Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Immunology ; 171(2): 235-249, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37947218

ABSTRACT

The incidence of systemic lupus erythematosus (SLE) is about nine times higher in women than in men, and the underlying mechanisms that contribute to this gender bias are not fully understood. Previously, using lupus-prone (SWR × NZB)F1 (SNF1) mice, we have shown that the intestinal immune system could play a role in the initiation and progression of disease in SLE, and depletion of gut microbiota produces more pronounced disease protection in females than in males. Here, we show that the gut permeability features of lupus-prone female SNF1 mice at juvenile ages directly correlate with the expression levels of pro-inflammatory factors, faecal IgA abundance and nAg reactivity and the eventual systemic autoantibody levels and proteinuria onset. Furthermore, we observed that the disease protection achieved in female SNF1 mice upon depletion of gut microbiota correlates with the diminished gut inflammatory protein levels, intestinal permeability and circulating microbial DNA levels. However, faecal microbiota transplant from juvenile male and females did not result in modulation of gut inflammatory features or permeability. Overall, these observations suggest that the early onset of intestinal inflammation, systemic autoantibody production and clinical stage disease in lupus-prone females is linked to higher gut permeability in them starting at as early as juvenile age. While the higher gut permeability in juvenile lupus-prone females is dependent on the presence of gut microbes, it appears to be independent of the composition of gut microbiota.


Subject(s)
Autoimmunity , Lupus Erythematosus, Systemic , Female , Humans , Male , Mice , Animals , Intestinal Barrier Function , Sexism , Mice, Inbred NZB , Autoantibodies , Disease Models, Animal
2.
Clin Immunol ; 242: 109107, 2022 09.
Article in English | MEDLINE | ID: mdl-36049603

ABSTRACT

Systemic lupus erythematosus (SLE) is characterized by the production of anti-nuclear autoantibodies. Here, for the first time, we show that the abundances of gut permeability marker Zonulin and IgA1- and IgA2- subclasses are significantly higher in the fecal samples of SLE patients compared to HCs. Importantly, IgA-total, and IgA1- and IgA2-subclasses from SLE patients showed higher nAg reactivity titers. Notably, we found that not only the nuclear antigen (nAg) reactive fecal IgA1:IgA2 ratio is higher in SLE patients, but also the abundance and nAg reactivity of fecal IgA and subclasses, IgA1 particularly, correlate with the fecal levels of Zonulin, which is produced primarily in the small intestine. These observations that higher amounts of nAg-reactive IgA and gut permeability marker are produced, particularly, in the proximal gut suggest a compromised epithelial barrier function and pro-inflammatory characteristics of small intestine in SLE patients.


Subject(s)
Immunoglobulin A , Lupus Erythematosus, Systemic , Antigens, Nuclear , Biomarkers , Feces , Humans , Permeability
3.
Front Toxicol ; 4: 937150, 2022.
Article in English | MEDLINE | ID: mdl-35846434

ABSTRACT

Background: Combination therapy of targeted drugs in cancer treatment is a field in constant flux, with research balancing side effects with efficacy. Efficacy from combination therapy is improved either through synthetic lethality or through prevention of recurrent clones. Previous research has shown (hydroxy-)chloroquine is insufficient to disrupt autophagy in tumors. Hence, either combinations or novel autophagy agents are desired. In vivo studies of ovarian cancer have revealed that chloroquine can be combined with up to four other autophagy drugs to suppress ovarian cancer growth. While cancer efficacy is now established for the autophagy drug combination, it is unclear what toxicities may require monitoring in human trials. Additive toxicity with chemotherapy is also unknown. Methods: To address toxicity in more depth than previous weight-monitoring studies, biochemical and histopathology studies were performed. Mouse groups were treated with autophagy drugs for 2 weeks, with or without the chemotherapy Doxil. After the last dose, mice were processed for blood biochemistry, white blood cell markers, and histopathology. Results: Data from a comprehensive blood biochemistry panel, flow cytometric measurements of blood cell markers, and histopathology are herein reported. While Doxil presented clear bone marrow and immunologic toxicity, autophagy drugs were overall less toxic and more variable in their presentation of potential toxicities. Only minor additive effects of autophagy drugs with Doxil were observed. Conclusion: Combinations of autophagy drugs may be considered for therapy in human oncology trials, with possible side effects to monitor informed by these murine pre-clinical data.

