Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters

Publication year range
1.
FASEB J ; 37(6): e22995, 2023 06.
Article in English | MEDLINE | ID: mdl-37219526

ABSTRACT

Immuno-oncology (IO)-based therapies such as checkpoint inhibitors, bi-specific antibodies, and CAR-T-cell therapies have shown significant success in the treatment of several cancer indications. However, these therapies can result in the development of severe adverse events, including cytokine release syndrome (CRS). Currently, there is a paucity of in vivo models that can evaluate dose-response relationships for both tumor control and CRS-related safety issues. We tested an in vivo PBMC humanized mouse model to assess both treatment efficacy against specific tumors and the concurrent cytokine release profiles for individual human donors after treatment with a CD19xCD3 bispecific T-cell engager (BiTE). Using this model, we evaluated tumor burden, T-cell activation, and cytokine release in response to bispecific T-cell-engaging antibody in humanized mice generated with different PBMC donors. The results show that PBMC engrafted NOD-scid Il2rgnull mice lacking expression of mouse MHC class I and II (NSG-MHC-DKO mice) and implanted with a tumor xenograft predict both efficacy for tumor control by CD19xCD3 BiTE and stimulated cytokine release. Moreover, our findings indicate that this PBMC-engrafted model captures variability among donors for tumor control and cytokine release following treatment. Tumor control and cytokine release were reproducible for the same PBMC donor in separate experiments. The PBMC humanized mouse model described here is a sensitive and reproducible platform that identifies specific patient/cancer/therapy combinations for treatment efficacy and development of complications.


Subject(s)
Leukocytes, Mononuclear , T-Lymphocytes , Humans , Animals , Mice , Mice, Inbred NOD , Treatment Outcome , Cytokine Release Syndrome , Cytokines , Disease Models, Animal , Mice, Knockout , Mice, SCID
2.
Hepatology ; 75(2): 266-279, 2022 02.
Article in English | MEDLINE | ID: mdl-34608663

ABSTRACT

BACKGROUND AND AIMS: The increased frequency of urinary tract infections in patients with primary biliary cholangitis (PBC) and the cross-reactivity between the lipoyl domains (LD) of human pyruvate dehydrogenase complex (hPDC-E2) and Escherichia coli PDC-E2 (ePDC-E2) have long suggested a role of E. coli in causality of PBC. This issue, however, has remained speculative. We hypothesized that by generating specific constructs of human and E. coli PDC-E2, we would be able to assess the specificity of autoantibody responses and define whether exposure to E. coli in susceptible hosts is the basis for the antimitochondrial antibody (AMA) response. APPROACH AND RESULTS: Importantly, the reactivity of hPDC-E2 LD (hPDC-E2LD) affinity-purified antibodies against hPDC-E2LD could only be removed by prior absorption with hPDC-E2LD and not ePDC-E2, suggesting the presence of unique human PDC-E2 epitopes distinct from E. coli PDC-E2. To identify the autoepitope(s) present in hPDC-E2LD, a more detailed study using a variety of PDC-E2 constructs was tested, including the effect of lipoic acid (LA) on ePDC-E2 conformation and AMA recognition. Individual recombinant ePDCE2 LD domains LD1, LD2 and LD3 did not react with either AMA or antibodies to LA (anti-LA), but in contrast, anti-LA was readily reactive against purified recombinant LD1, LD2, and LD3 expressed in tandem (LP); such reactivity increased when LP was precultured with LA. Moreover, when the three LD (LD1, LD2, LD3) domains were expressed in tandem in pET28a or when LD1 was expressed in another plasmid pGEX, they were lipoylated and reactive to PBC sera. CONCLUSIONS: In conclusion, our data are consistent with an exposure to E. coli that elicits specific antibody to ePDC-E2 resulting in determinant spreading and the classic autoantibody to hPDC-E2LD. We argue this is the first step to development of human PBC.


Subject(s)
Autoantigens/immunology , Dihydrolipoyllysine-Residue Acetyltransferase/immunology , Escherichia coli Infections/complications , Escherichia coli/immunology , Liver Cirrhosis, Biliary/microbiology , Mitochondria/immunology , Mitochondrial Proteins/immunology , Autoantibodies/blood , Case-Control Studies , Cross Reactions/immunology , Epitopes/immunology , Escherichia coli/enzymology , Hepatitis, Autoimmune/blood , Humans , Lipoylation , Molecular Conformation/drug effects , Thioctic Acid/immunology , Thioctic Acid/pharmacology
3.
J Autoimmun ; 113: 102503, 2020 09.
Article in English | MEDLINE | ID: mdl-32546343

ABSTRACT

Glycosylation of antibodies, particularly in the Fc domain, critically modulate the ability of antibodies to bind to FcRs, maintaining immune quiescence to achieve a finely orchestrated immune response. The removal of sialic acid and galactose residues dramatically alters the physiological function of IgGs, and alterations of Ig glycosylation have been associated with several autoimmune disorders. However, Ig glycosylation has not been extensively studied in autoimmune cholangitis. We applied triple quadruple mass spectroscopy with subsequent multiple reaction monitoring to elucidate the profile, composition and linkage of sugar residues of antibody glycans in patients with primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC) and healthy controls (HC). Agalactosylated, HexNAc terminated IgG1 glycoforms were enriched in both PBC and PSC. Levels of IgM glycans at site N439 and fucosylated glycans in J chain, were significantly decreased in PBC compared to PSC and HC. PSC patients had decreased bisecting glycoforms and increased biantennary glycoforms on IgA compared to PBC. Importantly, our data demonstrate the association of distinct branching and composition patterns of Ig glycoforms with disease severity and liver cirrhosis, which highlight the importance of glycan biology as a potential mechanism and/or a disease specific signal of inflammation.


