphocyte differentiation ,Glomerulonephritis ,medicine.disease ,Enzyme Activation ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Cytokine ,Pyrimidines ,Female ,Bone marrow - Abstract
We sought to determine if a selective HDAC6 inhibitor (ACY-738) decreases disease in NZB/W mice. From 22 to 38weeks-of-age, mice were injected intraperitoneally with 5 or 20mg/kg of ACY-738, or vehicle control. Body weight and proteinuria were measured every 2weeks, while sera anti-dsDNA, Ig isotypes, and cytokine levels were measured every 4weeks. Kidney disease was determined by evaluation of sera, urine, immune complex deposition, and renal pathology. Flow cytometric analysis assessed thymic, splenic, bone marrow, and peripheral lymphocyte differentiation patterns. Our results showed HDAC6 inhibition decreased SLE disease by inhibiting immune complex-mediated glomerulonephritis, sera anti-dsDNA levels, and inflammatory cytokine production and increasing splenic Treg cells. Inhibition of HDAC6 increased the percentage of cells in the early-stage developmental fractions of both pro- and pre-B cells. These results suggest that specific HDAC6 inhibition may be able to decrease SLE disease by altering aberrant T and B cell differentiation.
- Published
- 2016
- Full Text
- View/download PDF
46. Retinoic acid exerts disease stage-dependent and tissue-specific effects on pristane-induced lupus
- Author
-
Leila Abdelhamid, Xavier Cabana-Puig, Brianna Swartwout, Jiyoung Lee, Song Li, Sha Sun, Yaqi Li, A. Catharine Ross, Thomas E. Cecere, Tanya LeRoith, Haifeng Wang, Christopher M. Reilly, and Xin M. Luo
- Subjects
Immunology ,Immunology and Allergy - Abstract
We previously showed that all-trans-retinoic acid (tRA) exacerbated pre-existing autoimmunity in lupus; however, its effects before disease onset are unknown. Here, using a pristane-induced model, we show that tRA exerts tissue-specific effects when given at the initiation vs. continuation phase of lupus. Pre-pristane treatment with tRA aggravated glomerulonephritis through increasing renal expression of pro-fibrotic protein laminin β1, activating bone marrow CD11b- conventional dendritic cells, increasing the splenic CD4:CD8 ratio, and upregulating the interaction of ICAM-1 and LFA-1 that led to the infiltration of inflammatory cells to the spleen. Transcriptomic analysis revealed that prior to lupus induction, tRA significantly upregulated genes associated with cell differentiation, activation, and migration. Moreover, compared to pristane alone, tRA pre-treatment potentiated, whereas tRA post-treatment significantly suppressed, the renal expression of proinflammatory TNF-α, IL-1β, CCL2, and CCL3. While it may benefit the kidney, post-pristane treatment with tRA worsened the onset and severity of pristane-induced arthritis through promoting neutrophil and mononuclear cell infiltration into the joints. Together, these findings suggest that tRA supplementation regardless of the time of administration promotes chemokine-driven migration and tissue-specific infiltration of inflammatory cells, thereby exacerbating lupus-associated kidney or joint inflammation. Interestingly, both pre- and post-treatments with tRA modulated the gut microbiota in a similar fashion, suggesting a gut microbiota-independent mechanism by which tRA affects the initiation vs. continuation phase of lupus.
- Published
- 2020
47. The Effect of Dietary Fiber Intake on Systemic Lupus Erythematosus (SLE) Disease in NZB/W Lupus Mice
- Author
-
Eric James Panther, Jingjing Ren, Xavier Cabana-Puig, Leila Abdelhamid, Brianna Swartwout, Xin M. Luo, and Christopher M. Reilly
- Subjects
Immunology ,Immunology and Allergy - Abstract
Dysbiosis in the gut microbiota has been observed in a various autoimmune disease, including SLE, which could cause a leaky gut, triggering an immune response, and thus worsening autoimmune disease expression. In our current studies, we hypothesized that increasing dietary fiber would create a healthy microbiota environment in the gut, leading to decreased leakiness of the gut and to decreased disease expression in and NZB/NZW female mice. NZB/NZW mice were placed on standardized high fiber (HF 30%) or low fiber (LF 0.4%) for 12 weeks beginning at 10 weeks of age. Mice were assessed as they aged for various parameters of disease including proteinuria and anti-dsDNA antibody production. Alteration of the microbiota and short chain fatty acid levels were also assessed. At 36 weeks-of-age, the mice were euthanized and we assessed occlusion protein expression, splenocyte profiles, and kidney tissue. We found as the mice aged, their body weights, anti-dsDNA antibody levels, and proteinuria were not significantly different between the groups. Similarly, there were no differences in SCFA levels. In regard to the microbiota, Chlostridiales bacteria were consistently increased in the HF treated mice compared to the LF treated mice. In regard to disease progression, spleen weight, immune cell profiles, proteinuria, dsDNA levels, and kidney pathology, there was no difference between the HF and LF treated groups. Taken together, these results indicate that in the NZB/W female mouse, a HF diet may alter the microbiota but does not influence disease progression.