4.
Immunology ; 166(3): 341-356, 2022 07.
Article in English | MEDLINE | ID: mdl-35404483

ABSTRACT

Defective immune regulation has been recognized in type 1 diabetes (T1D). Immune regulatory T cell check-point receptors, which are generally upregulated on activated T cells, have been the molecules of attention as therapeutic targets for enhancing immune response in tumour therapy. Here, we show that pancreatic ß-cell antigen (BcAg) presentation by engineered tolerogenic dendritic cells (tDCs) that express CTLA4 selective ligand (B7.1wa) or a combination of CTLA4, PD1 and BTLA selective ligands (B7.1wa, PD-L1 and HVEM-CRD1 respectively; multiligand-DCs) causes an increase in regulatory cytokine and T cell (Treg) responses and suppression of the effector T cell function as compared with engineered control-DCs. Non-obese diabetic mice treated with BcAg-pulsed CTLA4-ligand-DCs and multiligand-DCs at pre-diabetic and early-hyperglycaemic stages showed significantly lower degree of insulitis, higher frequencies of insulin-positive islets, profound delay in and reversal of hyperglycaemia for a significant duration. Immune cells from the tDC-treated mice not only produced lower amounts of IFNγ and higher amounts of IL10 and TGFß1 upon BcAg challenge, but also failed to induce hyperglycaemia upon adoptive transfer. While both CTLA4-ligand-DCs and multiligand-DCs were effective in inducing tolerance, multiligand-DC treatment produced an overall higher suppressive effect on effector T cell function and disease outcome. These studies show that enhanced engagement of T cell checkpoint receptors during BcAg presentation can modulate T cell function and suppress autoimmunity and progression of the disease in T1D.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Hyperglycemia , Animals , Antigen Presentation , CTLA-4 Antigen/metabolism , Dendritic Cells , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/metabolism , Hyperglycemia/metabolism , Hyperglycemia/pathology , Immune Tolerance , Ligands , Mice , Receptors, Immunologic , T-Lymphocytes, Regulatory
5.
Immunology ; 165(4): 497-507, 2022 04.
Article in English | MEDLINE | ID: mdl-35138645

ABSTRACT

Systemic lupus erythematosus (SLE) is characterized by the production of pathogenic autoantibodies with nuclear antigen (nAg) specificity. Using (SWRxNZB)F1 (SNF1) mice, we showed higher levels of Immunoglobulin A (IgA) production in the intestine and the nAg reactivity of faecal IgA under lupus susceptibility. Here, we determined whether the faecal IgA abundance and nAg reactivity are higher in, different among, various lupus-prone preclinical models (MRL/lpr, NZBxNZW-F1, SNF1, NZM2410 and NZM2328). We also determined whether the faecal IgA nAg reactivity at preseropositive ages correlates with the eventual serum autoantibody levels in males and females of these mouse models. We show that age-dependent increase in the abundance and nAg reactivity of faecal IgA can vary among different lupus-prone mouse models. Importantly, faecal IgA in these mice show significant levels of nAg reactivity, starting as early as at juvenile age. Furthermore, the pre-seropositive stage nAg reactivity of faecal IgA in most lupus-prone strains correlates well with that of eventual, seropositive stage systemic autoantibody levels. Gender differences in serum autoantibody levels were preceded by similar differences in the faecal IgA abundance and nAg reactivity. These observations suggest that faecal IgA features, nAg reactivity particularly, could serve as a biomarker for early prediction of the eventual systemic autoimmunity in lupus-prone subjects.


Subject(s)
Immunoglobulin A , Lupus Erythematosus, Systemic , Animals , Antigens, Nuclear , Autoantibodies , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr
6.
Autoimmunity ; 55(2): 95-108, 2022 03.
Article in English | MEDLINE | ID: mdl-34882054