Subject(s)
Autoantibodies/metabolism , Autoimmune Diseases/diagnosis , Cholangitis, Sclerosing/diagnosis , Immunoglobulin G/metabolism , Liver Cirrhosis, Biliary/diagnosis , Adult , Aged , Autoantibodies/blood , Autoantibodies/immunology , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Biomarkers/blood , Biomarkers/metabolism , Case-Control Studies , Cholangitis, Sclerosing/blood , Cholangitis, Sclerosing/immunology , Female , Glycomics/methods , Glycosylation , Healthy Volunteers , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Liver Cirrhosis, Biliary/blood , Liver Cirrhosis, Biliary/immunology , Male , Middle Aged , Polysaccharides/immunology , Polysaccharides/metabolism , Severity of Illness Index
4.
J Autoimmun ; 101: 26-34, 2019 07.
Article in English | MEDLINE | ID: mdl-31027870

ABSTRACT

Primary biliary cholangitis (PBC) is a classic autoimmune disease in which humoral, cytotoxic, and innate immune responses have been implicated with the specific targeting of a mitochondrial antigen. The mainstay of treatment remains the bile acid ursodeoxycholic acid (UDCA). Corticosteroids may have some benefits, but to date, clinical trials of biologics targeting B cells and IL-12/23 have not shown any efficacy. Because activated T cells target the intrahepatic bile ducts in PBC and pre-clinical models suggested that blocking CD80/CD86 with CTLA-4 Ig might have therapeutic benefit in PBC, we performed an open-label trial to determine if CTLA-4 Ig (abatacept) is safe and potentially efficacious in PBC patients with an incomplete response to UDCA. PBC patients with an alkaline phosphatase (ALP) > 1.67 × the upper limit of normal after 6 months on UDCA treatment or who were intolerant of UDCA received abatacept 125 mg s.q. weekly for 24 weeks. The co-primary endpoint was ALP normalization or a >40% reduction from baseline. Among 16 subjects enrolled and who received at least 1 dose of abatacept, 1 (6.3%) met the co-primary endpoint. Absolute and percent changes in ALP [median (95% CI)] were +2.8 U/L (-90.9-96.6) and -0.28% (-21.1-15.5), respectively. No significant changes were observed in ALP, ALT, total bilirubin, albumin, immunoglobulins, or liver stiffness. Abatacept treatment decreased several non-terminally differentiated CD4+ but not CD8+ T cell populations, including decreases in CD4+ CCR5+ (p = 0.02) and CD4+ PD1+ (p = 0.03) lymphocytes. In contrast there were increases in CD4+ CCR7+ lymphocytes (p = 0.034). Treatment emergent adverse events occurred in 4 subjects. Abatacept was well tolerated in this population of PBC patients but like other biologics in PBC was ineffective in achieving biochemical responses associated with improved clinical outcomes.


Subject(s)
Biological Products/therapeutic use , Liver Cirrhosis, Biliary/drug therapy , Abatacept/administration & dosage , Abatacept/adverse effects , Abatacept/therapeutic use , Adult , Biological Products/administration & dosage , Biological Products/adverse effects , Biomarkers , Clinical Trials as Topic , Disease Susceptibility , Female , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Inflammation Mediators/metabolism , Liver Cirrhosis, Biliary/diagnosis , Liver Cirrhosis, Biliary/etiology , Liver Cirrhosis, Biliary/metabolism , Male , Middle Aged , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods
5.
Hepatology ; 67(4): 1408-1419, 2018 04.
Article in English | MEDLINE | ID: mdl-28921595

ABSTRACT

We have reported on a murine model of autoimmune cholangitis, generated by altering the AU-rich element (ARE) by deletion of the interferon gamma (IFN-γ) 3' untranslated region (coined ARE-Del-/- ), that has striking similarities to human primary biliary cholangitis (PBC) with female predominance. Previously, we suggested that the sex bias of autoimmune cholangitis was secondary to intense and sustained type I and II IFN signaling. Based on this thesis, and to define the mechanisms that lead to portal inflammation, we specifically addressed the hypothesis that type I IFNs are the driver of this disease. To accomplish these goals, we crossed ARE-Del-/- mice with IFN type I receptor alpha chain (Ifnar1) knockout mice. We report herein that loss of type I IFN receptor signaling in the double construct of ARE-Del-/- Ifnar1-/- mice dramatically reduces liver pathology and abrogated sex bias. More importantly, female ARE-Del-/- mice have an increased number of germinal center (GC) B cells as well as abnormal follicular formation, sites which have been implicated in loss of tolerance. Deletion of type I IFN signaling in ARE-Del-/- Ifnar1-/- mice corrects these GC abnormalities, including abnormal follicular structure. CONCLUSION: Our data implicate type I IFN signaling as a necessary component of the sex bias of this murine model of autoimmune cholangitis. Importantly these data suggest that drugs that target the type I IFN signaling pathway would have potential benefit in the earlier stages of PBC. (Hepatology 2018;67:1408-1419).