- Published
- 2020
48. Characterizing the role of Lactobacillus spp. in systemic lupus erythematosus
- Author
-
Xavier Cabana Puig, Qinghui Mu, Brianna Swartwout, Leila Abdelhamid, Meeta B. Prakash, Christopher M. Reilly, and Xin M. Luo
- Subjects
Immunology ,Immunology and Allergy - Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease with no known cure. The role of gut microbiota and its interaction with the immune system can direct tolerance induction to self-antigens not only in the intestinal mucosa but also at the systemic level. Our lab previously showed differences between the gut microbiota in lupus-prone MRL/Mp-Faslpr (lpr) mice compared to healthy controls, with lower Lactobacillaceae levels in lpr mice. Restoring the microbiota with a mix of 5 different Lactobacillus strains, L. reuteri, L. oris, L. johnsonii, L. gasseri, and L. rhamnosus significantly attenuated lupus-liked disease in lpr mice, by increasing regulatory T cells and balancing T helper-17 cells. Interestingly, we found L. reuteri and an uncultured Lactobacillus spp. were present based on 16S rRNA sequencing. To evaluate the effect of the main candidate L. reuteri in attenuating lupus, we treated lpr mice with the single bacterial strain. Surprisingly, L. reuteri did not reproduce the same results as the Lactobacillus mixture. Thus, we tested the remaining strains individually but none was able to recapitulate the effect of the 5 strains. Additionally, we treated lpr mice with the supernatant of the 5 strains containing secreted metabolites, which did not significantly attenuate lupus. These results suggest that communications among different strains of lactobacilli may be required in ameliorating the disease. Probiotic Lactobacillus spp. may be an alternative treatment against SLE, but further investigations are warranted to delineate the mechanisms of how multiple strains of lactobacilli can exert a beneficial effect as a community.
- Published
- 2020
49. Nutrients
- Author
-
Leila Abdelhamid and Xin M. Luo
- Subjects
leaky gut ,retinoic acid ,lcsh:TX341-641 ,autoimmune diseases ,lcsh:Nutrition. Foods and food supply - Abstract
A leaky gut has been observed in a number of autoimmune diseases including type 1 diabetes, multiple sclerosis, inflammatory bowel disease, and systemic lupus erythematosus. Previous studies from our laboratory have shown that lupus mice also bear a leaky gut and that the intestinal barrier function can be enhanced by gut colonization of probiotics such as Lactobacillus spp. Retinoic acid (RA) can increase the relative abundance of Lactobacillus spp. in the gut. Interestingly, RA has also been shown to strengthen the barrier function of epithelial cells in vitro and in the absence of probiotic bacteria. These reports bring up an interesting question of whether RA exerts protective effects on the intestinal barrier directly or through regulating the microbiota colonization. In this review, we will discuss the roles of RA in immunomodulation, recent literature on the involvement of a leaky gut in different autoimmune diseases, and how RA shapes the outcomes of these diseases. Published version
- Published
- 2018
50. Retinoic Acid, Leaky Gut, and Autoimmune Diseases
- Author
-
Leila, Abdelhamid and Xin M, Luo
- Subjects
Bacteria ,leaky gut ,Autoimmunity ,Tretinoin ,Review ,Permeability ,Autoimmune Diseases ,Gastrointestinal Microbiome ,Intestines ,Treatment Outcome ,Host-Pathogen Interactions ,retinoic acid ,Animals ,Humans ,Immunologic Factors - Abstract
A leaky gut has been observed in a number of autoimmune diseases including type 1 diabetes, multiple sclerosis, inflammatory bowel disease, and systemic lupus erythematosus. Previous studies from our laboratory have shown that lupus mice also bear a leaky gut and that the intestinal barrier function can be enhanced by gut colonization of probiotics such as Lactobacillus spp. Retinoic acid (RA) can increase the relative abundance of Lactobacillus spp. in the gut. Interestingly, RA has also been shown to strengthen the barrier function of epithelial cells in vitro and in the absence of probiotic bacteria. These reports bring up an interesting question of whether RA exerts protective effects on the intestinal barrier directly or through regulating the microbiota colonization. In this review, we will discuss the roles of RA in immunomodulation, recent literature on the involvement of a leaky gut in different autoimmune diseases, and how RA shapes the outcomes of these diseases.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.