ABSTRACT

Progressive destruction of pancreatic islet ß-cells by immune cells is a primary feature of type 1 diabetes (T1D) and therapies that can restore the functional ß-cell mass are needed to alleviate disease progression. Here, we report the use of mesenchymal stromal/stem cells (MSCs) for the production and delivery of Gastrin, a peptide hormone that is produced by intestinal cells and foetal islets and can increase ß-Cell mass, to promote protection from T1D. A single injection of syngeneic MSCs that were engineered to express Gastrin (Gastrin-MSCs) caused a significant delay in hyperglycaemia in non-obese diabetic (NOD) mice compared to engineered control-MSCs. Similar treatment of early-hyperglycaemic mice caused the restoration of euglycemia for a considerable duration, and these therapeutic effects were associated with the protection of, and/or higher frequencies of, insulin-producing islets and less severe insulitis. While the overall immune cell phenotype was not affected profoundly upon treatment using Gastrin-MSCs or upon in vitro culture, pancreatic lymph node cells from Gastrin-MSC treated mice, upon ex vivo challenge with self-antigen, showed a Th2 and Th17 bias, and diminished the diabetogenic property in NOD-Rag1 deficient mice suggesting a disease protective immune modulation under Gastrin-MSC treatment associated protection from hyperglycaemia. Overall, this study shows the potential of production and delivery of Gastrin in vivo, by MSCs, in protecting insulin-producing ß-cells and ameliorating the disease progression in T1D.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Gastrins , Islets of Langerhans , Mesenchymal Stem Cell Transplantation , Animals , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Gastrins/genetics , Mesenchymal Stem Cells , Mice , Mice, Inbred NOD
7.
Sci Rep ; 11(1): 12689, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34135376

ABSTRACT

Centrosomal P4.1-associated protein (CPAP) plays a critical role in restricting the centriole length in human cells. Here, we report a novel, positive regulatory influence for CPAP on endocytic vesicular transport (EVT) and lysosome targeting of internalized-cell surface receptor EGFR. We observed that higher CPAP levels cause an increase in the abundance of multi-vesicular body (MVB) and EGFR is detectable in CPAP-overexpression induced puncta. The surface and cellular levels of EGFR are higher under CPAP deficiency and lower under CPAP overexpression. While ligand-engagement induced internalization or routing of EGFR into early endosomes is not influenced by cellular levels of CPAP, we found that targeting of ligand-activated, internalized EGFR to lysosome is impacted by CPAP levels. Transport of ligand-bound EGFR from early endosome to late endosome/MVB and lysosome is diminished in CPAP-depleted cells. Moreover, CPAP depleted cells appear to show a diminished ability to form MVB structures upon EGFR activation. These observations suggest a positive regulatory effect of CPAP on EVT of ligand-bound EGFR-like cell surface receptors to MVB and lysosome. Overall, identification of a non-centriolar function of CPAP in endocytic trafficking provides new insights in understanding the non-canonical cellular functions of CPAP.


Subject(s)
Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism , Transport Vesicles/metabolism , Cell Line, Tumor , Endocytosis , Endosomes/metabolism , ErbB Receptors/metabolism , HEK293 Cells , HeLa Cells , Humans , Ligands , Microtubule-Associated Proteins/genetics , Multivesicular Bodies/metabolism , Protein Transport
8.
Oncotarget ; 12(8): 807-822, 2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33889303

ABSTRACT

Higher epidermal growth factor receptor (EGFR) signaling can contribute to tumor metastasis and resistance to therapies in oral squamous cell carcinoma (OSCC). EGFR signaling can promote epithelial-mesenchymal transition (EMT) in OSCC. EMT is a process by which epithelial cells acquire invasive properties and it can contribute to tumor metastasis. Not only do the abnormal functions of microtubule and microtubule-organizing centers (MTOC) such as centrosomes lead to cancers, but also the malignant tissues are characterized by aberrant centriolar features and amplified centrosomes. Microtubule inhibition therapies increase the sensitivity to EGFR targeting drugs in various cancers. In this study, we show that the loss of expression of a microtubule/tubulin binding protein, centrosomal protein 4.1-associated protein (CPAP), which is critical for centriole biogenesis and normal functioning of the centrosome, caused an increase in the EGFR levels and its signaling and, enhanced the EMT features and invasiveness of OSCC cells. Further, depletion of CPAP enhanced the tumorigenicity of these cells in a xeno-transplant model. Importantly, CPAP loss-associated EMT features and invasiveness of multiple OSCC cells were attenuated upon depletion of EGFR in them. On the other hand, we found that CPAP protein levels were higher in EGF treated OSCC cells as well as in oral cancer tissues, suggesting that the frequently reported aberrant centriolar features of tumors are potentially a consequence, but not the cause, of tumor progression. Overall, our novel observations show that, in addition to its known indispensable role in centrosome biogenesis, CPAP also plays a vital role in suppressing tumorigenesis in OSCC by facilitating EGFR homeostasis.