Subject(s)
Autoimmune Diseases/immunology , Cholangitis/immunology , Interferon Type I/genetics , Liver/pathology , Animals , B-Lymphocytes/immunology , Disease Models, Animal , Female , Flow Cytometry , Liver/immunology , Liver/metabolism , Male , Mice , Mice, Knockout , Sex Factors , Signal Transduction/immunology
6.
Hepatology ; 66(3): 885-895, 2017 09.
Article in English | MEDLINE | ID: mdl-28470667

ABSTRACT

A major problem in autoimmunity has been identification of the earliest events that lead to breach of tolerance. Although there have been major advances in dissecting effector pathways and the multilineage immune responses to mitochondrial self-antigens in primary biliary cholangitis, the critical links between environmental factors and tolerance remain elusive. We hypothesized that environmental xenobiotic modification of the E2 subunit of the pyruvate dehydrogenase (PDC-E2) inner lipoyl domain can lead to loss of tolerance to genetically susceptible hosts. Previously we demonstrated that serum anti-PDC-E2 autoantibodies cross-react with the chemical xenobiotics 2-octynoic acid and 6,8-bis (acetylthio) octanoic acid and further that there is a high frequency of PDC-E2-specific peripheral plasmablasts. Herein we generated 104 recombinant monoclonal antibodies (mAbs) based on paired heavy-chain and light-chain variable regions of individual plasmablasts derived from primary biliary cholangitis patients. We identified 32 mAbs reactive with native PDC-E2, including 20 specific for PDC-E2 and 12 cross-reactive with both PDC-E2 and 2-octynoic acid and 6,8-bis (acetylthio) octanoic acid. A lower frequency of replacement somatic hypermutations, indicating a lower level of affinity maturation, was observed in the complementarity-determining regions of the cross-reactive mAbs in comparison to mAbs exclusively recognizing PDC-E2 or those for irrelevant antigens. In particular, when the highly mutated heavy-chain gene of a cross-reactive mAb was reverted to the germline sequence, the PDC-E2 reactivity was reduced dramatically, whereas the xenobiotic reactivity was retained. Importantly, cross-reactive mAbs also recognized lipoic acid, a mitochondrial fatty acid that is covalently bound to PDC-E2. CONCLUSION: Our data reflect that chemically modified lipoic acid or lipoic acid itself, through molecular mimicry, is the initial target that leads to the development of primary biliary cholangitis. (Hepatology 2017;66:885-895).


Subject(s)
Antibodies, Monoclonal/immunology , Autoantigens/immunology , Autoimmunity/genetics , Cholangitis/immunology , Cholangitis/pathology , Xenobiotics/immunology , Antibodies, Monoclonal/metabolism , Autoantigens/genetics , Autoimmunity/immunology , Female , Gene Amplification , Humans , Immunoblotting , Male , Molecular Mimicry/genetics , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Thioctic Acid/immunology , Thioctic Acid/metabolism
7.
Hepatology ; 65(5): 1670-1682, 2017 05.
Article in English | MEDLINE | ID: mdl-28100006

ABSTRACT

The identification of environmental factors that lead to loss of tolerance has been coined the holy grail of autoimmunity. Our work has focused on the reactivity of antimitochondrial autoantibodies (AMA) to chemical xenobiotics and has hypothesized that a modified peptide within PDC-E2, the major mitochondrial autoantigen, will have been immunologically recognized at the time of loss of tolerance. Herein, we successfully applied intein technology to construct a PDC-E2 protein fragment containing amino acid residues 177-314 of PDC-E2 by joining a recombinant peptide spanning residues 177-252 (PDC-228) with a 62-residue synthetic peptide from 253 to 314 (PP), which encompasses PDC-E2 inner lipoyl domain (ILD). We named this intein-constructed fragment PPL. Importantly, PPL, as well as lipoic acid conjugated PPL (LA-PPL) and xenobiotic 2-octynoic acid conjugated PPL (2OA-PPL), are recognized by AMA. Of great importance, AMA has specificity for the 2OA-modified PDC-E2 ILD peptide backbone distinct from antibodies that react with native lipoylated PDC-E2 peptide. Interestingly, this unique AMA subfraction is of the immunoglobulin M isotype and more dominant in early-stage primary biliary cholangitis (PBC), suggesting that exposure to 2OA-PPL-like compounds occurs early in the generation of AMA. To understand the structural basis of this differential recognition, we analyzed PPL, LA-PPL, and 2OA-PPL using electron paramagnetic resonance spectroscopy, with confirmations by enzyme-linked immunosorbent assay, immunoblotting, and affinity antibody analysis. We demonstrate that the conformation of PDC-E2 ILD is altered when conjugated with 2OA, compared to conjugation with lipoic acid. CONCLUSION: A molecular understanding of the conformation of xenobiotic-modified PDC-E2 is critical for understanding xenobiotic modification and loss of tolerance in PBC with widespread implications for a role of environmental chemicals in the induction of autoimmunity. (Hepatology 2017;65:1670-1682).