9.
Sci Rep ; 10(1): 14258, 2020 08 31.
Article in English | MEDLINE | ID: mdl-32868790

ABSTRACT

Our recent studies, using (SWRxNZB)F1 (SNF1) mice, showed a potential contribution of the gut microbiota and pro-inflammatory immune responses of the gut mucosa to systemic autoimmunity in lupus. Here, using this mouse model, we determined the abundance and the nAg reactivity of IgA antibody produced in the intestine under lupus susceptibility. Intestinal lymphoid tissues from SNF1 mice, females particularly, showed significantly higher frequencies of nAg (dsDNA and nucleohistone) reactive IgA producing B cells compared to B6 females. Most importantly, younger age fecal IgA -abundance and -nAg reactivity of lupus-prone mice showed a positive correlation with eventual systemic autoimmunity and proteinuria onset. Depletion of gut microbiota in SNF1 mice resulted in the diminished production of IgA in the intestine and the nAg reactivity of these antibodies. Overall, these observations show that fecal IgA features, nuclear antigen reactivity particularly, at preclinical stages/in at-risk subjects could be predictive of autoimmune progression.


Subject(s)
Antigens, Nuclear/immunology , Autoimmunity/immunology , Immunoglobulin A/immunology , Intestinal Mucosa/immunology , Lupus Erythematosus, Systemic/immunology , Age Factors , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Feces , Female , Gastrointestinal Microbiome/immunology , Lymphoid Tissue/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
10.
Nutrients ; 12(8)2020 Jul 23.
Article in English | MEDLINE | ID: mdl-32717991

ABSTRACT

Previously, we have shown that oral administration of yeast derived ß-1,3/1,6-d-glucan enhances immune regulation and alters the composition of the gut microbiota. However, it is not known if other structurally distinct ß-glucans have similar properties. Here, using C57BL/6 mice, we show the potential of a microalgae derived ß-1,3-d-glucan, paramylon (PM), in shaping the gut microbiota and modulating the susceptibility to colitis. The community structure within the gut microbiota showed progressive changes including selective enrichment of specific communities and lowered community richness and diversity during prolonged oral treatment with PM. Compared to control mice, the gut microbiota of PM-treated mice had significantly higher abundance of Verrucomicrobia and lower abundance of Firmicutes. Specific taxa that were significantly more abundant in PM-treated mice include Akkermansia muciniphila and several Bacteroides members. Predictive functional analysis revealed overrepresentation of carbohydrate metabolism function in the fecal microbiota of PM recipients compared to controls, and this function was linked to Bacteroides spp. Prolonged pretreatment with PM not only diminished susceptibility to dextran sulfate sodium induced colitis severity, but also caused enhanced immune regulation. Overall, this study demonstrates the prebiotic properties of PM and the potential benefits of its prolonged oral consumption to gut health.


Subject(s)
Gastrointestinal Microbiome/drug effects , Glucans/pharmacology , Inflammation/drug therapy , Microalgae/chemistry , beta-Glucans/pharmacology , Animals , Bacteria/classification , Bacteria/genetics , Colitis , Feces/microbiology , Female , Mice , Mice, Inbred C57BL , Prebiotics , Verrucomicrobia
11.
J Autoimmun ; 108: 102420, 2020 03.
Article in English | MEDLINE | ID: mdl-32019684

ABSTRACT

The risk of developing systemic lupus erythematosus (SLE) is about 9 times higher in women as compared to men. Our recent report, which used (SWRxNZB) F1 (SNF1) mouse model of spontaneous lupus, showed a potential link between immune response initiated in the gut mucosa at juvenile age (sex hormone independent) and SLE susceptibility. Here, using this mouse model, we show that gut microbiota contributes differently to pro-inflammatory immune response in the intestine and autoimmune progression in lupus-prone males and females. We found that gut microbiota composition in male and female littermates are significantly different only at adult ages. However, depletion of gut microbes causes suppression of autoimmune progression only in females. In agreement, microbiota depletion suppressed the pro-inflammatory cytokine response of gut mucosa in juvenile and adult females. Nevertheless, microbiota from females and males showed, upon cross-transfer, contrasting abilities to modulate disease progression. Furthermore, orchidectomy (castration) not only caused changes in the composition of gut microbiota, but also a modest acceleration of autoimmune progression. Overall, our work shows that microbiota-dependent pro-inflammatory immune response in the gut mucosa of females initiated at juvenile ages and androgen-dependent protection of males contribute to gender differences in the intestinal immune phenotype and systemic autoimmune progression.


Subject(s)
Autoimmunity , Gastrointestinal Microbiome/immunology , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/pathology , Phenotype , Age Factors , Animals , Biomarkers , Disease Models, Animal , Disease Progression , Female , Inflammation Mediators/metabolism , Male , Metagenome , Metagenomics/methods , Mice , Sex Factors , Transcriptome
12.
J Nutr ; 150(5): 1291-1302, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31879786

ABSTRACT

BACKGROUND: ß-Glucans (BGs), a group of complex dietary polysaccharides (CDPs), are available as dietary supplements. However, the effects of orally administered highly purified BGs on gut inflammation are largely unknown. OBJECTIVES: The aim of this study was to investigate the impact of orally administering highly purified, yeast-derived BG (YBG; ß-1,3/1,6-d-glucan) on susceptibility to colitis. METHODS: Eight-week-old C57BL/6 (B6) mice were used in a series of experiments. Experiment (Expt) 1: male and female mice were treated every day, for 40 d, with saline (control) or 250 µg YBG, followed by 2.5% (wt:vol) dextran sulfate sodium (DSS) in drinking water during days 30-35; and colitis severity and intestinal immune phenotype were determined. Expt 2: female B6 mice were treated with saline or YBG for 30 d and intestinal immune phenotype, gut microbiota composition, and fecal SCFA concentrations were determined. Expt 3: female B6 mice were treated as in Expt 2, given drinking water with or without antibiotics [Abx; ampicillin (1 g/L), vancomycin (0.5 g/L), neomycin (1 g/L), and metronidazole (1 g/L)] during days 16-30, and gut immune phenotype and fecal SCFA concentrations were determined. Expt 4: female B6 Foxp3-green fluorescent protein (-GFP) reporter mice were treated as in Expt 3, and intestinal T-regulatory cell (Treg) frequencies and immune phenotypes were determined. Expt 5: female mice were treated as in Expt 1, given drinking water with or without antibiotics during days 16-40, and colitis severity and intestinal cytokine production were determined. RESULTS: Compared with controls, the YBG group in Expt 1 exhibited suppressive effects on features of colitis, such as loss of body weight (by 47%; P < 0.001), shortening of colon (by 24%; P = 0.016), and histopathology severity score (by 45%; P = 0.01). The YBG group of Expt 2 showed a shift in the abundance of gut microbiota towards Bacteroides (by 16%; P = 0.049) and Verrucomicrobia (mean ± SD: control = 7.8 ± 0.44 vs. YBG = 21.0 ± 9.6%) and a reduction in Firmicutes (by 66%; P < 0.001). The YBG group also showed significantly higher concentrations of fecal SCFAs such as acetic (by 37%; P = 0.016), propionic (by 47%; P = 0.026), and butyric (by 57%; P = 0.013) acids. Compared with controls, the YBG group of Expt 2 showed higher frequencies of Tregs (by 32%; P = 0.043) in the gut mucosa. Depletion of gut microbiota in the YBG group of mice caused diminished fecal SCFA concentrations (Expt 3) and intestinal Treg frequencies (Expt 4). Compared with the YBG group, the YBG-(Abx) group of Expt 5 showed aggravated colitis features including loss of body weight (by >100%; P < 0.01) and colonic inflammation score (by 42%; P = 0.04). CONCLUSIONS: Studies using B6 mice show that dietary BGs are beneficial for promoting intestinal health when the gut microbiota is intact. However, these CDPs may produce adverse effects if gut microbiota is compromised.


Subject(s)
Colitis/prevention & control , Gastrointestinal Microbiome/drug effects , Polysaccharides/administration & dosage , Saccharomyces cerevisiae/chemistry , beta-Glucans/administration & dosage , Animals , Colitis/chemically induced , Colitis/immunology , Dextran Sulfate/pharmacology , Fatty Acids, Volatile/analysis , Feces/chemistry , Feces/microbiology , Female , Forkhead Transcription Factors/genetics , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Immunity/drug effects , Intestines/immunology , Male , Mice , Mice, Inbred C57BL
13.
Sci Rep ; 9(1): 12065, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31427630