Subject(s)
Autoantibodies/blood , Cholangitis/chemically induced , Mitochondria/immunology , Pyruvate Dehydrogenase (Lipoamide)/drug effects , Xenobiotics/toxicity , Antibody Affinity , Case-Control Studies , Cholangitis/blood , Cholangitis/immunology , Electron Spin Resonance Spectroscopy , Enzyme-Linked Immunosorbent Assay , Humans , Inteins , Pyruvate Dehydrogenase (Lipoamide)/chemistry , Pyruvate Dehydrogenase (Lipoamide)/immunology
8.
Hepatology ; 64(4): 1189-201, 2016 10.
Article in English | MEDLINE | ID: mdl-27178326

ABSTRACT

UNLABELLED: In most autoimmune diseases the serologic hallmarks of disease precede clinical pathology by years. Therefore, the use of animal models in defining early disease events becomes critical. We took advantage of a "designer" mouse with dysregulation of interferon gamma (IFNγ) characterized by prolonged and chronic expression of IFNγ through deletion of the IFNγ 3'-untranslated region adenylate uridylate-rich element (ARE). The ARE-Del(-/-) mice develop primary biliary cholangitis (PBC) with a female predominance that mimics human PBC that is characterized by up-regulation of total bile acids, spontaneous production of anti-mitochondrial antibodies, and portal duct inflammation. Transfer of CD4 T cells from ARE-Del(-/-) to B6/Rag1(-/-) mice induced moderate portal inflammation and parenchymal inflammation, and RNA sequencing of liver gene expression revealed that up-regulated genes potentially define early stages of cholangitis. Interestingly, up-regulated genes specifically overlap with the gene expression signature of biliary epithelial cells in PBC, implying that IFNγ may play a pathogenic role in biliary epithelial cells in the initiation stage of PBC. Moreover, differentially expressed genes in female mice have stronger type 1 and type 2 IFN signaling and lymphocyte-mediated immune responses and thus may drive the female bias of the disease. CONCLUSION: Changes in IFNγ expression are critical for the pathogenesis of PBC. (Hepatology 2016;64:1189-1201).


Subject(s)
Autoimmune Diseases/etiology , Cholangitis/immunology , Interferon-gamma/biosynthesis , Animals , Autoimmune Diseases/metabolism , Cholangitis/metabolism , Female , Male , Mice , Sex Factors
9.
FASEB J ; 30(12): 4202-4213, 2016 12.
Article in English | MEDLINE | ID: mdl-27630169

ABSTRACT

Macrophage activation is an important feature of primary biliary cholangitis (PBC) pathogenesis and other cholestatic liver diseases. Galectin-3 (Gal3), a pleiotropic lectin, is produced by monocytic cells and macrophages. However, its role in PBC has not been addressed. We hypothesized that Gal3 is a key to induce NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome in macrophages and in turn to propagate proinflammatory IL-17 signaling. In liver tissues from patients with PBC and dnTGF-ßRII mice, a model of autoimmune cholangitis, the expression of Gal3, NLRP3, and the adaptor protein adaptor apoptosis-associated speck-like protein was induced, with the downstream activation of caspase-1 and IL-1ß. In wild-type hepatic macrophages, deoxycholic acid induced the association of Gal3 and NLRP3 with direct activation of the inflammasome, resulting in an increase in IL-1ß. Downstream retinoid-related orphan receptor C mRNA, IL-17A, and IL-17F were induced. In Gal3-/- macrophages, no inflammasome activation was detected. To confirm the key role of Gal3 in the pathogenesis of cholestatic liver injury, we generated dnTGF-ßRII/galectin-3-/- (dn/Gal3-/-) mice, which showed impaired inflammasome activation along with significantly improved inflammation and fibrosis. Taken together, our data point to a novel role of Gal3 as an initiator of inflammatory signaling in autoimmune cholangitis, mediating the activation of NLRP3 inflammasome and inducing IL-17 proinflammatory cascades. These studies provide a rationale to target Gal3 in autoimmune cholangitis and potentially other cholestatic diseases.-Tian, J., Yang, G., Chen, H.-Y., Hsu, D. K., Tomilov, A., Olson, K. A., Dehnad, A., Fish, S. R., Cortopassi, G., Zhao, B., Liu, F.-T., Gershwin, M. E., Török, N. J., Jiang, J. X. Galectin-3 regulates inflammasome activation in cholestatic liver injury.


Subject(s)
Galectin 3/metabolism , Inflammasomes/metabolism , Liver/metabolism , Macrophages/metabolism , Signal Transduction/physiology , Animals , Caspase 1/metabolism , Cells, Cultured , Galectin 3/genetics , Humans , Interleukin-17/metabolism , Interleukin-23/metabolism , Liver/injuries , Macrophage Activation/physiology , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
10.
Virol J ; 13(1): 190, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27884154

ABSTRACT

BACKGROUND: Adenoviruses are important pathogens with the potential for interspecies transmission between humans and non-human primates. Although many adenoviruses have been identified in monkeys, the knowledge of these viruses from the Colobinae members is quite limited. FINDINGS: We conducted a surveillance of viral infection in endangered golden snub-nosed monkeys (Rhinopithecus roxellana) in the subfamily Colobinae in China, and found that 5.1% of sampled individuals were positive for adenovirus. One of the adenoviruses (SAdV-WIV19) was successfully isolated and its full-length genome was sequenced. The full-length genome of WIV19 is 33,562 bp in size, has a G + C content of 56.2%, and encodes 35 putative genes. Sequence analysis revealed that this virus represents a novel species in the genus Mastadenovirus. Diverse cell lines, including those of human origin, were susceptible to WIV19. CONCLUSION: We report the first time the isolation and full-length genomic characterization of an adenovirus from the subfamily Colobinae.


Subject(s)
Adenoviridae Infections/veterinary , Adenoviridae/classification , Adenoviridae/isolation & purification , Colobinae/virology , Primate Diseases/epidemiology , Primate Diseases/virology , Adenoviridae/genetics , Adenoviridae Infections/epidemiology , Adenoviridae Infections/virology , Animals , Base Composition , China/epidemiology , Cluster Analysis , Gene Order , Genes, Viral , Genome, Viral , Phylogeny , Prevalence , Sequence Analysis, DNA
11.
Hepatology ; 60(5): 1708-16, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25043065

ABSTRACT

UNLABELLED: The serologic hallmark of primary biliary cirrhosis (PBC), the antimitochondrial response to the E2 component of the pyruvate dehydrogenase complex (PDC-E2), has unique features, including continuous high titers of immunoglobulin M (IgM) and IgG reactivity throughout all stages of disease, capable not only of target enzyme inhibition, but also crossreactive with chemical xenobiotics that share molecular homology with the inner lipoyl domain of PDC-E2; such chemicals have been proposed as potential etiological agents. We used flow cytometry and enzyme-linked immunospot assay (ELISPOT) to examine B-cell subsets in 59 subjects, including 28 with PBC, 13 with primary sclerosing cholangitis (PSC), and 18 healthy controls. Strikingly, in PBC, although there were no significant differences in B-cell phenotype subpopulations, 10% of the total IgG and IgA plasmablast population and 23% of the IgM plasmablast population were uniquely reactive with PDC-E2, detected in the CXCR7+ CCR10low plasmablast population. In contrast, plasmablast reactivity to a control antigen, tetanus toxoid, was minimal and similar in all groups. Additionally, we isolated plasmablast-derived polyclonal antibodies and compared reactivity with plasma-derived antibodies and noted a distinct noncirculating tissue source of xenobiotic crossreacting antibodies. The high levels of autoantigen specific peripheral plasmablasts indicate recent activation of naive or memory B cells and a continuous and robust activation. The presence of CXCR7+ CCR10low PDC-E2-specific ASCs suggests a mechanistic basis for the migration of circulating antigen specific plasmablasts to the mucosal epithelial ligands CXCL12 and CCL28. CONCLUSION: Our findings suggest a sustained rigorous B-cell response in PBC, likely activated and perpetuated by cognate autoantigen.


Subject(s)
Autoantigens/immunology , B-Lymphocytes/immunology , Dihydrolipoyllysine-Residue Acetyltransferase/immunology , Liver Cirrhosis, Biliary/immunology , Adult , Aged , Aged, 80 and over , Antibody Specificity , Antibody-Producing Cells/metabolism , Case-Control Studies , Female , Humans , Male , Middle Aged , Receptors, CCR10/metabolism , Receptors, CXCR/metabolism , Young Adult
12.
Hepatology ; 59(5): 1944-53, 2014 May.
Article in English | MEDLINE | ID: mdl-24375552

ABSTRACT

UNLABELLED: The interleukin (IL)-12/IL-23-mediated Th1/Th17 signaling pathway has been associated with the etiopathogenesis of primary biliary cirrhosis (PBC). To address the cytokine microenvironment specifically in the liver, we examined the localized expression of cytokine subunits and their corresponding receptors using previously optimized immunohistochemistry with an extensive panel of antibodies directed at IL-12p70, IL-12p35, interferon-gamma (IFN-γ), IL-12RB2, IL-23p40, IL-23p19, IL-17, and IL-23R using liver from PBC (n = 51) and non-PBC (n = 80) control liver disease patients. Multiple portal tracts in each patient were blindly evaluated and individually scored. We report herein that although IL-12/Th1 and IL-23/Th17 staining was detected in all of the liver sections, they were primarily localized around the damaged interlobular bile ducts in PBC. Most important, Th17 skewing was prominent in advanced PBC patients with intensive secretion of IL-23p19 by inflamed hepatocytes around IL-23R, IL-12RB2, and IFN-γ expressing degenerated cholangiocytes. Our novel finding on the direct association of Th17 skewing and disease severity illustrates the significance of the IL-23/Th17 pathway in the perpetuation of IL-12/Th1-mediated immunopathology in PBC. Furthermore, localized IL-23p19 production by hepatocytes may enhance profibrotic Th17 signaling and proinflammatory IFN-γ production that contribute to PBC pathology. CONCLUSION: Our data emphasize the pathogenic relevance of IL-12/Th1 and IL-23/Th17 in the evolution of PBC. Of significance, however, the shift from a Th1 to a Th17 imbalance at advanced stages of the disease suggests the necessity to consider modulation of the IL-23/Th17 pathway as a potential target for therapeutic intervention.


Subject(s)
Interleukin-12/physiology , Interleukin-23/physiology , Liver Cirrhosis, Biliary/etiology , Th1 Cells/physiology , Th17 Cells/physiology , Adult , Aged , Female , Humans , Immunohistochemistry , Interferon-gamma/metabolism , Liver Cirrhosis, Biliary/pathology , Liver Cirrhosis, Biliary/therapy , Male , Middle Aged , Signal Transduction/physiology
13.
J Environ Manage ; 161: 252-260, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26188990