ABSTRACT

Inhibitory/repressor-receptors are upregulated significantly on activated T cells, and have been the molecules of attention as targets for inducing immune tolerance. Induction of effective antigen specific tolerance depends on concurrent engagement of the TCR and one or more of these inhibitory receptors. Here, we show, for the first time that dendritic cells (DCs) can be efficiently engineered to express multiple T cell inhibitory ligands, and enhanced engagement of T cell inhibitory receptors, upon antigen presentation, by these DCs can induce effective CD4+ T cell tolerance and suppress autoimmunity. Compared to control DCs, antigen presentation by DCs that ectopically express CTLA4, PD1 and BTLA selective ligands (B7.1wa, PD-L1, and HVEM-CRD1 respectively) individually (mono-ligand DCs) or in combination (multi-ligand DCs) causes an inhibition of CD4+ T cell proliferation and pro-inflammatory cytokine response, as well as increase in Foxp3+ Treg frequency and immune regulatory cytokine production. Administration of self-antigen (mouse thyroglobulin; mTg) loaded multi-ligand DCs caused hyporesponsiveness to mTg challenge, suppression of autoantibody production, and amelioration of experimental autoimmune thyroiditis. Overall, this study shows that engineered DC-directed enhanced concurrent activation of multiple T cell coinhibitory pathways is an effective way to induce self-antigen specific T cell tolerance to suppress ongoing autoimmunity.


Subject(s)
Autoimmunity/immunology , Dendritic Cells/metabolism , Immune Tolerance/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation/immunology , Antigens/immunology , Autoimmunity/genetics , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Dendritic Cells/immunology , Immune Tolerance/genetics , Interleukin-10/immunology , Ligands , Lymphocyte Activation/immunology , Mice , Programmed Cell Death 1 Receptor/immunology , Receptors, Immunologic/immunology , Signal Transduction/genetics , T-Lymphocytes, Regulatory/immunology
14.
Diabetes ; 68(10): 1975-1989, 2019 10.
Article in English | MEDLINE | ID: mdl-31311801

ABSTRACT

Bacteroides fragilis (BF) is an integral component of the human colonic commensal microbiota. BF is also the most commonly isolated organism from clinical cases of intra-abdominal abscesses, suggesting its potential to induce proinflammatory responses upon accessing the systemic compartment. Hence, we examined the impact of mucosal and systemic exposures to BF on type 1 diabetes (T1D) incidence in NOD mice. The impact of intestinal exposure to BF under a chemically induced enhanced gut permeability condition, which permits microbial translocation, in T1D was also examined. While oral administration of heat-killed (HK) BF to prediabetic mice caused enhanced immune regulation and suppression of autoimmunity, resulting in delayed hyperglycemia, mice that received HK BF by intravenous injection showed rapid disease progression. Importantly, polysaccharide A-deficient BF failed to produce these opposing effects upon oral and systemic deliveries. Furthermore, BF-induced modulation of disease progression was observed in wild-type, but not TLR2-deficient, NOD mice. Interestingly, oral administration of BF under enhanced gut permeability conditions resulted in accelerated disease progression and rapid onset of hyperglycemia in NOD mice. Overall, these observations suggest that BF-like gut commensals can cause proinflammatory responses upon gaining access to the systemic compartment and contribute to T1D in at-risk subjects.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Gastrointestinal Microbiome/immunology , Animals , Autoimmunity/immunology , Bacteroides fragilis , Disease Progression , Humans , Mice , Polysaccharides
15.
Immunology ; 157(1): 70-85, 2019 05.
Article in English | MEDLINE | ID: mdl-30712258

ABSTRACT

The dietary supplement and prebiotic values of ß-glucan-rich products have been widely recognized and dietary approaches for modulating autoimmunity have been increasingly explored, we assess the impact of oral administration of high-purity yeast ß-glucan (YBG) on gut immune function, microbiota and type 1 diabetes (T1D) using mouse models. Oral administration of this non-digestible complex polysaccharide caused a dectin-1-dependent immune response involving increased expression of interleukin-10 (IL-10), retinaldehyde dehydrogenase (Raldh) and pro-inflammatory cytokines in the gut mucosa. YBG-exposed intestinal dendritic cells induced/expanded primarily Foxp3+ , IL-10+ and IL-17+ T cells, ex vivo. Importantly, prolonged oral administration of low-dose YBG at pre-diabetic stage suppressed insulitis and significantly delayed the appearance of T1D in non-obese diabetic (NOD) mice. Further, prolonged treatment with YBG showed increased Foxp3+ T-cell frequencies, and a significant change in the gut microbiota, particularly an increase in the abundance of Bacteroidetes and a decrease in the Firmicute members. Oral administration of YBG, together with Raldh-substrate and ß-cell antigen, resulted in better protection of NOD mice from T1D. These observations suggest that YBG not only has a prebiotic property, but also an oral tolerogenic-adjuvant-like effect, and these features could be exploited for modulating autoimmunity in T1D.