ABSTRACT

Best management practices (BMPs) can be used effectively to reduce nutrient loads transported from non-point sources to receiving water bodies. However, methodologies of BMP selection and placement in a cost-effective way are needed to assist watershed management planners and stakeholders. We developed a novel modeling-optimization framework that can be used to find cost-effective solutions of BMP placement to attain nutrient load reduction targets. This was accomplished by integrating a GIS-based BMP siting method, a WQM-TMDL-N modeling approach to estimate total nitrogen (TN) loading, and a multi-objective optimization algorithm. Wetland restoration and buffer strip implementation were the two BMP categories used to explore the performance of this framework, both differing greatly in complexity of spatial analysis for site identification. Minimizing TN load and BMP cost were the two objective functions for the optimization process. The performance of this framework was demonstrated in the Tippecanoe River watershed, Indiana, USA. Optimized scenario-based load reduction indicated that the wetland subset selected by the minimum scenario had the greatest N removal efficiency. Buffer strips were more effective for load removal than wetlands. The optimized solutions provided a range of trade-offs between the two objective functions for both BMPs. This framework can be expanded conveniently to a regional scale because the NHDPlus catchment serves as its spatial computational unit. The present study demonstrated the potential of this framework to find cost-effective solutions to meet a water quality target, such as a 20% TN load reduction, under different conditions.


Subject(s)
Conservation of Natural Resources/methods , Models, Theoretical , Nitrogen/analysis , Water Pollutants, Chemical/analysis , Wetlands , Algorithms , Conservation of Natural Resources/economics , Cost-Benefit Analysis , Environmental Restoration and Remediation , Geographic Information Systems , Indiana , Rivers/chemistry , Water Quality
14.
Semin Liver Dis ; 34(3): 285-96, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25057952

ABSTRACT

Within the last decade, several mouse models that manifest characteristic features of primary biliary cirrhosis (PBC) with antimitochondrial antibodies (AMAs) and immune-mediated biliary duct pathology have been reported. Here, the authors discuss the current findings on two spontaneous (nonobese diabetic autoimmune biliary disease [NOD.ABD] and dominant negative transforming growth factor-ß receptor II [dnTGFßRII]) and two induced (chemical xenobiotics and microbial immunization) models of PBC. These models exhibit the serological, immunological, and histopathological features of human PBC. From these animal models, it is evident that the etiology of PBC is multifactorial and requires both specific genetic predispositions and environmental insults (either xenobiotic chemicals or microbial), which lead to the breaking of tolerance and eventually liver pathology. Human PBC is likely orchestrated by multiple factors and hence no single model can fully mimic the immunopathophysiology of human PBC. Nevertheless, knowledge gained from these models has greatly advanced our understanding of the major immunological pathways as well as the etiology of PBC.


Subject(s)
Autoimmunity , Bile Ducts, Intrahepatic/immunology , Disease Models, Animal , Liver Cirrhosis, Biliary/immunology , Animals , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/microbiology , Bile Ducts, Intrahepatic/pathology , Biomarkers/blood , Escherichia coli/pathogenicity , Humans , Liver Cirrhosis, Biliary/blood , Liver Cirrhosis, Biliary/chemically induced , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Biliary/microbiology , Liver Cirrhosis, Biliary/pathology , Mice, Inbred NOD , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Sphingomonadaceae/pathogenicity , Xenobiotics
15.
J Autoimmun ; 50: 123-34, 2014 May.
Article in English | MEDLINE | ID: mdl-24556277

ABSTRACT

Primary biliary cirrhosis (PBC) is an enigmatic disease mediated by autoimmune destruction of cholangiocytes in hepatic bile ducts. The early immunological events leading to PBC are poorly understood; clinical signs of disease occur very late in the pathological process. We have used our unique murine model of PBC in dominant-negative TGF-ß receptor type II transgenic mice to delineate critical early immunopathological pathways, and previously showed that dnTGFßRII CD8 T cells transfer biliary disease. Herein we report significantly increased numbers of hepatic dnTGFßRII terminally differentiated (KLRG1(+)) CD8 T cells, a CD8 subset previously shown to be enriched in antigen specific cells during hepatic immune response to viral infections. We performed bone marrow chimera studies to assess whether dnTGFßRII CD8 mediated disease was cell intrinsic or extrinsic. Unexpectedly, mixed (dnTGFßRII and B6) bone marrow chimeric (BMC) mice were protected from biliary disease compared to dnTGFßRII single bone marrow chimerics. To define the protective B6 cell subset, we performed adoptive transfer studies, which showed that co-transfer of B6 Tregs prevented dnTGFßRII CD8 T cell mediated cholangitis. Treg mediated disease protection was associated with significantly decreased numbers of hepatic KLRG1(+) CD8 T cells. In contrast, co-transfer of dnTGFßRII Tregs offered no protection, and dnTGFßRII Treg cells were functionally defective in suppressing effector CD8 T cells in vitro compared to wild type B6 Tregs. In vitro cholangiocyte cytotoxicity assays demonstrated significantly increased numbers of cytotoxic hepatic dnTGFßRII KLRG1(+) CD8 cells compared to B6. Protection from disease by B6 Tregs was associated with elimination of hepatic dnTGFßRII CD8 mediated cholangiocyte cytotoxicity. These results emphasize that autoimmune cholangitis requires defects in both the T effector and regulatory compartments, and that an intrinsic T cell effector defect is not sufficient to mediate autoimmune biliary disease in the setting of intact immune regulation. These results have important implications for understanding the early pathogenesis of human PBC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cholangitis/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Autoimmune Diseases , Bone Marrow/immunology , Bone Marrow/pathology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Cell Lineage/immunology , Chimera/genetics , Chimera/immunology , Cholangitis/genetics , Cholangitis/pathology , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Gene Expression , Humans , Lectins, C-Type , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/immunology , Liver Cirrhosis, Biliary/pathology , Mice , Mice, Transgenic , Receptors, Immunologic/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/transplantation
16.
Hepatology ; 57(2): 708-15, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22996325

ABSTRACT

UNLABELLED: Collectively, the data in both humans and murine models of human primary biliary cirrhosis (PBC) suggest that activated T cells, particularly CD8 T cells, play a critical role in biliary cell destruction. Under physiological conditions, T-cell activation involves two critical signals that involve the major histocompatibility complex and a set of costimulatory molecules, which include a receptor on T cells termed cytotoxic T lymphocyte antigen 4 (CTLA-4). Germane to the studies reported herein, signaling by CTLA-4 has the potential to modulate costimulation and induce inhibitory signals. In this study, we have taken advantage of our well-defined murine model of PBC, in which mice are immunized with 2-octynoic acid coupled to bovine serum albumin (2OA-BSA), leading to the production of high-titer antimitochondrial autoantibodies (AMAs) and portal cellular infiltrates. To investigate the potential of CTLA-4-Ig (immunoglobulin) as an immunotherapeutic agent, we treated mice both before and after induction of autoimmune cholangitis. First, we demonstrate that CTLA-4-Ig treatment, begun 1 day before 2OA-BSA immunization, completely inhibits the manifestations of cholangitis, including AMA production, intrahepatic T-cell infiltrates, and bile duct damage. However, and more critically, treatment with CTLA-4-Ig, initiated after the development of autoimmune cholangitis in previously immunized mice, also resulted in significant therapeutic benefit, including reduced intrahepatic T-cell infiltrates and biliary cell damage, although AMA levels were not altered. CONCLUSION: These data suggest that an optimized regimen with CTLA-4-Ig has the potential to serve as an investigative therapeutic tool in patients with PBC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunoconjugates/therapeutic use , Liver Cirrhosis, Biliary/drug therapy , Abatacept , Animals , Cholangitis/drug therapy , Cholangitis/immunology , Disease Models, Animal , Fatty Acids, Monounsaturated , Female , Humans , Liver Cirrhosis, Biliary/immunology , Mice , Mitochondria/immunology
17.
Hepatology ; 58(3): 1094-104, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23532950

ABSTRACT

UNLABELLED: There are several murine models described with features similar to human primary biliary cirrhosis (PBC). Among these models, the one which has the closest serologic features to PBC is a mouse with a T-cell-restricted expression of the dominant negative transforming growth factor ß receptor type II (dnTGFßRII). Our work has demonstrated that CD8(+) T cells from dnTGFßRII mice transfer autoimmune cholangitis to Rag1(-/-) recipients. However, it remained unclear whether the autoimmune cholangitis was secondary to an intrinsic function within CD8(+) T cells or due to the abnormal TGFßR environment within which CD8(+) T cells were generated. To address this mechanistic issue, we used our dnTGFßRII, OT-I/Rag1(-/-) , OT-II/Rag1(-/-) mice and in addition generated OT-I/dnTGFßRII/Rag1(-/-) , and OT-II/dnTGFßRII/Rag1(-/-) mice in which the entire T-cell repertoire was replaced with ovalbumin (OVA)-specific CD8(+) or CD4(+) T cells, respectively. Importantly, neither the parental OT-I/dnTGFßRII/Rag1(-/-) mice and/or OT-II/dnTGFßRII/Rag1(-/-) mice developed cholangitis. However, adoptive transfer demonstrated that only transfer of CD8(+) T cells from dnTGFßRII mice but not CD8(+) T cells from OT-I/Rag1(-/-) mice or from OT-I/dnTGFßRII/Rag1(-/-) mice transferred disease. These data were not secondary to an absence of CD4(+) T cell help since a combination of CD8(+) T cells from OT-I/dnTGFßRII/Rag1(-/-) and CD4(+) T cells from OT II/dnTGFßRII/Rag1(-/-) or CD8(+) T cells from OT-I/dnTGFßRII/Rag1(-/-) with CD4(+) T cells from OT-II/Rag1(-/-) mice failed to transfer disease. CONCLUSION: Defective TGFßRII signaling, in addition to clonal CD8(+) T cells that target biliary cells, are required for induction of autoimmune cholangitis.


Subject(s)
Autoimmune Diseases/physiopathology , CD8-Positive T-Lymphocytes/pathology , Cholangitis/physiopathology , Disease Models, Animal , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Adoptive Transfer , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cholangitis/immunology , Cholangitis/metabolism , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , T-Cell Antigen Receptor Specificity/physiology
18.
Hepatology ; 57(2): 806-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22576253

ABSTRACT

Mice with a dominant-negative transforming growth factor ß receptor restricted to T cells (dnTGFßRII mice) develop an inflammatory biliary ductular disease that strongly resembles human primary biliary cirrhosis (PBC). Furthermore, deletion of the gene encoding interleukin (IL)-12p40 resulted in a strain (IL-12p40(-/-) dnTGFßRII) with dramatically reduced autoimmune cholangitis. To further investigate the role of the IL-12 cytokine family in dnTGFßRII autoimmune biliary disease, we deleted the gene encoding the IL-12p35 subunit from dnTGFßRII mice, resulting in an IL-12p35(-/-) dnTGFßRII strain which is deficient in two members of the IL-12 family, IL-12 and IL-35. In contrast to IL-12p40(-/-) mice, the IL-12p35(-/-) mice developed liver inflammation and bile duct damage with similar severity but delayed onset as the parental dnTGFßRII mice. The p35(-/-) mice also demonstrated a distinct cytokine profile characterized by a shift from a T-helper 1 (Th1) to a Th17 response. Strikingly, liver fibrosis was frequently observed in IL-12p35(-/-) mice. In conclusion, IL-12p35(-/-) dnTGFßRII mice, histologically and immunologically, reflect key features of PBC, providing a useful generic model to understand the immunopathology of human PBC.