Subject(s)
Bacteroidetes/physiology , Diabetes Mellitus, Type 1/immunology , Dietary Carbohydrates/therapeutic use , Gastrointestinal Microbiome/immunology , T-Lymphocytes, Regulatory/immunology , Aldehyde Dehydrogenase 1 Family , Animals , Diabetes Mellitus, Type 1/diet therapy , Diabetes Mellitus, Type 1/microbiology , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Immunity , Immunomodulation , Interleukin-10/metabolism , Isoenzymes/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Prebiotics , Retinal Dehydrogenase/metabolism
16.
Oncotarget ; 7(43): 69371-69383, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27750220

ABSTRACT

Advancements in adoptive cell transfer therapy (ACT) has led to the use of T cells engineered with tumor specific T cell receptors, which after rapid expansion can be obtained in sufficient numbers for treating patients. However, due to massive proliferation these cells are close to replicative senescence, exhibit exhausted phenotype, and also display increased susceptibility to activation induced cell death. We have previously shown that tumor reactive T cells undergo caspase-independent cell death upon TCR restimulation with cognate antigen, which involves reactive oxygen species and c-jun N-terminal kinase. Herein, we show that a large fraction of the human melanoma epitope tyrosinase reactive TCR transduced T cells that exhibit effector memory (TEM) phenotype and undergo programmed necrosis, or necroptosis, upon TCR restimulation. As compared to the T central memory (TCM) subsets, the TEM subset displayed an increased expression of genes involved in necroptotic cell death, and a necrotic phenotype upon TCR restimulation as confirmed by electron microscopy. Higher expression of receptor-interacting kinases (RIPK) that mediate necroptosis was also observed in the TEM fraction. Further, the TEM cells were rescued from undergoing necroptosis when pretreated with necroptotic inhibitor NecroX2 before TCR restimulation. Importantly, NecroX2 pretreated tumor reactive T cells also exhibited better tumor control and increased in vivo persistence when adoptively-transferred to treat subcutaneously established murine melanoma B16-F10. Thus, it is likely that the outcome of ACT could be vastly improved by interfering with the necroptotic cell death pathway in activated tumor reactive T cells used in immunotherapy.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/transplantation , Immunotherapy, Adoptive/methods , Melanoma, Experimental/therapy , Receptors, Antigen, T-Cell/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , L-Selectin/immunology , L-Selectin/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Necrosis/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
18.
J Biol Chem ; 290(11): 6890-902, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25616662

ABSTRACT

Microtubule-based centrioles in the centrosome mediate accurate bipolar cell division, spindle orientation, and primary cilia formation. Cellular checkpoints ensure that the centrioles duplicate only once in every cell cycle and achieve precise dimensions, dysregulation of which results in genetic instability and neuro- and ciliopathies. The normal cellular level of centrosomal protein 4.1-associated protein (CPAP), achieved by its degradation at mitosis, is considered as one of the major mechanisms that limits centriole growth at a predetermined length. Here we show that CPAP levels and centriole elongation are regulated by centrobin. Exogenous expression of centrobin causes abnormal elongation of centrioles due to massive accumulation of CPAP in the cell. Conversely, CPAP was undetectable in centrobin-depleted cells, suggesting that it undergoes degradation in the absence of centrobin. Only the reintroduction of full-length centrobin, but not its mutant form that lacks the CPAP binding site, could restore cellular CPAP levels in centrobin-depleted cells, indicating that persistence of CPAP requires its interaction with centrobin. Interestingly, inhibition of the proteasome in centrobin-depleted cells restored the cellular and centriolar CPAP expression, suggesting its ubiquitination and proteasome-mediated degradation when centrobin is absent. Intriguingly, however, centrobin-overexpressing cells also showed proteasome-independent accumulation of ubiquitinated CPAP and abnormal, ubiquitin-positive, elongated centrioles. Overall, our results show that centrobin interacts with ubiquitinated CPAP and prevents its degradation for normal centriole elongation function. Therefore, it appears that loss of centrobin expression destabilizes CPAP and triggers its degradation to restrict the centriole length during biogenesis.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/metabolism , Microtubule-Associated Proteins/metabolism , Cell Cycle Proteins/analysis , Cell Cycle Proteins/genetics , Cell Line , Centrioles/ultrastructure , Gene Deletion , Humans , Microtubule-Associated Proteins/analysis , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitination , Up-Regulation
19.
Diabetes ; 64(4): 1341-57, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25377877

ABSTRACT

The progression of autoimmune diseases is dictated by deviations in the fine balance between proinflammatory versus regulatory responses, and pathogen recognition receptors (PRRs) play a key role in maintaining this balance. Previously, we have reported that ligation of Toll-like receptor 2 (TLR2) and Dectin 1 on antigen-presenting cells by zymosan results in a regulatory immune response that prevents type 1 diabetes (T1D). Here, we show that TLR2 and Dectin 1 engagement by zymosan promotes regulatory T-cell (Treg) responses against the pancreatic ß-cell-specific antigen (Ag). Unlike the TLR4 ligand, bacterial lipopolysaccharide, which induced proinflammatory cytokines and pathogenic T cells, zymosan induced a mixture of pro- and anti-inflammatory factors and Tregs, both in vitro and in vivo. Ag-specific T cells that are activated using zymosan-exposed dendritic cells (DCs) expressed Foxp3 and produced large amounts of IL-10, TGF-ß1, and IL-17. NOD mice that received ß-cell-Ag-loaded, zymosan-exposed DCs showed delayed hyperglycemia. Injection of NOD mice at the prediabetic age and early hyperglycemic stage with ß-cell-Ag, along with zymosan, results in a superior protection of the NOD mice from diabetes as compared with mice that received zymosan alone. This therapeutic effect was associated with increased frequencies of IL-10-, IL-17-, IL-4-, and Foxp3-positive T cells, especially in the pancreatic lymph nodes. These results show that zymosan can be used as an immune regulatory adjuvant for modulating the T-cell response to pancreatic ß-cell-Ag and reversing early-stage hyperglycemia in T1D.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Immunity, Innate/immunology , Insulin-Secreting Cells/immunology , Lectins, C-Type/metabolism , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 2/metabolism , Animals , Antigen-Presenting Cells/immunology , Cytokines/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Insulin-Secreting Cells/drug effects , Mice , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Zymosan/pharmacology
20.
J Immunol ; 193(7): 3308-21, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25143443

ABSTRACT

ß-Glucans are naturally occurring polysaccharides in cereal grains, mushrooms, algae, or microbes, including bacteria, fungi, and yeast. Immune cells recognize these ß-glucans through a cell surface pathogen recognition receptor called Dectin-1. Studies using ß-glucans and other Dectin-1 binding components have demonstrated the potential of these agents in activating the immune cells for cancer treatment and controlling infections. In this study, we show that the ß-glucan from Saccharomyces cerevisiae induces the expression of immune regulatory cytokines (IL-10, TGF-ß1, and IL-2) and a tolerogenic enzyme (IDO) in bone marrow-derived dendritic cells as well as spleen cells. These properties can be exploited to modulate autoimmunity in the NOD mouse model of type 1 diabetes (T1D). Treatment of prediabetic NOD mice with low-dose ß-glucan resulted in a profound delay in hyperglycemia, and this protection was associated with increase in the frequencies of Foxp3(+), LAP(+), and GARP(+) T cells. Upon Ag presentation, ß-glucan-exposed dendritic cells induced a significant increase in Foxp3(+) and LAP(+) T cells in in vitro cultures. Furthermore, systemic coadministration of ß-glucan plus pancreatic ß cell Ag resulted in an enhanced protection of NOD mice from T1D as compared with treatment with ß-glucan alone. These observations demonstrate that the innate immune response induced by low-dose ß-glucan is regulatory in nature and can be exploited to modulate T cell response to ß cell Ag for inducing an effective protection from T1D.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Fungal Polysaccharides/pharmacology , Immunity, Innate/drug effects , Lectins, C-Type/immunology , Saccharomyces cerevisiae/chemistry , beta-Glucans/pharmacology , Animals , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Dose-Response Relationship, Immunologic , Fungal Polysaccharides/chemistry , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Lectins, C-Type/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/pathology , beta-Glucans/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...