Subject(s)
Interleukin-12 Subunit p35/genetics , Liver Cirrhosis/pathology , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Animals , Disease Models, Animal , Hepatitis, Animal/etiology , Hepatitis, Animal/pathology , Interleukin-12/deficiency , Interleukin-12/physiology , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p40/genetics , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/pathology , Mice , Receptor, Transforming Growth Factor-beta Type II , Th1 Cells/physiology , Th17 Cells/physiology
19.
Hepatology ; 57(4): 1498-508, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23184636

ABSTRACT

UNLABELLED: Antimitochondrial antibodies (AMAs) directed against the lipoyl domain of the E2 subunit of pyruvate dehydrogenase (PDC-E2) are detected in 95% of patients with primary biliary cirrhosis (PBC) and are present before the onset of clinical disease. The recent demonstration that AMAs recognize xenobiotic modified PDC-E2 with higher titers than native PDC-E2 raises the possibility that the earliest events involved in loss of tolerance are related to xenobiotic modification. We hypothesized that reactivity to such xenobiotics would be predominantly immunoglobulin M (IgM) and using sera from a large cohort of PBC patients and controls (n = 516), we examined in detail sera reactivity against either 6,8-bis(acetylthio) octanoic acid (SAc)-conjugated bovine serum albumin (BSA), recombinant PDC-E2 (rPDC-E2) or BSA alone. Further, we also defined the relative specificity to the SAc moiety using inhibition enzyme-linked immunosorbent assay (ELISA); SAc conjugate and rPDC-E2-specific affinity-purified antibodies were also examined for antigen specificity, isotype, and crossreactivity. Reactivity to SAc conjugates is predominantly IgM; such reactivity reflects a footprint of previous xenobiotic exposure. Indeed, this observation is supported by both direct binding, crossreactivity, and inhibition studies. In both early and late-stage PBC, the predominant Ig isotype to SAc is IgM, with titers higher with advanced stage disease. We also note that there was a higher level of IgM reactivity to SAc than to rPDC-E2 in early-stage versus late-stage PBC. Interestingly, this finding is particularly significant in light of the structural similarity between SAc and the reduced form of lipoic acid, a step which is similar to the normal physiological oxidation of lipoic acid. CONCLUSION: Specific modifications of the disulfide bond within the lipoic-acid-conjugated PDC-E2 moiety, i.e., by an electrophilic agent renders PDC-E2 immunogenic in a genetically susceptible host.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Liver Cirrhosis, Biliary/etiology , Liver Cirrhosis, Biliary/immunology , Mitochondria/immunology , Xenobiotics/adverse effects , Antibody Specificity , Autoantigens/immunology , Case-Control Studies , Cholangitis, Sclerosing/blood , Cholangitis, Sclerosing/immunology , Dihydrolipoyllysine-Residue Acetyltransferase/immunology , Hepatitis, Autoimmune/blood , Hepatitis, Autoimmune/immunology , Humans , Immunoglobulin M/blood , Liver Cirrhosis, Biliary/blood , Mitochondrial Proteins/immunology , Recombinant Proteins/immunology , Serum Albumin, Bovine/immunology
20.
J Environ Manage ; 135: 1-10, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24486566

ABSTRACT

This paper presents a screening-level modeling approach that can be used to rapidly estimate nutrient loading, assess numerical nutrient standard exceedance risk of surface waters leading to potential classification as impaired for designated use, and explore best management practice (BMP) implementation to reduce loading with a relatively low data requirement. The modeling framework uses a hybrid statistical and process based approach to estimate source of pollutants, their transport and decay in the terrestrial and aquatic parts of watersheds. The framework is developed in the ArcGIS environment and is based on the total maximum daily load (TMDL) balance model. Nitrogen (N) is currently addressed in the framework, referred to as WQM-TMDL-N. Loading for each catchment includes non-point sources (NPS) and point sources (PS). The probability of a nutrient load to exceed a target load is evaluated using probabilistic risk assessment, by including the uncertainty associated with export coefficients of various land uses. In an application of this modeling approach to the Tippecanoe River watershed in Indiana, USA, total nitrogen (TN) loading, confidence interval and risk of standard exceedance leading to potential impairment were estimated. Model results suggest that decay coefficients decrease, and delivery fractions increase with increasing stream order. The spatial distribution pattern of delivered incremental TN yield shows a trend similar to that of the delivery fraction in this watershed. The target TN exceedance risk increases considerably when switching from Indiana draft-N benchmark to far lower EPA-proposed TN criteria, suggesting that load reduction to meet the latter criteria may benefit from BMP implementation through source control and delivery reduction.


Subject(s)
Environmental Monitoring/methods , Nitrogen/analysis , Rivers/chemistry , Water Pollutants, Chemical/analysis , Indiana , Models, Theoretical , Risk Assessment , Uncertainty , Water Pollutants, Chemical/standards
SELECTION OF CITATIONS
SEARCH DETAIL